Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 380
Filter
1.
Int J Cancer ; 146(4): 999-1009, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31081934

ABSTRACT

Comparably little is known about breast cancer (BC) risks in women from families tested negative for BRCA1/2 mutations despite an indicative family history, as opposed to BRCA1/2 mutation carriers. We determined the age-dependent risks of first and contralateral breast cancer (FBC, CBC) both in noncarriers and carriers of BRCA1/2 mutations, who participated in an intensified breast imaging surveillance program. The study was conducted between January 1, 2005, and September 30, 2017, at 12 university centers of the German Consortium for Hereditary Breast and Ovarian Cancer. Two cohorts were prospectively followed up for incident FBC (n = 4,380; 16,398 person-years [PY], median baseline age: 39 years) and CBC (n = 2,993; 10,090 PY, median baseline age: 42 years). Cumulative FBC risk at age 60 was 61.8% (95% CI 52.8-70.9%) for BRCA1 mutation carriers, 43.2% (95% CI 32.1-56.3%) for BRCA2 mutation carriers and 15.7% (95% CI 11.9-20.4%) for noncarriers. FBC risks were significantly higher than in the general population, with incidence rate ratios of 23.9 (95% CI 18.9-29.8) for BRCA1 mutation carriers, 13.5 (95% CI 9.2-19.1) for BRCA2 mutation carriers and 4.9 (95% CI 3.8-6.3) for BRCA1/2 noncarriers. Cumulative CBC risk 10 years after FBC was 25.1% (95% CI 19.6-31.9%) for BRCA1 mutation carriers, 6.6% (95% CI 3.4-12.5%) for BRCA2 mutation carriers and 3.6% (95% CI 2.2-5.7%) for noncarriers. CBC risk in noncarriers was similar to women with unilateral BC from the general population. Further studies are needed to confirm whether less intensified surveillance is justified in women from BRCA1/2 negative families with elevated risk.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/epidemiology , Genetic Predisposition to Disease , Adult , Age Factors , Breast Neoplasms/genetics , Epidemiological Monitoring , Female , Follow-Up Studies , Germany/epidemiology , Heterozygote , Humans , Incidence , Medical History Taking , Middle Aged , Mutation , Prospective Studies , Risk Assessment , Risk Factors
2.
Hum Mutat ; 40(9): 1557-1578, 2019 09.
Article in English | MEDLINE | ID: mdl-31131967

ABSTRACT

The multifactorial likelihood analysis method has demonstrated utility for quantitative assessment of variant pathogenicity for multiple cancer syndrome genes. Independent data types currently incorporated in the model for assessing BRCA1 and BRCA2 variants include clinically calibrated prior probability of pathogenicity based on variant location and bioinformatic prediction of variant effect, co-segregation, family cancer history profile, co-occurrence with a pathogenic variant in the same gene, breast tumor pathology, and case-control information. Research and clinical data for multifactorial likelihood analysis were collated for 1,395 BRCA1/2 predominantly intronic and missense variants, enabling classification based on posterior probability of pathogenicity for 734 variants: 447 variants were classified as (likely) benign, and 94 as (likely) pathogenic; and 248 classifications were new or considerably altered relative to ClinVar submissions. Classifications were compared with information not yet included in the likelihood model, and evidence strengths aligned to those recommended for ACMG/AMP classification codes. Altered mRNA splicing or function relative to known nonpathogenic variant controls were moderately to strongly predictive of variant pathogenicity. Variant absence in population datasets provided supporting evidence for variant pathogenicity. These findings have direct relevance for BRCA1 and BRCA2 variant evaluation, and justify the need for gene-specific calibration of evidence types used for variant classification.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Computational Biology/methods , Mutation, Missense , Neoplasms/diagnosis , Alternative Splicing , Early Detection of Cancer , Female , Genetic Predisposition to Disease , Humans , Likelihood Functions , Male , Multifactorial Inheritance , Neoplasms/genetics
4.
Internist (Berl) ; 60(12): 1305-1310, 2019 Dec.
Article in German | MEDLINE | ID: mdl-31549186

