46 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. 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
7. 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
8. 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
9. 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
10. 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
11. 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
12. 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
13. 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
14. 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
15. 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
16. 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
17. 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
18. 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
19. 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
20. 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
21. 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
22. 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
23. 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
24. 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
25. 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
26. 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
27. 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
28. 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
29. 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
30. 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
31. 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
32. 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
33. 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
34. 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
35. 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
36. 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
37. Potential Involvement of NSD1, KRT24 and ACACA in the Genetic Predisposition to Colorectal Cancer
- Author
-
Isabel Quintana, Pilar Mur, Mariona Terradas, Sandra García-Mulero, Gemma Aiza, Matilde Navarro, Virginia Piñol, Joan Brunet, Victor Moreno, Rebeca Sanz-Pamplona, Gabriel Capellá, and Laura Valle
- Subjects
Cancer Research ,Cribratge genètic ,cancer predisposition ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,polyposis ,somatic second hit ,Colorectal cancer ,Oncology ,hereditary cancer ,Càncer colorectal ,Genetic screening ,hereditary colorectal cancer ,Malalties hereditàries ,RC254-282 ,Genetic diseases - Abstract
The ALFRED (Allelic Loss Featuring Rare Damaging) in silico method was developed to identify cancer predisposition genes through the identification of somatic second hits. By applying ALFRED to ~10,000 tumor exomes, 49 candidate genes were identified. We aimed to assess the causal association of the identified genes with colorectal cancer (CRC) predisposition. Of the 49 genes, NSD1, HDAC10, KRT24, ACACA and TP63 were selected based on specific criteria relevant for hereditary CRC genes. Gene sequencing was performed in 736 patients with familial/early onset CRC or polyposis without germline pathogenic variants in known genes. Twelve (predicted) damaging variants in 18 patients were identified. A gene-based burden test in 1596 familial/early-onset CRC patients, 271 polyposis patients, 543 TCGA CRC patients and >134,000 controls (gnomAD, non-cancer), revealed no clear association with CRC for any of the studied genes. Nevertheless, (non-significant) over-representation of disruptive variants in NSD1, KRT24 and ACACA in CRC patients compared to controls was observed. A somatic second hit was identified in one of 20 tumors tested, corresponding to an NSD1 carrier. In conclusion, most genes identified through the ALFRED in silico method were not relevant for CRC predisposition, although a possible association was detected for NSD1, KRT24 and ACACA.
- Published
- 2022
38. Potential Involvement of
- Author
-
Isabel, Quintana, Pilar, Mur, Mariona, Terradas, Sandra, García-Mulero, Gemma, Aiza, Matilde, Navarro, Virginia, Piñol, Joan, Brunet, Victor, Moreno, Rebeca, Sanz-Pamplona, Gabriel, Capellá, and Laura, Valle
- Abstract
The ALFRED (Allelic Loss Featuring Rare Damaging) in silico method was developed to identify cancer predisposition genes through the identification of somatic second hits. By applying ALFRED to ~10,000 tumor exomes, 49 candidate genes were identified. We aimed to assess the causal association of the identified genes with colorectal cancer (CRC) predisposition. Of the 49 genes
