204 results on '"Gabriel Capellá"'
Search Results
2. Tumor analysis of MMR genes in Lynch‐like syndrome: Challenges associated with results interpretation
- Author
-
Paula Rofes, Núria Dueñas, Jesús delValle, Matilde Navarro, Judith Balmaña, Teresa Ramón y Cajal, Noemí Tuset, Carmen Castillo, Sara González, Joan Brunet, Gabriel Capellá, Conxi Lázaro, and Marta Pineda
- Subjects
clinical management ,Lynch syndrome ,Lynch‐like syndrome ,mismatch repair genes ,mismatch repair‐deficiency ,tumor testing ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Up to 70% of suspected Lynch syndrome patients harboring MMR deficient tumors lack identifiable germline pathogenic variants in MMR genes, being referred to as Lynch‐like syndrome (LLS). Previous studies have reported biallelic somatic MMR inactivation in a variable range of LLS‐associated tumors. Moreover, translating tumor testing results into patient management remains controversial. Our aim is to assess the challenges associated with the implementation of tumoral MMR gene testing in routine workflows. Methods Here, we present the clinical characterization of 229 LLS patients. MMR gene testing was performed in 39 available tumors, and results were analyzed using two variant allele frequency (VAF) thresholds (≥5% and ≥10%). Results and Discussion More biallelic somatic events were identified at VAF ≥ 5% than ≥10% (35.9% vs. 25.6%), although the rate of nonconcordant results regarding immunohistochemical pattern increased (30.8% vs. 20.5%). Interpretation difficulties question the current utility of the identification of MMR somatic hits in the diagnostic algorithm of suspected LS cases.
- Published
- 2024
- Full Text
- View/download PDF
3. Recommendations for the classification of germline variants in the exonuclease domain of POLE and POLD1
- Author
-
Pilar Mur, Julen Viana-Errasti, Sandra García-Mulero, Lorena Magraner-Pardo, Inés G. Muñoz, Tirso Pons, Gabriel Capellá, Marta Pineda, Lidia Feliubadaló, and Laura Valle
- Subjects
Polymerase proofreading-associated polyposis ,PPAP ,Polymerase epsilon ,Polymerase delta ,Proofreading deficiency ,Mutational signatures ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background Germline variants affecting the proofreading activity of polymerases epsilon and delta cause a hereditary cancer and adenomatous polyposis syndrome characterized by tumors with a high mutational burden and a specific mutational spectrum. In addition to the implementation of multiple pieces of evidence for the classification of gene variants, POLE and POLD1 variant classification is particularly challenging given that non-disruptive variants affecting the proofreading activity of the corresponding polymerase are the ones associated with cancer. In response to an evident need in the field, we have developed gene-specific variant classification recommendations, based on the ACMG/AMP (American College of Medical Genetics and Genomics/Association for Molecular Pathology) criteria, for the assessment of non-disruptive variants located in the sequence coding for the exonuclease domain of the polymerases. Methods A training set of 23 variants considered pathogenic or benign was used to define the usability and strength of the ACMG/AMP criteria. Population frequencies, computational predictions, co-segregation data, phenotypic and tumor data, and functional results, among other features, were considered. Results Gene-specific variant classification recommendations for non-disruptive variants located in the exonuclease domain of POLE and POLD1 were defined. The resulting recommendations were applied to 128 exonuclease domain variants reported in the literature and/or public databases. A total of 17 variants were classified as pathogenic or likely pathogenic, and 17 as benign or likely benign. Conclusions Our recommendations, with room for improvement in the coming years as more information become available on carrier families, tumor molecular characteristics and functional assays, are intended to serve the clinical and scientific communities and help improve diagnostic performance, avoiding variant misclassifications.
- Published
- 2023
- Full Text
- View/download PDF
4. Wnt genes in colonic polyposis predisposition
- Author
-
Isabel Quintana, Mariona Terradas, Pilar Mur, Iris B.A.W. te Paske, Sophia Peters, Isabel Spier, Verena Steinke-Lange, Claudia Maestro, David Torrents, Montserrat Puiggròs, Romina Royo, Raul Tonda, Genís Parra, Davide Piscia, Sergi Beltrán, Matilde Navarro, Virginia Piñol, Joan Brunet, Noemi Gonzalez-Abuin, Gemma Aiza, Anna Sommer, Yasmijn van Herwaarden, Galuh Astuti, Elke Holinski-Feder, Nicoline Hoogerbrugge, Richarda M. de Voer, Stefan Aretz, Gabriel Capellá, and Laura Valle
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2023
- Full Text
- View/download PDF
5. RNA assay identifies a previous misclassification of BARD1 c.1977A>G variant
- Author
-
Paula Rofes, Marta Pineda, Lídia Feliubadaló, Mireia Menéndez, Rafael de Cid, Carolina Gómez, Eva Montes, Gabriel Capellá, Joan Brunet, Jesús del Valle, and Conxi Lázaro
- Subjects
Medicine ,Science - Abstract
Abstract Case–control studies have shown an association of BARD1 with hereditary breast and/or ovarian cancer (HBOC) predisposition. BARD1 alternatively spliced isoforms are abundant and some are highly expressed in different cancer types. In addition, a number of BARD1 germline pathogenic variants have been reported among HBOC patients. In previous reports, BARD1 c.1977A>G variant has been classified as pathogenic since it produces a frameshift transcript lacking exons 2 to 9. In the present study, we sought to validate the mRNA splicing results previously published and to contribute with new evidence to refine the classification of this substitution according to ACMG/AMP guidelines. The presence of the variant was screened in patients and controls. RT-PCR was performed in order to compare the transcriptional profiles of two variant carriers and ten non-carrier controls. In addition, allele-specific expression was assessed. No differences in variant frequency were detected between patients and controls. The RNA assay confirmed the presence of the shorter transcript lacking exons 2–9, but it was detected both in carriers and non-carriers. Furthermore, allelic imbalance was discarded and no significant differences in the proportion of full-length and shorter transcript were detected between carriers and controls. The shorter transcript detected corresponds to BARD1 isoform η, constituted by exons 1, 10 and 11. Our results support that this transcript is a constitutive splicing product rather than an aberrant transcript caused by BARD1 c.1977A>G variant, and for this reason this variant should be considered as likely benign following ACMG/AMP guidelines.
- Published
- 2021
- Full Text
- View/download PDF
6. A genetic variant in telomerase reverse transcriptase (TERT) modifies cancer risk in Lynch syndrome patients harbouring pathogenic MSH2 variants
- Author
-
Mariann Unhjem Wiik, Tiffany-Jane Evans, Sami Belhadj, Katherine A. Bolton, Dagmara Dymerska, Shantie Jagmohan-Changur, Gabriel Capellá, Grzegorz Kurzawski, Juul T. Wijnen, Laura Valle, Hans F. A. Vasen, Jan Lubinski, Rodney J. Scott, and Bente A. Talseth-Palmer
- Subjects
Medicine ,Science - Abstract
Abstract Individuals with Lynch syndrome (LS), have an increased risk of developing cancer. Common genetic variants of telomerase reverse transcriptase (TERT) have been associated with a wide range of cancers, including colorectal cancer (CRC) in LS. We combined genotype data from 1881 LS patients, carrying pathogenic variants in MLH1, MSH2 or MSH6, for rs2075786 (G>A, intronic variant), 1207 LS patients for rs2736108 (C>T, upstream variant) and 1201 LS patients for rs7705526 (C>A, intronic variant). The risk of cancer was estimated by heterozygous/homozygous odds ratio (OR) with mixed-effects logistic regression to adjust for gene/gender/country of sample origin considering family identity. The AA genotype of SNP rs2075786 is associated with 85% higher odds at developing cancer compared to GG genotype in MSH2 pathogenic variant carriers (p = 0.0160). Kaplan–Meier analysis also shows an association for rs2075786; the AA allele for MSH2 variant carriers confers risk for earlier diagnosis of LS cancer (log-rank p = 0.0011). We report a polymorphism in TERT to be a possible modifier of disease risk in MSH2 pathogenic variant carriers. The rs2075786 SNP in TERT is associated with a differential risk of developing cancer for MSH2 pathogenic variant carriers. Use of this information has the potential to personalise screening protocols for LS patients.
- Published
- 2021
- Full Text
- View/download PDF
7. Phase I, multicenter, open-label study of intravenous VCN-01 oncolytic adenovirus with or without nab-paclitaxel plus gemcitabine in patients with advanced solid tumors
- Author
-
Gabriel Capellá, Francisco X Real, Marta Gil-Martin, Rocio Garcia-Carbonero, Rafael Moreno, Ramon Alemany, Martí Farrera-Sal, Ramon Salazar, Teresa Macarulla, Jaime Martinez de Villarreal, Natalia del Pozo, Carmen Guillén-Ponce, Miriam Bazan-Peregrino, Rafael Álvarez, Ana Mato-Berciano, Emma Blasi, Carmen Blasco, Manel Cascallo, Maria C Riesco-Martinez, Helena Verdaguer, Maria Victoria Maliandi, Silvia Torres-Manjon, Marcel Costa, and Noemí Vidal
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background VCN-01 is an oncolytic adenovirus (Ad5 based) designed to replicate in cancer cells with dysfunctional RB1 pathway, express hyaluronidase to enhance virus intratumoral spread and facilitate chemotherapy and immune cells extravasation into the tumor. This phase I clinical trial was aimed to find the maximum tolerated dose/recommended phase II dose (RP2D) and dose-limiting toxicity (DLT) of the intravenous delivery of the replication-competent VCN-01 adenovirus in patients with advanced cancer.Methods Part I: patients with advanced refractory solid tumors received one single dose of VCN-01. Parts II and III: patients with pancreatic adenocarcinoma received VCN-01 (only in cycle 1) and nab-paclitaxel plus gemcitabine (VCN-concurrent on day 1 in Part II, and 7 days before chemotherapy in Part III). Patients were required to have anti-Ad5 neutralizing antibody (NAbs) titers lower than 1/350 dilution. Pharmacokinetic and pharmacodynamic analyses were performed.Results 26% of the patients initially screened were excluded based on high NAbs levels. Sixteen and 12 patients were enrolled in Part I and II, respectively: RP2D were 1×1013 viral particles (vp)/patient (Part I), and 3.3×1012 vp/patient (Part II). Fourteen patients were included in Part III: there were no DLTs and the RP2D was 1×1013 vp/patient. Observed DLTs were grade 4 aspartate aminotransferase increase in one patient (Part I, 1×1013 vp), grade 4 febrile neutropenia in one patient and grade 5 thrombocytopenia plus enterocolitis in another patient (Part II, 1×1013 vp). In patients with pancreatic adenocarcinoma overall response rate were 50% (Part II) and 50% (Part III). VCN-01 viral genomes were detected in tumor tissue in five out of six biopsies (day 8). A second viral plasmatic peak and increased hyaluronidase serum levels suggested replication after intravenous injection in all patients. Increased levels of immune biomarkers (interferon-γ, soluble lymphocyte activation gene-3, interleukin (IL)-6, IL-10) were found after VCN-01 administration.Conclusions Treatment with VCN-01 is feasible and has an acceptable safety. Encouraging biological and clinical activity was observed when administered in combination with nab-paclitaxel plus gemcitabine to patients with pancreatic adenocarcinoma.Trial registration number NCT02045602.