ABSTRACT

MEDICAL HISTORY AND INITIAL PRESENTATION: A 35-year-old patient with a previous history of persistent episodic fever, sore throat, myalgia, and cephalgia presented for evaluation of pancytopenia. He had no recent travel history, except for a stay in Italy 1 year prior to admission and in Spain several years in the past. DIAGNOSTIC WORKUP: Laboratory evaluation confirmed pancytopenia, agranulocytosis, and elevated infection parameters without indicative serological results en par with lymphadenitis colli. Computed tomography scanning revealed cervical lymphadenopathy, hepatosplenomegaly, and colitis with occult perforation of the sigmoid colon. Bone marrow biopsy showed an infiltration of polyclonal plasma cells. Lymph node biopsy was compatible with necrotizing lymphadenitis. DIAGNOSIS: Polymerase chain reaction analysis of a lymph node specimen confirmed the presence of Leishmania species, thereby enabling the diagnosis of visceral Leishmania. THERAPY COURSE: Treatment with liposomal amphotericin B was initiated. Both fever and lymphadenopathy quickly resolved. CONCLUSION: VL is a clinically pleiotropic, severe disease with fatal outcome if left untreated. It often presents with distinct similarities to hematologic malignancies. Exacerbation can occasionally occur as fulminant macrophage activation syndrome. Disease incidence is globally increasing and has not peaked as yet. A complex interplay between pathogen and the immune system is the key pathophysiological mechanism.


Subject(s)
Fever/etiology , Leishmania donovani/isolation & purification , Leishmaniasis, Visceral/diagnosis , Pancytopenia/etiology , Adult , Amphotericin B/administration & dosage , Amphotericin B/therapeutic use , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/therapeutic use , Diagnosis, Differential , Hepatomegaly/diagnostic imaging , Hepatomegaly/drug therapy , Hepatomegaly/microbiology , Humans , Leishmania donovani/genetics , Leishmaniasis, Visceral/drug therapy , Liposomes , Male , Pancytopenia/diagnosis , Splenomegaly/diagnostic imaging , Splenomegaly/drug therapy , Splenomegaly/microbiology , Tomography, X-Ray Computed , Treatment Outcome
5.
Int J Cancer ; 143(6): 1416-1425, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29659014

ABSTRACT

To evaluate the role of constitutive epigenetic changes in normal body cells of BRCA1/BRCA2-mutation negative patients, we have developed a deep bisulfite sequencing assay targeting the promoter regions of 8 tumor suppressor (TS) genes (BRCA1, BRCA2, RAD51C, ATM, PTEN, TP53, MLH1, RB1) and the estrogene receptor gene (ESR1), which plays a role in tumor progression. We analyzed blood samples of two breast cancer (BC) cohorts with early onset (EO) and high risk (HR) for a heterozygous mutation, respectively, along with age-matched controls. Methylation analysis of up to 50,000 individual DNA molecules per gene and sample allowed quantification of epimutations (alleles with >50% methylated CpGs), which are associated with epigenetic silencing. Compared to ESR1, which is representative for an average promoter, TS genes were characterized by a very low (< 1%) average methylation level and a very low mean epimutation rate (EMR; < 0.0001% to 0.1%). With exception of BRCA1, which showed an increased EMR in BC (0.31% vs. 0.06%), there was no significant difference between patients and controls. One of 36 HR BC patients exhibited a dramatically increased EMR (14.7%) in BRCA1, consistent with a disease-causing epimutation. Approximately one third (15 of 44) EO BC patients exhibited increased rates of single CpG methylation errors in multiple TS genes. Both EO and HR BC patients exhibited global underexpression of blood TS genes. We propose that epigenetic abnormalities in normal body cells are indicative of disturbed mechanisms for maintaining low methylation and appropriate expression levels and may be associated with an increased BC risk.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , CpG Islands/genetics , DNA Methylation , Epigenesis, Genetic , Mutation , Tumor Suppressor Proteins/genetics , Adult , Alleles , Breast/metabolism , Breast/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Case-Control Studies , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Prognosis , Promoter Regions, Genetic , Risk Factors
6.
Blood ; 127(17): 2101-12, 2016 04 28.
Article in English | MEDLINE | ID: mdl-26888258