- Published
- 2022
39. Quality of Colonoscopy Is Associated With Adenoma Detection and Postcolonoscopy Colorectal Cancer Prevention in Lynch Syndrome
- Author
-
Daniel Rodríguez-Alcalde, Angeles Pizarro, Maria Pellise, Cristina Romero, Berta Caballol, Lorena Moreno, Esteban Saperas, Eva Martinez de Castro, Gemma Llort, Laura Rivas, Catalina Garau, Ariadna Sánchez, Elena Aguirre, Sabela Carballal, Virginia Piñol, Eva Barreiro-Alonso, Matilde Navarro, Lorena Rodríguez-Alonso, Marta Garzon, Xavier Bessa, Teresa Ocaña, Marta Ponce, Carmen Poves, Inmaculada Salces, Gerhard Jung, Cristina Alvarez-Urrutia, Maite Herraiz, Liseth Rivero-Sánchez, Evelien Dekker, Joan Brunet, Judith Balmaña, Maribel Gonzalez-Acosta, Francesc Balaguer, Blai Morales-Romero, Luis Bujanda, Leticia Moreira, Joaquín Cubiella, Teresa Ramón y Cajal, Marta Pineda, Miquel Serra-Burriel, Victorine H. Roos, María Dolores Picó, Andres Dacal, Antoni Castells, Lucía Cid, Maria Jesus López-Arias, Rodrigo Jover, Francisco Rodríguez-Moranta, Gabriel Capellá, Marta Carrillo-Palau, Barbara A. J. Bastiaansen, Adrià López-Fernández, Gastroenterology and Hepatology, Graduate School, APH - Methodology, APH - Personalized Medicine, CCA - Imaging and biomarkers, CCA - Cancer Treatment and Quality of Life, and AGEM - Amsterdam Gastroenterology Endocrinology Metabolism
- Subjects
Adenoma ,medicine.medical_specialty ,Colorectal cancer ,Colonoscopy ,HNPCC ,Chromoendoscopy ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Interquartile range ,Internal medicine ,medicine ,Humans ,Cumulative incidence ,Colonoscopy Quality ,Colorectal Neoplasms, HNPCC, Hereditary Non-Polyposis Colorectal Cancer, Lynch syndrome, colonoscopy quality, colonoscopy, hereditary colorectal cancer ,neoplasms ,Early Detection of Cancer ,Hereditary Colorectal Cancer ,Hepatology ,medicine.diagnostic_test ,business.industry ,Incidence ,Incidence (epidemiology) ,Gastroenterology ,Hereditary Nonpolyposis Colorectal Cancer ,Odds ratio ,colonoscopy quality, colonoscopy ,medicine.disease ,Colorectal Neoplasms, Hereditary Nonpolyposis ,Lynch syndrome ,digestive system diseases ,Hereditary Non-Polyposis Colorectal Cancer ,030220 oncology & carcinogenesis ,Lynch Syndrome ,030211 gastroenterology & hepatology ,business ,Colorectal Neoplasms - Abstract
BACKGROUND & AIMS: Colonoscopy reduces colorectal cancer (CRC) incidence and mortality in Lynch syndrome (LS) carriers. However, a high incidence of postcolonoscopy CRC (PCCRC) has been reported. Colonoscopy is highly dependent on endoscopist skill and is subject to quality variability. We aimed to evaluate the impact of key colonoscopy quality indicators on adenoma detection and prevention of PCCRC in LS. METHODS: We conducted a multicenter study focused on LS carriers without previous CRC undergoing colonoscopy surveillance (n = 893). Incident colorectal neoplasia during surveillance and quality indicators of all colonoscopies were analyzed. We performed an emulated target trial comparing the results from the first and second surveillance colonoscopies to assess the effect of colonoscopy quality indicators on adenoma detection and PCCRC incidence. Risk analyses were conducted using a multivariable logistic regression model. RESULTS: The 10-year cumulative incidence of adenoma and PCCRC was 60.6% (95% CI, 55.5%-65.2%) and 7.9% (95% CI, 5.2%-10.6%), respectively. Adequate bowel preparation (odds ratio [OR], 2.07; 95% CI, 1.06-4.3), complete colonoscopies (20% vs 0%; P = .01), and panchromoendoscopy use (OR, 2.14; 95% CI, 1.15-3.95) were associated with significant improvement in adenoma detection. PCCRC risk was significantly lower when colonoscopies were performed during a time interval of less than every 3 years (OR, 0.35; 95% CI, 0.14-0.97). We observed a consistent but not significant reduction in PCCRC risk for a previous complete examination (OR, 0.16; 95% CI, 0.03-1.28), adequate bowel preparation (OR, 0.64; 95% CI, 0.17-3.24), or previous use of high-definition colonoscopy (OR, 0.37; 95% CI, 0.02-2.33). CONCLUSIONS: Complete colonoscopies with adequate bowel preparation and chromoendoscopy use are associated with improved adenoma detection, while surveillance intervals of less than 3 years are associated with a reduction of PCCRC incidence. In LS, high-quality colonoscopy surveillance is of utmost importance for CRC prevention.