- Published
- 2022
- Full Text
- View/download PDF
8. VCN-01 disrupts pancreatic cancer stroma and exerts antitumor effects
- Author
-
Gabriel Capellá, Rocio Garcia-Carbonero, Rafael Moreno, Ramon Alemany, Alba Rodriguez-Garcia, Ramon Salazar, Mireia M Ginestà, Miriam Bazan-Peregrino, Berta Laquente, Rafael Álvarez, Ana Mato-Berciano, Marta Gimenez-Alejandre, Sara Morgado, Maria V Maliandi, M Carmen Riesco, Mercedes Perez-Carreras, Joan B Gornals, Susana Prados, Sofía Perea, Emma Blasi, Carmen Blasco, and Manel Cascallo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
9. Genetic Screening for TLR7 Variants in Young and Previously Healthy Men With Severe COVID-19
- Author
-
Xavier Solanich, Gardenia Vargas-Parra, Caspar I. van der Made, Annet Simons, Janneke Schuurs-Hoeijmakers, Arnau Antolí, Jesús del Valle, Gemma Rocamora-Blanch, Fernando Setién, Manel Esteller, Simon V. van Reijmersdal, Antoni Riera-Mestre, Joan Sabater-Riera, Gabriel Capellá, Frank L. van de Veerdonk, Ben van der Hoven, Xavier Corbella, Alexander Hoischen, and Conxi Lázaro
- Subjects
COVID-19 ,SARS-CoV-2 ,host genetics ,TLR7 ,immunodeficiency ,genetic screening ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionLoss-of-function TLR7 variants have been recently reported in a small number of males to underlie strong predisposition to severe COVID-19. We aimed to determine the presence of these rare variants in young men with severe COVID-19.MethodsWe prospectively studied males between 18 and 50 years-old without predisposing comorbidities that required at least high-flow nasal oxygen to treat COVID-19. The coding region of TLR7 was sequenced to assess the presence of potentially deleterious variants.ResultsTLR7 missense variants were identified in two out of 14 patients (14.3%). Overall, the median age was 38 (IQR 30-45) years. Both variants were not previously reported in population control databases and were predicted to be damaging by in silico predictors. In a 30-year-old patient a maternally inherited variant [c.644A>G; p.(Asn215Ser)] was identified, co-segregating in his 27-year-old brother who also contracted severe COVID-19. A second variant [c.2797T>C; p.(Trp933Arg)] was found in a 28-year-old patient, co-segregating in his 24-year-old brother who developed mild COVID-19. Functional testing of this variant revealed decreased type I and II interferon responses in peripheral mononuclear blood cells upon stimulation with the TLR7 agonist imiquimod, confirming a loss-of-function effect.ConclusionsThis study supports a rationale for the genetic screening for TLR7 variants in young men with severe COVID-19 in the absence of other relevant risk factors. A diagnosis of TLR7 deficiency could not only inform on treatment options for the patient, but also enables pre-symptomatic testing of at-risk male relatives with the possibility of instituting early preventive and therapeutic interventions.
- Published
- 2021
- Full Text
- View/download PDF
10. Expression and Role of MicroRNAs from the miR-200 Family in the Tumor Formation and Metastatic Propensity of Pancreatic Cancer
- Author
-
Zamira Vanessa Diaz-Riascos, Mireia M. Ginesta, Joan Fabregat, Teresa Serrano, Juli Busquets, Louis Buscail, Pierre Cordelier, and Gabriel Capellá
- Subjects
Therapeutics. Pharmacology ,RM1-950 - Abstract
MicroRNAs from the miR-200 family are commonly associated with the inhibition of the metastatic potential of cancer cells, following inhibition of ZEB transcription factors expression and epithelial-to-mesenchymal transition. However, previous studies performed in pancreatic adenocarcinoma revealed a more complex picture challenging this canonical model. To gain better insights into the role of miR-200 family members in this disease, we analyzed the expression of miR-200a, miR-200b, miR-200c, miR-141, miR-429, and miR-205, and ZEB1, ZEB2, and CDH1 in pancreatic tumors and matching normal adjacent parenchyma and patient-derived xenografts. We found that miR-200a, miR-429, and miR-205 are frequently overexpressed in pancreatic tumors, whereas CDH1 is downregulated, and ZEB1 and ZEB2 levels remain unchanged. Furthermore, we measured a positive correlation between miR-200 family members and CDH1 expression, and a negative correlation between ZEB1 and miR-200c, miR-141, and miR-205 expression, respectively. Interestingly, we identified significant changes in expression of epithelial-to-mesenchymal transition regulators and miR-200 members in patient-derived xenografts. Lastly, functional studies revealed that miR-141 and miR-429 inhibit the tumorigenic potential of pancreatic cancer cells. Taken together, this comprehensive analysis strongly suggests that miRNAs from the miR-200 family, and in particular miR-429, may act as a tumor suppressor gene in pancreatic cancer. Keywords: pancreatic cancer, patient-derived xenograft, miR-200 family, miR-429
- Published
- 2019
- Full Text
- View/download PDF
11. Germline mutations in the spindle assembly checkpoint genes BUB1 and BUB3 are infrequent in familial colorectal cancer and polyposis
- Author
-
Pilar Mur, Richarda M. De Voer, Rubén Olivera-Salguero, Sandra Rodríguez-Perales, Tirso Pons, Fernando Setién, Gemma Aiza, Rafael Valdés-Mas, Angelo Bertini, Marta Pineda, Lilian Vreede, Matilde Navarro, Silvia Iglesias, Sara González, Joan Brunet, Alfonso Valencia, Manel Esteller, Conxi Lázaro, Geert J. P. L. Kops, Miguel Urioste, Xose S. Puente, Gabriel Capellá, and Laura Valle
- Subjects
Colorectal cancer predisposition ,Hereditary colorectal cancer ,High-penetrance genes ,Variegated aneuploidy ,Mechanism ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Germline mutations in BUB1 and BUB3 have been reported to increase the risk of developing colorectal cancer (CRC) at young age, in presence of variegated aneuploidy and reminiscent dysmorphic traits of mosaic variegated aneuploidy syndrome. We performed a mutational analysis of BUB1 and BUB3 in 456 uncharacterized mismatch repair-proficient hereditary non-polyposis CRC families and 88 polyposis cases. Four novel or rare germline variants, one splice-site and three missense, were identified in four families. Neither variegated aneuploidy nor dysmorphic traits were observed in carriers. Evident functional effects in the heterozygous form were observed for c.1965-1G>A, but not for c.2296G>A (p.E766K), in spite of the positive co-segregation in the family. BUB1 c.2473C>T (p.P825S) and BUB3 c.77C>T (p.T26I) remained as variants of uncertain significance. As of today, the rarity of functionally relevant mutations identified in familial and/or early onset series does not support the inclusion of BUB1 and BUB3 testing in routine genetic diagnostics of familial CRC.
- Published
- 2018
- Full Text
- View/download PDF
12. Computational Tools for Splicing Defect Prediction in Breast/Ovarian Cancer Genes: How Efficient Are They at Predicting RNA Alterations?
- Author
-
Alejandro Moles-Fernández, Laura Duran-Lozano, Gemma Montalban, Sandra Bonache, Irene López-Perolio, Mireia Menéndez, Marta Santamariña, Raquel Behar, Ana Blanco, Estela Carrasco, Adrià López-Fernández, Neda Stjepanovic, Judith Balmaña, Gabriel Capellá, Marta Pineda, Ana Vega, Conxi Lázaro, Miguel de la Hoya, Orland Diez, and Sara Gutiérrez-Enríquez
- Subjects
hereditary cancer genes ,NGS of gene-panel ,VUS classification ,in silico tools ,splicing ,RNA alteration ,Genetics ,QH426-470 - Abstract
In silico tools for splicing defect prediction have a key role to assess the impact of variants of uncertain significance. Our aim was to evaluate the performance of a set of commonly used splicing in silico tools comparing the predictions against RNA in vitro results. This was done for natural splice sites of clinically relevant genes in hereditary breast/ovarian cancer (HBOC) and Lynch syndrome. A study divided into two stages was used to evaluate SSF-like, MaxEntScan, NNSplice, HSF, SPANR, and dbscSNV tools. A discovery dataset of 99 variants with unequivocal results of RNA in vitro studies, located in the 10 exonic and 20 intronic nucleotides adjacent to exon–intron boundaries of BRCA1, BRCA2, MLH1, MSH2, MSH6, PMS2, ATM, BRIP1, CDH1, PALB2, PTEN, RAD51D, STK11, and TP53, was collected from four Spanish cancer genetic laboratories. The best stand-alone predictors or combinations were validated with a set of 346 variants in the same genes with clear splicing outcomes reported in the literature. Sensitivity, specificity, accuracy, negative predictive value (NPV) and Mathews Coefficient Correlation (MCC) scores were used to measure the performance. The discovery stage showed that HSF and SSF-like were the most accurate for variants at the donor and acceptor region, respectively. The further combination analysis revealed that HSF, HSF+SSF-like or HSF+SSF-like+MES achieved a high performance for predicting the disruption of donor sites, and SSF-like or a sequential combination of MES and SSF-like for predicting disruption of acceptor sites. The performance confirmation of these last results with the validation dataset, indicated that the highest sensitivity, accuracy, and NPV (99.44%, 99.44%, and 96.88, respectively) were attained with HSF+SSF-like or HSF+SSF-like+MES for donor sites and SSF-like (92.63%, 92.65%, and 84.44, respectively) for acceptor sites.We provide recommendations for combining algorithms to conduct in silico splicing analysis that achieved a high performance. The high NPV obtained allows to select the variants in which the study by in vitro RNA analysis is mandatory against those with a negligible probability of being spliceogenic. Our study also shows that the performance of each specific predictor varies depending on whether the natural splicing sites are donors or acceptors.