ABSTRACT

Induction therapy for childhood acute lymphoblastic leukemia (ALL) traditionally includes prednisone; yet, dexamethasone may have higher antileukemic potency, leading to fewer relapses and improved survival. After a 7-day prednisone prephase, 3720 patients enrolled on trial Associazione Italiana di Ematologia e Oncologia Pediatrica and Berlin-Frankfurt-Münster (AIEOP-BFM) ALL 2000 were randomly selected to receive either dexamethasone (10 mg/m(2) per day) or prednisone (60 mg/m(2) per day) for 3 weeks plus tapering in induction. The 5-year cumulative incidence of relapse (± standard error) was 10.8 ± 0.7% in the dexamethasone and 15.6 ± 0.8% in the prednisone group (P < .0001), showing the largest effect on extramedullary relapses. The benefit of dexamethasone was partially counterbalanced by a significantly higher induction-related death rate (2.5% vs 0.9%, P = .00013), resulting in 5-year event-free survival rates of 83.9 ± 0.9% for dexamethasone and 80.8 ± 0.9% for prednisone (P = .024). No difference was seen in 5-year overall survival (OS) in the total cohort (dexamethasone, 90.3 ± 0.7%; prednisone, 90.5 ± 0.7%). Retrospective analyses of predefined subgroups revealed a significant survival benefit from dexamethasone only for patients with T-cell ALL and good response to the prednisone prephase (prednisone good-response [PGR]) (dexamethasone, 91.4 ± 2.4%; prednisone, 82.6 ± 3.2%; P = .036). In patients with precursor B-cell ALL and PGR, survival after relapse was found to be significantly worse if patients were previously assigned to the dexamethasone arm. We conclude that, for patients with PGR in the large subgroup of precursor B-cell ALL, dexamethasone especially reduced the incidence of better salvageable relapses, resulting in inferior survival after relapse. This explains the lack of benefit from dexamethasone in overall survival that we observed in the total cohort except in the subset of T-cell ALL patients with PGR. This trial was registered at www.clinicaltrials.gov (BFM: NCT00430118, AIEOP: NCT00613457).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Child , Child, Preschool , Combined Modality Therapy , Cranial Irradiation , Dexamethasone/administration & dosage , Dexamethasone/pharmacokinetics , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Kaplan-Meier Estimate , Male , Methotrexate/administration & dosage , Neoplasm, Residual , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prednisone/administration & dosage , Proportional Hazards Models , Remission Induction , Treatment Outcome
7.
BMC Cancer ; 18(1): 265, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29514593

ABSTRACT

BACKGROUND: There is no international consensus up to which age women with a diagnosis of triple-negative breast cancer (TNBC) and no family history of breast or ovarian cancer should be offered genetic testing for germline BRCA1 and BRCA2 (gBRCA) mutations. Here, we explored the association of age at TNBC diagnosis with the prevalence of pathogenic gBRCA mutations in this patient group. METHODS: The study comprised 802 women (median age 40 years, range 19-76) with oestrogen receptor, progesterone receptor, and human epidermal growth factor receptor type 2 negative breast cancers, who had no relatives with breast or ovarian cancer. All women were tested for pathogenic gBRCA mutations. Logistic regression analysis was used to explore the association between age at TNBC diagnosis and the presence of a pathogenic gBRCA mutation. RESULTS: A total of 127 women with TNBC (15.8%) were gBRCA mutation carriers (BRCA1: n = 118, 14.7%; BRCA2: n = 9, 1.1%). The mutation prevalence was 32.9% in the age group 20-29 years compared to 6.9% in the age group 60-69 years. Logistic regression analysis revealed a significant increase of mutation frequency with decreasing age at diagnosis (odds ratio 1.87 per 10 year decrease, 95%CI 1.50-2.32, p < 0.001). gBRCA mutation risk was predicted to be > 10% for women diagnosed below approximately 50 years. CONCLUSIONS: Based on the general understanding that a heterozygous mutation probability of 10% or greater justifies gBRCA mutation screening, women with TNBC diagnosed before the age of 50 years and no familial history of breast and ovarian cancer should be tested for gBRCA mutations. In Germany, this would concern approximately 880 women with newly diagnosed TNBC per year, of whom approximately 150 are expected to be identified as carriers of a pathogenic gBRCA mutation.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Biomarkers, Tumor/genetics , Genetic Testing , Germ-Line Mutation , Triple Negative Breast Neoplasms/genetics , Unilateral Breast Neoplasms/genetics , Adult , Aged , Female , Follow-Up Studies , Germany/epidemiology , Humans , Middle Aged , Prevalence , Prognosis , Triple Negative Breast Neoplasms/epidemiology , Triple Negative Breast Neoplasms/pathology , Unilateral Breast Neoplasms/epidemiology , Unilateral Breast Neoplasms/pathology , Young Adult
8.
Carcinogenesis ; 38(3): 312-320, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28426874