- Published
- 2022
40. RNA assay identifies a previous misclassification of BARD1 c.1977A>G variant
- Author
-
Carolina Gómez, Joan Brunet, Gabriel Capellá, Eva Montes, Mireia Menéndez, Conxi Lázaro, Marta Pineda, Jesús del Valle, Rafael de Cid, Lídia Feliubadaló, and Paula Rofes
- Subjects
Gene isoform ,Heredity ,Science ,Ubiquitin-Protein Ligases ,Càncer d'ovari ,Breast Neoplasms ,Biology ,Polymerase Chain Reaction ,Germline ,Article ,Càncer de mama ,Frameshift mutation ,Exon ,Breast cancer ,Human genetics ,Ovarian cancer ,Risk Factors ,BARD1 ,Biomarkers, Tumor ,Humans ,Genetic Predisposition to Disease ,Cancer genetics ,Genetics ,Gynaecological cancer ,Ovarian Neoplasms ,Genètica humana ,Multidisciplinary ,Molecular medicine ,Tumor Suppressor Proteins ,RNA ,Genetic Variation ,Alternative Splicing ,Case-Control Studies ,RNA splicing ,Allelic Imbalance ,Medicine ,Female - 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
41. Incidences of colorectal adenomas and cancers under colonoscopy surveillance suggest an accelerated 'Big Bang' pathway to CRC in three of the four Lynch syndromes
- Author
-
Pål Møller, Saskia Haupt, Aysel Ahadova, Matthias Kloor, Julian R. Sampson, Lone Sunde, Toni Seppälä, John Burn, Inge Bernstein, Gabriel Capella, D. Gareth Evans, Annika Lindblom, Ingrid Winship, Finlay Macrae, Lior Katz, Ido Laish, Elez Vainer, Kevin Monahan, Elizabeth Half, Karoline Horisberger, Leandro Apolinário da Silva, Vincent Heuveline, Christina Therkildsen, Charlotte Lautrup, Louise L Klarskov, Giulia Martina Cavestro, Gabriela Möslein, Eivind Hovig, and Mev Dominguez-Valentin
- Subjects
MSI ,MLH1 ,MSH2 ,MSH6 ,PMS2 ,dMMR ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Genetics ,QH426-470 - Abstract
Abstract Background Colorectal cancers (CRCs) in the Lynch syndromes have been assumed to emerge through an accelerated adenoma-carcinoma pathway. In this model adenomas with deficient mismatch repair have an increased probability of acquiring additional cancer driver mutation(s) resulting in more rapid progression to malignancy. If this model was accurate, the success of colonoscopy in preventing CRC would be a function of the intervals between colonoscopies and mean sojourn time of detectable adenomas. Contrary to expectations, colonoscopy did not decrease incidence of CRC in the Lynch syndromes and shorter colonoscopy intervals have not been effective in reducing CRC incidence. The prospective Lynch Syndrome Database (PLSD) was designed to examine these issues in carriers of pathogenic variants of the mis-match repair (path_MMR) genes. Materials and methods We examined the CRC and colorectal adenoma incidences in 3,574 path_MLH1, path_MSH2, path_MSH6 and path_PMS2 carriers subjected to regular colonoscopy with polypectomy, and considered the results based on sojourn times and stochastic probability paradigms. Results Most of the path_MMR carriers in each genetic group had no adenomas. There was no association between incidences of CRC and the presence of adenomas. There was no CRC observed in path_PMS2 carriers. Conclusions Colonoscopy prevented CRC in path_PMS2 carriers but not in the others. Our findings are consistent with colonoscopy surveillance blocking the adenoma-carcinoma pathway by removing identified adenomas which might otherwise become CRCs. However, in the other carriers most CRCs likely arised from dMMR cells in the crypts that have an increased mutation rate with increased stochastic chaotic probabilities for mutations. Therefore, this mechanism, that may be associated with no or only a short sojourn time of MSI tumours as adenomas, could explain the findings in our previous and current reports.
