145 results on '"Frederik J. Hes"'
Search Results
2. Supplementary Figure 1 from Infiltration of Lynch Colorectal Cancers by Activated Immune Cells Associates with Early Staging of the Primary Tumor and Absence of Lymph Node Metastases
- Author
-
Hans Morreau, Tom van Wezel, Hans F.A. Vasen, Frederik J. Hes, Carli M.J. Tops, Ekaterina S. Jordanova, Danny Goudkade, and Noel F.C.C. de Miranda
- Abstract
PDF file - 289K
- Published
- 2023
3. Data from MAX Mutations Cause Hereditary and Sporadic Pheochromocytoma and Paraganglioma
- Author
-
Mercedes Robledo, Anne-Paule Gimenez-Roqueplo, Graeme Eisenhofer, Giuseppe Opocher, Patricia L. M. Dahia, Massimo Mannelli, Karel Pacak, Felix Beuschlein, Miguel Urioste, Carli M.J. Tops, Henri J.L.M. Timmers, Elisa Taschin, Carlos Suarez, Alexander P.A. Stegmann, Frank Schillo, Macarena Ruiz-Ferrer, Giovanna Roncador, Nicole Reisch, Victoria Raymond, Elena Rapizzi, Nan Qin, Miguel Quesada-Charneco, Tamara Prodanov, Pierre-François Plouin, Peggy Pierre, Arnaud Murat, Luigi Mori, Anna Merlo, Arjen R. Mensenkamp, Rocío Letón, Jacques W.M. Lenders, Esther Korpershoek, Emiliano Honrado, Frederik J. Hes, Isabelle Guilhem, Álvaro Gómez-Graña, Encarna B. Gómez-García, Xavier Girerd, Tonino Ercolino, Ronald R. de Krijger, Mara Giacchè, Eleonora P.M. Corssmit, María-Dolores Chiara, Philippe Chanson, Maurizio Castellano, Salud Borrego, Sara Bobisse, Marinus J. Blok, Yves-Jean Bignon, Jérôme Bertherat, Sandra Bernaldo de Quirós, Marta Barontini, Laurence Amar, Aguirre A. de Cubas, Lucía Inglada-Pérez, Nasséra Abermil, Iñaki Comino-Méndez, Nicole Paes Morales, Francesca Schiavi, Alberto Cascón, and Nelly Burnichon
- Abstract
Purpose: Pheochromocytomas (PCC) and paragangliomas (PGL) are genetically heterogeneous neural crest–derived neoplasms. Recently we identified germline mutations in a new tumor suppressor susceptibility gene, MAX (MYC-associated factor X), which predisposes carriers to PCC. How MAX mutations contribute to PCC/PGL and associated phenotypes remain unclear. This study aimed to examine the prevalence and associated phenotypic features of germline and somatic MAX mutations in PCC/PGL.Design: We sequenced MAX in 1,694 patients with PCC or PGL (without mutations in other major susceptibility genes) from 17 independent referral centers. We screened for large deletions/duplications in 1,535 patients using a multiplex PCR-based method. Somatic mutations were searched for in tumors from an additional 245 patients. The frequency and type of MAX mutation was assessed overall and by clinical characteristics.Results: Sixteen MAX pathogenic mutations were identified in 23 index patients. All had adrenal tumors, including 13 bilateral or multiple PCCs within the same gland (P < 0.001), 15.8% developed additional tumors at thoracoabdominal sites, and 37% had familial antecedents. Age at diagnosis was lower (P = 0.001) in MAX mutation carriers compared with nonmutated cases. Two patients (10.5%) developed metastatic disease. A mutation affecting MAX was found in five tumors, four of them confirmed as somatic (1.65%). MAX tumors were characterized by substantial increases in normetanephrine, associated with normal or minor increases in metanephrine.Conclusions: Germline mutations in MAX are responsible for 1.12% of PCC/PGL in patients without evidence of other known mutations and should be considered in the genetic work-up of these patients. Clin Cancer Res; 18(10); 2828–37. ©2012 AACR.
- Published
- 2023
4. Data from Infiltration of Lynch Colorectal Cancers by Activated Immune Cells Associates with Early Staging of the Primary Tumor and Absence of Lymph Node Metastases
- Author
-
Hans Morreau, Tom van Wezel, Hans F.A. Vasen, Frederik J. Hes, Carli M.J. Tops, Ekaterina S. Jordanova, Danny Goudkade, and Noel F.C.C. de Miranda
- Abstract
Purpose: Lynch syndrome colorectal cancers often lose human leukocyte antigen (HLA) class I expression. The outgrowth of clones with immune evasive phenotypes is thought to be positively selected by the action of cytotoxic T cells that target HLA class I–positive cancer cells. To investigate this hypothesis, we related the type and density of tumor lymphocytic infiltrate in Lynch colorectal cancers with their HLA class I phenotype and clinicopathologic stage.Experimental Design: HLA class I expression was assessed by means of immunohistochemistry. Characterization of tumor-infiltrating lymphocytes was carried out by using a triple immunofluorescence procedure that allowed the simultaneous detection of CD3-, CD8-, and granzyme B (GZMB)-positive cells. Additional markers were also used for further characterization of an elusive CD3−/CD8−/GZMB+ cell population.Results: We discovered that high tumor infiltration by activated CD8+ T cells correlated with aberrant HLA class I expression and associated with early tumor stages (P < 0.05). CD8+ T cells were most abundant in HLA class I heterogeneous tumors (P = 0.02) and frequent in HLA class I–negative cases (P = 0.04) when compared with HLA class I–positive carcinomas. An elusive immune cell population (CD45+/CD8−/CD56−/GZMB+) was characteristic for HLA class I–negative tumors lacking lymph node metastases (P < 0.01).Conclusions: The immune system assumes an important role in counteracting the progression of Lynch colorectal cancers and in selecting abnormal HLA class I phenotypes. Our findings support the development of clinical strategies that explore the natural antitumor immune responses occurring in Lynch syndrome carriers. Clin Cancer Res; 18(5); 1237–45. ©2012 AACR.
- Published
- 2023
5. Supplementary Table 1 from Enrichment of Low Penetrance Susceptibility Loci in a Dutch Familial Colorectal Cancer Cohort
- Author
-
Tom van Wezel, Hans Morreau, Juul T. Wijnen, Jeanine J. Houwing-Duistermaat, Ian Tomlinson, Richard Houlston, Frederik J. Hes, Hans F. A. Vasen, Peter Devilee, Carli Tops, Ronald van Eijk, Shantie Jagmohan-Changur, and Anneke Middeldorp
- Abstract
Supplementary Table 1 from Enrichment of Low Penetrance Susceptibility Loci in a Dutch Familial Colorectal Cancer Cohort
- Published
- 2023
6. Data from Enrichment of Low Penetrance Susceptibility Loci in a Dutch Familial Colorectal Cancer Cohort
- Author
-
Tom van Wezel, Hans Morreau, Juul T. Wijnen, Jeanine J. Houwing-Duistermaat, Ian Tomlinson, Richard Houlston, Frederik J. Hes, Hans F. A. Vasen, Peter Devilee, Carli Tops, Ronald van Eijk, Shantie Jagmohan-Changur, and Anneke Middeldorp
- Abstract
Recent genome-wide association studies have identified several loci that confer an increased risk of colorectal cancer (CRC). We studied the role of the 8q24.21 (rs6983267), 18q21.1 (rs12953717), 15q13.3 (rs4779584), 11q23.1 (rs3802842), 8q23.3 (rs16892766), and 10p14 (rs10795668) risk variants in a series of 995 Dutch CRC cases and 1340 controls. The CRC cases were selected on basis of having a family history of CRC and/or early-onset disease. The detailed clinical and molecular data available on the cases allowed us to examine the relationship between risk variants and clinicopathologic characteristics. We replicated the association with an increased risk of CRC cancer for all loci, except 10p14. The association with the variant on chromosome 15q13.3 was confirmed for the first time. The risks associated with variants in our series were higher (not significant) than those previously reported, consistent with our series reflecting genetic enrichment. Moreover, we show that familial CRC cases possess an increased number of risk alleles compared with solitary CRC cases (early-onset; mean age at diagnosis of 48.5 years). We also identified a significant increase in the number of risk alleles in families with early-onset disease (≤50 years) compared with late-onset families (>50 years). In solitary CRC patients, enrichment for risk alleles was not observed, suggesting that other causes of increased CRC risk play a role in these cases. Overall, our results suggest that clustering of low-risk variants may explain part of the excess risk in CRC families. (Cancer Epidemiol Biomarkers Prev 2009;18(11):3062–7)
- Published
- 2023
7. Supplementary Figure Legends 1-2 from Infiltration of Lynch Colorectal Cancers by Activated Immune Cells Associates with Early Staging of the Primary Tumor and Absence of Lymph Node Metastases
- Author
-
Hans Morreau, Tom van Wezel, Hans F.A. Vasen, Frederik J. Hes, Carli M.J. Tops, Ekaterina S. Jordanova, Danny Goudkade, and Noel F.C.C. de Miranda
- Abstract
PDF file - 34K
- Published
- 2023
8. A systematic review and evidence assessment of monogenic gene-disease relationships in human female infertility and differences in sex development
- Author
-
Annelore Van Der Kelen, Özlem Okutman, Elodie Javey, Münevver Serdarogullari, Charlotte Janssens, Manjusha S Ghosh, Bart J H Dequeker, Florence Perold, Claire Kastner, Emmanuelle Kieffer, Ingrid Segers, Alexander Gheldof, Frederik J Hes, Karen Sermon, Willem Verpoest, Stéphane Viville, Clinical sciences, Centre for Medical Genetics, Brussels Heritage Lab, Medical Genetics, Faculty of Medicine and Pharmacy, Basic (bio-) Medical Sciences, UZB Other, Reproduction and Genetics, and Centre for Reproductive Medicine - Gynaecology
- Subjects
systematic review ,Reproductive Medicine ,female infertility ,reproductive genetics ,medically assisted reproduction ,monogenic ,Obstetrics and Gynecology ,Genetics(clinical) ,gene–disease relationship ,clinical validation ,Art ,differences in sex development - Abstract
BACKGROUND As in other domains of medicine, high-throughput sequencing methods have led to the identification of an ever-increasing number of gene variants in the fields of both male and female infertility. The increasing number of recently identified genes allows an accurate diagnosis for previously idiopathic cases of female infertility and more appropriate patient care. However, robust evidence of the gene–disease relationships (GDR) allowing the proper translation to clinical application is still missing in many cases. OBJECTIVE AND RATIONALE An evidence-based curation of currently identified genes involved in female infertility and differences in sex development (DSD) would significantly improve both diagnostic performance and genetic research. We therefore performed a systematic review to summarize current knowledge and assess the available GDR. SEARCH METHODS PRISMA guidelines were applied to curate all available information from PubMed and Web of Science on genetics of human female infertility and DSD leading to infertility, from 1 January 1988 to 1 November 2021. The reviewed pathologies include non-syndromic as well as syndromic female infertility, and endocrine and reproductive system disorders. The evidence that an identified phenotype is caused by pathogenic variants in a specific gene was assessed according to a standardized scoring system. A final score (no evidence, limited, moderate, strong, or definitive) was assigned to every GDR. OUTCOMES A total of 45 271 publications were identified and screened for inclusion of which 1078 were selected for gene and variant extraction. We have identified 395 genes and validated 466 GDRs covering all reported monogenic causes of female infertility and DSD. Furthermore, we present a genetic diagnostic flowchart including 105 genes with at least moderate evidence for female infertility and suggest recommendations for future research. The study did not take into account associated genetic risk factor(s) or oligogenic/polygenic causes of female infertility. WIDER IMPLICATIONS We have comprehensively reviewed the existing research on the genetics of female infertility and DSD, which will enable the development of diagnostic panels using validated genes. Whole genome analysis is shifting from predominantly research to clinical application, increasing its diagnostic potential. These new diagnostic possibilities will not only decrease the number of idiopathic cases but will also render genetic counselling more effective for infertile patients and their families.
- Published
- 2022
9. Use of sanger and next-generation sequencing to screen for mosaic and intronic APC variants in unexplained colorectal polyposis patients
- Author
-
Fadwa A. Elsayed, Hans Morreau, Frederik J. Hes, Maartje Nielsen, Carli M. J. Tops, Tom van Wezel, Clinical sciences, and Medical Genetics
- Subjects
0301 basic medicine ,Cancer Research ,Genes, APC ,Unexplained colorectal polyposis ,Adenomatous Polyposis Coli Protein ,Colorectal polyposis ,030105 genetics & heredity ,Biology ,Germline ,DNA sequencing ,03 medical and health sciences ,symbols.namesake ,Mosaic variants ,Genetics ,Humans ,Gene ,Germ-Line Mutation ,Genetics (clinical) ,Sanger sequencing ,Intronic variants ,High-Throughput Nucleotide Sequencing ,Human genetics ,APC ,Pseudoexons ,030104 developmental biology ,Adenomatous Polyposis Coli ,Oncology ,Mutation ,Dutch Population ,symbols ,Original Article ,Colorectal Neoplasms - Abstract
In addition to classic germline APC gene variants, APC mosaicism and deep intronic germline APC variants have also been reported to be causes of adenomatous polyposis. In this study, we investigated 80 unexplained colorectal polyposis patients without germline pathogenic variants in known polyposis predisposing genes to detect mosaic and deep intronic APC variants. All patients developed more than 50 colorectal polyps, with adenomas being predominantly observed. To detect APC mosaicism, we performed next-generation sequencing (NGS) in leukocyte DNA. Furthermore, using Sanger sequencing, the cohort was screened for the following previously reported deep intronic pathogenic germline APC variants: c.1408 + 731C > T, p.(Gly471Serfs*55), c.1408 + 735A > T, p.(Gly471Serfs*55), c.1408 + 729A > G, p.(Gly471Serfs*55) and c.532-941G > A, p.(Phe178Argfs*22). We did not detect mosaic or intronic APC variants in the screened unexplained colorectal polyposis patients. The results of this study indicate that the deep intronic APC variants investigated in this study are not a cause of colorectal polyposis in this Dutch population. In addition, NGS did not detect any further mosaic variants in our cohort.