ABSTRACT

Breast cancer (BC) is the leading cancer in women worldwide. Changes in DNA methylation in peripheral blood could be associated with malignant diseases. Making use of screening results by llumina 27K Methylation Assay, we validated demethylation of five CpG sites of S100P gene in blood cell DNA of BC patients by three independent retrospective studies with subjects from different centers (Validation I: 235 familial BC case and 206 controls, odds ratio per -1% methylation > 1.03, and P < 6.00 × 10-8 for all five CpG sites; Validation II: 189 sporadic BC case and 189 controls, odds ratio per -1% methylation > 1.03, P < 8.0 × 10-5 for four CpG sites; Validation III: 156 sporadic BC case and 151 controls, odds ratio per -1% methylation > 1.03, P < 6.0 × 10-4 for four CpG sites). In addition, the blood-based S100P methylation pattern was similar among BC patients with differential clinical characteristics regardless of stage, receptor status and menopause status. The observed BC-associated decreased S100P methylation in blood mainly originates from the leucocytes subpopulations but not B cells. The methylation levels of most S100P CpG sites were inversely correlated with the expression of S100P in leucocytes (P < 1.2 × 10-4) and in tissue (P < 1.1 × 10-4). This study reveals significant association between blood-based decreased S100P methylation and BC, and provides another proof for the application of altered DNA methylation signatures from blood cells as potential markers for the detection of BC, especially for the early stage.


Subject(s)
Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , DNA Methylation/genetics , Genetic Predisposition to Disease , Neoplasm Proteins/genetics , Adult , Aged , Breast Neoplasms/blood , Breast Neoplasms/pathology , Calcium-Binding Proteins/blood , CpG Islands/genetics , Female , Genetic Association Studies , Humans , Middle Aged , Neoplasm Proteins/blood
10.
Mol Genet Metab ; 121(4): 297-307, 2017 08.
Article in English | MEDLINE | ID: mdl-28688840

ABSTRACT

Whole exome sequencing (WES) is well established in research and is now being introduced into clinically indicated diagnostics (so-called clinical exomes). We evaluated the diagnostic yield and clinical implications of WES in 72 patients from 60 families with undiagnosed neurodevelopmental disorders (NDD), neurometabolic disorders, and dystonias. Pathogenic or likely pathogenic variants leading to a molecular diagnosis could be identified in 21 of the 60 families (overall 35%, in 36% of patients with NDD, in 43% of patients with neurometabolic disorders, in 25% of patients with dystonias). In one family two coexisting autosomal recessive diseases caused by homozygous pathogenic variants in two different genes were diagnosed. In another family, a homozygous frameshift variant in STRADA was found to cause a severe NDD with early onset epilepsy, brain anomalies, hypotonia, heart defect, nephrocalcinosis, macrocephaly and distinctive facies so far designated as PMSE (polyhydramnios, megalencephaly, symptomatic epilepsy) syndrome. In 7 of the 21 families with a molecular diagnosis the pathogenic variants were only identified by clinical follow-up, manual reevaluation of the literature, a change of filter setting, and/or reconsideration of inheritance pattern. Most importantly, clinical implications included management changes in 8 cases and impact on family planning in 20 families with a molecular diagnosis. This study shows that reevaluation and follow-up can improve the diagnostic rate and that WES results have important implications on medical management and family planning. Furthermore, we could confirm STRADA as a gene associated with syndromic ID but find it questionable if the current designation as PMSE depicts the most important clinical features.


Subject(s)
Exome , Molecular Diagnostic Techniques/methods , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Family Planning Services , Female , Genetic Predisposition to Disease , Homozygote , Humans , Male , Neurodevelopmental Disorders/physiopathology , Pedigree , Pregnancy , Sequence Analysis, DNA , Exome Sequencing
11.
Strahlenther Onkol ; 193(6): 433-443, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28138949

ABSTRACT

BACKGROUND: Stereotactic body radiotherapy (SBRT) in pancreatic cancer can be limited by its proximity to organs at risk (OAR). In this analysis, we evaluated the toxicity and efficacy of two different treatment approaches in patients with locally recurrent or oligometastatic pancreatic cancer. MATERIALS AND METHODS: According to the prescription method, patients were divided in two cohorts (C1 and C2). The planning target volume (PTV) was created through a 4 mm expansion of the internal target volume. In C2, a subvolume was additionally created, a simultaneous integrated protection (SIP), which is the overlap of the PTV with the planning risk volume of an OAR to which we prescribed a reduced dose. RESULTS: In all, 18 patients were treated (7 with local recurrences, 9 for oligometastases, 2 for both). Twelve of 23 lesions were treated without SIP (C1) and 11 with SIP (C2). The median follow-up was 12.8 months. Median overall survival (OS) was 13.2 (95% confidence interval [CI] 9.8-14.6) months. The OS rates at 6 and 12 months were 87 and 58%, respectively. Freedom from local progression for combined cohorts at 6 and 12 months was 93 and 67% (95% CI 15-36), respectively. Local control was not statistically different between the two groups. One patient in C2 experienced grade ≥3 acute toxicities and 1 patient in C1 experienced a grade ≥3 late toxicity. CONCLUSION: The SIP approach is a useful prescription method for abdominal SBRT with a favorable toxicity profile which does not compromise local control and overall survival despite dose sacrifices in small subvolumes.