- Published
- 2024
- Full Text
- View/download PDF
42. Variants of uncertain significance (VUS) in cancer predisposing genes: What are we learning from multigene panels?
- Author
-
Maurizio Genuardi, Gabriel Capellá, Arianna Panfili, Marta Pineda, Simona Amenta, Emanuela Lucci-Cordisco, and Jesús del Valle
- Subjects
variants ,education.field_of_study ,medicine.medical_specialty ,medicine.diagnostic_test ,Cancer-Predisposing Gene ,Molecular pathology ,Population ,Genetic Variation ,General Medicine ,Computational biology ,Biology ,Settore MED/03 - GENETICA MEDICA ,Neoplasms ,Genetics ,medicine ,Humans ,Medical genetics ,Genetic Predisposition to Disease ,Identification (biology) ,education ,Gene ,Uncertain significance ,Genetics (clinical) ,Genetic testing - Abstract
One of the main factors influencing the clinical utility of genetic tests for cancer predisposition is the ability to provide actionable classifications (ie pathogenic or benign). However, a large fraction of the variants identified in cancer predisposing genes (CPGs) are of uncertain significance (VUS), and cannot be used for clinical purposes either to identify individuals at risk or to drive treatment. Here we analyze the current status of VUS identification in a subset of 24 CPGs included by the American College of Medical Genetics/Association for Molecular Pathology in the list of genes that should be considered for the return of incidental findings. To this purpose we retrieved published literature using different search strings according to the frequency of the condition and we extracted corresponding data from ClinVar. The total number of VUS has not decreased with time, due to widespread multigene panel testing, and the relative yield of VUS compared to pathogenic variants is higher in more recent studies, which tend to involve series not selected for the presence of specific high risk criteria. In addition, only few studies adopt gene specific interpretation criteria when these are available. Despite the large yield of VUS associated with multigene testing, the data obtained from such studies can be very useful for variant classification, especially for those variants that are more likely to be benign, since these are expected to be detected more frequently in a population that does not show gene specific manifestations. In addition, wider use of gene specific interpretation criteria should be promoted in order to optimize the interpretation process.
- Published
- 2022
43. Clinically relevant combined effect of polygenic background, rare pathogenic germline variants, and family history on colorectal cancer incidence
- Author
-
Emadeldin Hassanin, Isabel Spier, Dheeraj R. Bobbili, Rana Aldisi, Hannah Klinkhammer, Friederike David, Nuria Dueñas, Robert Hüneburg, Claudia Perne, Joan Brunet, Gabriel Capella, Markus M. Nöthen, Andreas J. Forstner, Andreas Mayr, Peter Krawitz, Patrick May, Stefan Aretz, and Carlo Maj
- Subjects
Colorectal cancer ,Family history ,Hereditary cancer ,Polygenic risk ,Risk stratification ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background and aims Summarised in polygenic risk scores (PRS), the effect of common, low penetrant genetic variants associated with colorectal cancer (CRC), can be used for risk stratification. Methods To assess the combined impact of the PRS and other main factors on CRC risk, 163,516 individuals from the UK Biobank were stratified as follows: 1. carriers status for germline pathogenic variants (PV) in CRC susceptibility genes (APC, MLH1, MSH2, MSH6, PMS2), 2. low ( 80%), and 3. family history (FH) of CRC. Multivariable logistic regression and Cox proportional hazards models were applied to compare odds ratios and to compute the lifetime incidence, respectively. Results Depending on the PRS, the CRC lifetime incidence for non-carriers ranges between 6 and 22%, compared to 40% and 74% for carriers. A suspicious FH is associated with a further increase of the cumulative incidence reaching 26% for non-carriers and 98% for carriers. In non-carriers without FH, but high PRS, the CRC risk is doubled, whereas a low PRS even in the context of a FH results in a decreased risk. The full model including PRS, carrier status, and FH improved the area under the curve in risk prediction (0.704). Conclusion The findings demonstrate that CRC risks are strongly influenced by the PRS for both a sporadic and monogenic background. FH, PV, and common variants complementary contribute to CRC risk. The implementation of PRS in routine care will likely improve personalized risk stratification, which will in turn guide tailored preventive surveillance strategies in high, intermediate, and low risk groups.
- Published
- 2023
- Full Text
- View/download PDF
44. Colorectal cancer incidences in Lynch syndrome: a comparison of results from the prospective lynch syndrome database and the international mismatch repair consortium
- Author
-
Pål Møller, Toni Seppälä, James G. Dowty, Saskia Haupt, Mev Dominguez-Valentin, Lone Sunde, Inge Bernstein, Christoph Engel, Stefan Aretz, Maartje Nielsen, Gabriel Capella, Dafydd Gareth Evans, John Burn, Elke Holinski-Feder, Lucio Bertario, Bernardo Bonanni, Annika Lindblom, Zohar Levi, Finlay Macrae, Ingrid Winship, John-Paul Plazzer, Rolf Sijmons, Luigi Laghi, Adriana Della Valle, Karl Heinimann, Elizabeth Half, Francisco Lopez-Koestner, Karin Alvarez-Valenzuela, Rodney J. Scott, Lior Katz, Ido Laish, Elez Vainer, Carlos Alberto Vaccaro, Dirce Maria Carraro, Nathan Gluck, Naim Abu-Freha, Aine Stakelum, Rory Kennelly, Des Winter, Benedito Mauro Rossi, Marc Greenblatt, Mabel Bohorquez, Harsh Sheth, Maria Grazia Tibiletti, Leonardo S. Lino-Silva, Karoline Horisberger, Carmen Portenkirchner, Ivana Nascimento, Norma Teresa Rossi, Leandro Apolinário da Silva, Huw Thomas, Attila Zaránd, Jukka-Pekka Mecklin, Kirsi Pylvänäinen, Laura Renkonen-Sinisalo, Anna Lepisto, Päivi Peltomäki, Christina Therkildsen, Lars Joachim Lindberg, Ole Thorlacius-Ussing, Magnus von Knebel Doeberitz, Markus Loeffler, Nils Rahner, Verena Steinke-Lange, Wolff Schmiegel, Deepak Vangala, Claudia Perne, Robert Hüneburg, Aída Falcón de Vargas, Andrew Latchford, Anne-Marie Gerdes, Ann-Sofie Backman, Carmen Guillén-Ponce, Carrie Snyder, Charlotte K. Lautrup, David Amor, Edenir Palmero, Elena Stoffel, Floor Duijkers, Michael J. Hall, Heather Hampel, Heinric Williams, Henrik Okkels, Jan Lubiński, Jeanette Reece, Joanne Ngeow, Jose G. Guillem, Julie Arnold, Karin Wadt, Kevin Monahan, Leigha Senter, Lene J. Rasmussen, Liselotte P. van Hest, Luigi Ricciardiello, Maija R. J. Kohonen-Corish, Marjolijn J. L. Ligtenberg, Melissa Southey, Melyssa Aronson, Mohd N. Zahary, N. Jewel Samadder, Nicola Poplawski, Nicoline Hoogerbrugge, Patrick J. Morrison, Paul James, Grant Lee, Rakefet Chen-Shtoyerman, Ravindran Ankathil, Rish Pai, Robyn Ward, Susan Parry, Tadeusz Dębniak, Thomas John, Thomas van Overeem Hansen, Trinidad Caldés, Tatsuro Yamaguchi, Verónica Barca-Tierno, Pilar Garre, Giulia Martina Cavestro, Jürgen Weitz, Silke Redler, Reinhard Büttner, Vincent Heuveline, John L. Hopper, Aung Ko Win, Noralane Lindor, Steven Gallinger, Loïc Le Marchand, Polly A. Newcomb, Jane Figueiredo, Daniel D. Buchanan, Stephen N. Thibodeau, Sanne W. ten Broeke, Eivind Hovig, Sigve Nakken, Marta Pineda, Nuria Dueñas, Joan Brunet, Kate Green, Fiona Lalloo, Katie Newton, Emma J. Crosbie, Miriam Mints, Douglas Tjandra, Florencia Neffa, Patricia Esperon, Revital Kariv, Guy Rosner, Walter Hernán Pavicic, Pablo Kalfayan, Giovana Tardin Torrezan, Thiago Bassaneze, Claudia Martin, Gabriela Moslein, Aysel Ahadova, Matthias Kloor, Julian R. Sampson, Mark A. Jenkins, and The European Hereditary Tumour Group (EHTG) and the International Mismatch Repair Consortium (IMRC)
- Subjects
Lynch Syndrome ,Epidemiology ,Prevention ,Penetrance ,Colorectal cancer ,Segregation analysis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Genetics ,QH426-470 - Abstract
Abstract Objective To compare colorectal cancer (CRC) incidences in carriers of pathogenic variants of the MMR genes in the PLSD and IMRC cohorts, of which only the former included mandatory colonoscopy surveillance for all participants. Methods CRC incidences were calculated in an intervention group comprising a cohort of confirmed carriers of pathogenic or likely pathogenic variants in mismatch repair genes (path_MMR) followed prospectively by the Prospective Lynch Syndrome Database (PLSD). All had colonoscopy surveillance, with polypectomy when polyps were identified. Comparison was made with a retrospective cohort reported by the International Mismatch Repair Consortium (IMRC). This comprised confirmed and inferred path_MMR carriers who were first- or second-degree relatives of Lynch syndrome probands. Results In the PLSD, 8,153 subjects had follow-up colonoscopy surveillance for a total of 67,604 years and 578 carriers had CRC diagnosed. Average cumulative incidences of CRC in path_MLH1 carriers at 70 years of age were 52% in males and 41% in females; for path_MSH2 50% and 39%; for path_MSH6 13% and 17% and for path_PMS2 11% and 8%. In contrast, in the IMRC cohort, corresponding cumulative incidences were 40% and 27%; 34% and 23%; 16% and 8% and 7% and 6%. Comparing just the European carriers in the two series gave similar findings. Numbers in the PLSD series did not allow comparisons of carriers from other continents separately. Cumulative incidences at 25 years were < 1% in all retrospective groups. Conclusions Prospectively observed CRC incidences (PLSD) in path_MLH1 and path_MSH2 carriers undergoing colonoscopy surveillance and polypectomy were higher than in the retrospective (IMRC) series, and were not reduced in path_MSH6 carriers. These findings were the opposite to those expected. CRC point incidence before 50 years of age was reduced in path_PMS2 carriers subjected to colonoscopy, but not significantly so.
- Published
- 2022
- Full Text
- View/download PDF
45. Clinical, genetic, epidemiologic, evolutionary, and functional delineation of TSPEAR-related autosomal recessive ectodermal dysplasia 14
- Author
-
Adam Jackson, Sheng-Jia Lin, Elizabeth A. Jones, Kate E. Chandler, David Orr, Celia Moss, Zahra Haider, Gavin Ryan, Simon Holden, Mike Harrison, Nigel Burrows, Wendy D. Jones, Mary Loveless, Cassidy Petree, Helen Stewart, Karen Low, Deirdre Donnelly, Simon Lovell, Konstantina Drosou, Gaurav K. Varshney, Siddharth Banka, J.C. Ambrose, P. Arumugam, R. Bevers, M. Bleda, F. Boardman-Pretty, C.R. Boustred, H. Brittain, M.A. Brown, M.J. Caulfield, G.C. Chan, A. Giess, J.N. Griffin, A. Hamblin, S. Henderson, T.J.P. Hubbard, R. Jackson, L.J. Jones, D. Kasperaviciute, M. Kayikci, A. Kousathanas, L. Lahnstein, A. Lakey, S.E.A. Leigh, I.U.S. Leong, F.J. Lopez, F. Maleady-Crowe, M. McEntagart, F. Minneci, J. Mitchell, L. Moutsianas, M. Mueller, N. Murugaesu, A.C. Need, P. O‘Donovan, C.A. Odhams, C. Patch, D. Perez-Gil, M.B. Pereira, J. Pullinger, T. Rahim, A. Rendon, T. Rogers, K. Savage, K. Sawant, R.H. Scott, A. Siddiq, A. Sieghart, S.C. Smith, A. Sosinsky, A. Stuckey, M. Tanguy, A.L. Taylor Tavares, E.R.A. Thomas, S.R. Thompson, A. Tucci, M.J. Welland, E. Williams, K. Witkowska, S.M. Wood, M. Zarowiecki, Olaf Riess, Tobias B. Haack, Holm Graessner, Birte Zurek, Kornelia Ellwanger, Stephan Ossowski, German Demidov, Marc Sturm, Julia M. Schulze-Hentrich, Rebecca Schüle, Christoph Kessler, Melanie Wayand, Matthis Synofzik, Carlo Wilke, Andreas Traschütz, Ludger Schöls, Holger Hengel, Peter Heutink, Han Brunner, Hans Scheffer, Nicoline Hoogerbrugge, Alexander Hoischen, Peter A.C. ’t Hoen, Lisenka E.L.M. Vissers, Christian Gilissen, Wouter Steyaert, Karolis Sablauskas, Richarda M. de Voer, Erik-Jan Kamsteeg, Bart van de Warrenburg, Nienke van Os, Iris te Paske, Erik Janssen, Elke de