- Published
- 2021
10. Chromosomal abnormalities after ICSI in relation to semen parameters: results in 1114 fetuses and 1391 neonates from a single center
- Author
-
Frederik J. Hes, A Van den Bogaert, K. Keymolen, A. Buysse, F Belva, H. Tournaye, G. Verheyen, Mathieu Roelants, M. Bonduelle, Clinical sciences, Medical Genetics, Reproduction and Genetics, Vriendenkring VUB, Faculty of Economic and Social Sciences and Solvay Business School, Faculty of Psychology and Educational Sciences, Biology, Centre for Reproductive Medicine - Gynaecology, Surgical clinical sciences, and Biology of the Testis
- Subjects
Male ,medicine.medical_specialty ,Sperm donation ,Population ,Chorionic villus sampling ,Prenatal diagnosis ,film.subject ,03 medical and health sciences ,Semen quality ,Fetus ,0302 clinical medicine ,Pregnancy ,Semen ,medicine ,Humans ,Sperm Injections, Intracytoplasmic ,Child ,education ,reproductive and urinary physiology ,Chromosome Aberrations ,education.field_of_study ,030219 obstetrics & reproductive medicine ,medicine.diagnostic_test ,urogenital system ,Obstetrics ,business.industry ,Rehabilitation ,Infant, Newborn ,Obstetrics and Gynecology ,medicine.disease ,Semen Analysis ,Reproductive Medicine ,film ,030220 oncology & carcinogenesis ,Chromosome abnormality ,Amniocentesis ,Female ,business ,Blood sampling - Abstract
STUDY QUESTION Is there a relationship between karyotype abnormalities in fetuses and children conceived by ICSI and their father’s semen parameters? SUMMARY ANSWER The de novo chromosomal abnormality rate in pre- and postnatal karyotypes of ICSI offspring was higher than in the general population and related to fathers’ sperm parameters. WHAT IS KNOWN ALREADY Several studies have reported a higher rate of de novo chromosomal anomalies in ICSI fetuses but recent data from large cohorts are limited. Overall, reported prevalences of non-inherited karyotype aberrations are increased in fetuses conceived after ICSI and vary between 1.6% and 4.2%. Only a few studies focus on the relation between karyotype anomalies in ICSI offspring and semen parameters of their fathers. Furthermore, an increased incidence of abnormal karyotypes in ICSI neonates has been described, but the rates vary widely across studies. STUDY DESIGN, SIZE, DURATION We report on karyotype results from prenatal testing by means of chorionic villus sampling and amniocentesis and results from postnatal blood sampling in offspring conceived by ICSI in a single center. Ongoing pregnancies resulting from an oocyte retrieval between January 2004 and December 2012 and after transfer of fresh ICSI embryos obtained using ejaculated or non-ejaculated sperm (fresh or frozen-thawed) were considered. Pregnancies following frozen embryo transfer, oocyte or sperm donation, IVF, preimplantation genetic testing and IVM were excluded. All abnormal prenatal results after sampling are reported irrespective of the outcome of the pregnancy. PARTICIPANTS/MATERIALS, SETTING, METHODS From the 4816 ongoing ICSI pregnancies, information on pregnancy outcome was available for 4267 pregnancies. Prenatal testing was performed in 22.3% of the pregnancies, resulting in a diagnosis in 1114 fetuses. A postnatal karyotype was obtained in 29.4% of the pregnancies in which no invasive prenatal diagnosis was performed, resulting in a total of 1391 neonates sampled. The prevalence of chromosomal anomalies according to maternal age and semen quality was analyzed with logistic regression. For definitions of normal semen quality, the World Health Organization reference values for human semen characteristics were adopted. MAIN RESULTS AND THE ROLE OF CHANCE An abnormal fetal karyotype was found in 29 singletons and 12 multiples (41/1114; 3.7%; 95% CI 2.7–4.9%): 36 anomalies were de novo (3.2%; 95% CI 2.3–4.4), either numerical (n = 25), sex (n = 6) or structural (n = 5), and five were inherited. Logistic regression analysis did not show a significant association between maternal age and a de novo chromosomal fetal abnormality (odds ratio (OR) 1.05; 95% CI 0.96–1.15; P = 0.24). In all but one case, fetuses with an abnormal karyotype were conceived by ICSI using ejaculated sperm. Abnormal karyotypes were found in 14 (1.0%; 95% CI 0.6–1.7) out of 1391 postnatal samples of children born after ICSI who were not tested prenatally: 12 were de novo anomalies and two were inherited balanced karyotypes. The 14 abnormal karyotypes were all found in children born after ICSI using ejaculated sperm. The odds of a de novo karyotype aberration increased with maternal age when combining pre- and postnatal data (OR 1.11; 95% CI 1.04–1.19). A higher rate of de novo chromosomal abnormalities was found in fetuses and children of couples with men having a sperm concentration LIMITATIONS, REASONS FOR CAUTION We cannot exclude that the observation of a higher prevalence of karyotype anomalies in ICSI offspring compared to literature data in the general population is due to enhanced surveillance after ART given the lack of a control group. Although we did not find more chromosomal anomalies after ICSI with non-ejaculated sperm, the small numbers do not allow firm conclusions. WIDER IMPLICATIONS OF THE FINDINGS The observed increased risk of a de novo karyotype anomaly after ICSI conception in couples with poor sperm warrants continued counseling toward prenatal testing. The current and widespread use of innovative non-invasive prenatal testing will result in larger datasets, adding to a balanced estimation of the prevalence of karyotype anomalies in ICSI offspring. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the Methusalem grants issued by the Vrije Universiteit Brussel. All authors declared no conflict of interest related to this study. TRIAL REGISTRATION NUMBER N/A
- Published
- 2020
11. Disease expression in juvenile polyposis syndrome: a retrospective survey on a cohort of 221 European patients and comparison with a literature-derived cohort of 473 SMAD4/BMPR1A pathogenic variant carriers
- Author
-
Maurizio Genuardi, Robert Blatter, Benjamin Tschupp, Hans F. A. Vasen, Karl Heinimann, Fiona Lalloo, Gabriela Moeslein, D. Gareth Evans, Robert Hüneburg, Frederik J. Hes, Laura Renkonen-Sinisalo, Inge Bernstein, Stefan Aretz, Chrystelle Colas, Isabel Spier, Andrew Latchford, Nicoletta Resta, Heikki Järvinen, Dora Varvara, HUS Abdominal Center, II kirurgian klinikka, Department of Surgery, University of Helsinki, Helsinki University Hospital Area, Clinical sciences, Medical Genetics, University Hospital Basel [Basel], University of Basel (Unibas), University Hospital Bonn, Aalborg University [Denmark] (AAU), Unité fonctionnelle d'Oncogénétique et Angiogénétique Moléculaire [CHU Pitié-Salpêtrière], CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Sorbonne Université (SU), Institut Curie [Paris], University of Manchester [Manchester], Fondazione 'Policlinico Universitario A. Gemelli' [Rome], Università cattolica del Sacro Cuore [Roma] (Unicatt), Leiden University Medical Center (LUMC), Universität Witten Herdecke, and University of Bari Aldo Moro (UNIBA)
- Subjects
0301 basic medicine ,polyposis ,030105 genetics & heredity ,PHENOTYPE ,Settore MED/03 - GENETICA MEDICA ,SMAD4 ,Surveys and Questionnaires ,Genetics(clinical) ,Juvenile polyposis syndrome ,Telangiectasia ,Genetics (clinical) ,Smad4 Protein ,Intestinal Polyposis ,Stomach ,1184 Genetics, developmental biology, physiology ,genotype–phenotype correlation ,syndrome ,PREVALENCE ,GENOTYPE ,3. Good health ,juvenile polyposis syndrome ,medicine.anatomical_structure ,Cohort ,medicine.symptom ,Adult ,medicine.medical_specialty ,Anemia ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,colorectal cancer ,genotype-phenotype correlation ,Article ,hereditary hemorrhagic telangiectasia ,03 medical and health sciences ,Neoplastic Syndromes, Hereditary ,Internal medicine ,medicine ,Humans ,Bone Morphogenetic Protein Receptors, Type I ,Germ-Line Mutation ,BMPR1A ,Retrospective Studies ,MUTATIONS ,business.industry ,Cancer ,Retrospective cohort study ,medicine.disease ,digestive system diseases ,DELETIONS ,030104 developmental biology ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,[SDV.SPEE]Life Sciences [q-bio]/Santé publique et épidémiologie ,3111 Biomedicine ,business - Abstract
International audience; Purpose: Juvenile polyposis syndrome (JPS) is a rare, autosomal-dominantly inherited cancer predisposition caused in approximately 50% of cases by pathogenic germline variants in SMAD4 and BMPR1A. We aimed to gather detailed clinical and molecular genetic information on JPS disease expression to provide a basis for management guidelines and establish open access variant databases.Methods: We performed a retrospective, questionnaire-based European multicenter survey on and established a cohort of SMAD4/BMPR1A pathogenic variant carriers from the medical literature.Results: We analyzed questionnaire-based data on 221 JPS patients (126 kindreds) from ten European centers and retrieved literature-based information on 473 patients. Compared with BMPR1A carriers, SMAD4 carriers displayed anemia twice as often (58% vs. 26%), and exclusively showed overlap symptoms with hemorrhagic telangiectasia (32%) and an increased prevalence (39% vs. 13%) of gastric juvenile polyps. Cancer, reported in 15% of JPS patients (median age 41 years), mainly occurred in the colorectum (overall: 62%, SMAD4: 58%, BMPR1A: 88%) and the stomach (overall: 21%; SMAD4: 27%, BMPR1A: 0%).Conclusion: This comprehensive retrospective study on genotype-phenotype correlations in 694 JPS patients corroborates previous observations on JPS in general and SMAD4 carriers in particular, facilitates recommendations for clinical management, and provides the basis for open access variant SMAD4 and BMPR1A databases.
- Published
- 2020
12. Declining detection rates for APC and biallelic MUTYH variants in polyposis patients, implications for DNA testing policy
- Author
-
Frederik J. Hes, Hans Morreau, Alexandra M. J. Langers, Sanne W. ten Broeke, Tom van Wezel, Carli M. J. Tops, Hans J. J. P. Gille, Yvonne J. Vos, Maartje Nielsen, Manon Suerink, Sunny S. Singh, Diantha Terlouw, Hans F. A. Vasen, Human genetics, CCA - Cancer biology and immunology, Internal Medicine, and Clinical sciences
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Adenoma ,polyposis ,Disease ,MOSAICISM ,Logistic regression ,Gastroenterology ,Article ,DNA Glycosylases ,Familial adenomatous polyposis ,COLORECTAL-CANCER ,MUTYH ,familial adenomatous polyposis ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Testing ,Child ,MUTYH-associated polyposis ,Alleles ,Genetics (clinical) ,Aged ,Genetic testing ,Medicine(all) ,RISK ,medicine.diagnostic_test ,IDENTIFICATION ,business.industry ,MUTYH-Associated Polyposis ,FAP ,Odds ratio ,Middle Aged ,medicine.disease ,GENE ,digestive system diseases ,GERM-LINE MUTATIONS ,PREVALENCE ,Adenomatous Polyposis Coli ,Mutation ,ONSET ,Female ,ADENOMATOUS POLYPOSIS ,business - Abstract
This study aimed to determine the prevalence of APC-associated familial adenomatous polyposis (FAP) and MUTYH-associated polyposis (MAP) in a large cohort, taking into account factors as adenoma count and year of diagnosis. All application forms used to send patients in for APC and MUTYH variant analysis between 1992 and 2017 were collected (n = 2082). Using the data provided on the application form, the APC and biallelic MUTYH prevalence was determined and possible predictive factors were examined using multivariate multinomial logistic regression analysis in SPSS. The prevalence of disease causing variants in the APC gene significantly increases with adenoma count while MAP shows a peak prevalence in individuals with 50–99 adenomas. Logistic regression analysis shows significant odds ratios for adenoma count, age at diagnosis, and, interestingly, a decline in the chance of finding a variant in either gene over time. Moreover, in 22% (43/200) of patients with FAP-related extracolonic manifestations a variant was identified. The overall detection rates are above 10% for patients with >10 adenomas aged 20 adenomas aged T variant in the tumor or a first-degree relative with >10 adenomas. Therefore, APC and MUTYH testing in patients with >10 adenomas aged 20 adenomas aged
- Published
- 2020
13. Lack of genotype-phenotype correlation in basal cell nevus syndrome: A Dutch multicenter retrospective cohort study
- Author
-
Antonius F.W. van Hout, Jasper J. van der Smagt, Cora M. Aalfs, Klara Mosterd, M.G.H.C. Reinders, Edward M. Leter, Frederik J. Hes, Lieke P.V. Berger, Anja Wagner, C. Marleen Kets, Lizet E. van der Kolk, Johan J.P. Gille, Michel van Geel, Peter M. Steijlen, B. Cosgun, Johanna M. van Hagen, Clinical Genetics, Clinical sciences, Medical Genetics, Human genetics, Amsterdam Reproduction & Development (AR&D), MUMC+: MA AIOS Dermatologie (9), RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, Dermatologie, MUMC+: MA Dermatologie (9), MUMC+: DA KG Lab Centraal Lab (9), MUMC+: MA Dermatologie (3), MUMC+: CONC Poli Dermatologie (9), MUMC+: DA KG Polikliniek (9), and Academic Medical Center
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,PTCH1 ,SUFU ,DNA Mutational Analysis ,Basal Cell Nevus Syndrome ,MEDLINE ,Dermatology ,heat map ,medulloblastoma ,Genotype phenotype ,basal cell carcinoma ,Internal medicine ,medicine ,Humans ,Basal cell carcinoma ,basal cell nevus syndrome ,Genetic Association Studies ,Netherlands ,Retrospective Studies ,Medulloblastoma ,Medicine(all) ,business.industry ,Retrospective cohort study ,medicine.disease ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,Gorlin syndrome ,genotype-phenotype ,Patched-1 Receptor ,odontogenic keratocyst ,diagnostic criteria ,Female ,business - Abstract
Contains fulltext : 225468.pdf (Publisher’s version ) (Closed access)
- Published
- 2020
14. A unique case of two somatic APC mutations in an early onset cribriform-morular variant of papillary thyroid carcinoma and overview of the literature
- Author
-
A. M. W. Van Den Ouweland, Hans Morreau, Mehtap Derya Aydemirli, Frederik J. Hes, K van der Tuin, T. van Wezel, Ellen Kapiteijn, Clinical Genetics, Clinical sciences, and Medical Genetics
- Subjects
0301 basic medicine ,Cancer Research ,Genes, APC ,endocrine system diseases ,Somatic cell ,DNA sequencing ,Deep sequencing ,Germline ,Familial adenomatous polyposis ,Thyroid carcinoma ,Young Adult ,03 medical and health sciences ,Wnt ,Rare Diseases ,0302 clinical medicine ,Genetics ,Humans ,Medicine ,Cribriform-morular ,Thyroid Neoplasms ,Family history ,Thyroid cancer ,beta Catenin ,Genetics (clinical) ,Medicine(all) ,business.industry ,FAP ,beta-catenin ,β-catenin ,medicine.disease ,APC ,030104 developmental biology ,Adenomatous Polyposis Coli ,Oncology ,Thyroid Cancer, Papillary ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Cribriform-morular variant papillary thyroid carcinoma ,Original Article ,Female ,business - Abstract
We report a case of a 22-year-old female patient who was diagnosed with a cribriform-morular variant of papillary thyroid carcinoma (CMV-PTC). While at early ages this thyroid cancer variant is highly suggestive for familial adenomatous polyposis (FAP), there was no family history of FAP. In the tumor biallelic, inactivating APC variants were identified. The patient tested negative for germline variants based on analysis of genomic DNA from peripheral blood leukocytes. Somatic mosaicism was excluded by subsequent deep sequencing of leukocyte and normal thyroid DNA using next generation sequencing (NGS). This report presents a rare sporadic case of CMV-PTC, and to the best of our knowledge the first featuring two somatic APC mutations underlying the disease, with an overview of CMV-PTC cases with detected APC and CTNNB1 pathogenic variants from the literature. Electronic supplementary material The online version of this article (10.1007/s10689-019-00146-4) contains supplementary material, which is available to authorized users.
- Published
- 2020
15. Variation in the risk of colorectal cancer in families with Lynch syndrome: a retrospective cohort study
- Author
-
Seçil Aksoy, Michael O. Woods, Heinric Williams, Bruno Buecher, Finlay A. Macrae, Lotte N. Krogh, Jay Qiu, Wan K.W. Juhari, Jan T. Lowery, Anne-Marie Gerdes, Magnus von Knebel Doeberitz, Luigi Ricciardiello, Karsten Schulmann, Jose Luis Soto, Kristina Lagerstedt-Robinson, Kiwamu Akagi, Raj Ramesar, Uffe Birk Jensen, Angel Alonso, Robert Hüneburg, Olivier Caron, Michel Longy, Jan Lubinski, Kate Green, Annabel Goodwin, D. Gareth Evans, Julie Wods, Leigha Senter, Matthew F. Kalady, Mark Clendenning, Barbara A. Leggett, Ravindran Ankathil, Swati G. Patel, Julian Barwell, Katherine M. Tucker, Grant Lee, Pascaline Berthet, Dawn M. Nixon, Sonia S. Kupfer, Naohiro Tomita, Susan Parry, Trinidad Caldés, Robert W. Haile, Edenir Inêz Palmero, Karin Alvarez, Cassandra B. Nichols, Mark A. Jenkins, N. Jewel Samadder, Loic LeMarchand, John Burn, Francisco Lopez, Rodney J. Scott, Pierre Laurent-Puig, Julie Arnold, Christina Therkildsen, Hans K. Schackert, Pilar Garre, Reinhard Buettner, Adriana Della Valle, Patricia Esperon, Wolff Schmiegel, Karl Heinimann, Inge Bernstein, Matthias Kloor, Nicoline Hoogerbrugge, Rui Manuel Reis, Fränzel J.B. Van Duijnhoven, Christoph Engel, Mohd Nizam Zahary, Sylviane Olschwang, Sapna Syngal, Valérie Bonadona, Nicholas Pachter, Matilde Navarro, Albert de la Chapelle, Beate Betz, Jukka-Pekka Mecklin, Catherine Noguès, Elena M. Stoffel, Toni T. Seppälä, Chrystelle Colas, Anneke Lucassen, Allan D. Spigelman, Youenn Drouet, Elisa J. Cops, Uri Ladabaum, Steve Thibodeau, Jeffrey N. Weitzel, Fiona Lalloo, Patrick J. Morrison, Maurizio Genuardi, Kohji Tanakaya, Patrick M. Lynch, Frederik J. Hes, William D. Foulkes, Carmen Guillén-Ponce, Jenny von Salomé, Emilia Rogoża-Janiszewska, Andrew Latchford, John L. Hopper, Carrie Snyder, Verónica Barca-Tierno, Gabriela Möslein, Lauren M. Gima, Melissa C. Southey, Paul A. James, Marion Dhooge, Claudia Perne, Steven Gallinger, Heather Hampel, Amanda B. Spurdle, Ingrid Winship, Emmanuelle Fourme, Rish K. Pai, Daniela Turchetti, Marta Pineda, Jürgen Weitz, James Hill, Daniel D. Buchanan, Carlos A. Vaccaro, Noralane M. Lindor, Rachel Pearlman, Pål Møller, Christian P. Strassburg, Jane C. Figueiredo, Aída Falcón de Vargas, Silke Zachariae, Karolin Bucksch, Joanne Ngeow, Silke Redler, Henrik Okkels, Maija R.J. Kohonen-Corish, Hans F. A. Vasen, Verena Steinke-Lange, Roselyne Guimbaud, Deepak Vangala, Isabelle Coupier, Nils Rahner, Berrin Tunca, Sanne W. Bajwa-ten Broeke, Niels de Wind, Sophie Lejeune, José Gaston Guillem, Karin Wadt, Polly A. Newcomb, Elke Holinski-Feder, Florencia Neffa, Rodrigo Santa Cruz Guindalini, Paul E. Wise, Julian R. Sampson, Graham Casey, Lene Juel Rasmussen, Rolf H. Sijmons, Tadeusz Dębniak, Ann-Sofie Backman, Joji Utsunomiya, Melyssa Aronson, Aung Ko Win, Yves-Jean Bignon, Judy W. C. Ho, Robyn L. Ward, Mev Dominguez-Valentin, Karolina Malińska, Elizabeth E. Half, John-Paul Plazzer, Marjolijn J. L. Ligtenberg, Rachel Austin, Nicola K. Poplawski, Marcia Cruz-Correa, Nagahide Matsubara, Charlotte Kvist Lautrup, Thomas Hansen, Tatsuro Yamaguchi, Thomas John, David J. Amor, Ilana Solomon, Yun-Hee Choi, Meghan J. van Wanzeele, Rakefet Shtoyerman, Vanessa Huntley, Maartje Nielsen, Deborah Neklason, Kevin J. Monahan, Gülçin Tezcan, Stefan Aretz, Talya Boisjoli, Sophie Giraud, Thierry Frebourg, Christophe Rosty, Heike Görgens, Lone Sunde, Allyson Templeton, Jacob Nattermann, Mala Pande, Joan Brunet, Nancy Uhrhammer, James M. Church, Florencia Spirandelli, Laurent Briollais, James G. Dowty, Jeanette C. Reece, Rachel Susman, Fay Kastrinos, Kirsi Pylvänäinen, Gabriel Capellá, Helène Schuster, Min H. Chew, Markus Loeffler, Christine Lasset, Michael J. Hall, Capuccine Delnatte, Floor A. Duijkers, Imagerie Moléculaire et Stratégies Théranostiques (IMoST), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Clermont Auvergne (UCA), Centre Jean Perrin [Clermont-Ferrand] (UNICANCER/CJP), UNICANCER, Digital Precision Cancer Medicine (iCAN), ATG - Applied Tumor Genomics, HUS Abdominal Center, Clinical sciences, Medical Genetics, Win A.K., Dowty J.G., Reece J.C., Lee G., Templeton A.S., Plazzer J.-P., Buchanan D.D., Akagi K., Aksoy S., Alonso A., Alvarez K., Amor D.J., Ankathil R., Aretz S., Arnold J.L., Aronson M., Austin R., Backman A.-S., Bajwa-ten Broeke S.W., Barca-Tierno V., Barwell J., Bernstein I., Berthet P., Betz B., Bignon Y.-J., Boisjoli T., Bonadona V., Briollais L., Brunet J., Bucksch K., Buecher B., Buettner R., Burn J., Caldes T., Capella G., Caron O., Casey G., Chew M.H., Choi Y.-H., Church J., Clendenning M., Colas C., Cops E.J., Coupier I., Cruz-Correa M., de la Chapelle A., de Wind N., Debniak T., Della Valle A., Delnatte C., Dhooge M., Dominguez-Valentin M., Drouet Y., Duijkers F.A., Engel C., Esperon P., Evans D.G., Falcon de Vargas A., Figueiredo J.C., Foulkes W., Fourme E., Frebourg T., Gallinger S., Garre P., Genuardi M., Gerdes A.-M., Gima L.M., Giraud S., Goodwin A., Gorgens H., Green K., Guillem J., Guillen-Ponce C., Guimbaud R., Guindalini R.S.C., Half E.E., Hall M.J., Hampel H., Hansen T.V.O., Heinimann K., Hes F.J., Hill J., Ho J.W.C., Holinski-Feder E., Hoogerbrugge N., Huneburg R., Huntley V., James P.A., Jensen U.B., John T., Juhari W.K.W., Kalady M., Kastrinos F., Kloor M., Kohonen-Corish M.R., Krogh L.N., Kupfer S.S., Ladabaum U., Lagerstedt-Robinson K., Lalloo F., Lasset C., Latchford A., Laurent-Puig P., Lautrup C.K., Leggett B.A., Lejeune S., LeMarchand L., Ligtenberg M., Lindor N., Loeffler M., Longy M., Lopez F., Lowery J., Lubinski J., Lucassen A.M., Lynch P.M., Malinska K., Matsubara N., Mecklin J.-P., Moller P., Monahan K., Morrison P.J., Nattermann J., Navarro M., Neffa F., Neklason D., Newcomb P.A., Ngeow J., Nichols C., Nielsen M., Nixon D.M., Nogues C., Okkels H., Olschwang S., Pachter N., Pai R.K., Palmero E.I., Pande M., Parry S., Patel S.G., Pearlman R., Perne C., Pineda M., Poplawski N.K., Pylvanainen K., Qiu J., Rahner N., Ramesar R., Rasmussen L.J., Redler S., Reis R.M., Ricciardiello L., Rogoza-Janiszewska E., Rosty C., Samadder N.J., Sampson J.R., Schackert H.K., Schmiegel W., Schulmann K., Schuster H., Scott R., Senter L., Seppala T.T., Shtoyerman R., Sijmons R.H., Snyder C., Solomon I.B., Soto J.L., Southey M.C., Spigelman A., Spirandelli F., Spurdle A.B., Steinke-Lange V., Stoffel E.M., Strassburg C.P., Sunde L., Susman R., Syngal S., Tanakaya K., Tezcan G., Therkildsen C., Thibodeau S., Tomita N., Tucker K.M., Tunca B., Turchetti D., Uhrhammer N., Utsunomiya J., Vaccaro C., van Duijnhoven F.J.B., van Wanzeele M.J., Vangala D.B., Vasen H.F.A., von Knebel Doeberitz M., von Salome J., Wadt K.A.W., Ward R.L., Weitz J., Weitzel J.N., Williams H., Winship I., Wise P.E., Wods J., Woods M.O., Yamaguchi T., Zachariae S., Zahary M.N., Hopper J.L., Haile R.W., Macrae F.A., Moslein G., and Jenkins M.A.
- Subjects
0301 basic medicine ,Proband ,Oncology ,Male ,Heredity ,DNA mismatch repair ,[SDV]Life Sciences [q-bio] ,SUSCEPTIBILITY ,Settore MED/03 - GENETICA MEDICA ,0302 clinical medicine ,Residence Characteristics ,Risk Factors ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,PMS2 ,ComputingMilieux_MISCELLANEOUS ,MLH1 ,Age Factors ,Middle Aged ,Penetrance ,Lynch syndrome ,3. Good health ,Pedigree ,Phenotype ,030220 oncology & carcinogenesis ,Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis ,Female ,Adult ,medicine.medical_specialty ,PENETRANCE ,congenital, hereditary, and neonatal diseases and abnormalities ,GENES ,3122 Cancers ,colorectal cancer ,BREAST ,Risk Assessment ,03 medical and health sciences ,Sex Factors ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Retrospective Studies ,business.industry ,MUTATIONS ,Cancer ,medicine.disease ,digestive system diseases ,MSH2 ,MSH6 ,MODEL ,INDIVIDUALS ,030104 developmental biology ,Lynch Syndrome ,Gene-Environment Interaction ,business - Abstract
Findings 5585 families with Lynch syndrome from 22 countries were eligible for the analysis. Of these, there were insufficient numbers to estimate penetrance for Asia and South America, and for those with EPCAM variants. Therefore, we used data (collected between July 11, 2014, and Dec 31, 2018) from 5255 families (1829 MLH1, 2179 MSH2, 798 MSH6, and 449 PMS2), comprising 79 809 relatives, recruited in 15 countries in North America, Europe, and Australasia. There was strong evidence of the existence of unknown familial risk factors modifying colorectal cancer risk for Lynch syndrome carriers (p 0 center dot 0001 for each of the three three continents). These familial risk factors resulted in a wide within-gene variation in the risk of colorectal cancer for men and women from each continent who all carried pathogenic variants in the same gene or the MSH2 c.942+3A T variant. The variation was especially prominent for MLH1 and MSH2 variant carriers, depending on gene, sex and continent, with 7-56% of carriers having a colorectal cancer penetrance of less than 20%, 9-44% having a penetrance of more than 80%, and onlyBackground Existing clinical practice guidelines for carriers of pathogenic variants of DNA mismatch repair genes (Lynch syndrome) are based on the mean age-specific cumulative risk (penetrance) of colorectal cancer for all carriers of pathogenic variants in the same gene. We aimed to estimate the variation in the penetrance of colorectal cancer between carriers of pathogenic variants in the same gene by sex and continent of residence. Methods In this retrospective cohort study, we sourced data from the International Mismatch Repair Consortium, which comprises 273 members from 122 research centres or clinics in 32 countries from six continents who are involved in Lynch syndrome research. Families with at least three members and at least one confirmed carrier of a pathogenic or likely pathogenic variant in a DNA mismatch repair gene (MLH1, MSH2, MSH6, or PMS2) were included. The families of probands with known de-novo pathogenic variants were excluded. Data were collected on the method of ascertainment of the family, sex, carrier status, cancer diagnoses, and ages at the time of pedigree collection and at last contact or death. We used a segregation analysis conditioned on ascertainment to estimate the mean penetrance of colorectal cancer and modelled unmeasured polygenic factors to estimate the variation in penetrance. The existence of unknown familial risk factors modifying colorectal cancer risk for Lynch syndrome carriers was tested by use of a Wald p value for the null hypothesis that the polygenic SD is zero. Findings 5585 families with Lynch syndrome from 22 countries were eligible for the analysis. Of these, there were insufficient numbers to estimate penetrance for Asia and South America, and for those with EPCAM variants. Therefore, we used data (collected between July 11, 2014, and Dec 31, 2018) from 5255 families (1829 MLH1, 2179 MSH2, 798 MSH6, and 449 PMS2), comprising 79 809 relatives, recruited in 15 countries in North America, Europe, and Australasia. There was strong evidence of the existence of unknown familial risk factors modifying colorectal cancer risk for Lynch syndrome carriers (pT variant. The variation was especially prominent for MLH1 and MSH2 variant carriers, depending on gene, sex and continent, with 7-56% of carriers having a colorectal cancer penetrance of less than 20%, 9-44% having a penetrance of more than 80%, and only 10-19% having a penetrance of 40-60%. Interpretation Our study findings highlight the important role of risk modifiers, which could lead to personalised risk assessments for precision prevention and early detection of colorectal cancer for people with Lynch syndrome. Funding National Health and Medical Research Council, Australia. Copyright (c) 2021 Elsevier Ltd. All rights reserved.Methods In this retrospective cohort study, we sourced data from the International Mismatch Repair Consortium, which comprises 273 members from 122 research centres or clinics in 32 countries from six continents who are involved in Lynch syndrome research. Families with at least three members and at least one confirmed carrier of a pathogenic or likely pathogenic variant in a DNA mismatch repair gene (MLH1, MSH2, MSH6, or PMS2) were included. The families of probands with known de-novo pathogenic variants were excluded. Data were collected on the method of ascertainment of the family, sex, carrier status, cancer diagnoses, and ages at the time of pedigree collection and at last contact or death. We used a segregation analysis conditioned on ascertainment to estimate the mean penetrance of colorectal cancer and modelled unmeasured polygenic factors to estimate the variation in penetrance. The existence of unknown familial risk factors modifying colorectal cancer risk for Lynch syndrome carriers was tested by use of a Wald p value for the null hypothesis that the polygenic SD is zero.
- Published
- 2021
16. Variant type is associated with disease characteristics in SDHB, SDHC and SDHD-linked phaeochromocytoma–paraganglioma
- Author
-
Birke Bausch, Hartmut P. H. Neumann, Frederik J. Hes, Eleonora P M Corssmit, Johannes A. Rijken, Jeroen C. Jansen, Leonie T. van Hulsteijn, David B. Ascher, Douglas E. V. Pires, Erik F. Hensen, Peter Devilee, Jean-Pierre Bayley, Clinical sciences, and Otolaryngology / Head & Neck Surgery
- Subjects
Adult ,Male ,0301 basic medicine ,Heterozygote ,SDHB ,Mutation, Missense ,Kaplan-Meier Estimate ,Pheochromocytoma ,030105 genetics & heredity ,Paraganglioma ,03 medical and health sciences ,Germline mutation ,Genetics ,medicine ,Humans ,Missense mutation ,Germ-Line Mutation ,Genetics (clinical) ,Medicine(all) ,Variant type ,business.industry ,Membrane Proteins ,Middle Aged ,medicine.disease ,SDHC ,Penetrance ,SDHD ,genotype-phenotype ,Succinate Dehydrogenase ,030104 developmental biology ,Head and Neck Neoplasms ,Female ,business - Abstract
BackgroundPathogenic germline variants in subunits of succinate dehydrogenase (SDHB, SDHC and SDHD) are broadly associated with disease subtypes of phaeochromocytoma–paraganglioma (PPGL) syndrome. Our objective was to investigate the role of variant type (ie, missense vs truncating) in determining tumour phenotype.MethodsThree independent datasets comprising 950 PPGL and head and neck paraganglioma (HNPGL) patients were analysed for associations of variant type with tumour type and age-related tumour risk. All patients were carriers of pathogenic germline variants in the SDHB, SDHC or SDHD genes.ResultsTruncating SDH variants were significantly over-represented in clinical cases compared with missense variants, and carriers of SDHD truncating variants had a significantly higher risk for PPGL (pSDHB truncating variants displayed a trend towards increased risk of PPGL, and all three SDH genes showed a trend towards over-representation of missense variants in HNPGL cases. Overall, variant types conferred PPGL risk in the (highest-to-lowest) sequence SDHB truncating, SDHB missense, SDHD truncating and SDHD missense, with the opposite pattern apparent for HNPGL (pConclusionsSDHD truncating variants represent a distinct group, with a clinical phenotype reminiscent of but not identical to SDHB. We propose that surveillance and counselling of carriers of SDHD should be tailored by variant type. The clinical impact of truncating SDHx variants is distinct from missense variants and suggests that residual SDH protein subunit function determines risk and site of disease.
- Published
- 2019
17. Genotype-phenotype correlations for pancreatic cancer risk in Dutch melanoma families with pathogenic CDKN2A variants
- Author
-
Theo A. M. van Os, Thomas P. Potjer, Peter C. van den Akker, Kasper A. Overbeek, Marco J. Bruno, Hans F. A. Vasen, Djuna L. Cahen, Frederik J. Hes, Jan C. Oosterwijk, Mar Rodríguez-Girondo, Anja Wagner, Lizet E. van der Kolk, Nienke van der Stoep, Gastroenterology & Hepatology, Clinical Genetics, Translational Immunology Groningen (TRIGR), Damage and Repair in Cancer Development and Cancer Treatment (DARE), Targeted Gynaecologic Oncology (TARGON), Clinical sciences, and Medical Genetics
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,endocrine system diseases ,gastroenterology ,pancreas and biliary tract ,03 medical and health sciences ,0302 clinical medicine ,p14arf ,SDG 3 - Good Health and Well-being ,CDKN2A ,Internal medicine ,Pancreatic cancer ,Genetics ,medicine ,Genetics(clinical) ,Genotype-Phenotype Correlations ,Genetics (clinical) ,business.industry ,Incidence (epidemiology) ,Melanoma ,genetic screening/counselling ,genetic screening ,Familial Melanoma ,medicine.disease ,digestive system diseases ,counselling ,030104 developmental biology ,030220 oncology & carcinogenesis ,oncology ,Medical genetics ,business ,clinical genetics - Abstract
BackgroundPathogenic variants in the CDKN2A gene are generally associated with the development of melanoma and pancreatic ductal adenocarcinoma (PDAC), but specific genotype-phenotype correlations might exist and the extent of PDAC risk is not well established for many variants.MethodsUsing the Dutch national familial melanoma database, we identified all families with a pathogenic CDKN2A variant and investigated the occurrence of PDAC within these families. We also estimated the standardised incidence ratio and lifetime PDAC risk for carriers of a highly prevalent variant in these families.ResultsWe identified 172 families in which 649 individuals carried 15 different pathogenic variants. The most prevalent variant was the founder mutation c.225_243del (p16-Leiden, 484 proven carriers). Second most prevalent was c.67G>C (55 proven carriers). PDAC developed in 95 of 163 families (58%, including 373 of 629 proven carriers) harbouring a variant with an effect on the p16INK4a protein, whereas PDAC did not occur in the 9 families (20 proven carriers) with a variant affecting only p14ARF. In the c.67G>C families, PDAC occurred in 12 of the 251 (5%) persons at risk. The standardised incidence ratio was 19.1 (95% CI 8.3 to 33.6) and the cumulative PDAC incidence at age 75 years (lifetime risk) was 19% (95% CI 7.5% to 30.1%).ConclusionsOur results support the notion that pathogenic CDKN2A variants affecting the p16INK4a protein, including c.67G>C, are associated with increased PDAC risk and carriers of such variants should be offered pancreatic cancer surveillance. There is no clinical evidence that impairment of only the p14ARF protein leads to an increased PDAC risk.
- Published
- 2021
18. Duodenal adenomas and cancer in MUTYH-associated polyposis: an international cohort study
- Author
-
Andrew Latchford, A. Alonso Sanchez, Patrick M. Lynch, Matthew Mort, Joanna J Hurley, Maria Teresa Ricci, E. Meuser, Marco Vitellaro, Hannah West, A. Backman, Yann Parc, Susan K. Clark, M. R. Aznárez, Frederik J. Hes, Sunil Dolwani, O. Vinet, H. Leon Brito, Maureen E. Mork, Chrystelle Colas, M. Navarro Garcia, S. Gonzalez Romero, Sarah-Jane Walton, Hala Jundi, Gabriel Capellá, S. Kelland, Zeinab Ghorbanoghli, K van der Tuin, Hans F. A. Vasen, Julian R. Sampson, Jan Björk, Evelien Dekker, Jean-Christophe Saurin, Eduardo Vilar, Laura E. Thomas, M. Gonn, Maartje Nielsen, Clinical sciences, Medical Genetics, Gastroenterology and Hepatology, APH - Quality of Care, and AGEM - Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
Adult ,Male ,medicine.medical_specialty ,Duodenum ,Gastroenterology ,DNA Glycosylases ,Familial adenomatous polyposis ,Adenomatous Polyps ,Young Adult ,Duodenal Neoplasms ,Internal medicine ,Prevalence ,medicine ,Humans ,Genetics(clinical) ,Prospective Studies ,Intestinal Mucosa ,Endoscòpia ,Polyposis ,Càncer ,Duodenoscopy ,Aged ,Cancer ,Aged, 80 and over ,Hepatology ,Spigelman Stage ,business.industry ,MUTYH-Associated Polyposis ,Endoscopy ,Middle Aged ,medicine.disease ,digestive system diseases ,Estudi de casos ,Female ,Case studies ,business ,Cohort study - Abstract
Although duodenal adenomas and cancer appear to occur significantly less frequently in autosomal recessive MUTYH-associated polyposis (MAP) than in autosomal dominant familial adenomatous polyposis (FAP),1 current guidelines recommend similar endoscopic surveillance for both disorders.2-4 This involves gastro-duodenoscopy starting at 25 to 35 years of age and repeated at intervals determined by Spigelman staging based on the number, size, histological type and degree of dysplasia of adenomas, and by ampullary staging. Case reports of duodenal cancers in MAP suggest that they may develop in the absence of advanced Spigelman stage benign disease and even without coexisting adenomas.1 Recent molecular analyses suggest thatMAPduodenal adenomashave a higher mutational burden than FAP adenomas and are more likely to harbor oncogenic drivermutations, such as those in KRAS.5 These apparent differences in the biology and natural history of duodenal polyposis in FAP and MAP challenge the assumption that the same surveillance should be applied in both conditions.
- Published
- 2021
19. Germline DLST variants promote epigenetic modifications in pheochromocytoma-paraganglioma
- Author
-
Judith Favier, Judith V.M.G. Bovée, Peter Devilee, Erik F. Hensen, Anne-Paule Gimenez-Roqueplo, Frederik J. Hes, Aurélien Morini, Heggert Rebel, Jean-Pierre Bayley, Juan Zhang, Alexandre Buffet, Eleonora P M Corssmit, Jeroen C. Jansen, Clinical sciences, Medical Genetics, Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Paris-Centre de Recherche Cardiovasculaire (PARCC (UMR_S 970/ U970)), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), and Université de Paris (UP)
- Subjects
0301 basic medicine ,Male ,dihydrolipoamide S-succinyltransferase ,[SDV]Life Sciences [q-bio] ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Adrenal Gland Neoplasms ,Compound heterozygosity ,medicine.disease_cause ,Biochemistry ,Germline ,Epigenesis, Genetic ,Exon ,0302 clinical medicine ,Endocrinology ,Paraganglioma ,Missense mutation ,Genetics(clinical) ,Genetics ,Prognosis ,3. Good health ,030220 oncology & carcinogenesis ,Female ,reproductive medicine ,Adult ,DLST ,medicine.medical_specialty ,2-oxoglutarate dehydrogenase ,Context (language use) ,Pheochromocytoma ,Biology ,Methylation ,03 medical and health sciences ,Young Adult ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Aged ,Biochemistry (medical) ,DNA Methylation ,medicine.disease ,030104 developmental biology ,Carcinogenesis ,Acyltransferases ,Biomarkers - Abstract
Context Pheochromocytomas and paragangliomas (PPGLs) are neuroendocrine tumors in which altered central metabolism appears to be a major driver of tumorigenesis, and many PPGL genes encode proteins involved in the tricarboxylic acid (TCA) cycle. Objective/design While about 40% of PPGL cases carry a variant in a known gene, many cases remain unexplained. In patients with unexplained PPGL showing clear evidence of a familial burden or multiple tumors, we aimed to identify causative factors using genetic analysis of patient DNA and functional analyses of identified DNA variants in patient tumor material and engineered cell lines. Patients and Setting Patients with a likely familial cancer burden of pheochromocytomas and/or paragangliomas and under investigation in a clinical genetic and clinical research setting in university hospitals. Results While investigating unexplained PPGL cases, we identified a novel variant, c.1151C>T, p.(Pro384Leu), in exon 14 of the gene encoding dihydrolipoamide S-succinyltransferase (DLST), a component of the multi-enzyme complex 2-oxoglutarate dehydrogenase. Targeted sequence analysis of further unexplained cases identified a patient carrying a tumor with compound heterozygous variants in DLST, consisting of a germline variant, c.1121G>A, p.(Gly374Glu), together with a somatic missense variant identified in tumor DNA, c.1147A>G, p.(Thr383Ala), both located in exon 14. Using a range of in silico and functional assays we show that these variants are predicted to be pathogenic, profoundly impact enzyme activity, and result in DNA hypermethylation. Conclusions The identification and functional analysis of these DLST variants further validates DLST as an additional PPGL gene involved in the TCA cycle.
- Published
- 2021
20. Erratum to: Health of 2-year-old children born after vitrified oocyte donation in comparison with peers born after fresh oocyte donation
- Author
-
H. Tournaye, Maryse Bonduelle, Greta Verheyen, Frederik J. Hes, A. Buysse, Christophe Blockeel, Florence Belva, Mathieu Roelants, Marjan Van Reckem, Surgical clinical sciences, Reproduction and Genetics, Centre for Reproductive Medicine - Gynaecology, Clinical sciences, Vriendenkring VUB, Medical Genetics, and Biology of the Testis
- Subjects
medicine.medical_specialty ,Obstetrics ,business.industry ,Offspring ,Reproductive medicine ,MEDLINE ,Anthropometry ,Oocyte ,AcademicSubjects/MED00905 ,vitrified oocyte donation ,medicine.anatomical_structure ,Donation ,Oocyte donation ,embryology ,medicine ,Genetics(clinical) ,Observational study ,Erratum ,fresh oocyte donation ,business ,reproductive medicine - Abstract
Study question Does oocyte vitrification adversely affect the health of 2-year-old children compared with peers born after use of fresh oocytes in a donation programme? Summary answer The growth and health of 2-year-old children born after oocyte vitrification are similar to those of peers born after use of fresh oocytes. What is known already Although oocyte vitrification is a well-established procedure in ART, the evidence on its safety for offspring is limited. Currently, no disadvantageous effects of oocyte vitrification have been shown in terms of obstetric and neonatal outcome. However, no data beyond the neonatal period are available to date. Study design size duration A combined retrospective and prospective observational study was performed in a tertiary reproductive centre. The retrospective data were available in our extensive database of children born after ART. Donor cycles with an oocyte retrieval between January 2010 and March 2017 and a fresh embryo transfer resulting in the livebirth of a singleton were selected from the established oocyte donation programme. Fresh or vitrified oocytes were used in the donor cycles and all pregnancies in oocyte recipients were achieved after ICSI. Only children residing in Belgium were eligible for follow-up. Participants/materials setting methods Biometric and health parameters of 72 children born after oocyte vitrification were compared with those of 41 children born after use of a fresh oocyte. Data were collected by means of questionnaires and physical examinations at the age of 21-30 months. The primary outcome measures were anthropometry and health at 2 years of age. Main results and the role of chance Length, weight, BMI, head circumference, left arm circumference and waist circumference at the age of 2 years were comparable between the vitrification and fresh group, also after adjustment for treatment, and maternal and neonatal characteristics (all P > 0.05). Health of the children in terms of hospital admission and surgical intervention rates were comparable between the vitrification and fresh group (both P > 0.05). Limitations reasons for caution Although the current study is the largest series describing health parameters beyond the neonatal period, the small numbers still preclude definite conclusions. Wider implications of the findings This study provides the first evidence indicating that oocyte vitrification does not adversely affect the growth and health of offspring beyond the neonatal period. Study funding/competing interests This study was supported by Methusalem grants and by grants from Wetenschappelijk Fonds Willy Gepts, all issued by the Vrije Universiteit Brussel. All co-authors declared no conflict of interest in relation to this work. Both the Centre for Reproductive Medicine and the Centre for Medical Genetics from the UZ Brussel have received several educational grants from IBSA, Ferring, MSD and Merck for either research on oocyte vitrification or for establishing the database for follow-up research and organizing the data collection.
- Published
- 2021
21. The complexity of screening PMS2 in DNA isolated from formalin-fixed paraffin-embedded material
- Author
-
Tom van Wezel, Ronald van Eijk, Juul T. Wijnen, Anne M.L. Jansen, Maartje Nielsen, Hans Morreau, Dina Ruano, Carli M. J. Tops, Frederik J. Hes, Sanne W. ten Broeke, and Clinical sciences
- Subjects
Adult ,Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Tissue Fixation ,Pseudogene ,DNA Fragmentation ,Biology ,Sensitivity and Specificity ,Article ,Frameshift mutation ,03 medical and health sciences ,Formaldehyde ,Genetics ,PMS2 ,Humans ,Genetic Testing ,Gene conversion ,Poly-ADP-Ribose Binding Proteins ,Gene ,Genetics (clinical) ,Aged ,DNA Polymerase III ,Mismatch Repair Endonuclease PMS2 ,Aged, 80 and over ,Medicine(all) ,0303 health sciences ,Paraffin Embedding ,POLD1 ,030305 genetics & heredity ,DNA Polymerase II ,Sequence Analysis, DNA ,DNA ,Middle Aged ,Amplicon ,MMR ,digestive system diseases ,Lynch syndrome ,Mutation ,Screening ,Female ,DNA mismatch repair ,Colorectal Neoplasms ,Pseudogenes - Abstract
Germline variants in the DNA mismatch repair (MMR) gene PMS2 cause 1-14% of all Lynch Syndrome cancers. Correct variant analysis of PMS2 is complex due to the presence of multiple pseudogenes and the occurrence of gene conversion. The analysis complexity increases in highly fragmented DNA from formalin-fixed paraffin-embedded (FFPE) tissue. Here we describe a reliable approach to detect true PMS2 variants in fragmented DNA. A custom NGS panel designed for FFPE tissue was used targeting four MMR genes, POLE and POLD1. Amplicon design for PMS2 was based on the position of paralogous sequence variants (PSVs) that distinguish PMS2 from its pseudogenes. PMS2 variants in exons 1-11 can be correctly curated based on this information. For exons 12-15 this is less reliable as these undergo gene conversion. Using this method, we screened PMS2 variants in 125 MMR-deficient tumors. Of the 125 tumors tested, six were unexplained MMR-deficient tumors with solitary PMS2 protein expression loss. In these six tumors two unclassified variants (class 3) and five variants likely affecting function (class 4/5) were detected in PMS2. One microsatellite unstable tumor with positive staining for all MMR proteins was found to carry a frameshift PMS2 variant (class 5). No class 4 or class 5 PMS2 variants were detected in tumors with other patterns of MMR protein expression loss.
- Published
- 2020
22. OUP accepted manuscript
- Author
-
Florence Belva, Sietske Vermaning, Jean De Schepper, Frederik J. Hes, Michel De Vos, Sonja Desmyttere, Mathieu Roelants, Herman Tournaye, and Maryse Bonduelle
- Subjects
0301 basic medicine ,medicine.medical_specialty ,030219 obstetrics & reproductive medicine ,Obstetrics ,business.industry ,medicine.medical_treatment ,Anthropometry ,medicine.disease ,Polycystic ovary ,Intracytoplasmic sperm injection ,Gestational diabetes ,03 medical and health sciences ,Birth order ,030104 developmental biology ,0302 clinical medicine ,Cohort ,medicine ,business ,Body mass index ,Cohort study - Abstract
Study question Does In vitro maturation (IVM) of immature oocytes affect health, including growth at 2 years of age, in singletons born to mothers with polycystic ovary syndrome (PCOS)? Summary answer This study of 92 singletons born after IVM in mothers with PCOS showed no significant differences in anthropometry and health outcome parameters in comparison with a cohort of 74 peers born after intracytoplasmic sperm injection (ICSI) and conventional controlled ovarian stimulation (COS) in mothers with PCOS. What is known already IVM has been used worldwide in women with PCOS. However, the paucity of available data related to children's health following IVM is an important impediment to a more widespread use of the technology. Although previous reports on the neonatal outcome after IVM are generally reassuring, these studies have flaws that hamper the interpretation of outcomes. Moreover, few studies have reported on health outcomes after IVM beyond infancy, and particularly growth data in children born after IVM of immature oocytes from mothers with PCOS are lacking. Study design size duration This single-center cohort study compared anthropometry and health outcomes in 92 singletons born after ICSI of in vitro matured oocytes with 74 singletons born after ICSI without IVM (COS). All participants were born to mothers who were diagnosed with PCOS phenotype A, B, C or D and reached the age of 2 years between November 2012 and June 2019. Singletons born after COS were randomly selected for follow-up until young adulthood. Participants/materials setting methods Anthropometric parameters and health status data were prospectively collected at birth, 4 months and 2 years in cohorts of singletons followed since birth. Results were adjusted for neonatal (birthweight z-score, birth order), treatment (day of transfer, number of embryos transferred, mode of transfer) and parental (maternal smoking, age, body mass index (BMI), anti-Mullerian hormone level, PCOS phenotype, gestational diabetes, hypertensive disorder and paternal BMI) characteristics. Main results and the role of chance Overall, no differences were found for bodyweight, height and head circumference z-score between IVM and COS children at birth, at 4 months or at 2 years (all P > 0.05). In addition, z-scores of waist and mid-upper arm circumference at 2 years were comparable in IVM and COS children. Adjustment for covariates did not change the conclusion. Surgical intervention rate as well as the hospital admission rate were comparable between the IVM and COS group (all P > 0.05). The proportion of children born to mothers with metabolically unfavorable PCOS phenotypes (A and C) was comparable in the two groups (52.1% in IVM and 45.9% in COS). Mothers giving birth to a child conceived using IVM were younger than mothers in the COS group but their BMI was comparable. Limitations reasons for caution Although our study describes the largest cohort to date of singletons born after IVM applied in mothers with well-defined PCOS phenotypes, the current sample size only allowed us to detect moderate differences in anthropometry. Also, follow-up of children born after IVM for indications other than PCOS, for example fertililty preservation after cancer diagnosis, is highly recommended. Wider implications of the findings We did not observe adverse effects of IVM on growth parameters in offspring ~2 years of age compared to COS, but future studies should focus on cardiovascular and metabolic outcomes in these children and adolescents given their mother's PCOS condition. Study funding/competing interests This study was supported by Methusalem grants and by grants from Wetenschappelijk Fonds Willy Gepts; all issued by the Vrije Universiteit Brussel (VUB). All co-authors, except M.B., M.D.V. and H.T. declared no conflict of interest. M.B. has received consultancy fees from MSD, Serono Symposia and Merck. M.D.V. has received fees for lectures from MSD, Ferring, Gedeon Richter and Cook Medical. H.T. has received consultancy fees from Gedeon Richter, Merck, Ferring, Abbott and ObsEva. The Universitair Ziekenhuis Brussel (UZ Brussel) and the Center for Medical Genetics have received several educational grants from IBSA, Ferring, MSD and Merck for establishing the database for follow-up research and organizing the data collection.
- Published
- 2020
23. Role of germline aberrations affecting CTNNA1, MAP3K6 and MYD88 in gastric cancer susceptibility
- Author
-
Encarna B. Gomez Garcia, Liesbeth Spruijt, Rolf H. Sijmons, Cora M. Aalfs, Margreet G. E. M. Ausems, Inga Bjørnevoll, Marjolijn J. L. Ligtenberg, Frederik J. Hes, Hans K. Schackert, Anna Jakubowska, Carla Oliveira, Rachel S. van der Post, Guglielmina Nadia Ranzani, Jan Lubinski, Annemieke Cats, Urszula Teodorczyk, Liselotte P. van Hest, Ingrid P. Vogelaar, Eveline J. Kamping, J. Han van Krieken, Maurizio Genuardi, Robbert D.A. Weren, Lizet E. van der Kolk, Elke Holinski-Feder, Anja Wagner, Nicoline Hoogerbrugge, Instituto de Investigação e Inovação em Saúde, Medical Genetics, Human genetics, CCA - Cancer biology and immunology, Clinical Genetics, RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Polikliniek (9), Klinische Genetica, and Human Genetics
- Subjects
0301 basic medicine ,Male ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,ACCURACY ,MAP Kinase Kinase Kinases / genetics ,heritability ,medicine.disease_cause ,GUIDELINES ,Germline ,Cohort Studies ,0302 clinical medicine ,Cadherins / genetics ,Genotype ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,ctnna1-map3k6-myd88 ,genetics ,Genetics(clinical) ,Genetics (clinical) ,Genetics ,Mutation ,education.field_of_study ,High-Throughput Nucleotide Sequencing ,Middle Aged ,Cadherins ,MAP Kinase Kinase Kinases ,Myeloid Differentiation Factor 88 / genetics ,Europe ,030220 oncology & carcinogenesis ,MOLECULAR INVERSION PROBES ,CARCINOMAS ,Female ,Antigens, CD / genetics ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adult ,Population ,Single-nucleotide polymorphism ,Biology ,cancer: gastric ,ctnna1 – map3k6 – myd88 ,next generation sequencing ,03 medical and health sciences ,Young Adult ,Germline mutation ,SDG 3 - Good Health and Well-being ,Antigens, CD ,Stomach Neoplasms ,Next generation sequencing ,medicine ,Cancer Genetics ,Humans ,Genetic Predisposition to Disease ,Stomach Neoplasms / genetics ,Allele ,alpha Catenin / genetics ,education ,Germ-Line Mutation ,Aged ,Settore MED/06 - ONCOLOGIA MEDICA ,CDH1 ,MUTATIONS ,Cancer ,Sequence Analysis, DNA ,medicine.disease ,030104 developmental biology ,Myeloid Differentiation Factor 88 ,alpha Catenin - Abstract
BackgroundIn approximately 10% of all gastric cancer (GC) cases, a heritable cause is suspected. A subset of these cases have a causative germline CDH1 mutation; however, in most cases the cause remains unknown. Our objective was to assess to what extent these remaining cases may be explained by germline mutations in the novel candidate GC predisposing genes CTNNA1, MAP3K6 or MYD88.MethodsWe sequenced a large cohort of unexplained young and/or familial patients with GC (n=286) without a CDH1germline mutation for germline variants affecting CTNNA1, MAP3K6 and MYD88 using a targeted next-generation sequencing approach based on single-molecule molecular inversion probes.ResultsPredicted deleterious germline variants were not encountered in MYD88, but recurrently observed in CTNNA1 (n=2) and MAP3K6 (n=3) in our cohort of patients with GC. In contrast to deleterious variants in CTNNA1, deleterious variants in MAP3K6 also occur frequently in the general population.ConclusionsBased on our results MAP3K6 should no longer be considered a GC predisposition gene, whereas deleterious CTNNA1 variants are confirmed as an infrequent cause of GC susceptibility. Biallelic MYD88 germline mutations are at most a very rare cause of GC susceptibility as no additional cases were identified.
- Published
- 2018
24. The penetrance of paraganglioma and pheochromocytoma in SDHB germline mutation carriers
- Author
-
Charles R. Leemans, Johannes A. Rijken, Peter H. Bisschop, N.D. Niemeijer, Eleonora P M Corssmit, A. van Berkel, A. N. A. van der Horst-Schrivers, Marianne A. Jonker, Erik F. Hensen, Jeroen C. Jansen, Frederik J. Hes, Michiel N. Kerstens, Koen M.A. Dreijerink, Henri J L M Timmers, M. F. van Dooren, Henricus P. M. Kunst, and Karin Eijkelenkamp
- Subjects
0301 basic medicine ,Oncology ,Genetics ,medicine.medical_specialty ,Hereditary Paraganglioma ,SDHB ,business.industry ,medicine.disease ,Penetrance ,Pheochromocytoma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Germline mutation ,Paraganglioma ,030220 oncology & carcinogenesis ,Internal medicine ,Mutation (genetic algorithm) ,medicine ,Medical genetics ,business ,Genetics (clinical) - Abstract
Germline mutations in succinate dehydrogenase B (SDHB) predispose to hereditary paraganglioma (PGL) syndrome type 4. The risk of developing PGL or pheochromocytoma (PHEO) in SDHB mutation carriers is subject of recent debate. In the present nationwide cohort study of SDHB mutation carriers identified by the clinical genetics centers of the Netherlands, we have calculated the penetrance of SDHB associated tumors using a novel maximum likelihood estimator. This estimator addresses ascertainment bias and missing data on pedigree size and structure. A total of 195 SDHB mutation carriers were included, carrying 27 different SDHB mutations. The 2 most prevalent SDHB mutations were Dutch founder mutations: a deletion in exon 3 (31% of mutation carriers) and the c.423+1G>A mutation (24% of mutation carriers). One hundred and twelve carriers (57%) displayed no physical, radiological or biochemical evidence of PGL or PHEO. Fifty-four patients had a head and neck PGL (28%), 4 patients had a PHEO (2%), 26 patients an extra-adrenal PGL (13%). The overall penetrance of SDHB mutations is estimated to be 21% at age 50 and 42% at age 70 when adequately corrected for ascertainment. These estimates are lower than previously reported penetrance estimates of SDHB-linked cohorts. Similar disease risks are found for different SDHB germline mutations as well as for male and female SDHB mutation carriers.
- Published
- 2017
25. The phenotype of SDHB germline mutation carriers
- Author
-
Johannes A. Rijken, Henricus P. M. Kunst, C. René Leemans, Michiel N. Kerstens, Eleonora P M Corssmit, Alberto M. Pereira, Peter H. Bisschop, Anouk N A van der Horst-Schrivers, Erik F. Hensen, Marieke F. van Dooren, Henri J L M Timmers, Jeroen C. Jansen, Koen M.A. Dreijerink, Karin Eijkelenkamp, Carli M. J. Tops, Frederik J. Hes, Nicolasine D. Niemeijer, Jean-Pierre Bayley, Anouk van Berkel, Clinical Genetics, Guided Treatment in Optimal Selected Cancer Patients (GUTS), Medical Genetics, Otolaryngology / Head & Neck Surgery, CCA - Cancer biology and immunology, APH - Quality of Care, Amsterdam Gastroenterology Endocrinology Metabolism, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, AMS - Amsterdam Movement Sciences, and Endocrinology
- Subjects
Male ,PHEOCHROMOCYTOMA ,SDHB ,Adrenal Gland Neoplasms/diagnosis ,Endocrinology, Diabetes and Metabolism ,NETHERLANDS ,CLINICAL-MANIFESTATIONS ,medicine.disease_cause ,Germ-Line Mutation/genetics ,0302 clinical medicine ,Endocrinology ,Renal cell carcinoma ,Paraganglioma ,Stromal tumor ,Child ,Medicine(all) ,Mutation ,Vascular damage Radboud Institute for Molecular Life Sciences [Radboudumc 16] ,General Medicine ,Middle Aged ,Phenotype ,PARAGANGLIOMA ,030220 oncology & carcinogenesis ,Cohort studies ,young adult ,Female ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Pheochromocytoma/diagnosis ,Adult ,medicine.medical_specialty ,Heterozygote ,Adolescent ,GENETICS ,030209 endocrinology & metabolism ,Netherlands/epidemiology ,Pheochromocytoma ,03 medical and health sciences ,Germline mutation ,Succinate Dehydrogenase/genetics ,Internal medicine ,Paraganglioma/diagnosis ,medicine ,Humans ,Aged ,Retrospective Studies ,business.industry ,Retrospective cohort study ,medicine.disease ,Head and Neck Neoplasms/diagnosis ,business ,Follow-Up Studies - Abstract
Objective Succinate dehydrogenase B subunit (SDHB) gene germline mutations predispose to pheochromocytomas, sympathetic paragangliomas, head and neck paragangliomas and non-paraganglionic tumors (e.g. renal cell carcinoma, gastrointestinal stromal tumor and pituitary neoplasia). The aim of this study was to determine phenotypical characteristics of a large Dutch cohort of SDHB germline mutation carriers and assess differences in clinical phenotypes related to specific SDHB mutations. Design Retrospective descriptive study. Methods Retrospective descriptive study in seven academic centers. Results We included 194 SDHB mutation carriers consisting 65 (33.5%) index patients and 129 (66.5%) relatives. Mean age was 44.8 ± 16.0 years. Median duration of follow-up was 2.6 years (range: 0–36). Sixty persons (30.9%) carried the exon 3 deletion and 46 (23.7%) the c.423 + 1G > A mutation. Fifty-four mutation carriers (27.8%) had one or multiple head and neck paragangliomas, 4 (2.1%) had a pheochromocytoma and 26 (13.4%) had one or more sympathetic paragangliomas. Fifteen patients (7.7%) developed metastatic paraganglioma and 17 (8.8%) developed non-paraganglionic tumors. At study close, there were 111 (57.2%) unaffected mutation carriers. Statistical analyses showed no significant differences in the number and location of head and neck paragangliomas, sympathetic paragangliomas or pheochromocytomas, nor in the occurrence of metastatic disease or other tumors between carriers of the two founder SDHB mutations (exon 3 deletion vs c.423 + 1G > A). Conclusions In this nationwide study of disease-affected and unaffected SDHB mutation carriers, we observed a lower rate of metastatic disease and a relatively high number of head and neck paragangliomas compared with previously reported referral-based cohorts.
- Published
- 2017
26. Age and Tumor Volume Predict Growth of Carotid and Vagal Body Paragangliomas
- Author
-
Berit M. Verbist, Frederik J. Hes, Andel G. L. van der Mey, Jeroen C. Jansen, Berdine L. Heesterman, Eleonora P M Corssmit, Peter Paul G. van Benthem, Lisa M. H. de Pont, and Medical Genetics
- Subjects
medicine.medical_specialty ,growth ,medicine.medical_treatment ,carotid body tumors ,Wait and scan ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,030223 otorhinolaryngology ,Medicine(all) ,Natural course ,medicine.diagnostic_test ,business.industry ,Proportional hazards model ,Magnetic resonance imaging ,Retrospective cohort study ,prediction ,succinate dehydrogenase ,medicine.disease ,paragangliomas ,SDHD ,Surgery ,Radiation therapy ,vagal body ,030220 oncology & carcinogenesis ,Referral center ,Neurology (clinical) ,Radiology ,business ,Progressive disease - Abstract
Objective Treatment for head and neck paragangliomas (HNGPL) can be more harmful than the disease. After diagnosis, an initial period of surveillance is often indicated, and surgery or radiotherapy is reserved for progressive disease. With the aim to optimize this “wait and scan” strategy, we studied growth and possible predictors. Design A retrospective cohort study was conducted. Setting This study was conducted at a tertiary referral center for patients with HNGPL. Methods Tumor volume was estimated for 184 SDHD-related carotid and vagal body paragangliomas using sequential magnetic resonance imaging. Cox regression was used to study predictors of tumor growth. Results The estimated fraction of growing tumors ranged from 0.42 after 1 year of follow-up to 0.85 after 11 years. A median growth rate of 10.4 and 12.0% per year was observed for carotid and vagal body tumors, respectively. Tumor location, initial volume, and age (p Conclusions We created a prediction model (available online), enabling a more individualized “wait and scan” strategy. The favorable natural course of carotid and vagal body paragangliomas was confirmed; although with long follow-up growth will be observed in most cases.
- Published
- 2017
27. A novel keratin 13 variant in a four‐generation family with white sponge nevus
- Author
-
W.H. Irwin McLean, Maarten H. Vermeer, Alexander A.W. Peters, Vincent T.H.B.M. Smit, Tjalling Bosse, Hakima El Idrissi, Stephanie B de Haseth, Frederik J. Hes, Egbert Bakker, Anna Terron-Kwiatkowski, and Medical Genetics
- Subjects
medicine.medical_specialty ,Pathology ,viruses ,mucosal neoplasia ,Case Report ,Case Reports ,Germline ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,White sponge nevus ,Keratin ,Female patient ,Medicine ,Basal cell ,Sex organ ,chemistry.chemical_classification ,Medicine(all) ,business.industry ,030206 dentistry ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Dermatology ,3. Good health ,Young age ,stomatognathic diseases ,chemistry ,KRT13 ,Dysplasia ,De novo variant ,business - Abstract
Key Clinical Message We report a novel KRT13 germ line variant that causes white sponge nevus (WSN) with mucosal dysplasia. Genital, vaginal, and cervical WSN were observed in four female patients, of whom two had premalignant cervical lesions at young age. Two of the 12 patients with oral WSN developed oral squamous cell carcinoma.
- Published
- 2017
28. Loss of maternal chromosome 11 is a signature event in SDHAF2, SDHD, and VHL-related paragangliomas, but less significant in SDHB-related paragangliomas
- Author
-
Esther Korpershoek, Eleonora P M Corssmit, Cees J. Cornelisse, Henri J L M Timmers, Jeroen C. Jansen, Adrian Bateman, Erik F. Hensen, Ekaterina S. Jordanova, Jean-Pierre Bayley, Peter Devilee, Diana Eccles, Henricus P. M. Kunst, Attje S. Hoekstra, Frederik J. Hes, Anouk N A van der Horst-Schrivers, Judith V.M.G. Bovée, Otolaryngology / Head & Neck Surgery, APH - Quality of Care, Obstetrics and gynaecology, CCA - Cancer biology and immunology, Amsterdam Reproduction & Development (AR&D), Pathology, Guided Treatment in Optimal Selected Cancer Patients (GUTS), and Medical Genetics
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cellular pathology ,Pathology ,SDHB ,Copy number analysis ,SDHA ,Chromosomes, Human, Pair 11/genetics ,Germ-Line Mutation/genetics ,Mitochondrial Proteins ,Von Hippel-Lindau ,paraganglioma ,03 medical and health sciences ,0302 clinical medicine ,Succinate Dehydrogenase/genetics ,Paraganglioma ,medicine ,Humans ,Alleles ,Germ-Line Mutation ,Medicine(all) ,Gynecology ,Paraganglioma/genetics ,business.industry ,Chromosomes, Human, Pair 11 ,Vascular damage Radboud Institute for Molecular Life Sciences [Radboudumc 16] ,Autosomal dominant trait ,succinate dehydrogenase ,medicine.disease ,Von Hippel-Lindau Tumor Suppressor Protein/genetics ,pheochromocytoma ,030104 developmental biology ,Oncology ,Von Hippel-Lindau Tumor Suppressor Protein ,Mitochondrial Proteins/genetics ,030220 oncology & carcinogenesis ,Medical genetics ,Female ,loss of heterozygosity ,SDHD ,business ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Research Paper - Abstract
// Attje S. Hoekstra 1 , Erik F. Hensen 2 , Ekaterina S. Jordanova 3 , Esther Korpershoek 4 , Anouk N.A. van der Horst-Schrivers 5 , Cees Cornelisse 3 , Eleonora P.M. Corssmit 6 , Frederik J. Hes 7 , Jeroen C. Jansen 8 , Henricus P.M. Kunst 9 , Henri J.L.M. Timmers 10 , Adrian Bateman 11 , Diana Eccles 12 , Judith V.M.G. Bovee 3 , Peter Devilee 1, 3 , Jean-Pierre Bayley 1 1 Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands 2 Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center, Amsterdam, The Netherlands 3 Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands 4 Department of Pathology, Josephine Nefkens Institute, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands 5 Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands 6 Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands 7 Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands 8 Department of Otorhinolaryngology, Leiden University Medical Center, Leiden, The Netherlands 9 Department of Otorhinolaryngology, Head and Neck Surgery, Radboud University Medical Centre, Nijmegen, The Netherlands 10 Department of Medicine, Division of Endocrinology, Radboud University Medical Centre, Nijmegen, The Netherlands 11 Department of Cellular Pathology, University Hospital Southampton, Southampton, UK 12 University of Southampton School of Medicine, Cancer Sciences Division, Somers Cancer Research Building, Southampton, UK Correspondence to: Jean-Pierre Bayley, email: J.P.L.Bayley@lumc.nl Peter Devilee, email: P.Devilee@lumc.nl Keywords: paraganglioma, pheochromocytoma, succinate dehydrogenase, Von Hippel-Lindau, loss of heterozygosity Received: September 06, 2016 Accepted: January 04, 2017 Published: January 14, 2017 ABSTRACT Germline mutations in the succinate dehydrogenase (SDHA, SDHB, SDHC, SDHD, SDHAF2) or Von Hippel-Lindau (VHL) genes cause hereditary paraganglioma/pheochromocytoma. While SDHB (1p36) and VHL (3p25) are associated with autosomal dominant disease, SDHD (11q23) and SDHAF2 (11q13) show a remarkable parent-of-origin effect whereby tumor formation is almost completely dependent on paternal transmission of the mutant allele. Loss of the entire maternal copy of chromosome 11 occurs frequently in SDHD -linked tumors, and has been suggested to be the basis for this typical inheritance pattern. Using fluorescent in situ hybridization, microsatellite marker and SNP array analysis, we demonstrate that loss of the entire copy of chromosome 11 is also frequent in SDHAF2 -related PGLs, occurring in 89% of tumors. Analysis of two imprinted differentially methylated regions (DMR) in 11p15, H19-DMR and KvDMR, showed that this loss always affected the maternal copy of chromosome 11. Likewise, loss of maternal chromosome 11p15 was demonstrated in 85% of SDHD and 75% of VHL -related PGLs/PCCs. By contrast, both copies of chromosome 11 were found to be retained in 62% of SDHB -mutated PGLs/PCCs, while only 31% showed loss of maternal chromosome 11p15. Genome-wide copy number analysis revealed frequent loss of 1p in SDHB mutant tumors and show greater genomic instability compared to SDHD and SDHAF2 . These results show that loss of the entire copy of maternal chromosome 11 is a highly specific and statistically significant event in SDHAF2 , SDHD and VHL -related PGLs/PCCs, but is less significant in SDHB -mutated tumors, suggesting that these tumors have a distinct genetic etiology.
- Published
- 2017
29. CDC73-Related Disorders: Clinical Manifestations and Case Detection in Primary Hyperparathyroidism
- Author
-
Frederik J. Hes, Jan Maarten Cobben, Hans Morreau, Romana T. Netea-Maier, Bruce H. R. Wolffenbuttel, Gerlof D. Valk, Karin van der Tuin, Fred H. Menko, Muriel A. Adank, Bernadette P M van Nesselrooij, Neveen A. T. Hamdy, Carli M. J. Tops, Jan C. Oosterwijk, Marjolijn C.J. Jongmans, Medical Genetics, ANS - Cellular & Molecular Mechanisms, Paediatric Genetics, Human genetics, ACS - Atherosclerosis & ischemic syndromes, Damage and Repair in Cancer Development and Cancer Treatment (DARE), Targeted Gynaecologic Oncology (TARGON), Lifestyle Medicine (LM), and Center for Liver, Digestive and Metabolic Diseases (CLDM)
- Subjects
Male ,Pediatrics ,Hyperparathyroidism, Primary/genetics ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Penetrance ,Jaw Neoplasms/genetics ,Biochemistry ,0302 clinical medicine ,Endocrinology ,Child ,Parathyroid adenoma ,Netherlands ,Medicine(all) ,FAMILIAL ISOLATED HYPERPARATHYROIDISM ,JAW TUMOR SYNDROME ,Parathyroid Neoplasms/genetics ,Middle Aged ,Hyperparathyroidism, Primary ,CANCER ,Jaw Neoplasms ,Parathyroid Neoplasms ,Parathyroid carcinoma ,HRPT2 GENE ,030220 oncology & carcinogenesis ,Cohort studies ,young adult ,Female ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,GENETIC ANALYSES ,Adult ,medicine.medical_specialty ,Heterozygote ,Adolescent ,Mutation/genetics ,Genetic counseling ,030209 endocrinology & metabolism ,Context (language use) ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Tumor Suppressor Proteins/genetics ,03 medical and health sciences ,Germline mutation ,All institutes and research themes of the Radboud University Medical Center ,Internal medicine ,GERMLINE ,medicine ,PARATHYROID CARCINOMA ,MANAGEMENT ,Humans ,Germ-Line Mutation ,Aged ,Retrospective Studies ,Hyperparathyroidism ,business.industry ,MUTATIONS ,Tumor Suppressor Proteins ,Biochemistry (medical) ,medicine.disease ,PARAFIBROMIN ,Mutation ,business ,aged, 80 and over ,Primary hyperparathyroidism - Abstract
Context: Heterozygous pathogenic germline variants in CDC73 predispose to the development of primary hyperparathyroidism (pHPT) and, less frequently, ossifying fibroma of the jaw and renal and uterine tumors. Clinical information on CDC73-related disorders has so far been limited to small case series.Objective: To assess the clinical manifestations and penetrance in CDC73-related disorders and to improve case detection in pHPT.Design: Nationwide retrospective Dutch cohort study.Setting: Tertiary referral center.Patients: We studied 89 patients with pHPT referred for germline CDC73 analysis and 43 subsequently tested relatives who proved to be mutation carriers.Investigation: Germline CDC73 mutation analysis.Mean Outcome: CDC73 mutation detection yield, referral rate, and CDC73-related disease penetrance.Results: Pathogenic germline CDC73 variants were identified in 11 of the 89 referred pHPT patients (12.4%), with (suspected) hyperparathyroidism-jaw tumor (HPT-JT) syndrome (n = 3), familial isolated pHPT (n = 5), apparently sporadic parathyroid carcinoma (n = 2), and apparently sporadic parathyroid adenoma (n = 1). The estimated penetrance of CDC73-related disorders was 65% at age 50 years (95% confidence interval, 48% to 82%) in 43 nonindex mutation carriers.Conclusions: Germline CDC73 analysis is recommended in individuals with (suspected) HPT-JT syndrome, familial isolated pHPT, atypical or malignant parathyroid histology, and young individuals with pHPT. These criteria would increase germline CDC73 mutation detection, enabling optimal clinical management of pHPT as well as genetic counseling and surveillance for family members at risk for developing CDC73-related disorders.
- Published
- 2017
30. Unraveling genetic predisposition to familial or early onset gastric cancer using germline whole-exome sequencing
- Author
-
Carla Oliveira, Roland P. Kuiper, Hildegunn Høberg-Vetti, James R. Lupski, Encarna B. Gomez Garcia, Margreet G. E. M. Ausems, Christian Gilissen, Liesbeth Spruijt, Elke Holinski-Feder, C. Marleen Kets, Urszula Teodorczyk, Jelle J. Goeman, Rachel S. van der Post, Ad Geurts van Kessel, Anja Wagner, Alexander Hoischen, Anna Jakubowska, Wendy A. G. van Zelst-Stams, Rolf H. Sijmons, Cora M. Aalfs, Nicoline Hoogerbrugge, Maartje van de Vorst, Marjolijn J. L. Ligtenberg, Maurizio Genuardi, Shalini N. Jhangiani, Jan Lubinski, Lisenka E.L.M. Vissers, Frederik J. Hes, Inga Bjørnevoll, J. Han van Krieken, Hugo Pinheiro, Hans K. Schackert, Joep de Ligt, Ingrid P. Vogelaar, Guglielmina Nadia Ranzani, Donna M. Muzny, Liselotte P. van Hest, Richard A. Gibbs, Lizet E. van der Kolk, Valeria Molinaro, CCA - Cancer biology and immunology, Human genetics, Medical Genetics, Human Genetics, RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Polikliniek (9), Klinische Genetica, Clinical Genetics, and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
0301 basic medicine ,Male ,Candidate gene ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,VARIANTS ,Gene mutation ,Bioinformatics ,Germline ,COLORECTAL-CANCER ,0302 clinical medicine ,CDH1 MUTATIONS ,Stomach Neoplasms/diagnosis ,HISTORY ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Genetics(clinical) ,Exome ,Early Detection of Cancer ,DIFFUSE ,Genetics (clinical) ,Exome sequencing ,RISK ,Genetics ,Medicine(all) ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Middle Aged ,Cadherins ,3. Good health ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,Sequence Analysis, DNA/methods ,030220 oncology & carcinogenesis ,Female ,Genetic Testing/methods ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adult ,medicine.medical_specialty ,MUTATION CARRIERS ,Biology ,Article ,CLASSIFICATION ,03 medical and health sciences ,Germline mutation ,SDG 3 - Good Health and Well-being ,Stomach Neoplasms ,HELICOBACTER-PYLORI ,Antigens, CD ,RESOURCE ,Molecular genetics ,Genetic predisposition ,medicine ,Journal Article ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Germ-Line Mutation ,Aged ,Settore MED/06 - ONCOLOGIA MEDICA ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,gastric cancer ,Cadherins/genetics ,Sequence Analysis, DNA ,030104 developmental biology ,Early Detection of Cancer/methods ,exome - Abstract
Contains fulltext : 182216.pdf (Publisher’s version ) (Open Access) Recognition of individuals with a genetic predisposition to gastric cancer (GC) enables preventive measures. However, the underlying cause of genetic susceptibility to gastric cancer remains largely unexplained. We performed germline whole-exome sequencing on leukocyte DNA of 54 patients from 53 families with genetically unexplained diffuse-type and intestinal-type GC to identify novel GC-predisposing candidate genes. As young age at diagnosis and familial clustering are hallmarks of genetic tumor susceptibility, we selected patients that were diagnosed below the age of 35, patients from families with two cases of GC at or below age 60 and patients from families with three GC cases at or below age 70. All included individuals were tested negative for germline CDH1 mutations before or during the study. Variants that were possibly deleterious according to in silico predictions were filtered using several independent approaches that were based on gene function and gene mutation burden in controls. Despite a rigorous search, no obvious candidate GC predisposition genes were identified. This negative result stresses the importance of future research studies in large, homogeneous cohorts.
- Published
- 2017
31. Estimating the penetrance of pathogenic gene variants in families with missing pedigree information
- Author
-
Erik F. Hensen, Marianne A. Jonker, Frederik J. Hes, Johannes A. Rijken, Hein Putter, Epidemiology and Data Science, Otolaryngology / Head & Neck Surgery, CCA - Cancer biology and immunology, CCA - Cancer Treatment and quality of life, Medical Genetics, and Ear, Nose and Throat
- Subjects
Male ,Statistics and Probability ,Epidemiology ,Pedigree information ,Penetrance ,Disease ,Biology ,01 natural sciences ,Germline ,010104 statistics & probability ,03 medical and health sciences ,missing data ,0302 clinical medicine ,Health Information Management ,Humans ,Genetic Predisposition to Disease ,030212 general & internal medicine ,0101 mathematics ,Gene ,conditional maximum likelihood method ,Germ-Line Mutation ,Aged ,Medicine(all) ,Genetics ,Models, Genetic ,Genetic Variation ,Estimator ,Articles ,Middle Aged ,Missing data ,SDHB ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,Pedigree ,Age-at-onset ,Cohort ,Female - Abstract
Contains fulltext : 208801.pdf (Publisher’s version ) (Open Access) Accurate assessment of the age-dependent disease risk conferred by germline variants in disease susceptibility genes is often hampered by the way the data are collected. Cohort-based data sets frequently contain an overrepresentation of patients (i.e. carriers of the gene variant of interest affected with the associated disease), and an underrepresentation of disease-free carriers. In order to overcome this problem, penetrance estimates can be based on family-based study designs, through the evaluation of index patients and their family members. This approach facilitates the identification of asymptomatic germline variant carriers. By adjusting for the way these family data are ascertained, an estimate for the penetrance of the pathogenic gene variant can be obtained. However, the family structure is often incomplete or missing. This complicates the estimation of the penetrance, because full adjustment of the likelihood is not possible. We present a conditional likelihood for the estimation of the penetrance of pathogenic gene variants, based on a cohort of multiple families comprising index patients, disease-free and affected non-index carriers, but with missing information on pedigree structure. The proposed estimator corrects for the ascertainment in a robust way and is shown to be more accurate than the frequently used Kaplan-Meier estimator of the penetrance function.
- Published
- 2019
32. Surveillance for familial melanoma: recommendations from a national centre of expertise
- Author
-
Thomas P. Potjer, Frederik J. Hes, Nicole A Kukutsch, Hans F. A. Vasen, A B Halk, R. van Doorn, Clinical sciences, and Medical Genetics
- Subjects
Adult ,Heterozygote ,medicine.medical_specialty ,Skin Neoplasms ,Adolescent ,Biopsy ,DNA Mutational Analysis ,Dermatology ,Young Adult ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Child ,Medical History Taking ,Melanoma ,Referral and Consultation ,familial melanoma ,Netherlands ,Skin ,Medicine(all) ,Academic Medical Centers ,business.industry ,Age Factors ,Middle Aged ,Familial Melanoma ,national centre of expertise ,Family medicine ,Mutation ,Practice Guidelines as Topic ,business - Published
- 2019
33. Genotype-phenotype correlations for pancreatic cancer risk in Dutch melanoma families with pathogenic
- Author
-
Kasper A, Overbeek, Mar Dm, Rodríguez-Girondo, Anja, Wagner, Nienke, van der Stoep, Peter C, van den Akker, Jan C, Oosterwijk, Theo A, van Os, Lizet E, van der Kolk, Hans F A, Vasen, Frederik J, Hes, Djuna L, Cahen, Marco J, Bruno, and Thomas P, Potjer
- Subjects
Adult ,Male ,endocrine system diseases ,Genotype-Phenotype Correlations ,gastroenterology ,genetic screening/counselling ,Middle Aged ,pancreas and biliary tract ,digestive system diseases ,Pancreatic Neoplasms ,Biliary Tract Neoplasms ,Risk Factors ,oncology ,Humans ,Female ,Genetic Predisposition to Disease ,Melanoma ,Pancreas ,Cyclin-Dependent Kinase Inhibitor p16 ,Genetic Association Studies ,clinical genetics ,Aged ,Netherlands - Abstract
Background Pathogenic variants in the CDKN2A gene are generally associated with the development of melanoma and pancreatic ductal adenocarcinoma (PDAC), but specific genotype-phenotype correlations might exist and the extent of PDAC risk is not well established for many variants. Methods Using the Dutch national familial melanoma database, we identified all families with a pathogenic CDKN2A variant and investigated the occurrence of PDAC within these families. We also estimated the standardised incidence ratio and lifetime PDAC risk for carriers of a highly prevalent variant in these families. Results We identified 172 families in which 649 individuals carried 15 different pathogenic variants. The most prevalent variant was the founder mutation c.225_243del (p16-Leiden, 484 proven carriers). Second most prevalent was c.67G>C (55 proven carriers). PDAC developed in 95 of 163 families (58%, including 373 of 629 proven carriers) harbouring a variant with an effect on the p16INK4a protein, whereas PDAC did not occur in the 9 families (20 proven carriers) with a variant affecting only p14ARF. In the c.67G>C families, PDAC occurred in 12 of the 251 (5%) persons at risk. The standardised incidence ratio was 19.1 (95% CI 8.3 to 33.6) and the cumulative PDAC incidence at age 75 years (lifetime risk) was 19% (95% CI 7.5% to 30.1%). Conclusions Our results support the notion that pathogenic CDKN2A variants affecting the p16INK4a protein, including c.67G>C, are associated with increased PDAC risk and carriers of such variants should be offered pancreatic cancer surveillance. There is no clinical evidence that impairment of only the p14ARF protein leads to an increased PDAC risk.
- Published
- 2019
34. Targetable gene fusions identified in radioactive iodine refractory advanced thyroid carcinoma
- Author
-
Frederik J. Hes, Theo S. Plantinga, D Ruano Neto, M N Khalifa, T. van Wezel, Thera P. Links, K van der Tuin, Hans Morreau, Johannes W. A. Smit, Wim E. Corver, M Ventayol Garcia, E. P. M. Corssmit, Ellen Kapiteijn, Medical Genetics, Damage and Repair in Cancer Development and Cancer Treatment (DARE), and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Sorafenib ,Male ,medicine.medical_specialty ,tumor ,Adolescent ,Endocrinology, Diabetes and Metabolism ,advanced radioactive iodine-refractory (RAI-R) thyroid carcinoma ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,ONCOGENES ,medicine.disease_cause ,Healthcare improvement science Radboud Institute for Health Sciences [Radboudumc 18] ,Fusion gene ,Thyroid carcinoma ,Endocrinology ,Internal medicine ,DRIVERS ,REVEALS ,medicine ,Humans ,HRAS ,Thyroid Neoplasms ,Thyroid cancer ,Advanced Thyroid Carcinoma ,Aged ,Retrospective Studies ,Medicine(all) ,Aged, 80 and over ,THERAPEUTIC TARGET ,business.industry ,DNA ,General Medicine ,Middle Aged ,medicine.disease ,targeted therapy ,CANCER ,REDIFFERENTIATION ,SORAFENIB ,ETV6 ,Gene Targeting ,Cancer research ,Female ,KRAS ,Gene Fusion ,business ,medicine.drug ,Iodine - Abstract
ObjectiveGene alterations leading to activation of the MAPK pathway are of interest for targeted therapy in patients with advanced radioactive iodine refractory (RAI-R) thyroid carcinoma. Due to technical reasons gene fusion analysis in RNA isolated from formalin-fixed tumor tissues has till now been limited. The objective of the present study was to identify targetable gene rearrangements in RNA isolated from formalin-fixed RAI-R thyroid carcinomas.DesignRetrospective study in 132 patients with RAI-R thyroid carcinoma (59 papillary-, 24 follicular-, 35 Hürthle cell- and 14 anaplastic thyroid carcinoma).MethodsTotal nucleic acid (undivided DNA and RNA) was isolated from formalin-fixed tissue. Extensive gene fusion analysis was performed in all samples that tested negative for pathogenicBRAF,NRAS,HRASandKRASvariants.ResultsSeven targetable gene fusions were identified in the remaining 60 samples without known DNA variants. This includes frequently reported gene fusions such asCCDC6/RET(PTC1),PRKAR1A/RET(PTC2) andETV6/NTRK3, and gene fusions that are less common in thyroid cancer (TPM3/NTRK1,EML4/ALKandEML4/NTRK3). Of note, most gene fusions were detected in papillary thyroid carcinoma and MAPK-associated alterations in Hürthle cell carcinomas are rare (2/35).ConclusionTargetable gene fusions were found in 12% of RAI-R thyroid carcinoma without DNA variants and can be effectively identified in formalin-fixed tissue. These gene fusions might provide a preclinical rationale to include specific kinase inhibitors in the treatment regimen for these patients. The latter intends to restore iodine transport and/or take advantage of the direct effect on tumor cell vitality once progressive disease is seen.
- Published
- 2019
35. Low frequency of POLD1 and POLE exonuclease domain variants in patients with multiple colorectal polyps
- Author
-
Tom van Wezel, Frederik J. Hes, Fadwa A. Elsayed, Maartje Nielsen, Carli M. J. Tops, Hans Morreau, Dina Ruano, and Hans F. A. Vasen
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Adolescent ,Colorectal cancer ,Mutation, Missense ,Colonic Polyps ,colorectal cancer ,Colorectal polyposis ,030105 genetics & heredity ,DNA polymerase ,POLD1 ,Germline ,03 medical and health sciences ,Germline mutation ,Mutation Rate ,Catalytic Domain ,Internal medicine ,Genetics ,Carcinoma ,medicine ,Humans ,Family history ,Poly-ADP-Ribose Binding Proteins ,Molecular Biology ,Germ-Line Mutation ,Genetics (clinical) ,Aged ,DNA Polymerase III ,Aged, 80 and over ,business.industry ,Endometrial cancer ,Original Articles ,DNA Polymerase II ,Middle Aged ,medicine.disease ,colorectal polyposis ,digestive system diseases ,030104 developmental biology ,germline mutations ,Original Article ,Female ,exonuclease domain ,Colorectal Neoplasms ,business - Abstract
Background Germline mutations affecting the exonuclease domains of POLE and POLD1 predispose to colorectal adenomas and carcinoma. Here, we aimed to screen the exonuclease domains to find the genetic causes of multiple colorectal polyps in unexplained cases. Methods Using a custom next‐generation sequencing panel, we sequenced the exonuclease domains of POLE and POLD1 in 332 index patients diagnosed with multiple colorectal polyps without germline alteration in colorectal polyposis predisposing genes. Results We identified two variants of unknown significance. One germline POLD1 c.961G>A, p.(Gly321Ser) variant was found in two cases. The first patient was diagnosed with multiple polyps at age 35 and colorectal cancer (CRC) at age 37, with no known family history of CRC. The second patient was diagnosed with CRC at age 44 and cumulatively developed multiple polyps; this patient had two sisters with endometrial cancer who did not carry the variant. Furthermore, we identified a novel POLD1 c.955 T>G, p.(Cys319Gly) variant in a patient diagnosed with multiple colorectal adenomas at age 40. Co‐segregation analysis showed that one sister who cumulatively developed multiple adenomas from age 34, and another sister who developed CRC at age 38 did not carry the variant. We did not identify pathogenic variants in POLE and POLD1. Conclusion This study confirms the low frequency of causal variants in these genes in the predisposition for multiple colorectal polyps, and also establishes that these genes are a rare cause of the disease.
- Published
- 2019
36. Mathematical Models for Tumor Growth and the Reduction of Overtreatment
- Author
-
Berit M. Verbist, Jeroen C. Jansen, Jean-Pierre Bayley, Peter Paul G. van Benthem, John-Melle Bokhorst, Lisa M. H. de Pont, Eleonora P M Corssmit, Andel G. L. van der Mey, Berdine L. Heesterman, Frederik J. Hes, and Medical Genetics
- Subjects
Coefficient of determination ,Mathematical model ,business.industry ,mathematics ,growth ,Gompertz function ,Clinical Neurology ,carotid body tumors ,Regression ,paragangliomas ,Exponential function ,03 medical and health sciences ,vagal body ,models ,0302 clinical medicine ,Goodness of fit ,Non-linear least squares ,Statistics ,Medicine ,Neurology (clinical) ,Analysis of variance ,business ,030217 neurology & neurosurgery - Abstract
Background To improve our understanding of the natural course of head and neck paragangliomas (HNPGL) and ultimately differentiate between cases that benefit from early treatment and those that are best left untreated, we studied the growth dynamics of 77 HNPGL managed with primary observation. Methods Using digitally available magnetic resonance images, tumor volume was estimated at three time points. Subsequently, nonlinear least squares regression was used to fit seven mathematical models to the observed growth data. Goodness of fit was assessed with the coefficient of determination (R 2) and root-mean-squared error. The models were compared with Kruskal–Wallis one-way analysis of variance and subsequent post-hoc tests. In addition, the credibility of predictions (age at onset of neoplastic growth and estimated volume at age 90) was evaluated. Results Equations generating sigmoidal-shaped growth curves (Gompertz, logistic, Spratt and Bertalanffy) provided a good fit (median R 2: 0.996–1.00) and better described the observed data compared with the linear, exponential, and Mendelsohn equations (p Conclusions Growth of HNPGL is best described by decelerating tumor growth laws, with a preference for the Bertalanffy model. To the best of our knowledge, this is the first time that this often-neglected model has been successfully fitted to clinically obtained growth data.
- Published
- 2019
37. Declining Detection Rates for APC and Biallelic MUTYH Pathogenic Variants in Polyposis Patients, Implications for DNA Testing Policy
- Author
-
C. Tops, Sunny S Singh, Frederik J. Hes, Manon Suerink, Johan J.P. Gille, Alexandra M. J. Langers, Diantha Terlouw, J. T. van Wezel, Hans F. A. Vasen, S. W. ten Broeke, Maartje Nielsen, Hans Morreau, and Yvonne J. Vos
- Subjects
Oncology ,medicine.medical_specialty ,medicine.diagnostic_test ,Adenoma ,Colorectal cancer ,business.industry ,MUTYH-Associated Polyposis ,Odds ratio ,medicine.disease ,medicine.disease_cause ,digestive system diseases ,Familial adenomatous polyposis ,MUTYH ,Internal medicine ,medicine ,KRAS ,business ,Genetic testing - Abstract
Background: This study aimed to determine the prevalence of APC-associated familial adenomatous polyposis (FAP) and MUTYH-associated polyposis (MAP) in a large cohort, taking into account several factors including adenoma count and year of diagnosis. Methods: All application forms used to send patients in for APC and MUTYH mutation analysis between 1992 and 2017 were collected (n=2082). Using this data, possible predictive factors were examined using multivariate multinomial logistic regression analysis in SPSS. Findings: The prevalence of APC mutations significantly increases with adenoma count while MAP shows a peak prevalence in individuals with 50-99 adenomas. Logistic regression analysis shows significant odds ratios for adenoma count, age at diagnosis and having a first degree family member with more than 10 polyps. Moreover, the chance of finding an APC or MUTYH mutation steadily declined over the past 23 years. Mutations in APC and MUTYH were found in 22% (43/200) of patients with FAP-related extracolonic manifestations, such as duodenal polyps and desmoid tumors. Mutation detection rate were 40% and 10% for respectively patients with >10 adenomas under age 60 and >20 adenomas under age 70. Mutation carriers not falling into these criteria, had FAP-related extracolonic manifestations, colorectal cancer (CRC) below the age of 40, somatic KRAS c.34G>T variant in the tumor or a first-degree family member with >10 adenomas. Interpretation: Adenoma count, age at diagnosis of the adenomas and year of analysis are important predictive factors for APC and MUTYH mutations. Based on our results, APC and MUTYH testing is advised in patients >10 adenomas under age 60 and >20 adenomas under age 70. Almost all FAP and MAP patients who did not meet these criteria showed other characteristics that can be used as an indication to prompt genetic testing. Funding Statement: The authors declare: "N/A." Declaration of Interests: The authors state: "No competing interest to declare." Ethics Approval Statement: The study was approved by local ethics review boards (P01.019).
- Published
- 2019
38. Clinical and Molecular Characteristics May Alter Treatment Strategies of Thyroid Malignancies in DICER1 Syndrome
- Author
-
Hans Morreau, Marie Jose M. Pouwels, Marjolijn C.J. Jongmans, Frederik J. Hes, William D. Foulkes, Tom van Wezel, Marek Niedziela, Eveline J. Kamping, Karin van der Tuin, Sabine E. Hannema, Leanne de Kock, Amsterdam Gastroenterology Endocrinology Metabolism, Pediatric surgery, and Medical Genetics
- Subjects
0301 basic medicine ,Male ,Ribonuclease III ,Pathology ,DICER1 Syndrome ,Somatic cell ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,DNA Mutational Analysis ,Disease ,tumours ,Biochemistry ,Germline ,DEAD-box RNA Helicases ,0302 clinical medicine ,Endocrinology ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Thyroid Nodule ,Child ,Medicine(all) ,Thyroid ,Hyperplasia ,Prognosis ,medicine.anatomical_structure ,autosomal-dominantly inherited disorder ,030220 oncology & carcinogenesis ,Monoclonal ,Female ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adult ,medicine.medical_specialty ,Adolescent ,Thyroid carcinoma ,Thyroid Malignancies ,03 medical and health sciences ,Young Adult ,Neoplastic Syndromes, Hereditary ,Internal medicine ,medicine ,Humans ,Thyroid Neoplasms ,differentiated thyroid carcinoma ,Retrospective Studies ,business.industry ,Biochemistry (medical) ,medicine.disease ,030104 developmental biology ,Mutation ,business ,Follow-Up Studies - Abstract
Context DICER1 syndrome is a rare autosomal-dominantly inherited disorder that predisposes to a variety of cancerous and noncancerous tumors of mostly pediatric and adolescent onset, including differentiated thyroid carcinoma (DTC). DTC has been hypothesized to arise secondarily to the increased prevalence of thyroid hyperplastic nodules in syndromic patients. Objective To determine somatic alterations in DICER1-associated DTC and to study patient outcomes. Design Retrospective series. Setting Tertiary referral centers. Patients Ten patients with germline pathogenic DICER1 variants and early-onset DTC. Methods Somatic DICER1 mutation analysis, extensive somatic DNA variant and gene fusion analyses were performed on all tumors. Results Median age at DTC diagnosis was 13.5 years and there was no recurrent or metastatic disease (median follow-up, 8 years). All thyroid specimens showed diffuse nodular hyperplasia with at least one focus suspicious of DTC but without infiltrative growth, extrathyroidal extension, vascular invasion, or lymph node metastasis. Most of the individual nodules (benign and malignant) sampled from the 10 tumors harbored distinct DICER1 RNase IIIb hotspot mutations, indicating a polyclonal composition of each tumor. Furthermore, nine of 10 DICER1-related DTCs lacked well-known oncogenic driver DNA variants and gene rearrangements. Conclusion On the basis of our clinical, histological, and molecular data, we consider that most DICER1-related DTCs form a low-risk subgroup. These tumors may arise within one of multiple benign monoclonal nodules; thus, hemi-thyroidectomy or, more likely, total thyroidectomy may often be required. However, radioiodine treatment may be unnecessary given the patients’ ages and the tumors’ low propensity for metastases.
- Published
- 2019
39. Increased Mortality in
- Author
-
Johannes A, Rijken, Leonie T, van Hulsteijn, Olaf M, Dekkers, Nicolasine D, Niemeijer, C René, Leemans, Karin, Eijkelenkamp, Anouk N A, van der Horst-Schrivers, Michiel N, Kerstens, Anouk, van Berkel, Henri J L M, Timmers, Henricus P M, Kunst, Peter H L T, Bisschop, Koen M A, Dreijerink, Marieke F, van Dooren, Frederik J, Hes, Jeroen C, Jansen, Eleonora P M, Corssmit, and Erik F, Hensen
- Subjects
paraganglioma ,SDHB ,mortality ,pheochromocytoma ,Article ,SDHD - Abstract
Germline mutations in succinate dehydrogenase subunit B and D (SDHB and SDHD) are predisposed to hereditary paraganglioma (PGL) and pheochromocytoma (PHEO). The phenotype of pathogenic variants varies according to the causative gene. In this retrospective study, we estimate the mortality of a nationwide cohort of SDHB variant carriers and that of a large cohort of SDHD variant carriers and compare it to the mortality of a matched cohort of the general Dutch population. A total of 192 SDHB variant carriers and 232 SDHD variant carriers were included in this study. The Standard Mortality Ratio (SMR) for SDHB mutation carriers was 1.89, increasing to 2.88 in carriers affected by PGL. For SDHD variant carriers the SMR was 0.93 and 1.06 in affected carriers. Compared to the general population, mortality seems to be increased in SDHB variant carriers, especially in those affected by PGL. In SDHD variant carriers, the mortality is comparable to that of the general Dutch population, even if they are affected by PGL. This insight emphasizes the significance of DNA-testing in all PGL and PHEO patients, since different clinical risks may warrant gene-specific management strategies.
- Published
- 2018
40. Comprehensive Mutation Analysis of PMS2 in a Large Cohort of Probands Suspected of Lynch Syndrome or Constitutional Mismatch Repair Deficiency Syndrome
- Author
-
Maartje Nielsen, Marjolijn J. L. Ligtenberg, Yvonne Tiersma, Juul T. Wijnen, Maran J. W. Olderode-Berends, Encarna Gomez Garcia, B. Redeker, José B. M. Zonneveld, Sanne W. ten Broeke, Frederik J. Hes, Carli M. J. Tops, Peter Devilee, Theo A. M. van Os, Christi J. van Asperen, Hans J. J. P. Gille, Niels de Wind, Heleen M. van der Klift, Arjen R. Mensenkamp, Tom G.W. Letteboer, Yvonne J. Vos, Elsa C. Bik, Mark Drost, S. Verhoef, Liselotte P. van Hest, Anja Wagner, Human Genetics, Human genetics, CCA - Cancer biology, RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Polikliniek (9), Klinische Genetica, Medical Genetics, and Clinical Genetics
- Subjects
0301 basic medicine ,DNA Mutational Analysis ,pseudogenes ,COLORECTAL-CANCER ,Cohort Studies ,0302 clinical medicine ,Mutation Carrier ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,PMS2 ,Missense mutation ,Genetics (clinical) ,Mismatch Repair Endonuclease PMS2 ,Netherlands ,Medicine(all) ,Genetics ,Brain Neoplasms ,MLH1 ,Neoplastic Syndromes, Hereditary/genetics ,Lynch syndrome ,CMMRD ,missense variants ,immunohistochemistry ,mismatch repair ,030220 oncology & carcinogenesis ,Mutation (genetic algorithm) ,DNA mismatch repair ,Microsatellite Instability ,Colorectal Neoplasms ,EUROPEAN CONSORTIUM CARE ,PSEUDOGENE INTERFERENCE ,congenital, hereditary, and neonatal diseases and abnormalities ,DNA Mutational Analysis/methods ,Biology ,03 medical and health sciences ,Germline mutation ,Neoplastic Syndromes, Hereditary ,Colorectal Neoplasms, Hereditary Nonpolyposis/genetics ,SYNDROME FAMILIES ,CFR PARTICIPANTS ,medicine ,Journal Article ,Humans ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Brain Neoplasms/genetics ,Microsatellite instability ,Genetic Variation ,Mismatch Repair Endonuclease PMS2/genetics ,medicine.disease ,Colorectal Neoplasms/genetics ,Colorectal Neoplasms, Hereditary Nonpolyposis ,GENE ,digestive system diseases ,030104 developmental biology ,PROMOTER HYPERMETHYLATION ,3' DELETIONS ,Cancer research ,NONPOLYPOSIS COLON-CANCER - Abstract
Monoallelic PMS2 germline mutations cause 5-15% of Lynch syndrome, a midlife cancer predisposition, whereas biallelic PMS2 mutations cause approximately 60% of constitutional MMR deficiency (CMMRD), a rare childhood cancer syndrome. Recently improved DNA and RNA-based strategies are applied to overcome problematic PMS2 mutation analysis due to the presence of pseudogenes and frequent gene conversion events. Here, we determined PMS2 mutation detection yield and mutation spectrum in a nationwide cohort of 396 probands. Furthermore, we studied concordance between tumor IHC/ MSI (immunohistochemistry/ microsatellite-instability) profile and mutation carrier state. Overall, we found 52 different pathogenic PMS2 variants explaining 121 Lynch syndrome and nine CMMRD patients. In vitro MMR assays suggested pathogenicity for three missense variants. Ninety-one PMS2 mutation carriers (70%) showed isolated loss of PMS2 in their tumors, for 31 (24%) no or inconclusive IHC was available, and eight carriers (6%) showed discordant IHC (presence of PMS2 or loss of both MLH1 and PMS2). Ten cases with isolated PMS2 loss (10%; 10/97) harbored MLH1 mutations. We confirmed that recently improved mutation analysis provides a high yield of PMS2 mutations in patients with isolated loss of PMS2 expression. Application of universal tumor pre-screening methods will however miss some PMS2 germline mutation carriers. This article is protected by copyright. All rights reserved.
- Published
- 2016
41. Clinical progression and metachronous paragangliomas in a large cohort of SDHD germline variant carriers
- Author
-
Berit M. Verbist, Lisa M. H. de Pont, Peter Paul G. van Benthem, Eleonora P M Corssmit, Berdine L. Heesterman, Jeroen C. Jansen, Andel G. L. van der Mey, Frederik J. Hes, Jean-Pierre Bayley, and Medical Genetics
- Subjects
Adult ,Male ,0301 basic medicine ,Heterozygote ,Pediatrics ,medicine.medical_specialty ,Logistic regression ,Asymptomatic ,Article ,Germline ,Paraganglioma ,multifocal paragangliomas ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,SDHD germline variant carriers ,Germ-Line Mutation ,Genetics (clinical) ,Aged ,Medicine(all) ,business.industry ,Neoplasms, Second Primary ,Middle Aged ,medicine.disease ,cranial nerve impairment ,Succinate Dehydrogenase ,030104 developmental biology ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Cohort ,Female ,SDHD ,metachronous paragangliomas ,medicine.symptom ,business ,SDHD variant carriers ,Clinical progression - Abstract
Although it is well established that paternally transmitted germline variants in SDHD are associated with multifocal paragangliomas and lifelong follow-up is generally advised, the risk of metachronous lesions is presently unknown. In a large Dutch cohort of SDHD variant carriers, we studied the development of new paragangliomas, and the evolution of symptoms and cranial nerve impairment. Recurrent event analysis and the Kaplan-Meier product limit estimator were used to study the risk of new lesions. The relation between several predictors and development of new symptoms was assessed using logistic regression. Of the 222 SDHD variant carriers included, 65% presented with symptoms and 11% with cranial nerve dysfunction. Over a median period of 8 years, 42% reported new symptoms, and new cranial nerve impairment was observed in 11% of subjects. The estimated fraction of subjects that developed new HNPGL increased to 73% (95% CI: 52-85%) after 22 years of follow-up. Males were more likely to develop new HNPGL compared to females (HR: 1.63, 95% CI: 1.10-2.40), as were subjects that presented with symptoms, compared to subjects that were asymptomatic at baseline (HR: 1.61, 95% CI: 1.01-2.55). In addition, the risk of new lesions decreased with number of HNPGL present at first diagnosis (HR: 0.68 and 95% CI: 0.56-0.82). Carriers of a paternally inherited SDHD variant face a considerable risk for new HNPGL. In addition, nearly 50% of subjects reported new symptoms. However, new cranial nerve deficits were observed in only 11%, which is less than reported in surgical series. These risks should be taken into account when considering treatment strategies and counseling.
- Published
- 2018
42. Excluding Lynch syndrome in a female patient with metachronous DNA mismatch repair deficient colon- and ovarian cancer
- Author
-
Juul T. Wijnen, Winand N.M. Dinjens, Maartje Nielsen, Anne M.L. Jansen, Hans Morreau, Marije Koopmans, Frederik J. Hes, Stijn Crobach, M.J.L. Ligtenberg, Tom van Wezel, Medical Genetics, and Pathology
- Subjects
0301 basic medicine ,Adult ,Pathology ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,Colorectal cancer ,Short Communication ,Germline mosaicism ,Biology ,MLH1 ,DNA Mismatch Repair ,Neoplasms, Multiple Primary ,03 medical and health sciences ,0302 clinical medicine ,All institutes and research themes of the Radboud University Medical Center ,SDG 3 - Good Health and Well-being ,Ovarian cancer ,Ovarian carcinoma ,PMS2 ,medicine ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Genetics ,Humans ,Genetic Predisposition to Disease ,Genetics(clinical) ,Genetics (clinical) ,Medicine(all) ,Ovarian Neoplasms ,Middle Aged ,medicine.disease ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Colorectal Neoplasms, Hereditary Nonpolyposis ,Lynch syndrome ,digestive system diseases ,3. Good health ,Colon cancer ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,DNA mismatch repair ,Female - Abstract
Patients synchronously or metachronously presenting with ovarian and colon cancer can pose diagnostic challenges. A primary colon carcinoma can metastasize to one or both ovaries, two independent primary tumors can arise or an ovarian carcinoma can metastasize to the colon. Clinical and immunohistochemical characterization can aid the diagnosis. Recently, we reported that in difficult cases finding pathogenic APC variants supports a colonic origin. In this case report we describe the clinical history of a female patient suspected for Lynch syndrome. She was diagnosed with a bilateral ovarian cancer at age 44, followed by the detection of a colon carcinoma 12.5 months later. Lesions of both sites showed a DNA mismatch repair deficiency with immunohistochemical loss of MLH1 and PMS2 expression without MLH1 promoter hypermethylation. In absence of germline MMR gene variants identical somatic MLH1 and CTNNB1 gene variants were found, indicating a clonal relation. MMR germline mosaicism was made unlikely by ultra deep sequencing of the MLH1 variant in DNA isolated from normal mucosa, blood, urine and saliva. Although initially being suspect for Lynch syndrome it was eventually concluded that a metachronously diagnosed colon carcinoma that metastasized to both ovaries was most likely.
- Published
- 2018
43. High Growth Rate of Pancreatic Ductal Adenocarcinoma in
- Author
-
Isaura S, Ibrahim, Martin N, Wasser, Yinghui, Wu, Akin, Inderson, Wouter H, de Vos Tot Nederveen Cappel, Hans, Morreau, Frederik J, Hes, Roeland A, Veenendaal, Hein, Putter, Shirin, Feshtali, Anneke M, van Mil, Nelleke A, Gruis, Rob A, Tollenaar, Wilma, Bergman, Bert A, Bonsing, and Hans F A, Vasen
- Subjects
Adult ,Male ,Heterozygote ,Time Factors ,Middle Aged ,Magnetic Resonance Imaging ,Founder Effect ,Cohort Studies ,Pancreatic Neoplasms ,Mutation ,Humans ,Mass Screening ,Female ,Genetic Predisposition to Disease ,Pancreatic Cyst ,Pancreas ,Precancerous Conditions ,Cyclin-Dependent Kinase Inhibitor p16 ,Early Detection of Cancer ,Aged ,Carcinoma, Pancreatic Ductal ,Follow-Up Studies ,Netherlands - Published
- 2018
44. Clinical Aspects of SDHA-Related Pheochromocytoma and Paraganglioma: A Nationwide Study
- Author
-
Henricus P. M. Kunst, Mirjam M. de Jong, Bernadette M P van Nesselrooij, Frederik J. Hes, Henri J L M Timmers, Arjen R. Mensenkamp, Edward M Leter, Hans Morreau, Benno Küsters, Winand N.M. Dinjens, Anouk N A van de Horst-Schrivers, Karin van der Tuin, Jeroen C. Jansen, Eleonora P M Corssmit, Liesbeth Spruijt, Carli M. J. Tops, Rogier A. Oldenburg, Pathology, Clinical Genetics, Medical Genetics, Guided Treatment in Optimal Selected Cancer Patients (GUTS), MUMC+: DA KG Polikliniek (9), and RS: FHML non-thematic output
- Subjects
Male ,Oncology ,Endocrinology, Diabetes and Metabolism ,DNA Mutational Analysis ,Clinical Biochemistry ,Adrenal Gland Neoplasms ,SDHA ,Penetrance ,Paraganglioma/epidemiology ,Biochemistry ,Germline ,0302 clinical medicine ,Endocrinology ,Paraganglioma ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Medicine ,Child ,Netherlands ,Medicine(all) ,Electron Transport Complex II ,GASTROINTESTINAL STROMAL TUMORS ,Vascular damage Radboud Institute for Molecular Life Sciences [Radboudumc 16] ,Middle Aged ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Pheochromocytoma/epidemiology ,030220 oncology & carcinogenesis ,Electron Transport Complex II/genetics ,young adult ,Female ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,Adult ,medicine.medical_specialty ,GENES ,Adolescent ,Genetic counseling ,SOCIETY ,Netherlands/epidemiology ,030209 endocrinology & metabolism ,Context (language use) ,Pheochromocytoma ,Adrenal Gland Neoplasms/epidemiology ,03 medical and health sciences ,Germline mutation ,All institutes and research themes of the Radboud University Medical Center ,Internal medicine ,Humans ,Genetic Predisposition to Disease ,HEAD ,Germ-Line Mutation ,Aged ,Retrospective Studies ,business.industry ,MUTATIONS ,Biochemistry (medical) ,medicine.disease ,NECK PARAGANGLIOMAS ,PRACTICE GUIDELINE ,aged, 80 and over ,business - Abstract
Context Paraganglioma (PGL) has the highest degree of heritability among human neoplasms. Current clinical understanding of germline SDHA mutation carriers is limited. Objective To estimate the contribution of SDHA mutations in PGL and to assess clinical manifestations and age-related penetrance. Design Nationwide retrospective cohort study. Setting Tertiary referral centers in the Netherlands (multicenter). Patients Germline SDHA analysis was performed in 393 patients with genetically unexplained PGL. Subsequently, 30 index SDHA mutation carriers and 56 nonindex carriers were studied. Main Outcome Measures SDHA mutation detection yield, clinical manifestations, and SDHA-related disease penetrance. Results Pathogenic germline SDHA variants were identified in 30 of the 393 referred patients with PGL (7.6%), who had head and neck PGL (21 of 174 [12%]), pheochromocytoma (4 of 191 [2%]), or sympathetic PGL (5 of 28 [18%]). The median age at diagnosis was 43 years (range, 17 to 81 years) in index SDHA mutation carriers compared with 52 years (range, 7 to 90 years) in nonmutation carriers (P = 0.002). The estimated penetrance of any SDHA-related manifestation was 10% at age 70 years (95% confidence interval, 0% to 21%) in nonindex mutation carriers. Conclusion Germline SDHA mutations are relatively common (7.6%) in patients with genetically unexplained PGL. Most index patients presented with apparently sporadic PGL. In this SDHA series, the largest assembled so far, we found the lowest penetrance of all major PGL predisposition genes. This suggests that recommendations for genetic counseling of at-risk relatives and stringency of surveillance for SDHA mutation carriers might need to be reassessed.
- Published
- 2018
45. Mutational Signature Analysis Reveals NTHL1 Deficiency to Cause a Multi-Tumor Phenotype Including a Predisposition to Colon and Breast Cancer
- Author
-
M. Elisa Vink-Börger, Ian Tomlinson, Janet R. Vos, Barbara Rivera, Dagmara Dymerska, Na Li, Sanne W. ten Broeke, Isabel Spier, Hildegunn Høberg-Vetti, Gabriel Capellá, Clara Ruiz-Ponte, Erik A. M. Jansen, Mark Clendenning, Renske A. Kuiper, Laura Valle, Stefan Aretz, Rolf H. Sijmons, Tom van Wezel, Claire Palles, Judith E. Grolleman, William D. Foulkes, Ian G. Campbell, Julian Adlard, Sue Kenwrick, Olivera Spasic-Boskovic, Frederik J. Hes, Eveline J. Kamping, Robert Hüneburg, Noel F C C de Miranda, Wenche Sjursen, Ad Geurts van Kessel, Richarda M. de Voer, Isabell Popp, Jan Lubinski, Fadwa A. Elsayed, Detlev Schindler, Marija Staninova, Hans K. Schackert, Robbert D.A. Weren, Aleksandar Dimovski, Nicoline Hoogerbrugge, Kevin Sweet, Helen Lindsay, Alois Lang, Roland P. Kuiper, Kornelia Neveling, David Cockburn, Maartje Nielsen, Daniel D. Buchanan, Hans Morreau, Marjolijn J. L. Ligtenberg, Marjolijn C.J. Jongmans, and Paul A. James
- Subjects
Breast cancer ,Germline mutation ,business.industry ,Colorectal cancer ,Tumor phenotype ,Cancer research ,Medicine ,Lifetime risk ,business ,medicine.disease ,Gene ,Phenotype - Abstract
Biallelic germline mutations affecting NTHL1 predispose to adenomatous polyposis and colorectal cancer, but the complete phenotype is unknown, which hampers patient recognition. We describe 29 individuals from 17 families, of which 26 developed one (n=10) or multiple (n=16) malignancies in 14 different tissues. An unexpected high breast cancer incidence was observed in women with biallelic NTHL1 mutations (60%). Mutational signature analysis of 14 tumors from seven organs revealed that deficient NTHL1 repair underlies the main mutational process in all but one of the tumors (93%). These results reveal NTHL1 as a multi-tumor predisposition gene with a high lifetime risk for extracolonic cancers, and a typical mutational signature observed across tumor types, which can assist in the recognition of this syndrome.
- Published
- 2018
46. CM-Score: a validated scoring system to predict
- Author
-
Thomas P, Potjer, Hildur, Helgadottir, Mirjam, Leenheer, Nienke, van der Stoep, Nelleke A, Gruis, Veronica, Höiom, Håkan, Olsson, Remco, van Doorn, Hans F A, Vasen, Christi J, van Asperen, Olaf M, Dekkers, Frederik J, Hes, and L, Berger
- Subjects
Adult ,Male ,Sweden ,Middle Aged ,Cohort Studies ,Europe ,Pancreatic Neoplasms ,Young Adult ,Logistic Models ,Research Design ,Humans ,Female ,Melanoma ,Cyclin-Dependent Kinase Inhibitor p16 ,Germ-Line Mutation ,Netherlands - Abstract
Several factors have been reported that influence the probability of a germlineFive clinical features and their association withAll five features were significantly associated (p0.05) with aWe developed a practical scoring system to predict
- Published
- 2017
47. Characterization of endolymphatic sac tumors and von Hippel-Lindau disease in the International Endolymphatic Sac Tumor Registry
- Author
-
Elisabetta Zanoletti, Patrice Tran Ba Huy, Claudio Letizia, Mathieu Peyre, Olivier Sterkers, Giuseppe Opocher, Gabriela Sanso, Stéphane Richard, Carlos Suárez, Ulrich F. Wellner, Charis Eng, Carsten Christof Boedeker, Jose M. de Campos, Hartmut P. H. Neumann, Marta Barontini, Birke Bausch, Mariagiulia Anglani, Sophie Giraud, Francesca Schiavi, Christian Offergeld, Angelica Malinoc, Tobias Krauss, Eamonn R. Maher, Hiroshi Kanno, S. Bobin, Claudia Hader, Sven Gläsker, Frederik J. Hes, Thera P. Links, and Stefan Zschiedrich
- Subjects
Gynecology ,medicine.medical_specialty ,Pathology ,business.industry ,Von Hippel-Lindau Tumor Suppressor Protein ,Cancer ,Ear neoplasm ,Von hippel lindau ,medicine.disease ,Endolymphatic sac ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Otorhinolaryngology ,030220 oncology & carcinogenesis ,medicine ,Von Hippel–Lindau disease ,business ,Endolymphatic sac tumor ,030217 neurology & neurosurgery - Abstract
Hartmut P.H. Neumann is supported by grants from the Deutsche Krebshilfe (Grant 107995 to H.P.H.N.). Stephane Richard is supported by grants from the Direction Generale de l’Organisation des Soins (French Department of Health), the Institut National du Cancer (INCa; French NCI), and the Ligue Nationale contre le Cancer (Comites du Cher et de l’Indre; French League against Cancer).
- Published
- 2015
48. Evidence for genetic association between chromosome 1q loci and predisposition to colorectal neoplasia
- Author
-
Arantza Farina Sarasqueta, Tom van Wezel, Clara Ruiz-Ponte, Stephanie A. Schubert, Juul T. Wijnen, Sergi Castellví-Bel, Rolf H. Sijmons, Hans Morreau, Bruce H. R. Wolffenbuttel, Stijn Crobach, Melanie M. van der Klauw, Arnoud Boot, Jan Oosting, Noel F C C de Miranda, Fadwa A. Elsayed, Frederik J. Hes, Hans F. A. Vasen, Maartje Nielsen, Pavel Vodicka, Rolf H. A. M. Vossen, Carli M. J. Tops, Ronald van Eijk, Malcolm G. Dunlop, Ian Tomlinson, Dina Ruano, Medical Genetics, Lifestyle Medicine (LM), Guided Treatment in Optimal Selected Cancer Patients (GUTS), Life Course Epidemiology (LCE), Center for Liver, Digestive and Metabolic Diseases (CLDM), and Pathology
- Subjects
0301 basic medicine ,Cancer Research ,MIA3 ,RNA, Messenger/metabolism ,Genetics & Genomics ,REGULATED NUCLEAR ,chromosome 1q ,0302 clinical medicine ,Neoplasm Proteins/genetics ,Precancerous Conditions/genetics ,Genotype ,Aryl Hydrocarbon Receptor Nuclear Translocator/genetics ,Exome ,Exome sequencing ,Genetics ,Medicine(all) ,0303 health sciences ,education.field_of_study ,GTPase-Activating Proteins ,COLON-CANCER ,Homozygote ,Chromosome Mapping ,Adenomatous Polyposis Coli/genetics ,GERMLINE MUTATIONS ,Disease gene identification ,FIELD SYNOPSIS ,Genetic linkage ,colorectal polyps ,Neoplasm Proteins ,3. Good health ,Adenomatous Polyposis Coli ,Oncology ,Chromosomes, Human, Pair 1/genetics ,homozygosity mapping ,Chromosomes, Human, Pair 1 ,030220 oncology & carcinogenesis ,hereditary colorectal cancer ,Erratum ,Colorectal Neoplasms ,CARCINOMA ,MATRIX-ASSOCIATED PROTEIN ,Population ,Single-nucleotide polymorphism ,Biology ,BREAST ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Chromosome (genetic algorithm) ,GTPase-Activating Proteins/genetics ,Journal Article ,Humans ,Genetic Predisposition to Disease ,linkage analysis ,RNA, Messenger ,education ,030304 developmental biology ,Genetic association ,CANCER RISK ,Aryl Hydrocarbon Receptor Nuclear Translocator ,Colorectal Neoplasms/genetics ,030104 developmental biology ,COLLAGEN SECRETION ,SYSTEMATIC METAANALYSES ,Precancerous Conditions ,exome sequencing ,Microsatellite Repeats - Abstract
BACKGROUND: A substantial fraction of familial colorectal cancer (CRC) and polyposis heritability remains unexplained. This study aimed to identify predisposing loci in patients with these disorders.METHODS: Homozygosity mapping was performed using 222 563 SNPs in 302 index patients with various colorectal neoplasms and 3367 controls. Linkage analysis, exome and whole-genome sequencing were performed in a family affected by microsatellite stable CRCs. Candidate variants were genotyped in 10 554 cases and 21 480 controls. Gene expression was assessed at the mRNA and protein level.RESULTS: Homozygosity mapping revealed a disease-associated region at 1q32.3 which was part of the linkage region 1q32.2-42.2 identified in the CRC family. This includes a region previously associated with risk of CRC. Sequencing identified the p.Asp1432Glu variant in the MIA3 gene (known as TANGO1 or TANGO) and 472 additional rare, shared variants within the linkage region. In both cases and controls the population frequency was 0.02% for this MIA3 variant. The MIA3 mutant allele showed predominant mRNA expression in normal, cancer and precancerous tissues. Furthermore, immunohistochemistry revealed increased expression of MIA3 in adenomatous tissues.CONCLUSIONS: Taken together, our two independent strategies associate genetic variations in chromosome 1q loci and predisposition to familial CRC and polyps, which warrants further investigation.British Journal of Cancer advance online publication: 25 July 2017; doi:10.1038/bjc.2017.240 www.bjcancer.com.
- Published
- 2017
49. Distinct Patterns of Somatic Mosaicism in the APC Gene in Neoplasms From Patients With Unexplained Adenomatous Polyposis
- Author
-
Anne M.L. Jansen, Frederik J. Hes, Marina Ventayol Garcia, Stijn Crobach, Dina Ruano, Brendy E.W.M. van den Akker, Carli M. J. Tops, Tom van Wezel, Juul T. Wijnen, Hans Morreau, Willemina R. R. Geurts-Giele, Winand N.M. Dinjens, Maartje Nielsen, Medical Genetics, and Pathology
- Subjects
0301 basic medicine ,Adenoma ,Adult ,Male ,Pathology ,medicine.medical_specialty ,Genes, APC ,Sequence analysis ,030105 genetics & heredity ,Biology ,medicine.disease_cause ,DNA sequencing ,Neoplasms, Multiple Primary ,03 medical and health sciences ,Intestinal mucosa ,medicine ,Humans ,unexplained polyposis ,Gene ,Wnt Signaling Pathway ,Aged ,Medicine(all) ,Mutation ,Hepatology ,Mosaicism ,Carcinoma ,Gastroenterology ,Wnt signaling pathway ,High-Throughput Nucleotide Sequencing ,Adenomatous Polyposis Coli/genetics ,Sequence Analysis, DNA ,Middle Aged ,Adenoma/genetics ,Colorectal Neoplasms/genetics ,Wnt signaling ,digestive system diseases ,Carcinoma/genetics ,colorectal carcinogenesis ,stomatognathic diseases ,030104 developmental biology ,Somatic mosaicism ,Adenomatous Polyposis Coli ,Colon neoplasm ,Neoplasms, Multiple Primary/genetics ,Female ,mutation ,Colorectal Neoplasms - Abstract
We investigated the presence and patterns of mosaicism in the APC gene in patients with colon neoplasms not associated with any other genetic variants; we performed deep sequence analysis of APC in at least 2 adenomas or carcinomas per patient. We identified mosaic variants in APC in adenomas from 9 of the 18 patients with 21 to approximately 100 adenomas. Mosaic variants of APC were variably detected in leukocyte DNA and/or non-neoplastic intestinal mucosa of these patients. In a comprehensive sequence analysis of 1 patient, we found no evidence for mosaicism in APC in non-neoplastic intestinal mucosa. One patient was found to carry a mosaic c.4666dupA APC variant in only 10 of 16 adenomas, indicating the importance of screening 2 or more adenomas for genetic variants.
- Published
- 2017
50. The phenotype of
- Author
-
Nicolasine D, Niemeijer, Johannes A, Rijken, Karin, Eijkelenkamp, Anouk N A, van der Horst-Schrivers, Michiel N, Kerstens, Carli M J, Tops, Anouk, van Berkel, Henri J L M, Timmers, Henricus P M, Kunst, C René, Leemans, Peter H, Bisschop, Koen M A, Dreijerink, Marieke F, van Dooren, Jean-Pierre, Bayley, Alberto M, Pereira, Jeroen C, Jansen, Frederik J, Hes, Erik F, Hensen, and Eleonora P M, Corssmit
- Subjects
Adult ,Male ,Heterozygote ,Adolescent ,Adrenal Gland Neoplasms ,Pheochromocytoma ,Middle Aged ,Cohort Studies ,Paraganglioma ,Succinate Dehydrogenase ,Young Adult ,Phenotype ,Head and Neck Neoplasms ,Humans ,Female ,Child ,Germ-Line Mutation ,Aged ,Follow-Up Studies ,Netherlands ,Retrospective Studies - Abstract
Succinate dehydrogenase B subunit (Retrospective descriptive study.Retrospective descriptive study in seven academic centers.We included 194In this nationwide study of disease-affected and unaffected
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.