Subject(s)
Adenocarcinoma/radiotherapy , Neoplasm Recurrence, Local/radiotherapy , Pancreatic Neoplasms/radiotherapy , Radiation Protection/methods , Radiosurgery/methods , Radiotherapy, Intensity-Modulated/methods , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Cohort Studies , Combined Modality Therapy , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Organs at Risk/radiation effects , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Positron Emission Tomography Computed Tomography , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted , Radiotherapy, Adjuvant/methods , Retrospective Studies , Survival Analysis , Tomography, X-Ray Computed
12.
Am J Med Genet A ; 173(7): 1878-1886, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28489334

ABSTRACT

Neurodegeneration with brain iron accumulation (NBIA) is a group of neurodegenerative disorders characterized by iron accumulation in the basal ganglia. Recently, mutations in CoA synthase (COASY) have been identified as a cause of a novel NBIA subtype (COASY Protein-Associated Neurodegeneration, CoPAN) in two patients with dystonic paraparesis, parkinsonian features, cognitive impairment, behavior abnormalities, and axonal neuropathy. COASY encodes an enzyme required for Coenzyme A (CoA) biosynthesis. Using whole exome sequencing (WES) we identified compound heterozygous COASY mutations in two siblings with intellectual disability, ataxic gait, progressive spasticity, and obsessive-compulsive behavior. The "eye-of-the tiger-sign," a characteristic hypointense spot within the hyperintense globi pallidi on MRI found in the most common subtype of NBIA (Pantothenate Kinase-Associated Neurodegeneration, PKAN), was not present. Instead, bilateral hyperintensity and swelling of caudate nucleus, putamen, and thalamus were found. In addition, our patients showed a small corpus callosum and frontotemporal and parietal white matter changes, expanding the brain phenotype of patients with CoPAN. Metabolic investigations showed increased free carnitine and decreased acylcarnitines in the patients dried blood samples. Carnitine palmitoyl transferase 1 (CPT1) deficiency was excluded by further enzymatic and metabolic investigations. As CoA and its derivate Acetyl-CoA play an essential role in fatty acid metabolism, we assume that abnormal acylcarnitine profiles are a result of the COASY mutations. This report not only illustrates that WES is a powerful tool to elucidate the etiology of rare genetic diseases, but also identifies unique neuroimaging and metabolic findings that may be key features for an early diagnosis of CoPAN.

13.
Am J Med Genet A ; 173(5): 1369-1373, 2017 May.
Article in English | MEDLINE | ID: mdl-28371085

ABSTRACT

Recently, de novo heterozygous variants in DDX3X have been reported in about 1.5% of 2659 females with previously unexplained intellectual disability (ID). We report on the identification of DDX3X variants in two unrelated girls with clinical features of Toriello-Carey Syndrome (T-CS). In patient 1, the recurrent variant c.1703C>T; p.(P568L) was identified when reconsidering X-linked de novo heterozygous variants in exome sequencing data. In patient 2, the DDX3X variant c.1600C>G; p.(R534G) was also detected by exome sequencing. Based on these data, de novo heterozygous DDX3X variants should be considered not only in females with unexplained ID, but also in individuals with a clinical diagnosis of T-CS.