Boer, Marloes Steehouwer, Burcu Yaldiz, Tjitske Kleefstra, Anthony J. Brookes, Colin Veal, Spencer Gibson, Marc Wadsley, Mehdi Mehtarizadeh, Umar Riaz, Greg Warren, Farid Yavari Dizjikan, Thomas Shorter, Ana Töpf, Volker Straub, Chiara Marini Bettolo, Sabine Specht, Jill Clayton-Smith, Elizabeth Alexander, Laurence Faivre, Christel Thauvin, Antonio Vitobello, Anne-Sophie Denommé-Pichon, Yannis Duffourd, Emilie Tisserant, Ange-Line Bruel, Christine Peyron, Aurore Pélissier, Sergi Beltran, Ivo Glynne Gut, Steven Laurie, Davide Piscia, Leslie Matalonga, Anastasios Papakonstantinou, Gemma Bullich, Alberto Corvo, Carles Garcia, Marcos Fernandez-Callejo, Carles Hernández, Daniel Picó, Ida Paramonov, Hanns Lochmüller, Gulcin Gumus, Virginie Bros-Facer, Ana Rath, Marc Hanauer, Annie Olry, David Lagorce, Svitlana Havrylenko, Katia Izem, Fanny Rigour, Giovanni Stevanin, Alexandra Durr, Claire-Sophie Davoine, Léna Guillot-Noel, Anna Heinzmann, Giulia Coarelli, Gisèle Bonne, Teresinha Evangelista, Valérie Allamand, Isabelle Nelson, Rabah Ben Yaou, Corinne Metay, Bruno Eymard, Enzo Cohen, Antonio Atalaia, Tanya Stojkovic, Milan Macek, Jr., Marek Turnovec, Dana Thomasová, Radka Pourová Kremliková, Vera Franková, Markéta Havlovicová, Vlastimil Kremlik, Helen Parkinson, Thomas Keane, Dylan Spalding, Alexander Senf, Peter Robinson, Daniel Danis, Glenn Robert, Alessia Costa, Christine Patch, Mike Hanna, Henry Houlden, Mary Reilly, Jana Vandrovcova, Francesco Muntoni, Irina Zaharieva, Anna Sarkozy, Vincent Timmerman, Jonathan Baets, Liedewei Van de Vondel, Danique Beijer, Peter de Jonghe, Vincenzo Nigro, Sandro Banfi, Annalaura Torella, Francesco Musacchia, Giulio Piluso, Alessandra Ferlini, Rita Selvatici, Rachele Rossi, Marcella Neri, Stefan Aretz, Isabel Spier, Anna Katharina Sommer, Sophia Peters, Carla Oliveira, Jose Garcia Pelaez, Ana Rita Matos, Celina São José, Marta Ferreira, Irene Gullo, Susana Fernandes, Luzia Garrido, Pedro Ferreira, Fátima Carneiro, Morris A. Swertz, Lennart Johansson, Joeri K. van der Velde, Gerben van der Vries, Pieter B. Neerincx, Dieuwke Roelofs-Prins, Sebastian Köhler, Alison Metcalfe, Alain Verloes, Séverine Drunat, Caroline Rooryck, Aurelien Trimouille, Raffaele Castello, Manuela Morleo, Michele Pinelli, Alessandra Varavallo, Manuel Posada De la Paz, Eva Bermejo Sánchez, Estrella López Martín, Beatriz Martínez Delgado, F. Javier Alonso García de la Rosa, Andrea Ciolfi, Bruno Dallapiccola, Simone Pizzi, Francesca Clementina Radio, Marco Tartaglia, Alessandra Renieri, Elisa Benetti, Peter Balicza, Maria Judit Molnar, Ales Maver, Borut Peterlin, Alexander Münchau, Katja Lohmann, Rebecca Herzog, Martje Pauly, Alfons Macaya, Anna Marcé-Grau, Andres Nascimiento Osorio, Daniel Natera de Benito, Rachel Thompson, Kiran Polavarapu, David Beeson, Judith Cossins, Pedro M. Rodriguez Cruz, Peter Hackman, Mridul Johari, Marco Savarese, Bjarne Udd, Rita Horvath, Gabriel Capella, Laura Valle, Elke Holinski-Feder, Andreas Laner, Verena Steinke-Lange, Evelin Schröck, and Andreas Rump
- Subjects
TSPEAR ,Ectodermal dysplasia ,Enamel knot ,WNT10A ,Hypodontia ,Conical teeth ,Genetics ,QH426-470 - Abstract