- Published
- 2018
- Full Text
- View/download PDF
13. Comprehensive establishment and characterization of orthoxenograft mouse models of malignant peripheral nerve sheath tumors for personalized medicine
- Author
-
Joan Castellsagué, Bernat Gel, Juana Fernández‐Rodríguez, Roger Llatjós, Ignacio Blanco, Yolanda Benavente, Diana Pérez‐Sidelnikova, Javier García‐del Muro, Joan Maria Viñals, August Vidal, Rafael Valdés‐Mas, Ernest Terribas, Adriana López‐Doriga, Miguel Angel Pujana, Gabriel Capellá, Xose S Puente, Eduard Serra, Alberto Villanueva, and Conxi Lázaro
- Subjects
MPNST ,NF1 ,patient‐derived tumor xenograft ,preclinical mouse models ,sorafenib ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Malignant peripheral nerve sheath tumors (MPNSTs) are soft‐tissue sarcomas that can arise either sporadically or in association with neurofibromatosis type 1 (NF1). These aggressive malignancies confer poor survival, with no effective therapy available. We present the generation and characterization of five distinct MPNST orthoxenograft models for preclinical testing and personalized medicine. Four of the models are patient‐derived tumor xenografts (PDTX), two independent MPNSTs from the same NF1 patient and two from different sporadic patients. The fifth model is an orthoxenograft derived from an NF1‐related MPNST cell line. All MPNST orthoxenografts were generated by tumor implantation, or cell line injection, next to the sciatic nerve of nude mice, and were perpetuated by 7–10 mouse‐to‐mouse passages. The models reliably recapitulate the histopathological properties of their parental primary tumors. They also mimic distal dissemination properties in mice. Human stroma was rapidly lost after MPNST engraftment and replaced by murine stroma, which facilitated genomic tumor characterization. Compatible with an origin in a catastrophic event and subsequent genome stabilization, MPNST contained highly altered genomes that remained remarkably stable in orthoxenograft establishment and along passages. Mutational frequency and type of somatic point mutations were highly variable among the different MPNSTs modeled, but very consistent when comparing primary tumors with matched orthoxenografts generated. Unsupervised cluster analysis and principal component analysis (PCA) using an MPNST expression signature of ~1,000 genes grouped together all primary tumor–orthoxenograft pairs. Our work points to differences in the engraftment process of primary tumors compared with the engraftment of established cell lines. Following standardization and extensive characterization and validation, the orthoxenograft models were used for initial preclinical drug testing. Sorafenib (a BRAF inhibitor), in combination with doxorubicin or rapamycin, was found to be the most effective treatment for reducing MPNST growth. The development of genomically well‐characterized preclinical models for MPNST allowed the evaluation of novel therapeutic strategies for personalized medicine.
- Published
- 2015
- Full Text
- View/download PDF
14. An alternative approach to establishing unbiased colorectal cancer risk estimation in Lynch syndrome
- Author
-
Suerink, Manon, Rodríguez-Girondo, Mar, van der Klift, Heleen M., Colas, Chrystelle, Brugieres, Laurence, Lavoine, Noémie, Jongmans, Marjolijn, Munar, Gabriel Capellá, Evans, D. Gareth, Farrell, Michael P., Genuardi, Maurizio, Goldberg, Yael, Gomez-Garcia, Encarna, Heinimann, Karl, Hoell, Jessica I., Aretz, Stefan, Jasperson, Kory W., Kedar, Inbal, Modi, Mitul B., Nikolaev, Sergey, van Os, Theo A. M., Ripperger, Tim, Rueda, Daniel, Senter, Leigha, Sjursen, Wenche, Sunde, Lone, Therkildsen, Christina, Tibiletti, Maria G., Trainer, Alison H., Vos, Yvonne J., Wagner, Anja, Winship, Ingrid, Wimmer, Katharina, Zimmermann, Stefanie Y., Vasen, Hans F., van Asperen, Christi J., Houwing-Duistermaat, Jeanine J., ten Broeke, Sanne W., and Nielsen, Maartje
- Published
- 2019
- Full Text
- View/download PDF
15. Data from Characterization of New Founder Alu-Mediated Rearrangements in MSH2 Gene Associated with a Lynch Syndrome Phenotype
- Author
-
Pérez-Cabornero, Lucia, primary, Borrás Flores, Ester, primary, Sanz, Mar Infante, primary, Sampedro, Eladio Velasco, primary, Becares, Alberto Acedo, primary, Aras, Enrique Lastra, primary, González, Jorge Cuevas, primary, Riu, Marta Pineda, primary, Asensio, Teresa Ramón y Cajal, primary, Munar, Gabriel Capellá, primary, Pino, Cristina Miner, primary, and Domínguez, Mercedes Durán, primary
- Published
- 2023
- Full Text
- View/download PDF
16. Perspective on This Article from Characterization of New Founder Alu-Mediated Rearrangements in MSH2 Gene Associated with a Lynch Syndrome Phenotype
- Author
-
Pérez-Cabornero, Lucia, primary, Borrás Flores, Ester, primary, Sanz, Mar Infante, primary, Sampedro, Eladio Velasco, primary, Becares, Alberto Acedo, primary, Aras, Enrique Lastra, primary, González, Jorge Cuevas, primary, Riu, Marta Pineda, primary, Asensio, Teresa Ramón y Cajal, primary, Munar, Gabriel Capellá, primary, Pino, Cristina Miner, primary, and Domínguez, Mercedes Durán, primary
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Materials from Characterization of New Founder Alu-Mediated Rearrangements in MSH2 Gene Associated with a Lynch Syndrome Phenotype
- Author
-
Pérez-Cabornero, Lucia, primary, Borrás Flores, Ester, primary, Sanz, Mar Infante, primary, Sampedro, Eladio Velasco, primary, Becares, Alberto Acedo, primary, Aras, Enrique Lastra, primary, González, Jorge Cuevas, primary, Riu, Marta Pineda, primary, Asensio, Teresa Ramón y Cajal, primary, Munar, Gabriel Capellá, primary, Pino, Cristina Miner, primary, and Domínguez, Mercedes Durán, primary
- Published
- 2023
- Full Text
- View/download PDF
18. Ability of a polygenic risk score to refine colorectal cancer risk in Lynch syndrome
- Author
-
Nuria Dueñas, Hannah Klinkhammer, Nuria Bonifaci, Isabel Spier, Andreas Mayr, Emadeldin Hassanin, Anna Díez-Villanueva, Victor Moreno, Marta Pineda, Carlo Maj, Gabriel Capellá, Stefan Aretz, and Joan Brunet
- Abstract
BackgroundPolygenic risk scores (PRS) have been used to stratify colorectal cancer (CRC) risk in the general population, whereas its role in Lynch syndrome (LS), the most common type of hereditary CRC, is still conflicting. We aimed to assess the ability of PRS to refine CRC risk prediction in European-descendant LS individuals.Methods1,465 LS individuals (557MLH1, 517MSH2/EPCAM, 299MSH6, and 92PMS2)and 5,656 CRC-free population-based controls from two independent cohorts were included. A 91-Single Nucleotide Polymorphism PRS was applied. A Cox proportional hazard regression model with “family” as a random effect and a logistic regression analysis, followed by a meta-analysis combining both cohorts were conducted.ResultsOverall, we did not observe a statistically significant association between PRS and CRC risk in the entire cohort. Nevertheless, PRS was significantly associated with a slightly increased risk of CRC or advanced adenoma (AA), in those with CRC diagnosed < 50 years, and in individuals with multiple CRCs or AAs diagnosed < 60 years.ConclusionThe PRS may slightly influence CRC risk in LS individuals, in particular in more extreme phenotypes such as early-onset disease. However, the study design and recruitment strategy strongly influence the results of PRS studies. A separate analysis by genes and its combination with other genetic and non-genetic risk factors will help refine its role as a risk modifier in LS.KEY MESSAGESWHAT IS ALREADY KNOWN ON THIS TOPIC?-Great variability in the incidence of CRC has been described in LS individuals, even within the same family.-Polygenic risk scores (PRS) can help stratify colorectal cancer risk and, thus, adjust surveillance or treatment procedures.WHAT THIS STUDY ADDS-PRS performed on family-based registries slightly influences CRC risk in subgroups of LS individuals, even though with weak effects.-Our study showed a weak association of PRS with multiple and young CRC cases, pointing to a possible risk-modifying role in extreme phenotypes.HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY-Gene-based PRS analysis and its combination with other genetic and non-genetic factors may contribute to refining cancer risk in LS patients.
- Published
- 2023
19. Data from Nanofluidic Digital PCR and Extended Genotyping of RAS and BRAF for Improved Selection of Metastatic Colorectal Cancer Patients for Anti-EGFR Therapies
- Author
-
Ramon Salazar, Gabriel Capellá, Josep Tabernero, Clara Montagut, Rodrigo Dienstmann, Elena Elez, Guillem Argiles, Beatriz Bellosillo, Victor Moreno, Robert Montal, Joana Vidal, Fátima Marin, Xavier Sanjuan, Marga Nadal, Julieta Grasselli, Adriana Lopez-Doriga, Cristina Santos, and Daniel Azuara
- Abstract
The clinical significance of low-frequent RAS pathway–mutated alleles and the optimal sensitivity cutoff value in the prediction of response to anti-EGFR therapy in metastatic colorectal cancer (mCRC) patients remains controversial. We aimed to evaluate the added value of genotyping an extended RAS panel using a robust nanofluidic digital PCR (dPCR) approach. A panel of 34 hotspots, including RAS (KRAS and NRAS exons 2/3/4) and BRAF (V600E), was analyzed in tumor FFPE samples from 102 mCRC patients treated with anti-EGFR therapy. dPCR was compared with conventional quantitative PCR (qPCR). Response rates, progression-free survival (PFS), and overall survival (OS) were correlated to the mutational status and the mutated allele fraction. Tumor response evaluations were not available in 9 patients and were excluded for response rate analysis. Twenty-two percent of patients were positive for one mutation with qPCR (mutated alleles ranged from 2.1% to 66.6%). Analysis by dPCR increased the number of positive patients to 47%. Mutated alleles for patients only detected by dPCR ranged from 0.04% to 10.8%. An inverse correlation between the fraction of mutated alleles and radiologic response was observed. ROC analysis showed that a fraction of 1% or higher of any mutated alleles offered the best predictive value for all combinations of RAS and BRAF analysis. In addition, this threshold also optimized prediction both PFS and OS. We conclude that mutation testing using an extended gene panel, including RAS and BRAF with a threshold of 1% improved prediction of response to anti-EGFR therapy. Mol Cancer Ther; 15(5); 1106–12. ©2016 AACR.
- Published
- 2023
20. Supplementary Table S5 from Nanofluidic Digital PCR and Extended Genotyping of RAS and BRAF for Improved Selection of Metastatic Colorectal Cancer Patients for Anti-EGFR Therapies
- Author
-
Ramon Salazar, Gabriel Capellá, Josep Tabernero, Clara Montagut, Rodrigo Dienstmann, Elena Elez, Guillem Argiles, Beatriz Bellosillo, Victor Moreno, Robert Montal, Joana Vidal, Fátima Marin, Xavier Sanjuan, Marga Nadal, Julieta Grasselli, Adriana Lopez-Doriga, Cristina Santos, and Daniel Azuara
- Abstract
Estimation of the prediction quality of mutational status for treatment response. First column (AUC) indicates the Area Under the Curve as a prediction quality measure for all mutation groups. As expected, the AUC increases for lower mutation frequency cut-off. Then, a symmetric matrix for each mutation group with five columns, one column for each cut-off, reports the p-values for all curves comparisons. Highlighted in black the p-values that indicate correlative comparison. Highlighted in red are the p-values close to significant levels that correspond to the comparison between 1% and 1.5% cut-off value. Differences between cut-off of 1% and 1.5% are bigger than differences between the other correlative cut-off comparisons. P-values are close to significance level with a confidence of 90%.
- Published
- 2023
21. Supplementary Materials from Characterization of New Founder Alu-Mediated Rearrangements in MSH2 Gene Associated with a Lynch Syndrome Phenotype
- Author
-
Mercedes Durán Domínguez, Cristina Miner Pino, Gabriel Capellá Munar, Teresa Ramón y Cajal Asensio, Marta Pineda Riu, Jorge Cuevas González, Enrique Lastra Aras, Alberto Acedo Becares, Eladio Velasco Sampedro, Mar Infante Sanz, Ester Borrás Flores, and Lucia Pérez-Cabornero
- Abstract
PDF file - 198K
- Published
- 2023
22. Perspective on This Article from Characterization of New Founder Alu-Mediated Rearrangements in MSH2 Gene Associated with a Lynch Syndrome Phenotype
- Author
-
Mercedes Durán Domínguez, Cristina Miner Pino, Gabriel Capellá Munar, Teresa Ramón y Cajal Asensio, Marta Pineda Riu, Jorge Cuevas González, Enrique Lastra Aras, Alberto Acedo Becares, Eladio Velasco Sampedro, Mar Infante Sanz, Ester Borrás Flores, and Lucia Pérez-Cabornero
- Abstract
Perspective on This Article from Characterization of New Founder Alu-Mediated Rearrangements in MSH2 Gene Associated with a Lynch Syndrome Phenotype
- Published
- 2023
23. Supplementary Tables 1-4 from DNA Methylation Biomarkers for Noninvasive Diagnosis of Colorectal Cancer
- Author
-
Victor Moreno, Manel Esteller, Gabriel Capellá, Jordi Guardiola, Mario F. Fraga, Alberto Villanueva, Ramón Salazar, Francisco Rodriguez-Moranta, Javier de Oca, Sebastiano Biondo, Agustin F. Fernández, Antonio Berenguer-Llergo, Daniel Azuara, and F. Javier Carmona
- Abstract
PDF File - 100K, Pyrosequencing primers used to quantify the methylation status of selected CG sites.
- Published
- 2023
24. Data from Characterization of New Founder Alu-Mediated Rearrangements in MSH2 Gene Associated with a Lynch Syndrome Phenotype
- Author
-
Mercedes Durán Domínguez, Cristina Miner Pino, Gabriel Capellá Munar, Teresa Ramón y Cajal Asensio, Marta Pineda Riu, Jorge Cuevas González, Enrique Lastra Aras, Alberto Acedo Becares, Eladio Velasco Sampedro, Mar Infante Sanz, Ester Borrás Flores, and Lucia Pérez-Cabornero
- Abstract
It has been reported that large genomic deletions in the MLH1 and MSH2 genes are a frequent cause of Lynch syndrome in certain populations. Here, a cohort has been screened and two new founder rearrangements have been found in the MSH2 gene. These mutations have been characterized by break point determination, haplotype analysis, and genotype–phenotype correlation. Mutations have been identified in the MLH1, MSH2, and MSH6 genes in 303 subjects from 160 suspected Lynch syndrome unrelated families. All subjects were tested using heteroduplex analysis by capillary array electrophoresis. Multiplex ligation-dependent probe amplification was used to detect rearrangements in mutation-negative index patients and confirmed by reverse transcriptase PCR. The break point of the deletions was further characterized by the array comparative genomic hybridization method. Immunohistochemical staining and microsatellite instability were studied in tumor samples. Hereditary nonpolyposis colorectal cancer–related phenotypes were evaluated. More than 16% (24 of 160) of the families had pathogenic mutations (8 MLH1, 15 MSH2, and 1 MSH6). Twelve of these families (50%) are carriers of a novel mutation. Seven of the 15 positive MSH2 families (47%) are carriers of a rearrangement. The exon 7 deletion and exon 4 to 8 deletion of MSH2 are new founder mutations. The segregation of a common haplotype, a similar phenotype, and anticipation effects were observed in these families. These findings will greatly simplify the diagnosis, counseling, and clinical care in suspected Lynch syndrome families and not just in specific geographic areas, so wide distribution may be explained by migration patterns. Cancer Prev Res; 4(10); 1546–55. ©2011 AACR.
- Published
- 2023
25. Supplementary Figures 1-6 from DNA Methylation Biomarkers for Noninvasive Diagnosis of Colorectal Cancer
- Author
-
Victor Moreno, Manel Esteller, Gabriel Capellá, Jordi Guardiola, Mario F. Fraga, Alberto Villanueva, Ramón Salazar, Francisco Rodriguez-Moranta, Javier de Oca, Sebastiano Biondo, Agustin F. Fernández, Antonio Berenguer-Llergo, Daniel Azuara, and F. Javier Carmona
- Abstract
PDF File - 916K, Representative pyrograms in a healthy colonic mucosa sample for the markers analyzed.
- Published
- 2023
26. Data from DNA Methylation Biomarkers for Noninvasive Diagnosis of Colorectal Cancer
- Author
-
Victor Moreno, Manel Esteller, Gabriel Capellá, Jordi Guardiola, Mario F. Fraga, Alberto Villanueva, Ramón Salazar, Francisco Rodriguez-Moranta, Javier de Oca, Sebastiano Biondo, Agustin F. Fernández, Antonio Berenguer-Llergo, Daniel Azuara, and F. Javier Carmona
- Abstract
DNA methylation biomarkers for noninvasive diagnosis of colorectal cancer (CRC) and precursor lesions have been extensively studied. Different panels have been reported attempting to improve current protocols in clinical practice, although no definite biomarkers have been established.In the present study, we have examined patient biopsies starting from a comprehensive analysis of DNA methylation differences between paired normal and tumor samples in known cancer-related genes aiming to select the best performing candidates informative for CRC diagnosis in stool samples.Five selected markers were considered for subsequent analyses in independent biologic cohorts and in silico data sets. Among the five selected genes, three of them (AGTR1, WNT2 and SLIT2) were validated in stool DNA of affected patients with a detection sensitivity of 78% [95% confidence interval (CI), 56%–89%]. As a reference, DNA methylation of VIM and SEPT9 was evaluated in a subset of stool samples yielding sensitivities of 55% and 20%, respectively. Moreover, our panel may complement histologic and endoscopic diagnosis of inflammatory bowel disease (IBD)-associated neoplasia, as it was also efficient detecting aberrant DNA methylation in non-neoplastic tissue samples from affected patients.This novel panel of specific methylation markers can be useful for early diagnosis of CRC using stool DNA and may help in the follow-up of high-risk patients with IBD. Cancer Prev Res; 6(7); 656–65. ©2013 AACR.
- Published
- 2023
27. Supplementary Methods from DNA Methylation Biomarkers for Noninvasive Diagnosis of Colorectal Cancer
- Author
-
Victor Moreno, Manel Esteller, Gabriel Capellá, Jordi Guardiola, Mario F. Fraga, Alberto Villanueva, Ramón Salazar, Francisco Rodriguez-Moranta, Javier de Oca, Sebastiano Biondo, Agustin F. Fernández, Antonio Berenguer-Llergo, Daniel Azuara, and F. Javier Carmona
- Abstract
PDF file - 574K, Microarray pre-processing.
- Published
- 2023
28. Lack of evidence for germline WWP1 pathogenic variants in gastrointestinal polyposis and other phenotypes suggestive of PTEN-hamartoma-tumor syndrome
- Author
-
Noemi Gonzalez-Abuin, Tirso Pons, Teresa Fuster, Isabel Quintana, Mariona Terradas, Gemma Aiza, Joan Brunet, Gabriel Capellá, Heather Hampel, and Laura Valle
- Subjects
Cell Biology ,Molecular Biology ,Biochemistry ,Genetics (clinical) - Published
- 2023
29. Supplementary Table 1 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Author
-
Carlos López-Otín, Santiago Cal, Gabriel Capellá, Aurora Astudillo, María Llamazares, Angela Moncada-Pazos, Alvaro J. Obaya, and Cristina G. Viloria
- Abstract
Supplementary Table 1 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Published
- 2023
30. Supplementary Figure 1 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Author
-
Carlos López-Otín, Santiago Cal, Gabriel Capellá, Aurora Astudillo, María Llamazares, Angela Moncada-Pazos, Alvaro J. Obaya, and Cristina G. Viloria
- Abstract
Supplementary Figure 1 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Published
- 2023
31. Supplementary Table 2 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Author
-
Carlos López-Otín, Santiago Cal, Gabriel Capellá, Aurora Astudillo, María Llamazares, Angela Moncada-Pazos, Alvaro J. Obaya, and Cristina G. Viloria
- Abstract
Supplementary Table 2 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Published
- 2023
32. Data from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Author
-
Gabriel Capellá, Conxi Lázaro, Bhramar Mukherjee, Noah A. Rosenberg, Stephen B. Gruber, Víctor Moreno, Sara González, Ángel Lanas, Ángel Alonso, Sergi Castellví-Bel, Lucía Pérez-Cabornero, Judit Sanz, Teresa Ramón y Cajal, Asunción Torres, Judith Balmaña, Joan Brunet, Cristina Martínez-Bouzas, Miguel Urioste, Trinidad Caldés, Àlex Teulé, Fei Wang, Ethan M. Jewett, Ignacio Blanco, Marta Pineda, and Ester Borràs
- Abstract
The variants c.306+5G>A and c.1865T>A (p.Leu622His) of the DNA repair gene MLH1 occur frequently in Spanish Lynch syndrome families. To understand their ancestral history and clinical effect, we performed functional assays and a penetrance analysis and studied their genetic and geographic origins. Detailed family histories were taken from 29 carrier families. Functional analysis included in silico and in vitro assays at the RNA and protein levels. Penetrance was calculated using a modified segregation analysis adjusted for ascertainment. Founder effects were evaluated by haplotype analysis. The identified MLH1 c.306+5G>A and c.1865T>A (p.Leu622His) variants are absent in control populations and segregate with the disease. Tumors from carriers of both variants show microsatellite instability and loss of expression of the MLH1 protein. The c.306+5G>A variant is a pathogenic mutation affecting mRNA processing. The c.1865T>A (p.Leu622His) variant causes defects in MLH1 expression and stability. For both mutations, the estimated penetrance is moderate (age-cumulative colorectal cancer risk by age 70 of 20.1% and 14.1% for c.306+5G>A and of 6.8% and 7.3% for c.1865T>A in men and women carriers, respectively) in the lower range of variability estimated for other pathogenic Spanish MLH1 mutations. A common haplotype was associated with each of the identified mutations, confirming their founder origin. The ages of c.306+5G>A and c.1865T>A mutations were estimated to be 53 to 122 and 12 to 22 generations, respectively. Our results confirm the pathogenicity, moderate penetrance, and founder origin of the MLH1 c.306+5G>A and c.1865T>A mutations. These findings have important implications for genetic counseling and molecular diagnosis of Lynch syndrome. Cancer Res; 70(19); 7379–91. ©2010 AACR.
- Published
- 2023
33. Supplementary Figure 2 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Author
-
Carlos López-Otín, Santiago Cal, Gabriel Capellá, Aurora Astudillo, María Llamazares, Angela Moncada-Pazos, Alvaro J. Obaya, and Cristina G. Viloria
- Abstract
Supplementary Figure 2 from Genetic Inactivation of ADAMTS15 Metalloprotease in Human Colorectal Cancer
- Published
- 2023
34. Supplementary Table 1 from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Author
-
Gabriel Capellá, Conxi Lázaro, Bhramar Mukherjee, Noah A. Rosenberg, Stephen B. Gruber, Víctor Moreno, Sara González, Ángel Lanas, Ángel Alonso, Sergi Castellví-Bel, Lucía Pérez-Cabornero, Judit Sanz, Teresa Ramón y Cajal, Asunción Torres, Judith Balmaña, Joan Brunet, Cristina Martínez-Bouzas, Miguel Urioste, Trinidad Caldés, Àlex Teulé, Fei Wang, Ethan M. Jewett, Ignacio Blanco, Marta Pineda, and Ester Borràs
- Abstract
Supplementary Table 1 from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Published
- 2023
35. Supplementary Figure 1 from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Author
-
Gabriel Capellá, Conxi Lázaro, Bhramar Mukherjee, Noah A. Rosenberg, Stephen B. Gruber, Víctor Moreno, Sara González, Ángel Lanas, Ángel Alonso, Sergi Castellví-Bel, Lucía Pérez-Cabornero, Judit Sanz, Teresa Ramón y Cajal, Asunción Torres, Judith Balmaña, Joan Brunet, Cristina Martínez-Bouzas, Miguel Urioste, Trinidad Caldés, Àlex Teulé, Fei Wang, Ethan M. Jewett, Ignacio Blanco, Marta Pineda, and Ester Borràs
- Abstract
Supplementary Figure 1 from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Published
- 2023
36. Supplementary Table 2 from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Author
-
Gabriel Capellá, Conxi Lázaro, Bhramar Mukherjee, Noah A. Rosenberg, Stephen B. Gruber, Víctor Moreno, Sara González, Ángel Lanas, Ángel Alonso, Sergi Castellví-Bel, Lucía Pérez-Cabornero, Judit Sanz, Teresa Ramón y Cajal, Asunción Torres, Judith Balmaña, Joan Brunet, Cristina Martínez-Bouzas, Miguel Urioste, Trinidad Caldés, Àlex Teulé, Fei Wang, Ethan M. Jewett, Ignacio Blanco, Marta Pineda, and Ester Borràs
- Abstract
Supplementary Table 2 from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Published
- 2023
37. Supplementary Methods from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Author
-
Gabriel Capellá, Conxi Lázaro, Bhramar Mukherjee, Noah A. Rosenberg, Stephen B. Gruber, Víctor Moreno, Sara González, Ángel Lanas, Ángel Alonso, Sergi Castellví-Bel, Lucía Pérez-Cabornero, Judit Sanz, Teresa Ramón y Cajal, Asunción Torres, Judith Balmaña, Joan Brunet, Cristina Martínez-Bouzas, Miguel Urioste, Trinidad Caldés, Àlex Teulé, Fei Wang, Ethan M. Jewett, Ignacio Blanco, Marta Pineda, and Ester Borràs
- Abstract
Supplementary Methods from MLH1 Founder Mutations with Moderate Penetrance in Spanish Lynch Syndrome Families
- Published
- 2023
38. vaRHC: an R package for semi-automation of variant classification in hereditary cancer genes according to ACMG/AMP and gene-specific ClinGen guidelines
- Author
-
Elisabet Munté, Lidia Feliubadaló, Marta Pineda, Eva Tornero, Maribel Gonzalez, José Marcos Moreno-Cabrera, Carla Roca, Joan Bales Rubio, Laura Arnaldo, Gabriel Capellá, Jose Luis Mosquera, and Conxi Lázaro
- Subjects
Statistics and Probability ,Computational Mathematics ,Computational Theory and Mathematics ,Molecular Biology ,Biochemistry ,Computer Science Applications - Abstract
Motivation Germline variant classification allows accurate genetic diagnosis and risk assessment. However, it is a tedious iterative process integrating information from several sources and types of evidence. It should follow gene-specific (if available) or general updated international guidelines. Thus, it is the main burden of the incorporation of next-generation sequencing into the clinical setting. Results We created the vaRiants in HC (vaRHC) R package to assist the process of variant classification in hereditary cancer by: (i) collecting information from diverse databases; (ii) assigning or denying different types of evidence according to updated American College of Molecular Genetics and Genomics/Association of Molecular Pathologist gene-specific criteria for ATM, CDH1, CHEK2, MLH1, MSH2, MSH6, PMS2, PTEN, and TP53 and general criteria for other genes; (iii) providing an automated classification of variants using a Bayesian metastructure and considering CanVIG-UK recommendations; and (iv) optionally printing the output to an .xlsx file. A validation using 659 classified variants demonstrated the robustness of vaRHC, presenting a better criteria assignment than Cancer SIGVAR, an available similar tool. Availability and implementation The source code can be consulted in the GitHub repository (https://github.com/emunte/vaRHC) Additionally, it will be submitted to CRAN soon.
- Published
- 2023
39. Genotype-first approach to identify associations between CDH1 germline variants and cancer phenotypes: a multicentre study by the European Reference Network on Genetic Tumour Risk Syndromes
- Author
-
José Garcia-Pelaez, Rita Barbosa-Matos, Silvana Lobo, Alexandre Dias, Luzia Garrido, Sérgio Castedo, Sónia Sousa, Hugo Pinheiro, Liliana Sousa, Rita Monteiro, Joaquin J Maqueda, Susana Fernandes, Fátima Carneiro, Nádia Pinto, Carolina Lemos, Carla Pinto, Manuel R Teixeira, Stefan Aretz, Svetlana Bajalica-Lagercrantz, Judith Balmaña, Ana Blatnik, Patrick R Benusiglio, Maud Blanluet, Vincent Bours, Hilde Brems, Joan Brunet, Daniele Calistri, Gabriel Capellá, Sergio Carrera, Chrystelle Colas, Karin Dahan, Robin de Putter, Camille Desseignés, Elena Domínguez-Garrido, Conceição Egas, D Gareth Evans, Damien Feret, Eleanor Fewings, Rebecca C Fitzgerald, Florence Coulet, María Garcia-Barcina, Maurizio Genuardi, Lisa Golmard, Karl Hackmann, Helen Hanson, Elke Holinski-Feder, Robert Hüneburg, Mateja Krajc, Kristina Lagerstedt-Robinson, Conxi Lázaro, Marjolijn J L Ligtenberg, Cristina Martínez-Bouzas, Sonia Merino, Geneviève Michils, Srdjan Novaković, Ana Patiño-García, Guglielmina Nadia Ranzani, Evelin Schröck, Inês Silva, Catarina Silveira, José L Soto, Isabel Spier, Verena Steinke-Lange, Gianluca Tedaldi, María-Isabel Tejada, Emma R Woodward, Marc Tischkowitz, Nicoline Hoogerbrugge, Carla Oliveira, Institut Català de la Salut, [Garcia-Pelaez J] Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal. Faculty of Medicine, University of Porto, Porto, Portugal. Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal. Porto Comprehensive Cancer Center Raquel Seruca, Porto, Portugal. [Barbosa-Matos R, Lobo S] Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal. Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal. Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal. Porto Comprehensive Cancer Center Raquel Seruca, Porto, Portugal. [Dias A] Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal. Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal. Porto Comprehensive Cancer Center Raquel Seruca, Porto, Portugal. [Garrido L] Centro Hospitalar Universitário São João, Porto, Portugal. [Castedo S] Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal. Faculty of Medicine, University of Porto, Porto, Portugal. Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal. Centro Hospitalar Universitário São João, Porto, Portugal. Porto Comprehensive Cancer Center Raquel Seruca, Porto, Portugal. European Reference Network on Genetic Tumour Risk Syndromes (ERN GENTURIS), Porto, Portugal. [Balmaña J] Vall d'Hebron Hospital Universitari, Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. ERN GENTURIS, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
neoplasias::neoplasias por localización::neoplasias del sistema digestivo::neoplasias gastrointestinales::neoplasias gástricas [ENFERMEDADES] ,Genotype ,Neoplasms::Neoplasms by Site::Breast Neoplasms [DISEASES] ,enfermedades de la piel y tejido conjuntivo::enfermedades de la piel::enfermedades de la mama [ENFERMEDADES] ,Genetic Phenomena::Genotype::Genetic Predisposition to Disease [PHENOMENA AND PROCESSES] ,Mutation, Missense ,Breast Neoplasms ,All institutes and research themes of the Radboud University Medical Center ,Germ Cells/pathology ,Antigens, CD ,Stomach Neoplasms ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Humans ,Genetic Predisposition to Disease ,Genotip ,Càncer ,Germ-Line Mutation ,fenómenos genéticos::genotipo::predisposición genética a la enfermedad [FENÓMENOS Y PROCESOS] ,Cancer ,Retrospective Studies ,Breast Neoplasms/epidemiology ,Cadherins/genetics ,Antigens, CD/genetics ,Stomach Neoplasms/epidemiology ,Cadherins ,Pedigree ,Fenotip ,Carcinoma, Lobular ,Germ Cells ,Phenotype ,Oncology ,Aparell digestiu - Càncer - Aspectes genètics ,Mama - Càncer - Aspectes genètics ,Neoplasms::Neoplasms by Site::Digestive System Neoplasms::Gastrointestinal Neoplasms::Stomach Neoplasms [DISEASES] ,Female - Abstract
Genotype; Cancer phenotypes; Genetic tumour Genotip; Fenotips del càncer; Tumor genètic Genotipo; Fenotipos del cáncer; Tumor genético Background Truncating pathogenic or likely pathogenic variants of CDH1 cause hereditary diffuse gastric cancer (HDGC), a tumour risk syndrome that predisposes carrier individuals to diffuse gastric and lobular breast cancer. Rare CDH1 missense variants are often classified as variants of unknown significance. We conducted a genotype–phenotype analysis in families carrying rare CDH1 variants, comparing cancer spectrum in carriers of pathogenic or likely pathogenic variants (PV/LPV; analysed jointly) or missense variants of unknown significance, assessing the frequency of families with lobular breast cancer among PV/LPV carrier families, and testing the performance of lobular breast cancer-expanded criteria for CDH1 testing. Methods This genotype-first study used retrospective diagnostic and clinical data from 854 carriers of 398 rare CDH1 variants and 1021 relatives, irrespective of HDGC clinical criteria, from 29 institutions in ten member-countries of the European Reference Network on Tumour Risk Syndromes (ERN GENTURIS). Data were collected from Oct 1, 2018, to Sept 20, 2022. Variants were classified by molecular type and clinical actionability with the American College of Medical Genetics and Association for Molecular Pathology CDH1 guidelines (version 2). Families were categorised by whether they fulfilled the 2015 and 2020 HDGC clinical criteria. Genotype–phenotype associations were analysed by Student's t test, Kruskal-Wallis, χ2, and multivariable logistic regression models. Performance of HDGC clinical criteria sets were assessed with an equivalence test and Youden index, and the areas under the receiver operating characteristic curves were compared by Z test. Findings From 1971 phenotypes (contributed by 854 probands and 1021 relatives aged 1–93 years), 460 had gastric and breast cancer histology available. CDH1 truncating PV/LPVs occurred in 176 (21%) of 854 families and missense variants of unknown significance in 169 (20%) families. Multivariable logistic regression comparing phenotypes occurring in families carrying PV/LPVs or missense variants of unknown significance showed that lobular breast cancer had the greatest positive association with the presence of PV/LPVs (odds ratio 12·39 [95% CI 2·66–57·74], p=0·0014), followed by diffuse gastric cancer (8·00 [2·18–29·39], p=0·0017) and gastric cancer (7·81 [2·03–29·96], p=0·0027). 136 (77%) of 176 families carrying PV/LPVs fulfilled the 2015 HDGC criteria. Of the remaining 40 (23%) families, who did not fulfil the 2015 criteria, 11 fulfilled the 2020 HDGC criteria, and 18 had lobular breast cancer only or lobular breast cancer and gastric cancer, but did not meet the 2020 criteria. No specific CDH1 variant was found to predispose individuals specifically to lobular breast cancer, although 12 (7%) of 176 PV/LPV carrier families had lobular breast cancer only. Addition of three new lobular breast cancer-centred criteria improved testing sensitivity while retaining high specificity. The probability of finding CDH1 PV/LPVs in patients fulfilling the lobular breast cancer-expanded criteria, compared with the 2020 criteria, increased significantly (AUC 0·92 vs 0·88; Z score 3·54; p=0·0004). Interpretation CDH1 PV/LPVs were positively associated with HDGC-related phenotypes (lobular breast cancer, diffuse gastric cancer, and gastric cancer), and no evidence for a positive association with these phenotypes was found for CDH1 missense variants of unknown significance. CDH1 PV/LPVs occurred often in families with lobular breast cancer who did not fulfil the 2020 HDGC criteria, supporting the expansion of lobular breast cancer-centred criteria. Funding European Reference Network on Genetic Tumour Risk Syndromes, European Regional Development Fund, Fundação para a Ciência e a Tecnologia (Portugal), Cancer Research UK, and European Union's Horizon 2020 research and innovation programme. European Reference Network on Genetic Tumour Risk Syndromes, European Regional Development Fund, Fundação para a Ciência e a Tecnologia (Portugal), Cancer Research UK, and European Union's Horizon 2020 research and innovation programme.
- Published
- 2023
40. Paired Somatic-Germline Testing of 15 Polyposis and Colorectal Cancer–Predisposing Genes Highlights the Role of APC Mosaicism in de Novo Familial Adenomatous Polyposis
- Author
-
Conxi Lázaro, Estela Carrasco, Mónica Salinas, Mar Varela, Elia Grau, Napoleón de la Ossa Merlano, Esther Darder, Ares Solanes, Angela Velasco, Teresa Ramón y Cajal, Silvia Iglesias, Sara González, Jesús del Valle, Marta Pineda, Noemí Tuset, Gemma Llort, Judith Balmaña, José Marcos Moreno-Cabrera, Barbara Rivera, Carolina Gómez, Matilde Navarro, Gabriel Capellá, Xavier Matias-Guiu, Joan Brunet, Paula Rofes, and Nuria Dueñas
- Subjects
Adult ,Male ,Genes, APC ,Somatic cell ,Colorectal cancer ,Adenomatous Polyposis Coli Protein ,Germline ,Pathology and Forensic Medicine ,Familial adenomatous polyposis ,Cohort Studies ,Young Adult ,medicine ,Humans ,Blood test ,Genetic Predisposition to Disease ,Genetic Testing ,Gene ,Germ-Line Mutation ,Aged ,medicine.diagnostic_test ,Mosaicism ,business.industry ,High-Throughput Nucleotide Sequencing ,Middle Aged ,medicine.disease ,Phenotype ,digestive system diseases ,Adenomatous Polyposis Coli ,Cancer research ,Etiology ,Molecular Medicine ,Female ,Colorectal Neoplasms ,business - Abstract
Familial adenomatous polyposis (FAP) is an autosomal dominant syndrome responsible for 1% of colorectal cancers (CRCs). Up to 90% of classic FAPs are caused by inactivating mutations in APC, and mosaicism has been previously reported in 20% of de novo cases, usually linked to milder phenotypic manifestations. This study aimed to explore the prevalence of mosaicism in 11 unsolved cases of classic FAP and to evaluate the diagnostic yield of somatic testing. Paired samples of colorectal polyps, tumors, and/or mucosa were analyzed using a custom next-generation sequencing panel targeting 15 polyposis and CRC-predisposing genes. Whenever possible, the extension of mosaicism to blood or sperm was also examined. Of 11 patients with classic adenomatous polyposis, a mosaic pathogenic variant in APC was identified in 7 (64%). No other altered genes were identified. In two of seven patients (29%), mosaicism was found restricted to colonic tissues, whereas in five of seven patients (71%), it was extended to the blood. Germline affectation was confirmed in one patient. We report the first analysis at a somatic level of 15 genes associated with CRC susceptibility, which highlights the role of APC mosaicism in classic FAP etiology. The results further reinforce the importance of testing target tissues when blood test results are negative.
- Published
- 2021
41. Highly Sensitive Microsatellite Instability and Immunohistochemistry Assessment in Endometrial Aspirates as a Tool for Cancer Risk Individualization in Lynch Syndrome
- Author
-
Júlia Canet-Hermida, Fátima Marín, Eduard Dorca, Núria Dueñas, Laura Costas, Mònica Salinas, Àngela Velasco, Paula Peremiquel-Trillas, Sònia Paytubi, Jordi Ponce, Sergi Fernández, José Manuel Martínez, Laura Cárdenas, Anna Taltavull, Laia Alemany, Cristina Meléndez, Glòria Oliveras, August Vidal, Gabriel Capellá, Eugeni López-Bonet, Joan Brunet, Xavier Matias-Guiu, and Marta Pineda
- Subjects
Pathology and Forensic Medicine - Published
- 2023
42. Solving the enigma of POLD1 p.V295M as a potential cause of increased cancer risk
- Author
-
Rebeca Sanz-Pamplona, Conxi Lázaro, Tirso Pons, Sandra García-Mulero, Laura Valle, Jesús del Valle, Elsa Ezquerro, Lorena Magraner-Pardo, Pilar Mur, Gabriel Capellá, Victor Moreno, and Anna Díez-Villanueva
- Subjects
Adenoma ,Genetics ,Exonuclease ,POLD1 ,biology ,Cancer ,DNA Polymerase II ,Brief Communication ,medicine.disease ,Germline ,Frameshift mutation ,biology.protein ,medicine ,Humans ,Missense mutation ,Proofreading ,Colorectal Neoplasms ,Poly-ADP-Ribose Binding Proteins ,Germ-Line Mutation ,Genetics (clinical) ,Polymerase ,DNA Polymerase III - Abstract
Germline variants that affect the proofreading activity of polymerases epsilon (POLE) and delta (POLD1) predispose to colorectal adenomas and carcinomas, among other cancers. All cancer-associated pathogenic variants reported to date consist of non-disruptive genetic changes affecting the sequence that codifies the exonuclease domain (ED). Generally, disruptive (frameshift, stop-gain) POLE and POLD1 variants and missense variants outside the ED do not predispose to cancer. However, this statement may not be true for some, very specific variants that would indirectly affect the proofreading activity of the corresponding polymerase. We evaluated, by using multiple approaches, the possibility that POLD1 c.883G>A; p.(Val295Met), -a variant located 9 amino acids upstream the ED and present in ~0.25% of hereditary cancer patients-, affects POLD1 proofreading activity. Our findings show cumulative evidence that support no alteration of the proofreading activity and lack of association with cancer. The variant is classified as likely benign according to the ACMG/AMP guidelines.
- Published
- 2021
43. Clonal relationship and directionality of progression of synchronous endometrial and ovarian carcinomas in patients with DNA mismatch repair-deficiency associated syndromes
- Author
-
Fresia Pareja, Arnaud Da Cruz Paula, Karen Cadoo, Timothy Hoang, Nadeem R. Abu-Rustum, Lora H. Ellenson, Gabriel Capellá, August Vidal Bel, Lea A. Moukarzel, Xavier Matias-Guiu, Lorenzo Ferrando, Kay J. Park, Jorge S. Reis-Filho, Ana Paula Martins Sebastiao, Achim A. Jungbluth, Britta Weigelt, Marta Pineda, Jeffrey Levin, and Zsofia K. Stadler
- Subjects
Adult ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Somatic cell ,clonality ,Ovary ,Endometrium ,DNA Mismatch Repair ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Neoplastic Syndromes, Hereditary ,medicine ,Sequencing ,Humans ,PTEN ,Ovarian Neoplasms ,Synchronous endometrial and ovarian cancer ,Massive parallel sequencing ,biology ,Brain Neoplasms ,sequencing ,Syndrome ,synchronous endometrial and ovarian cancer ,Middle Aged ,medicine.disease ,Primary tumor ,Lynch syndrome ,Endometrial Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Disease Progression ,Cancer research ,biology.protein ,Female ,DNA mismatch repair ,Colorectal Neoplasms ,Clonality - Abstract
Sporadic synchronous endometrial (ECs) and ovarian cancers (OCs), although clinically considered to be independent primaries, have been shown to be clonally related and likely constitute metastases from each other. We sought to define whether synchronous ECs/OCs in patients with DNA mismatch repair (MMR)-deficiency syndromes would be clonally-related. We subjected synchronous ECs/OCs from four patients (LS3-LS6) with clinically confirmed Lynch syndrome (LS) and one patient with constitutional mismatch repair deficiency syndrome (CMMRD) to massively parallel sequencing targeting 468 cancer-related genes. Somatic mutations, copy number alterations (CNAs), clonal relatedness and clonal decomposition analyses were performed using previously described bioinformatics methods. All synchronous ECs/OCs analyzed were considered independent primaries based on clinicopathologic criteria. Sequencing analysis revealed that the ECs/OCs of three cases (LS2-CMMRD, L3, L4) harbored similar repertoires of somatic mutations and CNAs and were clonally-related. In these three cases, a subset of subclonal mutations in the EC became clonal in the OC, suggesting that the EC was likely the substrate from which the OC developed. LS5’s EC/OC had distinct mutational profiles but shared specific CNAs. In contrast, LS6’s EC/OC harbored distinct somatic mutation and lacked CNAs, consistent with each tumor constituting an independent primary lesion. In LS5 and LS6, PTEN mutations and PTEN loss of protein expression were found to be restricted to the EC. Finally, re-analysis of sequencing data of sporadic synchronous ECs/OCs supported the observations made in the current study that the directionality of progression is likely from the endometrium to the ovary. In conclusion, contrary to sporadic synchronous ECs/OCs, which are almost invariably clonally-related, ECs/OCs simultaneously involving the uterus and ovary in LS patients may represent distinct primary tumors. A subset of MMR-deficiency syndrome-related synchronous ECs/OCs, however, may originate from a single primary tumor at variance with their clinical diagnosis, with the endometrium being the likeliest site of origin. JSR-F was funded in part by a Breast Cancer Research Foundation grant, BW in part by Breast Cancer Research Foundation, Cycle for Survival and Stand Up To Cancer grants, F.P. partially by a National Institutes of Health/ National Cancer Institute K12 CA184746 grant, XM-G by AECC (grupos estables) and GC and MP by the Spanish Ministry of Economy and Competitiveness and co-funded by FEDER funds -a way to build Europe- (grant SAF2015-68016-R), CIBERONC and the Government of Catalonia (grant 2017SGR1282). XM-G, GC and MP thank CERCA Programme for institutional support. Research reported in this publication was supported in part by a Cancer Center Support Grant of the NIH/NCI (Grant No. P30CA008748).
- Published
- 2021
44. Position statement of the International Society for Gastrointestinal Hereditary Tumours (InSiGHT) onAPCI1307K and cancer risk
- Author
-
Laura Valle, Lior H Katz, Andrew Latchford, Pilar Mur, Victor Moreno, Ian M Frayling, Brandie Heald, and Gabriel Capellá
- Subjects
Genetics ,Genetics (clinical) - Abstract
While constitutional pathogenic variants in theAPCgene cause familial adenomatous polyposis,APCc.3920T>A; p.Ile1307Lys (I1307K) has been associated with a moderate increased risk of colorectal cancer (CRC), particularly in individuals of Ashkenazi Jewish descent. However, published data include relatively small sample sizes, generating inconclusive results regarding cancer risk, particularly in non-Ashkenazi populations. This has led to different country/continental-specific guidelines regarding genetic testing, clinical management and surveillance recommendations for I1307K. A multidisciplinary international expert group endorsed by the International Society for Gastrointestinal Hereditary Tumours (InSiGHT), has generated a position statement on theAPCI1307K allele and its association with cancer predisposition. Based on a systematic review and meta-analysis of the evidence published, the aim of this document is to summarise the prevalence of theAPCI1307K allele and analysed the evidence of the associated cancer risk in different populations. Here we provide recommendations on the laboratory classification of the variant, define the role of predictive testing for I1307K, suggest recommendations for cancer screening in I1307K heterozygous and homozygous individuals and identify knowledge gaps to be addressed in future research studies. Briefly, I1307K, classified as pathogenic, low penetrance, is a risk factor for CRC in individuals of Ashkenazi Jewish origin and should be tested in this population, offering carriers specific clinical surveillance. There is not enough evidence to support an increased risk of cancer in other populations/subpopulations. Therefore, until/unless future evidence indicates otherwise, individuals of non-Ashkenazi Jewish descent harbouring I1307K should be enrolled in national CRC screening programmes for average-risk individuals.
- Published
- 2023
45. Role of POLE and POLD1 in familial cancer
- Author
-
Pau M. Munoz-Torres, Joan Brunet, Rebeca Sanz-Pamplona, August Vidal, Gemma Llort, Esther Darder, Victor Moreno, Teresa Ramón y Cajal, Laura Valle, Judith Balmaña, Marta Pineda, Tirso Pons, Xavier Matias-Guiu, Jesús del Valle, Lorena Magraner-Pardo, Rosa Aligué, Pilar Mur, Giacomo Cinnirella, Josep M. Piulats, Elia Grau, Lídia Feliubadaló, Sami Belhadj, Adriana Lopez-Doriga, Matilde Navarro, Conxi Lázaro, Sandra García-Mulero, Judit Sanz, Gabriel Capellá, Edgar Martin-Ramos, [Mur P, Del Valle J, Pineda M] Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain. Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain. Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain. [García-Mulero S] Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain. Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain. [Magraner-Pardo L] Prostate Cancer Clinical Research Unit. Spanish National Cancer Research Center (CNIO), Madrid, Spain. [Vidal A] Department of Pathology, Bellvitge University Hospital, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain. [Balmana J] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, Vall d'Hebron Barcelona Hospital Campus, Ministerio de Economía y Competitividad (España), Instituto de Salud Carlos III, Generalitat de Catalunya, Fundación Olga Torres, and European Cooperation in Science and Technology
- Subjects
fenómenos genéticos::variación genética::mutación::mutación de la línea germinal [FENÓMENOS Y PROCESOS] ,Proband ,Polymerase proofreading–associated polyposis ,Recte - Càncer - Aspectes genètics ,Colorectal cancer ,Genetic counseling ,Population ,Còlon - Càncer - Aspectes genètics ,Biology ,Article ,Genetic Phenomena::Genetic Variation::Mutation::Germ-Line Mutation [PHENOMENA AND PROCESSES] ,Germline ,PPAP ,Endometrial cancer ,Càncer colorectal ,Malalties hereditàries ,Ultramutated phenotype ,medicine ,Humans ,Missense mutation ,Poly-ADP-Ribose Binding Proteins ,education ,Allele frequency ,Germ-Line Mutation ,Genetics (clinical) ,Exonuclease domain ,DNA Polymerase III ,Genetics ,education.field_of_study ,Malalties transmissibles - Teoria germinal ,POLD1 ,ultramutated phenotype ,DNA Polymerase II ,medicine.disease ,polymerase proofreading–associated polyposis ,Càncer d'endometri ,Neoplasms::Neoplasms::Neoplastic Syndromes, Hereditary::Colorectal Neoplasms, Hereditary Nonpolyposis [DISEASES] ,Hereditary colorectal cancer ,Mutation ,hereditary colorectal cancer ,neoplasias::neoplasias::síndromes neoplásicos hereditarios::neoplasias colorrectales hereditarias sin poliposis [ENFERMEDADES] ,exonuclease domain ,Colorectal Neoplasms ,Genetic diseases - Abstract
[Purpose]: Germline pathogenic variants in the exonuclease domain (ED) of polymerases POLE and POLD1 predispose to adenomatous polyps, colorectal cancer (CRC), endometrial tumors, and other malignancies, and exhibit increased mutation rate and highly specific associated mutational signatures. The tumor spectrum and prevalence of POLE and POLD1 variants in hereditary cancer are evaluated in this study. [Methods]: POLE and POLD1 were sequenced in 2813 unrelated probands referred for genetic counseling (2309 hereditary cancer patients subjected to a multigene panel, and 504 patients selected based on phenotypic characteristics). Cosegregation and case–control studies, yeast-based functional assays, and tumor mutational analyses were performed for variant interpretation. [Results]: Twelve ED missense variants, 6 loss-of-function, and 23 outside-ED predicted-deleterious missense variants, all with population allele frequencies, This work was funded by the Spanish Ministry of Science and Innovation, cofunded by FEDER funds (SAF2016-80888-R, SAF2015-68016-R, Severo Ochoa SVP-2014-068895 contract [L.M.-P.]); Instituto de Salud Carlos III (PI16/00563, PI16/00588, PI14/00613, PI19/00553, CIBERONC CB16/12/00234, CIBERESP, Sara Borrell contract [P.M.]); Government of Catalonia [AGAUR 2017SGR1282, CERCA Program]; and Fundación Olga Torres. This study was facilitated by COST Action CA17118, supported by COST (European Cooperation in Science and Technology).
- Published
- 2020
46. Improving Genetic Testing in Hereditary Cancer by RNA Analysis
- Author
-
Conxi Lázaro, Mireia Menéndez, Eva Tornero, Marta Pineda, Jesús del Valle, Lídia Feliubadaló, Paula Rofes, Mónica Salinas, Gardenia Vargas-Parra, Alex Teulé, Joan Brunet, Eva Montes, Carolina Gómez, Sara González, Gabriel Capellá, and Ares Solanes
- Subjects
0301 basic medicine ,Sanger sequencing ,medicine.medical_specialty ,medicine.diagnostic_test ,Genetic counseling ,Genomics ,Computational biology ,Biology ,Pathology and Forensic Medicine ,03 medical and health sciences ,Exon ,symbols.namesake ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,RNA splicing ,medicine ,symbols ,Molecular Medicine ,Medical genetics ,Gene ,Genetic testing - Abstract
RNA analyses are a potent tool to identify spliceogenic effects of DNA variants, although they are time-consuming and cannot always be performed. We present splicing assays of 20 variants that represent a variety of mutation types in 10 hereditary cancer genes and attempt to incorporate these results into American College of Medical Genetics and Genomics (ACMG) classification guidelines. Sixteen single-nucleotide variants, 3 exon duplications, and 1 single-exon deletion were selected and prioritized by in silico algorithms. RNA was extracted from short-term lymphocyte cultures to perform RT-PCR and Sanger sequencing, and allele-specific expression was assessed whenever possible. Aberrant transcripts were detected in 14 variants (70%). Variant interpretation was difficult, especially comparing old classification standards to generic ACMG guidelines and a proposal was devised to weigh functional analyses at RNA level. According to the ACMG guidelines, only 12 variants were reclassified as pathogenic/likely pathogenic because the other two variants did not gather enough evidence. This study highlights the importance of RNA studies to improve variant classification. However, it also indicates the challenge of incorporating these results into generic ACMG guidelines and the need to refine these criteria gene specifically. Nevertheless, 60% of variants were reclassified, thus improving genetic counseling and surveillance for carriers of these variants.
- Published
- 2020
47. Lessons learnt from the implementation of a colorectal cancer screening programme for lynch syndrome in a tertiary public hospital
- Author
-
Nuria Dueñas, Matilde Navarro, Xavier Sanjuán, Núria Ruiz, Silvia Iglesias, Xavier Matias-Guiu, Jordi Guardiola, Esther Kreisler, Sebastiano Biondo, Sara González, Raquel Legido, Ana Blanco, Silvia Navarro, Leyre Asiain, Cristina Santos, Gabriel Capellá, Marta Pineda, and Joan Brunet
- Subjects
Cancer Research ,Oncology ,Epidemiology - Abstract
Lynch syndrome (LS) is the first cause of inherited colorectal cancer (CRC), being responsible for 2-4% of all diagnoses. Identification of affected individuals is important as they have an increased lifetime risk of multiple CRC and other neoplasms, however, LS is consistently underdiagnosed at the population level. We aimed to evaluate the yield of LS screening in CRC in a single-referral centre and to identify the barriers to its effective implementation.LS screening programme included individuals with CRC 70 years, multiple CRC, or endometrial cancer at any age. Mismatch repair (MMR) protein immunohistochemistry (IHC) analysis was performed in routine practice on the surgical specimen and, if MLH1 IHC was altered, MLH1 gene promoter methylation was analysed. Results were collected in the CRC multidisciplinary board database. LS suspected individuals (altered MMR IHC without MLH1 promoter methylation) were referred to the Cancer Genetic Counselling Unit (CGCU). If accepted, a genetic study was performed. Two checkpoints were included: review of the pathology data and verification of patient referral by a genetic counsellor.Between 2016 and 2019, 381 individuals were included. MMR IHC analysis was performed in 374/381 (98.2 %) CRC cases and MLH1 promoter methylation in 18/21 (85.7 %). Seventeen of the 20 LS suspected individuals were invited for referral at the CGCU. Two cases were not invited and the remaining patient died of cancer before completion of tumour screening. Fifteen individuals attended and a genetic analysis was performed in 15/20 (75 %) LS suspected individuals. Ten individuals were diagnosed with LS, in concordance with the IHC profile (2.7 % of the total cohort). This led to cascade testing in 58/75 (77.3 %) of the available adult relatives at risk, identifying 26 individuals with LS.Establishing a standardized institutional LS screening programme with checkpoints in the workflow is key to increasing the yield of LS identification.
- Published
- 2022
48. Characteristics of Adrenocortical Carcinoma Associated With Lynch Syndrome
- Author
-
Marta Domenech, Cristina Carrato, Conxi Lázaro, Gabriel Capellá, Angel Izquierdo, Jesús del Valle, Joan Brunet, Matilde Navarro, Marta Pineda, Nuria Dueñas, Ares Solanes, Magda Pujol, and Elia Grau
- Subjects
Oncology ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Biochemistry ,Loss of heterozygosity ,Endocrinology ,Germline mutation ,Internal medicine ,Adrenocortical Carcinoma ,Carcinoma ,Humans ,Medicine ,Adrenocortical carcinoma ,business.industry ,Endometrial cancer ,Biochemistry (medical) ,Prognosis ,medicine.disease ,Colorectal Neoplasms, Hereditary Nonpolyposis ,Adrenal Cortex Neoplasms ,Lynch syndrome ,MSH6 ,MSH2 ,business - Abstract
Context Lynch syndrome (LS) is the most common inherited colorectal and endometrial cancer syndrome, caused by germline mutations in DNA mismatch repair (MMR) genes. It is also characterized by an increased risk of other tumors with lower prevalence, such as adrenal cortical carcinoma (ACC), an endocrine tumor with an incidence of Evidence Acquisition The objective of this study is to determine the prevalence of ACC in a Spanish LS cohort and their molecular and histological characteristics. This retrospective study includes 634 patients from 220 LS families registered between 1999 and 2018. Evidence Synthesis During the follow-up period, 3 patients were diagnosed with ACC (0.47%); all were carriers of a MSH2 germline mutation. The 3 ACC patients presented loss of expression of MSH2 and MSH6 proteins. One tumor analysis showed loss of heterozygosity of the MSH2 wildtype allele. Our findings support previous data that considered ACC as a LS spectrum tumor. Conclusion MMR protein immunohistochemistry screening could be an efficient strategy to detect LS in patients with ACC.
- Published
- 2020
49. TP53, a gene for colorectal cancer predisposition in the absence of Li-Fraumeni-associated phenotypes
- Author
-
Emma R. Woodward, Conxi Lázaro, D. Gareth Evans, Pau M. Munoz-Torres, George J Burghel, Gabriel Capellá, Mariona Terradas, Matilde Navarro, Marta Pineda, Isabel Quintana, Laura Valle, Victor Moreno, Pilar Mur, Sami Belhadj, and Joan Brunet
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,education.field_of_study ,medicine.diagnostic_test ,Molecular pathology ,business.industry ,Colorectal cancer ,Population ,Gastroenterology ,Cancer ,Gene mutation ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Medical genetics ,Missense mutation ,business ,education ,Genetic testing - Abstract
ObjectiveGermline TP53 pathogenic (P) variants cause Li-Fraumeni syndrome (LFS), an aggressive multitumor-predisposing condition. Due to the implementation of multigene panel testing, TP53 variants have been detected in individuals without LFS suspicion, for example, patients with colorectal cancer (CRC). We aimed to decipher whether these findings are the result of detecting the background population prevalence or the aetiological basis of CRC.DesignWe analysed TP53 in 473 familial/early-onset CRC cases and evaluated the results together with five additional studies performed in patients with CRC (total n=6200). Control population and LFS data were obtained from Genome Aggregation Database (gnomAD V.2.1.1) and the International Agency for Research on Cancer (IARC) TP53 database, respectively. All variants were reclassified according to the guidelines of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP), following the ClinGen TP53 Expert Panel specifications.ResultsP or likely pathogenic (LP) variants were identified in 0.05% of controls (n=27/59 095) and 0.26% of patients with CRC (n=16/6200) (pTP53 testing. This association was still detected when patients with CRC diagnosed at more advanced ages (>50 and>60 years) were excluded from the analysis to minimise the inclusion of variants caused by clonal haematopoiesis. Loss-of-function and missense variants were strongly associated with CRC as compared with controls (OR=25.44, 95% CI 6.10 to 149.03, for loss of function and splice-site alleles, and OR=3.58, 95% CI 1.46 to 7.98, for missense P or LP variants).ConclusionTP53 P variants should not be unequivocally associated with LFS. Prospective follow-up of carriers of germline TP53 P variants in the absence of LFS phenotypes will define how surveillance and clinical management of these individuals should be performed.
- Published
- 2020
50. Candidate genes for hereditary colorectal cancer: Mutational screening and systematic review
- Author
-
Gabriel Capellá, Laura Valle, Matilde Navarro, Mariona Terradas, Gemma Aiza, Sami Belhadj, and Pau M. Munoz-Torres
- Subjects
Adult ,Candidate gene ,Adolescent ,Colorectal cancer ,DNA Mutational Analysis ,Biology ,Cohort Studies ,Young Adult ,03 medical and health sciences ,Germline mutation ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,Promoter Regions, Genetic ,Allele frequency ,Interleukin 12 receptor, beta 1 subunit ,Gene ,Early Detection of Cancer ,Genetics (clinical) ,Aged ,030304 developmental biology ,Aged, 80 and over ,0303 health sciences ,FAN1 ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,030305 genetics & heredity ,DNA Methylation ,Middle Aged ,medicine.disease ,Colorectal Neoplasms - Abstract
Genome-wide approaches applied for the identification of new hereditary colorectal cancer (CRC) genes, identified several potential causal genes, including RPS20, IL12RB1, LIMK2, POLE2, MRE11, POT1, FAN1, WIF1, HNRNPA0, SEMA4A, FOCAD, PTPN12, LRP6, POLQ, BLM, MCM9, and the epigenetic inactivation of PTPRJ. Here we attempted to validate the association between variants in these genes and nonpolyposis CRC by performing a mutational screening of the genes and PTPRJ promoter methylation analysis in 473 familial/early-onset CRC cases, a systematic review of the published cases, and assessment of allele frequencies in control population. In the studied cohort, 24 (5%) carriers of (predicted) deleterious variants in the studied genes and no constitutional PTPRJ epimutations were identified. Assessment of allele frequencies in controls compared with familial/early-onset patients with CRC showed association with increased nonpolyposis CRC risk of disruptive variants in RPS20, IL12RB1, POLE2, MRE11 and POT1, and of FAN1 c.149T>G (p.Met50Arg). Lack of association was demonstrated for LIMK2, PTPN12, LRP6, PTPRJ, POLQ, BLM, MCM9 and FOCAD variants. Additional studies are required to provide conclusive evidence for SEMA4A, WIF1, HNRNPA0 c.-110G>C, and FOCAD large deletions.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.