Subject(s)
Agenesis of Corpus Callosum/genetics , Craniofacial Abnormalities/genetics , DEAD-box RNA Helicases/genetics , Heart Defects, Congenital/genetics , Intellectual Disability/genetics , Limb Deformities, Congenital/genetics , Pierre Robin Syndrome/genetics , Urogenital Abnormalities/genetics , Agenesis of Corpus Callosum/diagnosis , Agenesis of Corpus Callosum/physiopathology , Child , Child, Preschool , Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/physiopathology , Exome/genetics , Female , Genes, X-Linked , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/physiopathology , Heterozygote , Humans , Intellectual Disability/diagnosis , Intellectual Disability/physiopathology , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/physiopathology , Mutation , Phenotype , Pierre Robin Syndrome/diagnosis , Pierre Robin Syndrome/physiopathology , Urogenital Abnormalities/diagnosis , Urogenital Abnormalities/physiopathology
14.
Contact Dermatitis ; 76(3): 160-166, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28090651

ABSTRACT

BACKGROUND: Hairdressing is one of the professions with the highest risk of occupational skin and respiratory diseases. The incidence of these diseases in hairdressing apprentices has been studied only sparsely. OBJECTIVE: To determine the incidence of skin and respiratory diseases in hairdressing apprentices, and to explore whether hairdressing apprentices leave the trade during training because of these diseases. METHODS: A 3-year follow-up questionnaire study was conducted among 248 hairdressing apprentices and a control group comprising 816 young adults from the general population. RESULTS: The incidence rate ratios (IRRs) for contact urticaria (IRR 4.7, 95%CI: 2.6-8.6), hand eczema (IRR 1.7, 95%CI: 1.1-2.6) and rhinitis symptoms (IRR 1.6, 95%CI: 1.2-2.2) were significantly increased in the hairdressing apprentices, whereas wheezing was similar between groups. During the follow-up period, 21.8% of the hairdressing apprentices had left the trade, and 70.3% of these had left because of health complaints. The most frequently reported reasons for leaving were musculoskeletal pain (47.4%) and skin diseases (47.4%), followed by respiratory symptoms (23.7%). CONCLUSIONS: Hairdressing apprentices are at increased risk for contact urticaria, hand eczema and rhinitis symptoms compared with the general population, and a substantial proportion leave the trade because of these diseases, causing a 'healthy worker survivor effect.'


Subject(s)
Beauty Culture/education , Dermatitis, Contact/epidemiology , Dermatitis, Occupational/epidemiology , Occupational Exposure/adverse effects , Respiration Disorders/epidemiology , Adult , Denmark , Female , Hair Preparations/adverse effects , Humans , Incidence , Male , Occupational Exposure/statistics & numerical data , Prevalence , Young Adult
15.
Hum Mol Genet ; 23(3): 590-601, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24045615

ABSTRACT

Acute lymphoblastic leukemia (ALL) accounts for ∼25% of pediatric malignancies. Of interest, the incidence of ALL is observed ∼20% higher in males relative to females. The mechanism behind the phenomenon of sex-specific differences is presently not understood. Employing genome-wide genetic aberration screening in 19 ALL samples, one of the most recurrent lesions identified was monoallelic deletion of the 5' region of SLX4IP. We characterized this deletion by conventional molecular genetic techniques and analyzed its interrelationships with biological and clinical characteristics using specimens and data from 993 pediatric patients enrolled into trial AIEOP-BFM ALL 2000. Deletion of SLX4IP was detected in ∼30% of patients. Breakpoints within SLX4IP were defined to recurrent positions and revealed junctions with typical characteristics of illegitimate V(D)J-mediated recombination. In initial and validation analyses, SLX4IP deletions were significantly associated with male gender and ETV6/RUNX1-rearranged ALL (both overall P < 0.0001). For mechanistic validation, a second recurrent deletion affecting TAL1 and caused by the same molecular mechanism was analyzed in 1149 T-cell ALL patients. Validating a differential role by sex of illegitimate V(D)J-mediated recombination at the TAL1 locus, 128 out of 1149 T-cell ALL samples bore a deletion and males were significantly more often affected (P = 0.002). The repeatedly detected association of SLX4IP deletion with male sex and the extension of the sex bias to deletion of the TAL1 locus suggest that differential illegitimate V(D)J-mediated recombination events at specific loci may contribute to the consistent observation of higher incidence rates of childhood ALL in boys compared with girls.