Summary: TSPEAR variants cause autosomal recessive ectodermal dysplasia (ARED) 14. The function of TSPEAR is unknown. The clinical features, the mutation spectrum, and the underlying mechanisms of ARED14 are poorly understood. Combining data from new and previously published individuals established that ARED14 is primarily characterized by dental anomalies such as conical tooth cusps and hypodontia, like those seen in individuals with WNT10A-related odontoonychodermal dysplasia. AlphaFold-predicted structure-based analysis showed that most of the pathogenic TSPEAR missense variants likely destabilize the β-propeller of the protein. Analysis of 100000 Genomes Project (100KGP) data revealed multiple founder TSPEAR variants across different populations. Mutational and recombination clock analyses demonstrated that non-Finnish European founder variants likely originated around the end of the last ice age, a period of major climatic transition. Analysis of gnomAD data showed that the non-Finnish European population TSPEAR gene-carrier rate is ∼1/140, making it one of the commonest AREDs. Phylogenetic and AlphaFold structural analyses showed that TSPEAR is an ortholog of drosophila Closca, an extracellular matrix-dependent signaling regulator. We, therefore, hypothesized that TSPEAR could have a role in enamel knot, a structure that coordinates patterning of developing tooth cusps. Analysis of mouse single-cell RNA sequencing (scRNA-seq) data revealed highly restricted expression of Tspear in clusters representing enamel knots. A tspeara−/−;tspearb−/− double-knockout zebrafish model recapitulated the clinical features of ARED14 and fin regeneration abnormalities of wnt10a knockout fish, thus suggesting interaction between tspear and wnt10a. In summary, we provide insights into the role of TSPEAR in ectodermal development and the evolutionary history, epidemiology, mechanisms, and consequences of its loss of function variants.
- Published
- 2023
- Full Text
- View/download PDF
46. Molecular Nodal Restaging Based on CEACAM5, FGFR2b and PTPN11 Expression Adds No Relevant Clinical Information in Resected Non-Small Cell Lung Cancer
- Author
-
Ivan Macia, Gemma Aiza, Ricard Ramos, Ignacio Escobar, Francisco Rivas, Anna Ureña, Samantha Aso, Gabriela Rosado, Pau Rodriguez-Taboada, Carlos Deniz, Ernest Nadal, and Gabriel Capella
- Subjects
lung development ,molecular marker ,non-small cell lung cancer ,nodal staging ,Surgery ,RD1-811 - Abstract
Background The relapse rate in non-small cell lung cancer (NSCLC) is high, even in localized disease, suggesting that the current approach to pathological staging is insufficiently sensitive to detect occult micrometastases present in resected lymph nodes. Therefore, we aimed to determine the prognostic value of the expression of embryonic molecular markers in histologically-negative lymph nodes of completely-resected NSCLC. Methods 76 completely-resected NSCLC patients were included: 60 pN0 and 16 pN1. Primary tumors and 347 lymph node were studied. CEACAM5, FGFR2b, and PTPN11 expression levels were evaluated through mRNA analysis using real-time RT-qPCR assay. Statistical analyses included the Kruskal-Wallis test, Kaplan Meier curves, and log-rank tests. Results CEACAM5 expression levels were scored as high in of 90 lymph nodes (26%). The molecular-positive lymph nodes lead to the restaging of 37 (62%) pN0 patients as molecular N1 or N2 and 5 (31%) pN1 cases were reclassified as molecular-positive N2. Surprisingly, molecular-positive patients associated with a better OS (overall survival, p = 0,04). FGFR2b overexpression was observed in 41 (12%) lymph nodes leading to the restaging of 17 patients (22%). Again a trend was observed toward a better DFS (disease-free survival) in the restaged patients (p = 0,09). Accordingly, high expression levels of CEACAM5 or FGFR2b in the primary were related to better DFS (p = 0,06; p
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.