Subject(s)
Carrier Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recombinases/genetics , V(D)J Recombination , Adolescent , Basic Helix-Loop-Helix Transcription Factors/genetics , Child , Child, Preschool , Cohort Studies , Core Binding Factor Alpha 2 Subunit/genetics , Female , Gene Deletion , Humans , Infant , Male , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1 , ETS Translocation Variant 6 Protein
16.
Br J Haematol ; 173(5): 742-8, 2016 06.
Article in English | MEDLINE | ID: mdl-26913693

ABSTRACT

Minimal residual disease (MRD) at the end of induction therapy is important for risk stratification of acute lymphoblastic leukaemia (ALL), but bone marrow (BM) aspiration is often postponed or must be repeated to fulfil qualitative and quantitative criteria for morphological assessment of haematological remission and/or MRD analysis. The impact of BM aspiration delay on measured MRD levels and resulting risk stratification is currently unknown. We analysed paired MRD data of 289 paediatric ALL patients requiring a repeat BM aspiration. MRD levels differed in 108 patients (37%) with a decrease in the majority (85/108). This would have resulted in different risk group allocation in 64 of 289 patients (23%) when applying the ALL-Berlin-Frankfurt-Münster 2000 criteria. MRD change was associated with the duration of delay; 40% of patients with delay ≥7 days had a shift to lower MRD levels compared to only 18% after a shorter delay. Patients MRD-positive at the original but MRD-negative at the repeat BM aspiration (n = 50) had a worse 5-year event-free survival than those already negative at first aspiration (n = 115) (86 ± 5% vs. 94 ± 2%; P = 0·024). We conclude that BM aspirations should be pursued as scheduled in the protocol because delayed MRD sampling at end of induction may result in false-low MRD load and distort MRD-based risk assessment.


Subject(s)
Bone Marrow Examination/methods , Diagnostic Errors/prevention & control , Neoplasm, Residual/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adolescent , Biopsy, Fine-Needle , Child , Child, Preschool , Delayed Diagnosis , Female , Humans , Infant , Male , Neoplasm, Residual/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Remission Induction , Risk Assessment , Time Factors , Treatment Outcome
17.
Am J Med Genet A ; 170(6): 1502-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27016154

ABSTRACT

Intellectual disability (ID) with cerebellar ataxia comprises a genetically heterogeneous group of neurodevelopmental disorders. We identified a homozygous frameshift mutation in CWF19L1 (c.467delC; p.(P156Hfs*33)) by a combination of linkage analysis and Whole Exome Sequencing in a consanguineous Turkish family with a 9-year-old boy affected by early onset cerebellar ataxia and mild ID. Serial MRI showed mildly progressive cerebellar atrophy. Absent C19L1 protein expression in lymphoblastoid cell lines strongly suggested that c.467delC is a disease-causing alteration. One further pregnancy of the mother had been terminated at 22 weeks of gestation because of a small cerebellum and agenesis of corpus callosum. The homozygous CWF19L1 variant was also present in the fetus. Postmortem examination of the fetus in addition showed unilateral hexadactyly and vertebral malformations. These features have not been reported and may represent an expansion of the CWF19L1-related phenotypic spectrum, but could also be due to another, possibly autosomal recessive disorder. The exact function of the evolutionarily highly conserved C19L1 protein is unknown. So far, homozygous or compound heterozygous mutations in CWF19L1 have been identified in two Turkish siblings and a Dutch girl, respectively, affected by cerebellar ataxia and ID. A zebrafish model showed that CWF19L1 loss-of-function mutations result in abnormal cerebellar morphology and movement disorders. Our report corroborates that loss-of-function mutations in CWF19Ll lead to early onset cerebellar ataxia and (progressive) cerebellar atrophy. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Cycle Proteins/genetics , Cerebellum/abnormalities , Exome , High-Throughput Nucleotide Sequencing , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Mutation , Nervous System Malformations/diagnosis , Nervous System Malformations/genetics , Brain/abnormalities , Child , Comparative Genomic Hybridization , Consanguinity , Developmental Disabilities/diagnosis , Developmental Disabilities/genetics , Genetic Association Studies , Genetic Linkage , Humans , Magnetic Resonance Imaging , Male , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Radiography
18.
Int Urogynecol J ; 27(8): 1137-47, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26590137

ABSTRACT

Bladder pain syndrome/interstitial cystitis (BPS/IC) is a chronic disease characterised by persistent irritating micturition symptoms and pain. The objective was to compare the clinical efficacy of currently available products for intravesical therapy of BPS/IC and to assess their pharmacoeconomic impact. A Pubmed/Medline database search was performed for articles on intravesical therapy for BPS/IC. A total of 345 publications were identified, from which 326 were excluded. Statistical evaluation was performed with effect size (ES) assessment of symptom reduction and response rates. The final set of 19 articles on intravesical BPS/IC therapy included 5 prospective controlled trials (CTs), the remaining were classified as uncontrolled clinical studies. The total number of patients included was 801, 228 of whom had been evaluated in a CT. For CTs, the largest ES for symptom reduction as well as response rate was observed for high molecular weight hyaluronic acid (HMW-HA), with similar findings in two uncontrolled studies with HMW-HA. The number needed to treat to achieve a response to intravesical therapy was 2.67 for intravesical pentosan polysulphate and 1.31 for HMW-HA which were superior to all other instillates. HMW-HA was significantly superior in cost effectiveness and cost efficacy to all other instillation regimes. The present meta-analysis combined medical and pharmacoeconomic aspects and demonstrated an advantage of HMW-HA over other instillation agents; however, direct comparisons between the different products have not been performed to date in properly designed controlled studies.


Subject(s)
Cystitis, Interstitial/drug therapy , Hyaluronic Acid/administration & dosage , Administration, Intravesical , Adult , Clinical Trials as Topic , Female , Humans , Male , Treatment Outcome
19.
Int J Cancer ; 136(8): 1845-55, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25213452

ABSTRACT

Breast cancer (BC) is the leading cause of cancer-related mortality in women worldwide. Changes in DNA methylation in peripheral blood could be associated with malignancy at early stage. However, the BC-associated DNA methylation signatures in peripheral blood were largely unknown. Here, we performed a genome-wide methylation screening and identified a BC-associated differentially methylated CpG site cg27091787 in the hyaluronoglucosaminidase 2 gene (HYAL2) (discovery round with 72 BC case and 24 controls: p = 2.61 × 10(-9) adjusted for cell-type proportions). The substantially decreased methylation of cg27091787 in BC cases was confirmed in two validation rounds (first validation round with 338 BC case and 507 controls: p < 0.0001; second validation round with 189 BC case and 189 controls: p < 0.0001). In addition to cg27091787, the decreased methylation of a 650-bp CpG island shore of HYAL2 was also associated with increased risk of BC. Moreover, the expression and methylation of HYAL2 were inversely correlated with a p-value of 0.006. To note, the BC-associated decreased HYAL2 methylation was replicated in the T-cell fraction (p = 0.034). The cg27091787 methylation level enabled a powerful discrimination of early-stage BC cases (stages 0 and I) from healthy controls [area under curve (AUC) = 0.89], and was robust for the detection of BC in younger women as well (age < 50, AUC = 0.87). Our study reveals a strong association between decreased HYAL2 methylation in peripheral blood and BC, and provides a promising blood-based marker for the detection of early BC.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cell Adhesion Molecules/genetics , DNA Methylation/genetics , Genetic Predisposition to Disease/genetics , Hyaluronoglucosaminidase/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Breast Neoplasms/blood , Case-Control Studies , Cell Adhesion Molecules/blood , CpG Islands/genetics , Early Detection of Cancer/methods , Female , GPI-Linked Proteins/blood , GPI-Linked Proteins/genetics , Humans , Hyaluronoglucosaminidase/blood , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Young Adult
20.
Am J Hum Genet ; 90(4): 701-7, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22424600

ABSTRACT

Kohlschütter-Tönz syndrome (KTS) is an autosomal-recessive disease characterized by the combination of epilepsy, psychomotor regression, and amelogenesis imperfecta. The molecular basis has not yet been elucidated. Here, we report that KTS is caused by mutations in ROGDI. Using a combination of autozygosity mapping and exome sequencing, we identified a homozygous frameshift deletion, c.229_230del (p.Leu77Alafs(∗)64), in ROGDI in two affected individuals from a consanguineous family. Molecular studies in two additional KTS-affected individuals from two unrelated Austrian and Swiss families revealed homozygosity for nonsense mutation c.286C>T (p.Gln96(∗)) and compound heterozygosity for the splice-site mutations c.531+5G>C and c.532-2A>T in ROGDI, respectively. The latter mutation was also found to be heterozygous in the mother of the Swiss affected individual in whom KTS was reported for the first time in 1974. ROGDI is highly expressed throughout the brain and other organs, but its function is largely unknown. Possible interactions with DISC1, a protein involved in diverse cytoskeletal functions, have been suggested. Our finding that ROGDI mutations cause KTS indicates that the protein product of this gene plays an important role in neuronal development as well as amelogenesis.


Subject(s)
Amelogenesis Imperfecta/genetics , Dementia/genetics , Epilepsy/genetics , Membrane Proteins/genetics , Mutation , Nuclear Proteins/genetics , Base Sequence , Chromosome Mapping , Exome , Exons , Female , Heterozygote , Homozygote , Humans , Infant , Male , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL