154 results on '"Paola Ghiorzo"'
Search Results
2. Comprehensive genomic profiling on metastatic Melanoma: results from a network screening from 7 Italian Cancer Centres
- Author
-
Matteo Pallocca, Ivan Molineris, Enrico Berrino, Benedetta Marcozzi, Martina Betti, Lauretta Levati, Stefania D’Atri, Chiara Menin, Gabriele Madonna, Paola Ghiorzo, Jenny Bulgarelli, Virgina Ferraresi, Tiziana Venesio, Monica Rodolfo, Licia Rivoltini, Luisa Lanfrancone, Paolo Antonio Ascierto, Luca Mazzarella, Pier Giuseppe Pelicci, Ruggero De Maria, Gennaro Ciliberto, Enzo Medico, and Giandomenico Russo
- Subjects
Comprehensive genomic profiling ,Network trial ,Alleanza Contro il Cancro ,Melanoma ,SKCM ,Immuno-checkpoint inhibitors ,Medicine - Abstract
Abstract Background The current therapeutic algorithm for Advanced Stage Melanoma comprises of alternating lines of Targeted and Immuno-therapy, mostly via Immune-Checkpoint blockade. While Comprehensive Genomic Profiling of solid tumours has been approved as a companion diagnostic, still no approved predictive biomarkers are available for Melanoma aside from BRAF mutations and the controversial Tumor Mutational Burden. This study presents the results of a Multi-Centre Observational Clinical Trial of Comprehensive Genomic Profiling on Target and Immuno-therapy treated advanced Melanoma. Methods 82 samples, collected from 7 Italian Cancer Centres of FFPE-archived Metastatic Melanoma and matched blood were sequenced via a custom-made 184-gene amplicon-based NGS panel. Sequencing and bioinformatics analysis was performed at a central hub. Primary analysis was carried out via the Ion Reporter framework. Secondary analysis and Machine Learning modelling comprising of uni and multivariate, COX/Lasso combination, and Random Forest, was implemented via custom R/Python scripting. Results The genomics landscape of the ACC-mela cohort is comparable at the somatic level for Single Nucleotide Variants and INDELs aside a few gene targets. All the clinically relevant targets such as BRAF and NRAS have a comparable distribution thus suggesting the value of larger scale sequencing in melanoma. No comparability is reached at the CNV level due to biotechnological biases and cohort numerosity. Tumour Mutational Burden is slightly higher in median for Complete Responders but fails to achieve statistical significance in Kaplan–Meier survival analysis via several thresholding strategies. Mutations on PDGFRB, NOTCH3 and RET were shown to have a positive effect on Immune-checkpoint treatment Overall and Disease-Free Survival, while variants in NOTCH4 were found to be detrimental for both endpoints. Conclusions The results presented in this study show the value and the challenge of a genomics-driven network trial. The data can be also a valuable resource as a validation cohort for Immunotherapy and Target therapy genomic biomarker research.
- Published
- 2024
- Full Text
- View/download PDF
3. Special Issue 'Molecular Advances in Cancer Genetics 3.0'
- Author
-
William Bruno and Paola Ghiorzo
- Subjects
n/a ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
The third volume of this Special Issue focuses on new advances in cancer genetics studies and collates papers reporting on a variety of mechanisms of tumorigenesis, the need to explore them from multiple perspectives, and the difficulties in exploring them, as well as the challenge of integrating them into a unifying but still different model for each tumor type [...]
- Published
- 2024
- Full Text
- View/download PDF
4. Birth cohort-specific trends of sun-related behaviors among individuals from an international consortium of melanoma-prone families
- Author
-
John Charles A. Lacson, Shawn A. Zamani, Luis Alberto Ribeiro Froes, Nandita Mitra, Lu Qian, Scarlet H. Doyle, Esther Azizi, Claudia Balestrini, D. Timothy Bishop, William Bruno, Blanca Carlos-Ortega, Francisco Cuellar, Anne E. Cust, David E. Elder, Anne-Marie Gerdes, Paola Ghiorzo, Thais C. Grazziotin, Nelleke A. Gruis, Johan Hansson, Marko Hočevar, Veronica Höiom, Elizabeth A. Holland, Christian Ingvar, Gilles Landman, Alejandra Larre-Borges, Graham J. Mann, Montserrat Molgo, Luciana Facure Moredo, Håkan Olsson, Jacoba J. Out-Luiting, Barbara Perić, Dace Pjanova, Susana Puig, Julio Salas-Alanis, Helen Schmid, Karin A. W. Wadt, Julia A. Newton-Bishop, Peter A. Kanetsky, and on behalf of the GenoMEL Study Group
- Subjects
Trends ,Sun-related behaviors ,Sunscreen use ,Sun exposure ,Sunburn ,Sunbed ,Public aspects of medicine ,RA1-1270 - Abstract
Abstract Background Individuals from melanoma-prone families have similar or reduced sun-protective behaviors compared to the general population. Studies on trends in sun-related behaviors have been temporally and geographically limited. Methods Individuals from an international consortium of melanoma-prone families (GenoMEL) were retrospectively asked about sunscreen use, sun exposure (time spent outside), sunburns, and sunbed use at several timepoints over their lifetime. Generalized linear mixed models were used to examine the association between these outcomes and birth cohort defined by decade spans, after adjusting for covariates. Results A total of 2407 participants from 547 families across 17 centers were analyzed. Sunscreen use increased across subsequent birth cohorts, and although the likelihood of sunburns increased until the 1950s birth cohort, it decreased thereafter. Average sun exposure did not change across the birth cohorts, and the likelihood of sunbed use increased in more recent birth cohorts. We generally did not find any differences in sun-related behavior when comparing melanoma cases to non-cases. Melanoma cases had increased sunscreen use, decreased sun exposure, and decreased odds of sunburn and sunbed use after melanoma diagnosis compared to before diagnosis. Conclusions Although sunscreen use has increased and the likelihood of sunburns has decreased in more recent birth cohorts, individuals in melanoma-prone families have not reduced their overall sun exposure and had an increased likelihood of sunbed use in more recent birth cohorts. These observations demonstrate partial improvements in melanoma prevention and suggest that additional intervention strategies may be needed to achieve optimal sun-protective behavior in melanoma-prone families.
- Published
- 2021
- Full Text
- View/download PDF
5. Quality assessment of a clinical next-generation sequencing melanoma panel within the Italian Melanoma Intergroup (IMI)
- Author
-
Irene Vanni, Milena Casula, Lorenza Pastorino, Antonella Manca, Bruna Dalmasso, Virginia Andreotti, Marina Pisano, Maria Colombino, Italian Association for Cancer Research (AIRC) Study Group, Ulrich Pfeffer, Enrica Teresa Tanda, Carla Rozzo, Panagiotis Paliogiannis, Antonio Cossu, Paola Ghiorzo, Giuseppe Palmieri, and for the Italian Melanoma Intergroup (IMI)
- Subjects
Melanoma ,Gene panel testing ,Next generation sequencing (NGS) ,Somatic mutations ,Quality controls ,BRAF ,Pathology ,RB1-214 - Abstract
Abstract Background Identification of somatic mutations in key oncogenes in melanoma is important to lead the effective and efficient use of personalized anticancer treatment. Conventional methods focus on few genes per run and, therefore, are unable to screen for multiple genes simultaneously. The use of Next-Generation Sequencing (NGS) technologies enables sequencing of multiple cancer-driving genes in a single assay, with reduced costs and DNA quantity needed and increased mutation detection sensitivity. Methods We designed a customized IMI somatic gene panel for targeted sequencing of actionable melanoma mutations; this panel was tested on three different NGS platforms using 11 metastatic melanoma tissue samples in blinded manner between two EMQN quality certificated laboratory. Results The detection limit of our assay was set-up to a Variant Allele Frequency (VAF) of 10% with a coverage of at least 200x. All somatic variants detected by all NGS platforms with a VAF ≥ 10%, were also validated by an independent method. The IMI panel achieved a very good concordance among the three NGS platforms. Conclusion This study demonstrated that, using the main sequencing platforms currently available in the diagnostic setting, the IMI panel can be adopted among different centers providing comparable results.
- Published
- 2020
- Full Text
- View/download PDF
6. Clinical, pathological and dermoscopic phenotype of MITF p.E318K carrier cutaneous melanoma patients
- Author
-
Giulia Ciccarese, Bruna Dalmasso, William Bruno, Paola Queirolo, Lorenza Pastorino, Virginia Andreotti, Francesco Spagnolo, Enrica Tanda, Giovanni Ponti, Cesare Massone, Francesco Drago, Aurora Parodi, Giovanni Ghigliotti, Maria Antonietta Pizzichetta, Paola Ghiorzo, and Italian Melanoma Intergroup (I.M.I.)
- Subjects
Melanocyte Inducing Transcription Factor ,E318K ,Cutaneous melanoma ,Renal cell carcinoma ,Nevi ,Dysplastic nevi ,Medicine - Abstract
Abstract Background The p.E318K variant of the Melanocyte Inducing Transcription Factor (MITF) has been implicated in genetic predisposition to melanoma as an intermediate penetrance allele. However, the impact of this variant on clinico-phenotypic, as well as on dermoscopic patterns features of affected patients is not entirely defined. The purpose of our study was to assess the association between the p.E318K germline variant and clinic-phenotypical features of MITF+ compared to non-carriers (MITF−), including dermoscopic findings of melanomas and dysplastic nevi. Methods we retrospectively analyzed a consecutive series of 1386 patients recruited between 2000 and 2017 who underwent genetic testing for CDKN2A, CDK4, MC1R and MITF germline variants in our laboratory for diagnostic/research purposes. The patients were probands of melanoma-prone families and apparently sporadic single or multiple primary melanoma patients. For all, we collected clinical, pathological information and dermoscopic images of the histopathologically diagnosed melanomas and dysplastic nevi, when available. Results After excluding patients positive for CDKN2A/CDK4 pathogenic variants and those affected by non-cutaneous melanomas, our study cohort comprised 984 cutaneous melanoma patients, 22 MITF+ and 962 MITF−. MITF+ were more likely to develop dysplastic nevi and multiple primary melanomas. Nodular melanoma was more common in MITF+ patients (32% compared to 19% in MITF−). MITF+ patients showed more frequently dysplastic nevi and melanomas with uncommon dermoscopic patterns (unspecific), as opposed to MITF− patients, whose most prevalent pattern was the multicomponent. Conclusions MITF+ patients tend to develop melanomas and dysplastic nevi with histopathological features, frequency and dermoscopic patterns often different from those prevalent in MITF− patients. Our results emphasize the importance of melanoma prevention programs for MITF+ patients, including dermatologic surveillance with digital follow-up.
- Published
- 2020
- Full Text
- View/download PDF
7. Whole-Exome Sequencing and cfDNA Analysis Uncover Genetic Determinants of Melanoma Therapy Response in a Real-World Setting
- Author
-
Irene Vanni, Lorenza Pastorino, Enrica Teresa Tanda, Virginia Andreotti, Bruna Dalmasso, Nicola Solari, Matteo Mascherini, Francesco Cabiddu, Antonio Guadagno, Simona Coco, Eleonora Allavena, William Bruno, Gabriella Pietra, Michela Croce, Rosaria Gangemi, Michele Piana, Gabriele Zoppoli, Lorenzo Ferrando, Francesco Spagnolo, Paola Queirolo, and Paola Ghiorzo
- Subjects
melanoma ,whole-exome sequencing ,circulating free DNA ,BRAF V600 ,targeted therapy ,immunotherapy ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Although several studies have explored the molecular landscape of metastatic melanoma, the genetic determinants of therapy resistance are still largely unknown. Here, we aimed to determine the contribution of whole-exome sequencing and circulating free DNA (cfDNA) analysis in predicting response to therapy in a consecutive real-world cohort of 36 patients, undergoing fresh tissue biopsy and followed during treatment. Although the underpowered sample size limited statistical analysis, samples from non-responders had higher copy number variations and mutations in melanoma driver genes compared to responders in the BRAF V600+ subset. In the BRAF V600− subset, Tumor Mutational Burden (TMB) was twice that in responders vs. non-responders. Genomic layout revealed commonly known and novel potential intrinsic/acquired resistance driver gene variants. Among these, RAC1, FBXW7, GNAQ mutations, and BRAF/PTEN amplification/deletion were present in 42% and 67% of patients, respectively. Both Loss of Heterozygosity (LOH) load and tumor ploidy were inversely associated with TMB. In immunotherapy-treated patients, samples from responders showed higher TMB and lower LOH and were more frequently diploid compared to non-responders. Secondary germline testing and cfDNA analysis proved their efficacy in finding germline predisposing variants carriers (8.3%) and following dynamic changes during treatment as a surrogate of tissue biopsy, respectively.
- Published
- 2023
- Full Text
- View/download PDF
8. A Glance at Molecular Advances in Cancer Genetics: A Baffling Puzzle Still to Be Solved
- Author
-
Paola Ghiorzo and William Bruno
- Subjects
n/a ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
The purpose of this first Special Issue is to provide a glance at the molecular advances in cancer genetics to untangle the complexity of tumorigenesis [...]
- Published
- 2023
- Full Text
- View/download PDF
9. Ataxia-Telangiectasia Mutated Loss of Heterozygosity in Melanoma
- Author
-
Lorenza Pastorino, Bruna Dalmasso, Eleonora Allavena, Irene Vanni, Filippo Ugolini, Gianna Baroni, Michela Croce, Antonio Guadagno, Francesco Cabiddu, Virginia Andreotti, William Bruno, Gabriele Zoppoli, Lorenzo Ferrando, Enrica Teresa Tanda, Francesco Spagnolo, Chiara Menin, Rosaria Gangemi, Daniela Massi, and Paola Ghiorzo
- Subjects
germline variant ,loss of heterozygosity ,melanoma ,ATM ,susceptibility ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
ATM germline pathogenic variants were recently found enriched in high-risk melanoma patients. However, ATM loss of heterozygosity (LOH) has never been investigated in melanoma and, therefore, a causal association with melanoma development has not been established yet. The purpose of this study was to functionally characterize 13 germline ATM variants found in high-risk melanoma patients—and classified by in silico tools as pathogenic, uncertain significance, or benign—using multiple assays evaluating ATM/pATM expression and/or LOH in melanoma tissues and cell lines. We assessed ATM status by Immunohistochemistry (IHC), Western Blot, Whole-Exome Sequencing/Copy Number Variation analysis, and RNA sequencing, supported by Sanger sequencing and microsatellite analyses. For most variants, IHC results matched those obtained with in silico classification and LOH analysis. Two pathogenic variants (p.Ser1135_Lys1192del and p.Ser1993ArgfsTer23) showed LOH and complete loss of ATM activation in melanoma. Two variants of unknown significance (p.Asn358Ile and p.Asn796His) showed reduced expression and LOH, suggestive of a deleterious effect. This study, showing a classic two-hit scenario in a well-known tumor suppressor gene, supports the inclusion of melanoma in the ATM-related cancer spectrum.
- Published
- 2022
- Full Text
- View/download PDF
10. Prevalence of pathogenic/likely pathogenic variants in the 24 cancer genes of the ACMG Secondary Findings v2.0 list in a large cancer cohort and ethnicity-matched controls
- Author
-
Jung Kim, Wen Luo, Mingyi Wang, Talia Wegman-Ostrosky, Megan N. Frone, Jennifer J. Johnston, Michael L. Nickerson, Melissa Rotunno, Shengchao A. Li, Maria I. Achatz, Seth A. Brodie, Michael Dean, Kelvin C. de Andrade, Fernanda P. Fortes, Matthew Gianferante, Payal Khincha, Mary L. McMaster, Lisa J. McReynolds, Alexander Pemov, Maisa Pinheiro, Karina M. Santiago, Blanche P. Alter, Neil E. Caporaso, Shahinaz M. Gadalla, Lynn R. Goldin, Mark H. Greene, Jennifer Loud, Xiaohong R. Yang, Neal D. Freedman, Susan M. Gapstur, Mia M. Gaudet, Donato Calista, Paola Ghiorzo, Maria Concetta Fargnoli, Eduardo Nagore, Ketty Peris, Susana Puig, Maria Teresa Landi, Belynda Hicks, Bin Zhu, Jia Liu, Joshua N. Sampson, Stephen J. Chanock, Lisa J. Mirabello, Lindsay M. Morton, Leslie G. Biesecker, Margaret A. Tucker, Sharon A. Savage, Alisa M. Goldstein, and Douglas R. Stewart
- Subjects
ACMG secondary findings ,Familial cancer exome ,Population study ,Variant classification ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background Prior research has established that the prevalence of pathogenic/likely pathogenic (P/LP) variants across all of the American College of Medical Genetics (ACMG) Secondary Findings (SF) genes is approximately 0.8–5%. We investigated the prevalence of P/LP variants in the 24 ACMG SF v2.0 cancer genes in a family-based cancer research cohort (n = 1173) and in cancer-free ethnicity-matched controls (n = 982). Methods We used InterVar to classify variants and subsequently conducted a manual review to further examine variants of unknown significance (VUS). Results In the 24 genes on the ACMG SF v2.0 list associated with a cancer phenotype, we observed 8 P/LP unique variants (8 individuals; 0.8%) in controls and 11 P/LP unique variants (14 individuals; 1.2%) in cases, a non-significant difference. We reviewed 115 VUS. The median estimated per-variant review time required was 30 min; the first variant within a gene took significantly (p = 0.0009) longer to review (median = 60 min) compared with subsequent variants (median = 30 min). The concordance rate was 83.3% for the variants examined by two reviewers. Conclusion The 115 VUS required database and literature review, a time- and labor-intensive process hampered by the difficulty in interpreting conflicting P/LP determinations. By rigorously investigating the 24 ACMG SF v2.0 cancer genes, our work establishes a benchmark P/LP variant prevalence rate in a familial cancer cohort and controls.
- Published
- 2018
- Full Text
- View/download PDF
11. Beyond BRCA: The Emerging Significance of DNA Damage Response and Personalized Treatment in Pancreatic and Prostate Cancer Patients
- Author
-
Bruna Dalmasso, Alberto Puccini, Fabio Catalano, Roberto Borea, Maria Laura Iaia, William Bruno, Giuseppe Fornarini, Stefania Sciallero, Sara Elena Rebuzzi, and Paola Ghiorzo
- Subjects
DNA damage response ,BRCA ,mismatch repair ,homologous recombination ,genetics ,PARP inhibitors ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
The BRCA1/2 germline and/or somatic pathogenic variants (PVs) are key players in the hereditary predisposition and therapeutic response for breast, ovarian and, more recently, pancreatic and prostate cancers. Aberrations in other genes involved in homologous recombination and DNA damage response (DDR) pathways are being investigated as promising targets in ongoing clinical trials. However, DDR genes are not routinely tested worldwide. Due to heterogeneity in cohort selection and dissimilar sequencing approaches across studies, neither the burden of PVs in DDR genes nor the prevalence of PVs in genes in common among pancreatic and prostate cancer can be easily quantified. We aim to contextualize these genes, altered in both pancreatic and prostate cancers, in the DDR process, to summarize their hereditary and somatic burden in different studies and harness their deficiency for cancer treatments in the context of currently ongoing clinical trials. We conclude that the inclusion of DDR genes, other than BRCA1/2, shared by both cancers considerably increases the detection rate of potentially actionable variants, which are triplicated in pancreatic and almost doubled in prostate cancer. Thus, DDR alterations are suitable targets for drug development and to improve the outcome in both pancreatic and prostate cancer patients. Importantly, this will increase the detection of germline pathogenic variants, thereby patient referral to genetic counseling.
- Published
- 2022
- Full Text
- View/download PDF
12. Non-BRAF Mutant Melanoma: Molecular Features and Therapeutical Implications
- Author
-
Irene Vanni, Enrica Teresa Tanda, Bruna Dalmasso, Lorenza Pastorino, Virginia Andreotti, William Bruno, Andrea Boutros, Francesco Spagnolo, and Paola Ghiorzo
- Subjects
melanoma ,non-BRAF mutation ,targeted therapy ,driver mutations ,genetic ,heterogeneity ,Biology (General) ,QH301-705.5 - Abstract
Melanoma is one of the most aggressive tumors of the skin, and its incidence is growing worldwide. Historically considered a drug resistant disease, since 2011 the therapeutic landscape of melanoma has radically changed. Indeed, the improved knowledge of the immune system and its interactions with the tumor, and the ever more thorough molecular characterization of the disease, has allowed the development of immunotherapy on the one hand, and molecular target therapies on the other. The increased availability of more performing technologies like Next-Generation Sequencing (NGS), and the availability of increasingly large genetic panels, allows the identification of several potential therapeutic targets. In light of this, numerous clinical and preclinical trials are ongoing, to identify new molecular targets. Here, we review the landscape of mutated non-BRAF skin melanoma, in light of recent data deriving from Whole-Exome Sequencing (WES) or Whole-Genome Sequencing (WGS) studies on melanoma cohorts for which information on the mutation rate of each gene was available, for a total of 10 NGS studies and 992 samples, focusing on available, or in experimentation, targeted therapies beyond those targeting mutated BRAF. Namely, we describe 33 established and candidate driver genes altered with frequency greater than 1.5%, and the current status of targeted therapy for each gene. Only 1.1% of the samples showed no coding mutations, whereas 30% showed at least one mutation in the RAS genes (mostly NRAS) and 70% showed mutations outside of the RAS genes, suggesting potential new roads for targeted therapy. Ongoing clinical trials are available for 33.3% of the most frequently altered genes.
- Published
- 2020
- Full Text
- View/download PDF
13. Current State of Target Treatment in BRAF Mutated Melanoma
- Author
-
Enrica Teresa Tanda, Irene Vanni, Andrea Boutros, Virginia Andreotti, William Bruno, Paola Ghiorzo, and Francesco Spagnolo
- Subjects
melanoma ,BRAF mutation ,targeted therapy ,MAPK pathway ,metastatic disease ,Biology (General) ,QH301-705.5 - Abstract
Incidence of melanoma has been constantly growing during the last decades. Although most of the new diagnoses are represented by thin melanomas, the number of melanoma-related deaths in 2018 was 60,712 worldwide (Global Cancer Observatory, 2019). Until 2011, no systemic therapy showed to improve survival in patients with advanced or metastatic melanoma. At that time, standard of care was chemotherapy, with very limited results. The identification of BRAF V600 mutation, and the subsequent introduction of BRAF targeting drugs, radically changed the clinical practice and dramatically improved outcomes. In this review, we will retrace the development of molecular-target drugs and the current therapeutic scenario for patients with BRAF mutated melanoma, from the introduction of BRAF inhibitors as single agents to modern clinical practice. We will also discuss the resistance mechanisms identified so far, and the future therapeutic perspectives in BRAF mutated melanoma.
- Published
- 2020
- Full Text
- View/download PDF
14. The Current State of Molecular Testing in the BRAF-Mutated Melanoma Landscape
- Author
-
Irene Vanni, Enrica Teresa Tanda, Francesco Spagnolo, Virginia Andreotti, William Bruno, and Paola Ghiorzo
- Subjects
melanoma ,BRAF mutation ,targeted therapy ,liquid biopsy ,molecular techniques ,NGS ,Biology (General) ,QH301-705.5 - Abstract
The incidence of melanoma, among the most lethal cancers, is widespread and increasing. Metastatic melanoma has a poor prognosis, representing about 90% of skin cancer mortality. The increased knowledge of tumor biology and the greater understanding of the immune system role in the anti-tumor response has allowed us to develop a more rational approach to systemic therapies. The discovery of activating BRAF mutations in half of all melanomas has led to the development of molecularly targeted therapy with BRAF and MEK inhibitors, which dramatically improved outcomes of patients with stage IV BRAF-mutant melanoma. More recently, the results of clinical phase III studies conducted in the adjuvant setting led to the combined administration of BRAF and MEK inhibitors also in patients with resected high-risk melanoma (stage III). Therefore, BRAF mutation testing has become a priority to determine the oncologist's choice and course of therapy. In this review, we will report the molecular biology-based strategies used for BRAF mutation detection with the main advantages and disadvantages of the most commonly used diagnostic strategies. The timing of such molecular assessment in patients with cutaneous melanoma will be discussed, and we will also examine considerations and approaches for accurate and effective BRAF testing.
- Published
- 2020
- Full Text
- View/download PDF
15. Insights into Mechanisms of Tumorigenesis in Neuroendocrine Neoplasms
- Author
-
Lorenza Pastorino, Federica Grillo, Manuela Albertelli, Paola Ghiorzo, and William Bruno
- Subjects
neuroendocrine tumors ,tumorigenesis ,genomic integrity ,chromatin stability ,splicing ,animal models ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Genomic studies have identified some of the most relevant genetic players in Neuroendocrine Neoplasm (NEN) tumorigenesis. However, we are still far from being able to draw a model that encompasses their heterogeneity, elucidates the different biological effects consequent to the identified molecular events, or incorporates extensive knowledge of molecular biomarkers and therapeutic targets. Here, we reviewed recent insights in NEN tumorigenesis from selected basic research studies on animal models, highlighting novel players in the intergenic cooperation and peculiar mechanisms including splicing dysregulation, chromatin stability, or cell dedifferentiation. Furthermore, models of tumorigenesis based on composite interactions other than a linear progression of events are proposed, exemplified by the involvement in NEN tumorigenesis of genes regulating complex functions, such as MEN1 or DAXX. Although limited by interspecies differences, animal models have proved helpful for the more in-depth study of every facet of tumorigenesis, showing that the identification of driver mutations is only one of the many necessary steps and that other mechanisms are worth investigating.
- Published
- 2021
- Full Text
- View/download PDF
16. Contribution of Common Genetic Variants to Familial Aggregation of Disease and Implications for Sequencing Studies.
- Author
-
Andrew Schlafly, Ruth M Pfeiffer, Eduardo Nagore, Susana Puig, Donato Calista, Paola Ghiorzo, Chiara Menin, Maria Concetta Fargnoli, Ketty Peris, Lei Song, Tongwu Zhang, Jianxin Shi, Maria Teresa Landi, and Joshua Neil Sampson
- Subjects
Genetics ,QH426-470 - Abstract
Despite genetics being accepted as the primary cause of familial aggregation for most diseases, it is still unclear whether afflicted families are likely to share a single highly penetrant rare variant, many minimally penetrant common variants, or a combination of the two types of variants. We therefore use recent estimates of SNP heritability and the liability threshold model to estimate the proportion of afflicted families likely to carry a rare, causal variant. We then show that Polygenic Risk Scores (PRS) may be useful for identifying families likely to carry such a rare variant and therefore for prioritizing families to include in sequencing studies with that aim. Specifically, we introduce a new statistic that estimates the proportion of individuals carrying causal rare variants based on the family structure, disease pattern, and PRS of genotyped individuals. Finally, we consider data from the MelaNostrum consortium and show that, despite an estimated PRS heritability of only 0.05 for melanoma, families carrying putative causal variants had a statistically significantly lower PRS, supporting the idea that PRS prioritization may be a useful future tool. However, it will be important to evaluate whether the presence of rare mendelian variants are generally associated with the proposed test statistic or lower PRS in future and larger studies.
- Published
- 2019
- Full Text
- View/download PDF
17. Functional analysis of a CDKN2A 5'UTR germline variant associated with pancreatic cancer development.
- Author
-
William Bruno, Virginia Andreotti, Alessandra Bisio, Lorenza Pastorino, Giuseppe Fornarini, Stefania Sciallero, Giovanna Bianchi-Scarrà, Alberto Inga, and Paola Ghiorzo
- Subjects
Medicine ,Science - Abstract
CDKN2A coding region germline variants are associated with pancreatic adenocarcinoma (PC) susceptibility. Recently, we described functional germline 5'UTR CDKN2A variants from melanoma patients affecting the post-transcriptional regulation of p16INK4a mRNA that is dependent, at least in part, on an Internal Ribosome Entry Site (IRES) in the 5'UTR region. Here we describe a 5'UTR c.-201_-198delinsCTTT CDKN2A variant (frequency 0.0028 based on 350 PC patients), which seems to be private to PC, since it has never been found in public databases nor in thousands of melanoma patients tested. Functional analyses confirmed IRES activity of the 5'UTR in BX-PC3 PC cells and revealed a functional impact of the identified variant. Using gene reporter assays we observed reduced translation potential in cells treated with the mTOR inhibitor Torin1, a condition that favors the assessment of IRES activity. At the endogenous gene level we quantified allelic imbalance among polysome-associated mRNAs using a patient-derived cell line heterozygous for the c.-201_-198delinsCTTT. Overall, we conclude that this very rare private variant can be considered a potential mutation, specifically associated with PC. Our data indicate that sequencing of the entire 5'UTR of CDKN2A should be included in routine screening of PC cases with suspected inherited susceptibility.
- Published
- 2017
- Full Text
- View/download PDF
18. PCCR: Pancreatic Cancer Collaborative Registry
- Author
-
Simon Sherman, Oleg Shats, Marsha A. Ketcham, Michelle A. Anderson, David C. Whitcomb, Henry T. Lynch, Paola Ghiorzo, Wendy S. Rubinstein, Aaron R. Sasson, William E. Grizzle, Gleb Haynatzki, Jianmin Feng, Alexander Sherman, Leo Kinarsky, and Randall E. Brand
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2011
19. On the interplay of telomeres, nevi and the risk of melanoma.
- Author
-
Clara Bodelon, Ruth M Pfeiffer, Valentina Bollati, Julien Debbache, Donato Calista, Paola Ghiorzo, Maria Concetta Fargnoli, Giovanna Bianchi-Scarra, Ketty Peris, Mirjam Hoxha, Amy Hutchinson, Laurie Burdette, Laura Burke, Shenying Fang, Margaret A Tucker, Alisa M Goldstein, Jeffrey E Lee, Qingyi Wei, Sharon A Savage, Xiaohong R Yang, Christopher Amos, and Maria Teresa Landi
- Subjects
Medicine ,Science - Abstract
The relationship between telomeres, nevi and melanoma is complex. Shorter telomeres have been found to be associated with many cancers and with number of nevi, a known risk factor for melanoma. However, shorter telomeres have also been found to decrease melanoma risk. We performed a systematic analysis of telomere-related genes and tagSNPs within these genes, in relation to the risk of melanoma, dysplastic nevi, and nevus count combining data from four studies conducted in Italy. In addition, we examined whether telomere length measured in peripheral blood leukocytes is related to the risk of melanoma, dysplastic nevi, number of nevi, or telomere-related SNPs. A total of 796 cases and 770 controls were genotyped for 517 SNPs in 39 telomere-related genes genotyped with a custom-made array. Replication of the top SNPs was conducted in two American populations consisting of 488 subjects from 53 melanoma-prone families and 1,086 cases and 1,024 controls from a case-control study. We estimated odds ratios for associations with SNPs and combined SNP P-values to compute gene region-specific, functional group-specific, and overall P-value using an adaptive rank-truncated product algorithm. In the Mediterranean population, we found suggestive evidence that RECQL4, a gene involved in genome stability, RTEL1, a gene regulating telomere elongation, and TERF2, a gene implicated in the protection of telomeres, were associated with melanoma, the presence of dysplastic nevi and number of nevi, respectively. However, these associations were not found in the American samples, suggesting variable melanoma susceptibility for these genes across populations or chance findings in our discovery sample. Larger studies across different populations are necessary to clarify these associations.
- Published
- 2012
- Full Text
- View/download PDF
20. Novel PTCH1 mutations in patients with keratocystic odontogenic tumors screened for nevoid basal cell carcinoma (NBCC) syndrome.
- Author
-
Lorenza Pastorino, Annamaria Pollio, Giovanni Pellacani, Carmelo Guarneri, Paola Ghiorzo, Caterina Longo, William Bruno, Francesca Giusti, Sara Bassoli, Giovanna Bianchi-Scarrà, Cristel Ruini, Stefania Seidenari, Aldo Tomasi, and Giovanni Ponti
- Subjects
Medicine ,Science - Abstract
Keratocystic odontogenic tumors (KCOTs) are cystic tumors that arise sporadically or associated with nevoid basal cell carcinoma syndrome (NBCCS). NBCCS is a rare autosomal dominantly inherited disease mainly characterized by multiple basal cell carcinomas, KCOTs of the jaws and a variety of other tumors. PTCH1 mutation can be found both in sporadic or NBCCS associated KCOTs. The aim of the current study was to assess whether a combined clinical and bio-molecular approach could be suitable for the detection of NBCCS among patients with a diagnosis of keratocystic odontogenic tumors (KCOTs). The authors collected keratocystic odontogenic tumors recorded in the database of the Pathology Department of the University of Modena and Reggio Emilia during the period 1991-2011. Through interviews and examinations, family pedigrees were drawn for all patients affected by these odontogenic lesions. We found out that 18 of the 70 patients with KCOTs and/or multiple basal cell carcinomas actually met the clinical criteria for the diagnosis of NBCCS. A wide inter- and intra-familial phenotypic variability was evident in the families. Ameloblastomas (AMLs) were reported in two probands that are also carriers of the PCTH1 germline mutations. Nine germline mutations in the PTCH1 gene, 5 of them novel, were evident in 14 tested probands. The clinical evaluation of the keratocystic odontogenic tumors can be used as screening for the detection of families at risk of NBCCS. Keratocystic odontogenic lesions are uncommon, and their discovery deserves the search for associated cutaneous basal cell carcinomas and other benign and malignant tumors related to NBCCS.
- Published
- 2012
- Full Text
- View/download PDF
21. The challenge of the Molecular Tumor Board empowerment in clinical oncology practice: A Position Paper on behalf of the AIOM- SIAPEC/IAP-SIBioC-SIC-SIF-SIGU-SIRM Italian Scientific Societies
- Author
-
Antonio Russo, 1, Lorena Incorvaia, 2, Ettore Capoluongo, 3, Pierosandro Tagliaferri, 4, Antonio Galvano, 2, Marzia Del Re, 5, Umberto Malapelle, 6, Rita Chiari, 7, Pierfranco Conte, 8, Romano Danesi, 5, Matteo Fassan, 9, Roberto Ferrara, 10, Genuardi, M, Paola Ghiorzo, 12, Stefania Gori, 13, Fiorella Guadagni, 14, Antonio Marchetti, 15, Paolo Marchetti, 16, Massimo Midiri, 17, Nicola Normanno, 18, Francesco Passiglia, 19, Carmine Pinto, 20, Nicola Silvestris, 21, Giovanni Tallini, 22, Simona Vatrano, 23, Bruno Vincenzi, 24, Saverio Cinieri, 25, Giordano Beretta, 26, Antonio Russo 1, Lorena Incorvaia 2, Ettore Capoluongo 3, Pierosandro Tagliaferri 4, Antonio Galvano 2, Marzia Del Re 5, Umberto Malapelle 6, Rita Chiari 7, Pierfranco Conte 8, Romano Danesi 5, Matteo Fassan 9, Roberto Ferrara 10, Genuardi M (ORCID:0000-0002-7410-8351), Paola Ghiorzo 12, Stefania Gori 13, Fiorella Guadagni 14, Antonio Marchetti 15, Paolo Marchetti 16, Massimo Midiri 17, Nicola Normanno 18, Francesco Passiglia 19, Carmine Pinto 20, Nicola Silvestris 21, Giovanni Tallini 22, Simona Vatrano 23, Bruno Vincenzi 24, Saverio Cinieri 25, Giordano Beretta 26, Antonio Russo, 1, Lorena Incorvaia, 2, Ettore Capoluongo, 3, Pierosandro Tagliaferri, 4, Antonio Galvano, 2, Marzia Del Re, 5, Umberto Malapelle, 6, Rita Chiari, 7, Pierfranco Conte, 8, Romano Danesi, 5, Matteo Fassan, 9, Roberto Ferrara, 10, Genuardi, M, Paola Ghiorzo, 12, Stefania Gori, 13, Fiorella Guadagni, 14, Antonio Marchetti, 15, Paolo Marchetti, 16, Massimo Midiri, 17, Nicola Normanno, 18, Francesco Passiglia, 19, Carmine Pinto, 20, Nicola Silvestris, 21, Giovanni Tallini, 22, Simona Vatrano, 23, Bruno Vincenzi, 24, Saverio Cinieri, 25, Giordano Beretta, 26, Antonio Russo 1, Lorena Incorvaia 2, Ettore Capoluongo 3, Pierosandro Tagliaferri 4, Antonio Galvano 2, Marzia Del Re 5, Umberto Malapelle 6, Rita Chiari 7, Pierfranco Conte 8, Romano Danesi 5, Matteo Fassan 9, Roberto Ferrara 10, Genuardi M (ORCID:0000-0002-7410-8351), Paola Ghiorzo 12, Stefania Gori 13, Fiorella Guadagni 14, Antonio Marchetti 15, Paolo Marchetti 16, Massimo Midiri 17, Nicola Normanno 18, Francesco Passiglia 19, Carmine Pinto 20, Nicola Silvestris 21, Giovanni Tallini 22, Simona Vatrano 23, Bruno Vincenzi 24, Saverio Cinieri 25, and Giordano Beretta 26
- Abstract
The development of innovative technologies and the advances in the genetics and genomics, have offered new opportunities for personalized treatment in oncology. Although the selection of the patient based on the molecular characteristics of the neoplasm has the potential to revolutionize the therapeutic scenario of oncology, this approach is extremely challenging. The access, homogeneity, and economic sustainability of the required genomic tests should be warranted in the clinical practice, as well as the specific scientific and clinical expertise for the choice of medical therapies. All these elements make essential the collaboration of different specialists within the Molecular Tumor Boards (MTBs). In this position paper, based on experts' opinion, the AIOM-SIAPEC/IAP-SIBioC-SIC-SIF-SIGU-SIRM Italian Scientific Societies critically discuss the available molecular profiling technologies, the proposed criteria for the selection of patients candidate for evaluation by the MTB, the criteria for the selection and analysis of biological samples, and the regulatory and pharmaco-economic issues.
- Published
- 2022
22. Gene-Level Associations in Patients With and Without Pathogenic Germline Variants in
- Author
-
Esteban, Astiazaran-Symonds, Cole, Graham, Jung, Kim, Margaret A, Tucker, Christian, Ingvar, Hildur, Helgadottir, Lorenza, Pastorino, Remco, van Doorn, Joshua N, Sampson, Bin, Zhu, William, Bruno, Paola, Queirolo, Giuseppe, Fornarini, Stefania, Sciallero, Brian, Carter, Belynda, Hicks, Amy, Hutchinson, Kristine, Jones, Douglas R, Stewart, Stephen J, Chanock, Neal D, Freedman, Maria Teresa, Landi, Veronica, Höiom, Susana, Puig, Nelleke, Gruis, Xiaohong R, Yang, Paola, Ghiorzo, and Alisa M, Goldstein
- Subjects
Pancreatic Neoplasms ,Germ Cells ,Humans ,Genetic Predisposition to Disease ,Cyclin-Dependent Kinase Inhibitor p16 ,Carcinoma, Pancreatic Ductal ,Cyclin-Dependent Kinase Inhibitor Proteins - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a component of familial melanoma due to germline pathogenic variants (GPVs) inWe analyzed 189 cancer predisposition genes using parametric rare-variant association (RVA) tests and nonparametric permutation tests to identify gene-level associations in PDAC for patients with (In RVA tests,These results suggest that variants in other genes likely play a role in PDAC in all patients and that PDAC in
- Published
- 2022
23. PCCR Study for Italy: Meat Consumption, Preparation, and Meat-Derived Carcinogens on the Risk of Sporadic Pancreatic Cancer.
- Author
-
Gleb R. Haynatzki, Vera R. Haynatzka, Paola Ghiorzo, and Simon Sherman
- Published
- 2009
- Full Text
- View/download PDF
24. Novel MAPK/AKT-impairing germline NRAS variant identified in a melanoma-prone family
- Author
-
Jung Kim, Ketty Peris, Kristie Jones, Eduardo Nagore, Alex Stratigos, Paola Ghiorzo, Bin Zhu, Kevin M. Brown, Belynda Hicks, Mai Xu, Hyunbum Jang, Donato Calista, Dario Consonni, Laura Mendoza, Ruth Nussinov, Douglas R. Stewart, Maria Teresa Landi, Xiaohong Rose Yang, Mingzhen Zhang, Nicholas K. Hayward, Thorkell Andresson, Chiara Menin, Maria Concetta Fargnoli, Michael R. Sargen, Alisa M. Goldstein, Margaret A. Tucker, Susana Puig, Angela Cecilia Pesatori, and Tongwu Zhang
- Subjects
MAPK/ERK pathway ,Proto-Oncogene Proteins B-raf ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Skin Neoplasms ,Familial cancer ,In vitro characterization ,Melanoma ,Molecular modeling ,NRAS gene ,Rare variant ,GTPase ,Biology ,Article ,Germline ,GTP Phosphohydrolases ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Gene ,Protein kinase B ,Germ-Line Mutation ,Genetics (clinical) ,Exome sequencing ,Membrane Proteins ,medicine.disease ,Human genetics ,Oncology ,Cancer research ,Guanosine Triphosphate ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,Proto-Oncogene Proteins c-akt - Abstract
While several high-penetrance melanoma risk genes are known, variation in these genes fail to explain melanoma susceptibility in a large proportion of high-risk families. As part of a melanoma family sequencing study, including 435 families from Mediterranean populations, we identified a novel NRAS variant (c.170A>C, p.D57A) in a melanoma-prone family. This variant is absent in exomes in gnomAD, ESP, UKBiobank, and the 1000 Genomes Project, as well as in 11 273 Mediterranean individuals and 109 melanoma-prone families from the US and Australia. This variant occurs in the GTP-binding pocket of NRAS. Differently from other RAS activating alterations, NRAS D57A expression is unable to activate MAPK-pathway both constitutively and after stimulation but enhances EGF-induced PI3K-pathway signaling in serum starved conditions in vitro. Consistent with in vitro data demonstrating that NRAS D57A does not enrich GTP binding, molecular modeling suggests that the D57A substitution would be expected to impair Mg2+ binding and decrease nucleotide-binding and GTPase activity of NRAS. While we cannot firmly establish NRAS c.170A>C (p.D57A) as a melanoma susceptibility variant, further investigation of NRAS as a familial melanoma gene is warranted.
- Published
- 2021
25. Clinical Significance of Germline Pathogenic Variants among 51 Cancer Predisposition Genes in an Unselected Cohort of Italian Pancreatic Cancer Patients
- Author
-
Alberto Puccini, Marta Ponzano, Bruna Dalmasso, Irene Vanni, Annalice Gandini, Silvia Puglisi, Roberto Borea, Malvina Cremante, William Bruno, Virginia Andreotti, Eleonora Allavena, Valentino Martelli, Fabio Catalano, Massimiliano Grassi, Maria Laura Iaia, Chiara Pirrone, Alessandro Pastorino, Giuseppe Fornarini, Stefania Sciallero, Paola Ghiorzo, and Lorenza Pastorino
- Subjects
pancreatic cancer ,genetics ,DNA Damage Repair—Homologous Recombination Deficiency (DDR-HRD) ,hereditary cancer syndromes ,germline ,multigene panel testing ,BRCA ,CDKN2A ,overall survival ,Cancer Research ,Oncology - Abstract
Multigene germline panel testing is recommended for Pancreatic Cancer (PC) patients; however, for non-BRCA1/2 genes, the clinical utility is unclear. A comprehensive multi-gene assessment in unselected Italian PC patients is missing. We evaluated the prevalence and impact of Pathogenic Variants (PV) in 51 PC susceptibility genes in a real-world series of 422 Italian PC patients unselected for Family History (FH), compared the clinical characteristics and conducted survival analyses. 17% of patients had PVs (70/422), mainly in BRCA1/2 (4.5%, all 50 y), ATM (2.1%). PV carriers were younger (64 vs. 67; p = 0.02) and had more frequent personal/FH of PC, melanoma and breast/ovarian cancer (all p < 0.05). The Overall Survival (OS) was longer in patients carrying PVs (HR 0.78; p = 0.090), comprising ATM carriers (HR 0.33; p = 0.054). In the oxaliplatin-treated subset, PV carriers showed better control of the disease, although this was not statistically significant (67% vs. 56%). CDKN2A, BRCA2 and ATM were the most frequently altered genes. ATM PVs were positively associated with OS in 41% of PV carriers, 60% of whom carried CDKN2A,BRCA2 or ATM PVs, had negative FH and would have been missed by traditional referral. Thus, CDKN2A and ATM should be added to BRCA1/2 testing regardless of FH.
- Published
- 2022
26. Implementation of preventive and predictive BRCA testing in patients with breast, ovarian, pancreatic, and prostate cancer: a position paper of Italian Scientific Societies
- Author
-
Russo, A, Incorvaia, L, Capoluongo, E, Tagliaferri, P, Gori, S, Cortesi, L, Genuardi, M, Turchetti, D, De Giorgi, U, Di Maio, M, Barberis, M, Dessena, M, Del Re, M, Lapini, A, Luchini, C, Jereczek-Fossa, B A, Sapino, A, Cinieri, S, Italian Scientific Societies Collaborators, Giordano, Beretta, Maria Angela Bella, Sergio, Bracarda, Nicoletta, Colombo, Vincenza, Conteduca, Lucia Del Mastro, Antonio, Galvano, Valerio, Gristina, Valentina, Guarneri, Nicla La Verde, Domenica, Lorusso, Paolo, Marchetti, Nicola, Normanno, Laura, Ottini, Matilde, Pensabene, Sandro, Pignata, Giuseppe, Procopio, Enrico, Ricevuto, Nicola, Silvestris, Pierfrancesco, Tassone, Marcello, Tucci, Vittorio, Donato, Silvia, Carrara, Paiella, Salvatore, Oreste, Gentilini, Roberta, Gunelli, Fabrizio, Nicolis, Fiamma, Buttitta, Maurizio, Colecchia, Matteo, Fassan, Umberto, Malapelle, Antonio, Marchetti, Caterina, Marchiò, Scarpa, Aldo, Mauro, Truini, Zamboni, Giuseppe, Massimo, Gion, Chiara, Trevisiol, Alessandro, Gronchi, Romano, Danesi, Vito Di Marco, Paola, Carrera, Paola, Ghiorzo, Barbara, Pasini, Liliana, Varesco, Walter, Artibani, Giuseppe, Ludovico, Ornella, Campanella, Simona, Vatrano, Enrico, Tagliafico, Russo A., Incorvaia L., Capoluongo E., Tagliaferri P., Gori S., Cortesi L., Genuardi M., Turchetti D., De Giorgi U., Di Maio M., Barberis M., Dessena M., Del Re M., Lapini A., Luchini C., Jereczek-Fossa B.A., Sapino A., Cinieri S., Beretta G., Bella M.A., Bracarda S., Colombo N., Conteduca V., Del Mastro L., Galvano A., Gristina V., Guarneri V., La Verde N., Lorusso D., Marchetti P., Normanno N., Ottini L., Pensabene M., Pignata S., Procopio G., Ricevuto E., Silvestris N., Tassone P., Tucci M., Donato V., Carrara S., Paiella S., Gentilini O., Gunelli R., Nicolis F., Buttitta F., Colecchia M., Fassan M., Malapelle U., Marchetti A., Marchio C., Scarpa A., Truini M., Zamboni G., Gion M., Trevisiol C., Gronchi A., Danesi R., Di Marco V., Carrera P., Ghiorzo P., Pasini B., Varesco L., Artibani W., Ludovico G., Campanella O., Vatrano S., Tagliafico E., Russo, A, Incorvaia, L, Capoluongo, E, Tagliaferri, P, Gori, S, Cortesi, L, Genuardi, M, Turchetti, D, De Giorgi, U, Di Maio, M, Barberis, M, Dessena, M, Del Re, M, Lapini, A, Luchini, C, Jereczek-Fossa, B A, Sapino, A, Cinieri, S, Jereczek-Fossa, B, Beretta, G, Bella, M, Bracarda, S, Colombo, N, Conteduca, V, Del Mastro, L, Galvano, A, Gristina, V, Guarneri, V, La Verde, N, Lorusso, D, Marchetti, P, Normanno, N, Ottini, L, Pensabene, M, Pignata, S, Procopio, G, Ricevuto, E, Silvestris, N, Tassone, P, Tucci, M, Donato, V, Carrara, S, Paiella, S, Gentilini, O, Gunelli, R, Nicolis, F, Buttitta, F, Colecchia, M, Fassan, M, Malapelle, U, Marchetti, A, Marchio, C, Scarpa, A, Truini, M, Zamboni, G, Gion, M, Trevisiol, C, Gronchi, A, Danesi, R, Di Marco, V, Carrera, P, Ghiorzo, P, Pasini, B, Varesco, L, Artibani, W, Ludovico, G, Campanella, O, Vatrano, S, and Tagliafico, E
- Subjects
Societies, Scientific ,Male ,Ovarian Neoplasms ,Cancer Research ,genetic counseling ,BRCA ,BRCA testing ,BRCA-related cancer ,BRCA1 ,BRCA2 ,PARP inhibitors ,pancreatic ductal adenocarcinoma ,Prostatic Neoplasms ,Scientific ,Settore MED/03 - GENETICA MEDICA ,Female ,Humans ,Italy ,Pancreatic Neoplasms ,PARP inhibitor ,Oncology ,Societies - Abstract
Constitutional BRCA1/BRCA2 pathogenic or likely pathogenic variants (PVs) are associated with an increased risk for developing breast and ovarian cancers. Current evidence indicates that BRCA1/2 PVs are also associated with pancreatic cancer, and that BRCA2 PVs are associated with prostate cancer risk. The identification of carriers of constitutional PVs in the BRCA1/2 genes allows the implementation of individual and family prevention pathways, through validated screening programs and risk-reducing strategies. According to the relevant and increasing therapeutic predictive implications, the inclusion of BRCA testing in the routine management of patients with breast, ovarian, pancreatic and prostate cancers represent a key requirement to optimize medical or surgical therapeutic and prevention decision-making, and access to specific anticancer therapies. Therefore, accurate patient selection, the use of standardized and harmonized procedures, and adherence to homogeneous testing criteria, are essential elements to implement BRCA testing in clinical practice. This consensus position paper has been developed and approved by a multidisciplinary Expert Panel of 64 professionals on behalf of the AIOM-AIRO-AISP-ANISC-AURO-Fondazione AIOM-SIAPEC/IAP-SIBioC-SICO-SIF-SIGE-SIGU-SIU-SIURO-UROP Italian Scientific Societies, and a patient association (aBRCAdaBRA Onlus). The working group included medical, surgical and radiation oncologists, medical and molecular geneticists, clinical molecular biologists, surgical and molecular pathologists, organ specialists such as gynecologists, gastroenterologists and urologists, and pharmacologists. The manuscript is based on the expert consensus and reports the best available evidence, according to the current eligibility criteria for BRCA testing and counseling, it also harmonizes with current Italian National Guidelines and Clinical Recommendations.
- Published
- 2022
27. Functional annotation and investigation of the 10q24.33 melanoma risk locus identifies a common variant that influences transcriptional regulation of OBFC1
- Author
-
Massimiliano Scalvenzi, Alessandro Testori, Michiel Vermeulen, Annalaura Montella, Nicola Zambrano, Marijke P. Baltissen, Vito Alessandro Lasorsa, Marianna Avitabile, Kevin M. Brown, Sueva Cantalupo, Flora Cimmino, Antonella Cardinale, Mai Xu, Mark M. Iles, Achille Iolascon, Paola Ghiorzo, Fabrizio Ayala, Ferdinando Bonfiglio, Mariangela Succoio, Matthew Law, Daniela Formicola, Matteo Esposito, Mario Capasso, Cardinale, Antonella, Cantalupo, Sueva, Lasorsa, Vito Alessandro, Montella, Annalaura, Cimmino, Flora, Succoio, Mariangela, Vermeulen, Michiel, Baltissen, Marijke P, Esposito, Matteo, Avitabile, Marianna, Formicola, Daniela, Testori, Alessandro, Bonfiglio, Ferdinando, Ghiorzo, Paola, Scalvenzi, Massimiliano, Ayla, Fabrizio, Zambrano, Nicola, Iles, Mark M, Xu, Mai, Law, Matthew H, Brown, Kevin M, Iolascon, Achille, and Capasso, Mario
- Subjects
Genetics ,Skin Neoplasms ,Single-nucleotide polymorphism ,Locus (genetics) ,General Medicine ,Biology ,Polymorphism, Single Nucleotide ,Minor allele frequency ,Humans ,Genetic Predisposition to Disease ,General Article ,Allele ,Enhancer ,Gene ,Transcription factor ,Molecular Biology ,Melanoma ,Genetics (clinical) ,Epigenomics - Abstract
The 10q24.33 locus is known to be associated with susceptibility to cutaneous malignant melanoma (CMM), but the mechanisms underlying this association have been not extensively investigated. We carried out an integrative genomic analysis of 10q24.33 using epigenomic annotations and in vitro reporter gene assays to identify regulatory variants. We found two putative functional single nucleotide polymorphisms (SNPs) in an enhancer and in the promoter of OBFC1, respectively, in neural crest and CMM cells, one, rs2995264, altering enhancer activity. The minor allele G of rs2995264 correlated with lower OBFC1 expression in 470 CMM tumors and was confirmed to increase the CMM risk in a cohort of 484 CMM cases and 1801 controls of Italian origin. Hi-C and chromosome conformation capture (3C) experiments showed the interaction between the enhancer-SNP region and the promoter of OBFC1 and an isogenic model characterized by CRISPR-Cas9 deletion of the enhancer-SNP region confirmed the potential regulatory effect of rs2995264 on OBFC1 transcription. Moreover, the presence of G-rs2995264 risk allele reduced the binding affinity of the transcription factor MEOX2. Biologic investigations showed significant cell viability upon depletion of OBFC1, specifically in CMM cells that were homozygous for the protective allele. Clinically, high levels of OBFC1 expression associated with histologically favorable CMM tumors. Finally, preliminary results suggested the potential effect of decreased OBFC1 expression on telomerase activity in tumorigenic conditions. Our results support the hypothesis that reduced expression of OBFC1 gene through functional heritable DNA variation can contribute to malignant transformation of normal melanocytes.
- Published
- 2022
28. The challenge of the Molecular Tumor Board empowerment in clinical oncology practice: A Position Paper on behalf of the AIOM- SIAPEC/IAP-SIBioC-SIC-SIF-SIGU-SIRM Italian Scientific Societies
- Author
-
Antonio Russo, Lorena Incorvaia, Ettore Capoluongo, Pierosandro Tagliaferri, Antonio Galvano, Marzia Del Re, Umberto Malapelle, Rita Chiari, Pierfranco Conte, Romano Danesi, Matteo Fassan, Roberto Ferrara, Maurizio Genuardi, Paola Ghiorzo, Stefania Gori, Fiorella Guadagni, Antonio Marchetti, Paolo Marchetti, Massimo Midiri, Nicola Normanno, Francesco Passiglia, Carmine Pinto, Nicola Silvestris, Giovanni Tallini, Simona Vatrano, Bruno Vincenzi, Saverio Cinieri, Giordano Beretta, Russo, Antonio, Incorvaia, Lorena, Capoluongo, Ettore, Tagliaferri, Pierosandro, Galvano, Antonio, Del Re, Marzia, Malapelle, Umberto, Chiari, Rita, Conte, Pierfranco, Danesi, Romano, Fassan, Matteo, Ferrara, Roberto, Genuardi, Maurizio, Ghiorzo, Paola, Gori, Stefania, Guadagni, Fiorella, Marchetti, Antonio, Marchetti, Paolo, Midiri, Massimo, Normanno, Nicola, Passiglia, Francesco, Pinto, Carmine, Silvestris, Nicola, Tallini, Giovanni, Vatrano, Simona, Vincenzi, Bruno, Cinieri, Saverio, Beretta, Giordano, Russo, A., Incorvaia, L., Capoluongo, E., Tagliaferri, P., Galvano, A., Del Re, M., Malapelle, U., Chiari, R., Conte, P., Danesi, R., Fassan, M., Ferrara, R., Genuardi, M., Ghiorzo, P., Gori, S., Guadagni, F., Marchetti, A., Marchetti, P., Midiri, M., Normanno, N., Passiglia, F., Pinto, C., Silvestris, N., Tallini, G., Vatrano, S., Vincenzi, B., Cinieri, S., Beretta, G., Russo A., Incorvaia L., Capoluongo E., Tagliaferri P., Galvano A., Del Re M., Malapelle U., Chiari R., Conte P., Danesi R., Fassan M., Ferrara R., Genuardi M., Ghiorzo P., Gori S., Guadagni F., Marchetti A., Marchetti P., Midiri M., Normanno N., Passiglia F., Pinto C., Silvestris N., Tallini G., Vatrano S., Vincenzi B., Cinieri S., and Beretta G.
- Subjects
Societies, Scientific ,Molecular ,Scientific ,Precision oncology ,Hematology ,Genomics ,Molecular profiling ,Molecular tumor board ,Mutational oncology ,Settore MED/03 - GENETICA MEDICA ,Medical Oncology ,Oncology ,Italy ,Neoplasms ,Humans ,Genomic ,Neoplasm ,Societies ,Human - Abstract
The development of innovative technologies and the advances in the genetics and genomics, have offered new opportunities for personalized treatment in oncology. Although the selection of the patient based on the molecular characteristics of the neoplasm has the potential to revolutionize the therapeutic scenario of oncology, this approach is extremely challenging. The access, homogeneity, and economic sustainability of the required genomic tests should be warranted in the clinical practice, as well as the specific scientific and clinical expertise for the choice of medical therapies. All these elements make essential the collaboration of different specialists within the Molecular Tumor Boards (MTBs). In this position paper, based on experts' opinion, the AIOM-SIAPEC/IAP-SIBioC-SIC-SIF-SIGU-SIRM Italian Scientific Societies critically discuss the available molecular profiling technologies, the proposed criteria for the selection of patients candidate for evaluation by the MTB, the criteria for the selection and analysis of biological samples, and the regulatory and pharmaco-economic issues.
- Published
- 2021
29. NFB-09. Treatment of cardiac fibroma in aPTCH1-mutated Gorlin syndrome with medulloblastoma
- Author
-
Gianluca Piccolo, Antonio Verrico, Gianluca Trocchio, Maria Derchi, Alessandra Siboldi, Nicola Stagnaro, Marco Crocco, Angela Di Giannatale, Paola Ghiorzo, Claudia Milanaccio, and Maria Luisa Garrè
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
A 2,5-year-old girl presented vomiting episodes associated with severe motor delay, macrocephaly, and distal hypotonia. Brain MRI showed hydrocephalus and a non-metastatic lesion in the posterior fossa. A pre-operative ECG showed isodifasic T waves in leads V4R and V1, follow-up was recommended. Histology after gross total resection was consistent with Desmoplastic/Nodular Medulloblastoma. At chest X-rays a bifid rib was noted, leading to the diagnosis of Gorlin syndrome (GS; c.3306 + 1G>T in PTCH1). During chemotherapy, ventricular tachycardia (VT) occurred, requiring synchronized electrical cardioversion. An echocardiogram revealed an echogenic mass of the left ventricle free wall, but normal coronaries and ventricular function. Cardiac MRI (CMR) confirmed a 21x40x38mm mass, with eccentric development and intense homogeneous enhancement, indicative of cardiac fibroma (CF). A computed tomography excluded local calcifications. Therapy with amiodarone and beta-blocker was initiated. TREATMENT STRATEGY: priority to chemotherapy vs cardiac surgery; full-dose chemotherapy, preferring drugs with minor cardiotoxicity; administration in ICU under continuous vital parameters and ECG-monitoring. Other three VTs occurred during treatment or anesthesia, resolved after electrical cardioversion (unsuccessful attempts with i.v. adenosine and amiodarone). Lacking specific guidelines concerning CF in GS, a wait-and-see approach was preferred with close tumor follow-up and regular cardiological assessment (ECG, stress-test, Holter monitoring, CMR). No further arrhythmias were recorded in a 10-year-long follow-up and CMR confirmed CF stability. Medulloblastoma has never recurred. PTCH1 variants are rarely associated with medulloblastoma (
- Published
- 2022
30. Insights into Mechanisms of Tumorigenesis in Neuroendocrine Neoplasms
- Author
-
Federica Grillo, Lorenza Pastorino, Manuela Albertelli, William Bruno, and Paola Ghiorzo
- Subjects
QH301-705.5 ,chromatin stability ,Carcinogenesis ,Computational biology ,Review ,Biology ,medicine.disease_cause ,Catalysis ,Inorganic Chemistry ,splicing ,Death-associated protein 6 ,Basic research ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,MEN1 ,Biology (General) ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,Spectroscopy ,genomic integrity ,Cell dedifferentiation ,Tumor ,Organic Chemistry ,General Medicine ,Cell Dedifferentiation ,animal models ,Chromatin ,Computer Science Applications ,Chemistry ,Animal models ,Chromatin stability ,Genomic integrity ,Neuroendocrine tumors ,Splicing ,Tumorigenesis ,Mutation ,Neuroendocrine Tumors ,tumorigenesis ,RNA splicing ,Identification (biology) ,Biomarkers - Abstract
Genomic studies have identified some of the most relevant genetic players in Neuroendocrine Neoplasm (NEN) tumorigenesis. However, we are still far from being able to draw a model that encompasses their heterogeneity, elucidates the different biological effects consequent to the identified molecular events, or incorporates extensive knowledge of molecular biomarkers and therapeutic targets. Here, we reviewed recent insights in NEN tumorigenesis from selected basic research studies on animal models, highlighting novel players in the intergenic cooperation and peculiar mechanisms including splicing dysregulation, chromatin stability, or cell dedifferentiation. Furthermore, models of tumorigenesis based on composite interactions other than a linear progression of events are proposed, exemplified by the involvement in NEN tumorigenesis of genes regulating complex functions, such as MEN1 or DAXX. Although limited by interspecies differences, animal models have proved helpful for the more in-depth study of every facet of tumorigenesis, showing that the identification of driver mutations is only one of the many necessary steps and that other mechanisms are worth investigating.
- Published
- 2021
31. Efficacy of BRAF and MEK inhibition in patients with BRAF-mutant advanced melanoma and germline CDKN2A pathogenic variants
- Author
-
Francesco Spagnolo, Enrica Teresa Tanda, Miriam Potrony, Remco van Doorn, Ellen Kapiteijn, Susana Puig, Paola Queirolo, Bruna Dalmasso, Hildur Helgadottir, and Paola Ghiorzo
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Tumor suppressor gene ,melanoma susceptibility ,medicine.medical_treatment ,Genetic counseling ,Germline ,Targeted therapy ,03 medical and health sciences ,CDKN2A ,0302 clinical medicine ,Internal medicine ,melanoma ,Medicine ,neoplasms ,MEK inhibitors ,RC254-282 ,genetic counseling ,business.industry ,Communication ,Melanoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,BRAF inhibitors ,medicine.disease ,targeted therapy ,Clinical trial ,030104 developmental biology ,Melanoma susceptibility ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,business - Abstract
Simple Summary In our study, we retrospectively collected data of patients with germline CDKN2A pathogenic variants who received targeted therapy for advanced melanoma across four European centers. Since loss of CDKN2A function may intrinsically limit the activity of MAPK-directed targeted therapy, we decided to assess whether patients with germline CDKN2A pathogenic variants may achieve suboptimal results with BRAF and MEK inhibitors. To the best of our knowledge, this is the first study reporting on patients with BRAF-mutant advanced melanoma and a germline CDKN2A pathogenic variant who received treatment with BRAF with or without MEK inhibitors. Despite the limitations of our study, mostly due to the rare frequency of CDKN2A pathogenic variants, a challenge for the conduction of prospective trials with proper sample size, our results support treatment with targeted therapy in this subset of patients. Abstract Inherited pathogenic variants (PVs) in the CDKN2A tumor suppressor gene are among the strongest risk factors for cutaneous melanoma. Dysregulation of the p16/RB1 pathway may intrinsically limit the activity of MAPK-directed therapy due to the interplay between the two pathways. In our study, we assessed, for the first time, whether patients with germline CDKN2A PVs achieve suboptimal results with BRAF inhibitors (BRAFi)+/−MEK inhibitors (MEKi). We compared the response rate of nineteen CDKN2A PVs carriers who received first-line treatment with BRAFi+/−MEKi with an expected rate derived from phase III trials and “real-world” studies. We observed partial response in 16/19 patients (84%), and no complete responses. The overall response rate was higher than that expected from phase III trials (66%), although not statistically significant (p-value = 0.143; 95% CI = 0.60–0.97); the difference was statistically significant (p-value = 0.019; 95% CI = 0.62–0.97) in the comparison with real-world studies (57%). The clinical activity of BRAFi+/−MEKi in patients with germline CDKN2A PV was not inferior to that of clinical trials and real-world studies, which is of primary importance for clinical management and genetic counseling of this subgroup of patients.
- Published
- 2021
32. Birth cohort-specific trends of sun-related behaviors among individuals from an international consortium of melanoma-prone families
- Author
-
Julia Newton-Bishop, Montserrat Molgó, Julio C. Salas-Alanis, David E. Elder, Alejandra Larre-Borges, Johan Hansson, Blanca Carlos-Ortega, Lu Qian, Graham J. Mann, Veronica Höiom, Helen Schmid, John Charles A. Lacson, Luis Alberto Ribeiro Froes, Shawn A. Zamani, Nandita Mitra, Anne E. Cust, Karin Wadt, Scarlet H. Doyle, D. Timothy Bishop, Francisco Cuellar, Nelleke A. Gruis, Barbara Perić, Thaís Corsetti Grazziotin, Anne-Marie Gerdes, Claudia Balestrini, Håkan Olsson, Paola Ghiorzo, Esther Azizi, Christian Ingvar, Jacoba J. Out-Luiting, Marko Hočevar, Susana Puig, Elizabeth A. Holland, Dace Pjanova, Luciana Facure Moredo, Gilles Landman, Peter A. Kanetsky, William Bruno, Froes, Luis Alberto Ribeiro [0000-0002-1140-3046], Kanetsky, Peter A [0000-0002-5567-9618], and Apollo - University of Cambridge Repository
- Subjects
Risk-taking (Psychology) ,medicine.medical_specialty ,Radiació solar ,Family medicine ,Skin Neoplasms ,Sunbed ,Risk factors in diseases ,Sun exposure ,Population ,Sunburn ,High-risk families ,Melanoma ,Skin Cancer ,Sun-related behaviors ,Sunscreen use ,Trends ,Humans ,Retrospective Studies ,Sunscreening Agents ,Odds ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Conducta de risc (Psicologia) ,Epidemiology ,medicine ,Solar radiation ,Skin cancer ,030212 general & internal medicine ,education ,skin and connective tissue diseases ,Càncer de pell ,education.field_of_study ,business.industry ,Factors de risc en les malalties ,Public health ,Public Health, Environmental and Occupational Health ,Retrospective cohort study ,medicine.disease ,Medicina familiar ,Biostatistics ,Public aspects of medicine ,RA1-1270 ,business ,Research Article ,Demography - Abstract
Funder: Coordenação de Aperfeiçoamento de Pessoal de Nível Superior; doi: http://dx.doi.org/10.13039/501100002322, Funder: Radiumhemmets Forskningsfonder; doi: http://dx.doi.org/10.13039/501100007232, Funder: Swedish Cancer Society, Funder: Lunds Universitet Paulsson Trust, Funder: CIBER de Enfermedades Raras of the Instituto de Salud Carlos III, Funder: European Regional Development Fund; doi: http://dx.doi.org/10.13039/501100008530, Funder: Diagnoptics, Funder: CERCA Programme Generalitat de Catalunya, Funder: Esther Koplowitz Center, Barcelona, Spain, Funder: Comision Honoraria de Lucha Contra el Cancer, CSIC, Fundacion Manuel Perez, Montevideo, Uruguay, BACKGROUND: Individuals from melanoma-prone families have similar or reduced sun-protective behaviors compared to the general population. Studies on trends in sun-related behaviors have been temporally and geographically limited. METHODS: Individuals from an international consortium of melanoma-prone families (GenoMEL) were retrospectively asked about sunscreen use, sun exposure (time spent outside), sunburns, and sunbed use at several timepoints over their lifetime. Generalized linear mixed models were used to examine the association between these outcomes and birth cohort defined by decade spans, after adjusting for covariates. RESULTS: A total of 2407 participants from 547 families across 17 centers were analyzed. Sunscreen use increased across subsequent birth cohorts, and although the likelihood of sunburns increased until the 1950s birth cohort, it decreased thereafter. Average sun exposure did not change across the birth cohorts, and the likelihood of sunbed use increased in more recent birth cohorts. We generally did not find any differences in sun-related behavior when comparing melanoma cases to non-cases. Melanoma cases had increased sunscreen use, decreased sun exposure, and decreased odds of sunburn and sunbed use after melanoma diagnosis compared to before diagnosis. CONCLUSIONS: Although sunscreen use has increased and the likelihood of sunburns has decreased in more recent birth cohorts, individuals in melanoma-prone families have not reduced their overall sun exposure and had an increased likelihood of sunbed use in more recent birth cohorts. These observations demonstrate partial improvements in melanoma prevention and suggest that additional intervention strategies may be needed to achieve optimal sun-protective behavior in melanoma-prone families.
- Published
- 2021
33. Definition and management of colorectal polyposis not associated with APC/MUTYH germline pathogenic variants: AIFEG consensus statement
- Author
-
Emanuele Damiano Luca Urso, Maurizio Ponz de Leon, Marco Vitellaro, Guglielmo Niccolò Piozzi, Quoc Riccardo Bao, Aline Martayan, Andrea Remo, Vittoria Stigliano, Cristina Oliani, Emanuela Lucci Cordisco, Salvatore Pucciarelli, Guglielmina Nadia Ranzani, Alessandra Viel, Francesca Adami, Elisa Alducci, Lucia Amadori, Valentina Arcangeli, Luisa Balestrino, Daniela Barana, Lucio Bertario, Bernardo Bonanni, Stefania Boni, Pierluigi Bullian, Fiorella Carbonardi, Ileana Carnevali, Paola Castelli, Francesco Celotto, Giulia Cini, Gino Crivellari, Duilio Della Libera, Anastasia Dell'elice, Maria Digennaro, Alessandra D'urso, Antonella Fabretto, Daniele Fanale, Irene Feroce, Daniela Furlan, Paola Ghiorzo, Mara Giacché, Milena Gusella, Barbara Liserre, Isabella Mammi, Stefania Massuras, Daniela Mazzà, Eleonora Mollica, Alberto Morabito, Giorgia Nardo, Flavia Palermo, Elena Panizza, Margherita Patruno, Monica Pedroni, Valeria Grazia Maria Pensotti, Guglielmo Niccolo Piozzi, Simonetta Pozzi, Silvia Presi, Marta Puzzono, Mila Ravegnani, Maria Teresa Ricci, Luca Roncucci, Giovanni Battsita Rossi, Elena Maria Sala, Lupe Sanchez Mete, Daniele Sandonà, Stefania Sciallero, Davide Serrano, Stefano Signoroni, Francesca Spina, Monica Taborelli, Gianluca Tedaldi, Maria Grazia Tibiletti, Silvia Tognazzo, Gianluca Tolva, Cristina Maria Concetta Trovato, Daniela Turchetti, Dora Varvara, Caterina Vivanet, Stefania Zovato, Raffaella Alessia Zuppardo, Urso E.D.L., Ponz de Leon M., Vitellaro M., Piozzi G.N., Bao Q.R., Martayan A., Remo A., Stigliano V., Oliani C., Lucci Cordisco E., Pucciarelli S., Ranzani G.N., Viel A., Adami F., Alducci E., Amadori L., Arcangeli V., Balestrino L., Barana D., Bertario L., Bonanni B., Boni S., Bullian P., Carbonardi F., Carnevali I., Castelli P., Celotto F., Cini G., Crivellari G., Libera D.D., Dell'elice A., Digennaro M., D'urso A., Fabretto A., Fanale D., Feroce I., Furlan D., Ghiorzo P., Giacche M., Gusella M., Liserre B., Mammi I., Massuras S., Mazza D., Mollica E., Morabito A., Nardo G., Palermo F., Panizza E., Patruno M., Pedroni M., Pensotti V.G.M., Pozzi S., Presi S., Puzzono M., Ravegnani M., Ricci M.T., Roncucci L., Rossi G.B., Sala E.M., Mete L.S., Sandona D., Sciallero S., Serrano D., Signoroni S., Spina F., Taborelli M., Tedaldi G., Tibiletti M.G., Tognazzo S., Tolva G., Trovato C.M.C., Turchetti D., Varvara D., Vivanet C., Zovato S., and Zuppardo R.A.
- Subjects
Oncology ,medicine.medical_specialty ,Gastrointestinal tumors ,Colorectal cancer ,Surgical Management ,Colorectal polyposis ,Germline ,03 medical and health sciences ,Cancer Genetic ,0302 clinical medicine ,MUTYH ,Internal medicine ,medicine ,Cancer Genetics ,Polyposis coli ,Hepatology ,Pathogenic mutation ,business.industry ,Colorectal polyposis not associated with APC/MUTYH mutation ,Polyposis management guideline ,Gastroenterology ,Expert consensus ,Endoscopic surveillance ,medicine.disease ,Consensus development conference ,030220 oncology & carcinogenesis ,030211 gastroenterology & hepatology ,business - Abstract
An expert consensus panel convened by the Italian Association for Inherited and Familial Gastrointestinal Tumors (Associazione Italiana per lo Studio della Familiarita ed Ereditarieta dei Tumori Gastrointestinali, AIFEG) reviewed the literature and agreed on a number of position statements regarding the definition and management of polyposis coli without an identified pathogenic mutation on the APC or MUTYH genes, defined in the document as NAMP (non-APC/MUTYH polyposis).
- Published
- 2021
34. MC1R variants in relation to naevi in melanoma cases and controls: a pooled analysis from the M-SKIP project
- Author
-
M C Fargnoli, Hongmei Nan, Sara Gandini, Evangelos Evangelou, Francesco Sera, José C. García-Borrón, A. De Nicolo, Alexandros Stratigos, Nelleke A. Gruis, Patrick Maisonneuve, David Polsky, DeAnn Lazovich, Cristina Pellegrini, Sara Raimondi, Julia Newton-Bishop, Julian Little, Paola Ghiorzo, I. Stefanaki, Gabriella Guida, Gloria Ribas, Peter A. Kanetsky, Maria Teresa Landi, Katerina P. Kypreou, and S. Puig
- Subjects
Oncology ,medicine.medical_specialty ,Nevus, Pigmented ,Skin Neoplasms ,business.industry ,Melanoma ,Case-control study ,Dermatology ,medicine.disease ,Article ,Infectious Diseases ,Pooled analysis ,Genetic epidemiology ,Risk Factors ,Internal medicine ,Meta-analysis ,Case-Control Studies ,Cutaneous melanoma ,medicine ,Dysplastic nevus ,Nevus ,Humans ,business ,Receptor, Melanocortin, Type 1 - Published
- 2020
35. Efficacy of novel immunotherapy regimens in patients with metastatic melanoma with germline CDKN2A mutations
- Author
-
Miriam Potrony, Håkan Olsson, Richard F. Kefford, Cristina Carrera, Susana Puig, Remco van Doorn, Paola Queirolo, Ellen Kapiteijn, Veronica Höiom, Lorenza Pastorino, Hildur Helgadottir, Göran Jönsson, Max Levin, Martin Lauss, and Paola Ghiorzo
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,cdkn2a ,Immunoteràpia ,Phases of clinical research ,030105 genetics & heredity ,medicine.disease_cause ,Germline ,Metastasis ,CDKN2A ,CTLA-4 Antigen ,Neoplasm Metastasis ,Immune Checkpoint Inhibitors ,Melanoma ,familial melanoma ,Genetics (clinical) ,Clinical Trials as Topic ,Mutation ,Middle Aged ,immunotherapy ,metastatic melanoma ,tumor mutation burden ,Female ,Adult ,medicine.medical_specialty ,Immunotheraphy ,03 medical and health sciences ,Metàstasi ,Internal medicine ,Cancer Genetics ,Genetics ,medicine ,Humans ,Risk factor ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,Germ-Line Mutation ,Aged ,business.industry ,Immunotherapy ,Ipilimumab ,Clinical trial ,030104 developmental biology ,Cutaneous melanoma ,business - Abstract
BackgroundInherited CDKN2A mutation is a strong risk factor for cutaneous melanoma. Moreover, carriers have been found to have poor melanoma-specific survival. In this study, responses to novel immunotherapy agents in CDKN2A mutation carriers with metastatic melanoma were evaluated.MethodsCDKN2A mutation carriers that have developed metastatic melanoma and undergone immunotherapy treatments were identified among carriers enrolled in follow-up studies for familial melanoma. The carriers’ responses were compared with responses reported in phase III clinical trials for CTLA-4 and PD-1 inhibitors. From publicly available data sets, melanomas with somatic CDKN2A mutation were analysed for association with tumour mutational load.ResultsEleven of 19 carriers (58%) responded to the therapy, a significantly higher frequency than observed in clinical trials (p=0.03, binomial test against an expected rate of 37%). Further, 6 of the 19 carriers (32%) had complete response, a significantly higher frequency than observed in clinical trials (p=0.01, binomial test against an expected rate of 7%). In 118 melanomas with somatic CDKN2A mutations, significantly higher total numbers of mutations were observed compared with 761 melanomas without CDKN2A mutation (Wilcoxon test, pConclusionPatients with CDKN2A mutated melanoma may have improved immunotherapy responses due to increased tumour mutational load, resulting in more neoantigens and stronger antitumorous immune responses.
- Published
- 2020
36. Factors Affecting Sentinel Node Metastasis in Thin (T1) Cutaneous Melanomas: Development and External Validation of a Predictive Nomogram
- Author
-
Ilaria Mattavelli, Dorothy C. Bennett, Paolo Broganelli, Caterina Longo, Konstantinos Lasithiotakis, Alessandra Chiarugi, Roberta Mortarini, Massimo Milione, Francesca Consoli, Serena Sestini, Antonio Florita, Sara Fortunato, Giovanni Pellacani, Lorenzo Borgognoni, Paolo Nardini, Andrea Anichini, David J. Adams, Hanna Eriksson, May Chan, Ausilia Maria Manganoni, Odysseas Zoras, Pietro Quaglino, Catherine A. Harwood, Umberto Cortinovis, Andrea Leva, Corrado Del Forno, Francesco Barretta, Andrew J. Hayes, Simone Ribero, Mara Cossa, Barbara Valeri, Giancarlo Pruneri, Daniele Bergamaschi, Kara Heelan, Cristina Mangas, Roberto Patuzzo, Andrea Maurichi, Vittoria Espeli, Mario Santinami, Consuelo Barbieri, Bruna Dalmasso, Julia Newton-Bishop, Rosalba Miceli, Paola Ghiorzo, G. Gallino, Nicola Pimpinelli, Jérémie Nsengimana, and Elena Morittu
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,Errata ,business.industry ,External validation ,Sentinel node metastasis ,Nomogram ,Sentinel node ,Breslow Thickness ,03 medical and health sciences ,Predictive nomogram ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Original Reports ,Biopsy ,Cutaneous melanoma ,medicine ,030212 general & internal medicine ,Radiology ,business - Abstract
PURPOSE Thin melanomas (T1; ≤ 1 mm) constitute 70% of newly diagnosed cutaneous melanomas. Regional node metastasis determined by sentinel node biopsy (SNB) is an important prognostic factor for T1 melanoma. However, current melanoma guidelines do not provide clear indications on when to perform SNB in T1 disease and stress an individualized approach to SNB that considers all clinicopathologic risk factors. We aimed to identify determinants of sentinel node (SN) status for incorporation into an externally validated nomogram to better select patients with T1 disease for SNB. PATIENTS AND METHODS The development cohort comprised 3,666 patients with T1 disease consecutively treated at the Istituto Nazionale Tumori (Milan, Italy) between 2001 and 2018; 4,227 patients with T1 disease treated at 13 other European centers over the same period formed the validation cohort. A random forest procedure was applied to the development data set to select characteristics associated with SN status for inclusion in a multiple binary logistic model from which a nomogram was elaborated. Decision curve analyses assessed the clinical utility of the nomogram. RESULTS Of patients in the development cohort, 1,635 underwent SNB; 108 patients (6.6%) were SN positive. By univariable analysis, age, growth phase, Breslow thickness, ulceration, mitotic rate, regression, and lymphovascular invasion were significantly associated with SN status. The random forest procedure selected 6 variables (not growth phase) for inclusion in the logistic model and nomogram. The nomogram proved well calibrated and had good discriminative ability in both cohorts. Decision curve analyses revealed the superior net benefit of the nomogram compared with each individual variable included in it as well as with variables suggested by current guidelines. CONCLUSION We propose the nomogram as a decision aid in all patients with T1 melanoma being considered for SNB.
- Published
- 2020
37. Genome-wide association meta-analyses combining multiple risk phenotypes provide insights into the genetic architecture of cutaneous melanoma susceptibility
- Author
-
Eduardo Nagore, Caroline Hayward, Christopher I. Amos, Douglas F. Easton, Zaida García-Casado, Julie Lang, Anjali K. Henders, Veronica Höiom, Lisa Bowdler, Kathryn P. Burdon, Laura Del Regno, Nick Orr, Per Arne Andresen, Tongwu Zhang, Rose Yang, Myriam Brossard, Eric K. Moses, Dirk Schadendorf, Laura Cattaneo, Casey Rowe, Essen-Heidelberg Investigators, Hans-Joachim Schulze, Jamie E Craig, D. Timothy Bishop, Anne E. Cust, Johan Hansson, David E. Elder, Nelleke A. Gruis, Donato Calista, Wei V. Chen, Abrar A. Qureshi, Amy Hutchinson, Pilar Galan, Leanne Wallace, Jennifer H. Barrett, Nilesh J. Samani, Maria Teresa Landi, Per Helsing, Andreas Hadjisavvas, Fengju Song, Celia Requena, Elizabeth M. Gillanders, Arantxa Rodriguez, Joan Anton Puig-Butille, Blair H. Smith, Mark Smithers, Michael Malasky, Marianna Sanna, Miriam Potrony, Maria A. Loizidou, Evangelos Evangelou, Richard A. Scolyer, Karen A. Pooley, Rachel E. Neale, Mario Falchi, Adèle C. Green, Monica Rodolfo, Ketty Peris, Licia Rivoltini, Mark M. Iles, Catherine M. Olsen, Alexander J. Stratigos, Tadeusz Dębniak, Weiyin Zhou, H. Peter Soyer, Xin Li, Margaret A. Tucker, Rajesh Kumar, Håkan Olsson, Anders Molven, Nicholas G. Martin, Anthony J. Swerdlow, Aurelie Vogt, Lars A. Akslen, Stuart MacGregor, Sarah V. Ward, Hamida Mohamdi, Bruna Dalmasso, Grant W. Montgomery, Rona M. MacKie, Esther Azizi, Gonçalo R. Abecasis, Chiara Menin, Alison M. Dunning, Ibd investigators, Kevin M. Brown, Jiali Han, Veryan Codd, Graham J. Mann, Nicholas K. Hayward, Marko Hočevar, Eitan Friedman, Richard A. Sturm, Paola Queirolo, Qtwin Investigators, Lawrie Wheeler, Lars G. Fritsche, Shenying Fang, Kiarash Khosrotehrani, Nicole A Kukutsch, Pol Gimenez-Xavier, Belynda Hicks, Matthew Zawistowski, Alessia Visconti, Jessica Harris, Chad M. Brummett, Paola Ghiorzo, andMe, David G. Hunter, Veronique Bataille, Julia Newton-Bishop, Srdjan Novaković, Amfs Investigators, Siranoush Manoukian, Jianxin Shi, Mitchell J. Machiela, G. Mark Lathrop, Josep Malvehy, Katerina P. Kypreou, Susana Puig, Dale R. Nyholt, I. Stefanaki, Maria Concetta Fargnoli, Lisa A. Simms, Kerrie McAloney, M. Malt, Adam J. Trower, Matthew Law, Lei Song, Paul D.P. Pharoah, Christian Ingvar, Jiyeon Choi, Alisa M. Goldstein, Jeffrey E. Lee, Mark Harland, Cristina Pellegrini, Daniela Massi, Sarah Simi, Scott D. Gordon, Jacobo Martinez, Florence Demenais, Kristine Jones, Graham L. Radford-Smith, David C. Whiteman, Lorenza Pastorino, Lisa Elefanti, Arcangela De Nicolo, Mario Mandalà, Juliette Randerson-Moor, Q-Mega, Jan Lubinski, Stephen J. Chanock, Marie-Françoise Avril, David L. Duffy, Helen Gogas, Nienke van der Stoep, Peter A. Kanetsky, Georgina V. Long, National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), University of Leeds, QIMR Berghofer Medical Research Institute, National and Kapodistrian University of Athens (NKUA), Ospedale Policlinico San Martino [Genoa], Universita degli studi di Genova, Toxicité environnementale, cibles thérapeutiques, signalisation cellulaire (T3S - UMR_S 1124), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Maurizio Bufalini Hospital, University of L'Aquila [Italy] (UNIVAQ), Fondazione IRCCS Istituto Nazionale Tumori - National Cancer Institute [Milan], Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana [Espagne] (FISABIO), Cyprus Institute of Neurology and Genetics, University of Athens Medical School [Athens], The University of Sydney, Universitat de Barcelona (UB), Azienda Ospedaliera Ospedale Papa Giovanni XXIII [Bergamo, Italy], University of Michigan [Ann Arbor], University of Michigan System, Instituto Valenciano de Oncologia, Veneto Institute of Oncology IOV-IRCCS [Padua, Italy], IRCCS Istituto Nazionale dei Tumori [Milano], Ninewells Hospital and Medical School [Dundee], Cyprus Institute (CyI), Fondazione 'Policlinico Universitario A. Gemelli' [Rome], Università degli Studi di Firenze = University of Florence [Firenze] (UNIFI), University of Queensland [Brisbane], Haukeland University Hospital, University of Bergen (UiB), Geisel School of Medicine at Dartmouth, Oslo University Hospital [Oslo], Service de Dermatologie [CHU Cochin], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Sackler Faculty of Medicine, Tel Aviv University [Tel Aviv], King‘s College London, Menzies School of Health Research [Australia], Charles Darwin University, The University of Texas M.D. Anderson Cancer Center [Houston], University of Leicester, Flinders University [Adelaide, Australia], West Pomeranian University of Technology Szczecin, Equipe 3: EREN- Equipe de Recherche en Epidémiologie Nutritionnelle (CRESS - U1153), Université Sorbonne Paris Nord-Centre de Recherche Épidémiologie et Statistique Sorbonne Paris Cité (CRESS (U1153 / UMR_A_1125 / UMR_S_1153)), Conservatoire National des Arts et Métiers [CNAM] (CNAM)-Université Sorbonne Paris Cité (USPC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Conservatoire National des Arts et Métiers [CNAM] (CNAM)-Université Sorbonne Paris Cité (USPC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), National Human Genome Research Institute (NHGRI), Leiden University Medical Center (LUMC), Karolinska Institutet [Stockholm], Queensland University of Technology [Brisbane] (QUT), Institute of Oncology Ljubljana, Harvard T.H. Chan School of Public Health, Lund University [Lund], German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), University of Glasgow, Statistical Genetics, and Intramural Research Program of the Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health (NIH) United States Department of Health & Human ServicesNational Institutes of Health (NIH) - USAU19CA148112SEARCH team (Cancer Research UK) C490/A16561AOCS (US Army Medical Research and Material Command) DAMD17-01-1-0729Canadian Institutes of Health Research (CIHR)Cancer Council Victoria Queensland Cancer Fund Cancer Council New South Wales Cancer Council South Australia Cancer Council Western Australia Cancer Council Tasmania National Health and Medical Research Council of AustraliaID400413ID400281National Health and Medical Research Council of Australia National Health and Medical Research Council of Australia National Health and Medical Research Council of Australia National Health and Medical Research Council of Australia Intramural Research Program of the Division of Cancer Epidemiology and Genetics, NCI, NIH, DHHS Ministry of Health, Italy Associazione Italiana per la Ricerca sul Cancro (AIRC)IG 15460Spanish Fondo de Investigaciones Sanitarias grant - FEDER 'Una manera de hacer Europa' PI15/00716PI15/00956CIBER de Enfermedades Raras of the Instituto de Salud Carlos III, Spain - European Development Regional Fund 'A way to achieve Europe' ERDF AGAUR of the Catalan Government, Spain 2014_SGR_603European CommissionEuropean Commission Joint Research CentreLSHC-CT-2006-018702European Union (EU) 'Fundacio La Marato de TV3', Catalonia, Spain 201331-30'Fundacion Cientifica de la Asociacion Espanola Contra el Cancer', Spain GCB15152978SOENCERCA Programme/Generalitat de Catalunya Italian Ministry of the University and Scientific Research (PRIN-2012 grant) 2012JJX494Melanoma Research Alliance United States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Cancer Institute (NCI)CA88363CA83115CA122838CA87969CA055075CA100264CA133996CA49449National Health and Medical Research Council of Australia200071241944339462380385389927389875389891389892389938443036442915442981496610496675496739552485552498APP1049894Cancer Council Queensland Cancer Institute New South Wales Australian GovernmentDepartment of Industry, Innovation and ScienceCooperative Research Centres (CRC) Programme Australian Cancer Research Foundation Wellcome TrustWT084766/Z/08/ZNational Health and Medical Research Council of Australia Australian Research Council Department of Health and Human Services (DHHS)
- Subjects
Sequence Variants ,Male ,medicine.medical_specialty ,Skin Neoplasms ,Genotype ,Medizin ,Identifies 3 ,Genome-wide association study ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Malignant-melanoma ,0302 clinical medicine ,Genetics ,medicine ,Genetic predisposition ,Nevus ,Humans ,Hla Class-i ,Genetic Predisposition to Disease ,Polymorphism ,Melanoma ,030304 developmental biology ,Telomere Length ,Cancer ,0303 health sciences ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Loci ,Pigmentation ,E-cadherin ,Single Nucleotide ,medicine.disease ,Genetic architecture ,Attributable Fraction ,3. Good health ,Phenotype ,Female ,Genetic Loci ,Genome-Wide Association Study ,Cutaneous melanoma ,Medical genetics ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,030217 neurology & neurosurgery ,Familial Melanoma - Abstract
Meta-analysis of 36,760 cases and 375,188 controls identifies 54 loci associated with susceptibility to cutaneous melanoma. Further analysis combining nevus count and hair color GWAS results provide insights into the genetic architecture of melanoma.Most genetic susceptibility to cutaneous melanoma remains to be discovered. Meta-analysis genome-wide association study (GWAS) of 36,760 cases of melanoma (67% newly genotyped) and 375,188 controls identified 54 significant (P < 5 x 10(-8)) loci with 68 independent single nucleotide polymorphisms. Analysis of risk estimates across geographical regions and host factors suggests the acral melanoma subtype is uniquely unrelated to pigmentation. Combining this meta-analysis with GWAS of nevus count and hair color, and transcriptome association approaches, uncovered 31 potential secondary loci for a total of 85 cutaneous melanoma susceptibility loci. These findings provide insights into cutaneous melanoma genetic architecture, reinforcing the importance of nevogenesis, pigmentation and telomere maintenance, together with identifying potential new pathways for cutaneous melanoma pathogenesis.
- Published
- 2020
38. Neural crest-derived tumor neuroblastoma and melanoma share 1p13.2 as susceptibility locus that shows a long-range interaction with the SLC16A1 gene
- Author
-
Mark M. Iles, Massimiliano Scalvenzi, Annalaura Montella, Zalman Vaksman, Mario Capasso, Daniela Formicola, Stefania Staibano, Alessandro Testori, Vito Alessandro Lasorsa, Hakon Hakonarson, Matteo Esposito, Sueva Cantalupo, Jan Koster, Paola Ghiorzo, Fabrizio Ayala, Sharon J. Diskin, Marcella Devoto, Antonella Cardinale, Marianna Avitabile, Flora Cimmino, Maria Fiammetta Romano, Maria Valeria Corrias, Mariangela Succoio, Nicola Zambrano, Virginia Andreotti, Matthew Law, Kevin M. Brown, Achille Iolascon, Avitabile, Marianna, Succoio, Mariangela, Testori, Alessandro, Cardinale, Antonella, Vaksman, Zalman, Lasorsa, Vito Alessandro, Cantalupo, Sueva, Esposito, Matteo, Cimmino, Flora, Montella, Annalaura, Formicola, Daniela, Koster, Jan, Andreotti, Virginia, Ghiorzo, Paola, Romano, Maria Fiammetta, Staibano, Stefania, Scalvenzi, Massimiliano, Ayala, Fabrizio, Hakonarson, Hakon, Corrias, Maria Valeria, Devoto, Marcella, Law, Matthew H, Iles, Mark M, Brown, Kevin, Diskin, Sharon, Zambrano, Nicola, Iolascon, Achille, Capasso, Mario, Oncogenomics, and CCA - Cancer biology and immunology
- Subjects
0301 basic medicine ,Male ,Monocarboxylic Acid Transporters ,Cancer Research ,Neuroblastoma ,malignant cutaneous melanoma ,neural crest cells ,GWAS ,Skin Neoplasms ,Adrenal Gland Neoplasms ,Genome-wide association study ,Locus (genetics) ,Biology ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Melanoma ,Cancer Biomarkers and Molecular Epidemiology ,Symporters ,Neural crest ,Cell Differentiation ,General Medicine ,Cell cycle ,medicine.disease ,030104 developmental biology ,Chromosomes, Human, Pair 1 ,Neural Crest ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Carcinogenesis ,Genome-Wide Association Study - Abstract
Neuroblastoma (NB) and malignant cutaneous melanoma (CMM) are neural crest cells (NCC)-derived tumors and may have a shared genetic basis, but this has not been investigated systematically by genome-wide association studies (GWAS). We took a three-staged approach to conduct cross-disease meta-analysis of GWAS for NB and CMM (2101 NB cases and 4202 controls; 12 874 CMM cases and 23 203 controls) to identify shared loci. Findings were replicated in 1403 NB cases and 1403 controls of European ancestry and in 636 NB, 508 CMM cases and 2066 controls of Italian origin. We found a cross-association at locus 1p13.2 (rs2153977, odds ratio = 0.91, P = 5.36 × 10−8). We also detected a suggestive (P < 10−7) NB-CMM cross-association at 2q37.1 with opposite effect on cancer risk. Pathway analysis of 110 NB-CMM risk loci with P < 10−4 demonstrated enrichment of biological processes such as cell migration, cell cycle, metabolism and immune response, which are essential of human NCC development, underlying both tumors. In vitro and in silico analyses indicated that the rs2153977-T protective allele, located in an NB and CMM enhancer, decreased expression of SLC16A1 via long-range loop formation and altered a T-box protein binding site. Upon depletion of SLC16A1, we observed a decrease of cellular proliferation and invasion in both NB and CMM cell lines, suggesting its role as oncogene. This is the largest study to date examining pleiotropy across two NC cell-derived tumors identifying 1p13.2 as common susceptibility locus for NB and CMM risk. We demonstrate that combining genome-wide association studies results across cancers with same origins can identify new loci common to neuroblastoma and melanoma arising from tissues which originate from neural crest cells. Our results also show 1p13.2 confer risk to neuroblastoma and melanoma by regulating SLC16A1.
- Published
- 2020
39. Reply to E. Hindié
- Author
-
Andrea Maurichi, Rosalba Miceli, Hanna Eriksson, Julia Newton-Bishop, Jérémie Nsengimana, May Chan, Andrew J. Hayes, Kara Heelan, David Adams, Roberto Patuzzo, Francesco Barretta, Gianfranco Gallino, Catherine Harwood, Daniele Bergamaschi, Dorothy Bennett, Konstantinos Lasithiotakis, Paola Ghiorzo, Bruna Dalmasso, Ausilia Manganoni, Francesca Consoli, Ilaria Mattavelli, Consuelo Barbieri, Andrea Leva, Umberto Cortinovis, Vittoria Espeli, Cristina Mangas, Pietro Quaglino, Simone Ribero, Paolo Broganelli, Giovanni Pellacani, Caterina Longo, Corrado Del Forno, Lorenzo Borgognoni, Serena Sestini, Nicola Pimpinelli, Sara Fortunato, Alessandra Chiarugi, Paolo Nardini, Elena Morittu, Antonio Florita, Mara Cossa, Barbara Valeri, Massimo Milione, Giancarlo Pruneri, Odysseas Zoras, Andrea Anichini, Roberta Mortarini, and Mario Santinami
- Subjects
Cancer Research ,Oncology - Published
- 2020
40. MC1R variants and cutaneous melanoma risk according to histological type, body site, and Breslow thickness: A pooled analysis from the M-SKIP project
- Author
-
Julia Newton-Bishop, Chiara Menin, Sara Raimondi, Maria Concetta Fargnoli, Saverio Caini, Patrick Maisonneuve, Paola Ghiorzo, David Polsky, Julian Little, DeAnn Lazovich, Rajesh Kumar, Alexander J. Stratigos, Gloria Ribas, Elena Tagliabue, Susana Puig, José C. García-Borrón, Francesca Botta, Ines Zanna, Giuseppe Palmieri, Sara Gandini, Francesco Sera, Hongmei Nan, Eduardo Nagore, Gabriella Guida, Peter A. Kanetsky, Maria Teresa Landi, and Veronica Höiom
- Subjects
Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Skin Neoplasms ,Dermatology ,Acral lentiginous melanoma ,Gastroenterology ,Article ,Breslow Thickness ,03 medical and health sciences ,cutaneous melanoma ,melanocortin 1 receptor ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Melanoma ,Histological type ,business.industry ,Odds ratio ,Middle Aged ,medicine.disease ,body site ,Breslow thickness ,histological subtype ,pooled analysis ,Confidence interval ,030104 developmental biology ,Pooled analysis ,Oncology ,Case-Control Studies ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Female ,business ,Receptor, Melanocortin, Type 1 ,Melanocortin 1 receptor - Abstract
BACKGROUND: Little is known on whether melanocortin 1 receptor (MC1R) associated cutaneous melanoma (CM) risk varies depending on histological subtype and body site, and whether tumour thickness at diagnosis (the most important prognostic factor for CM patients) differs between MC1R variant carriers and wild-type individuals. OBJECTIVE: We studied the association between MC1R variants and CM risk by histological subtype, body site, and Breslow thickness, using the database of the M-SKIP project. METHODS: We pooled individual data from fifteen case-control studies conducted during 2005–2015 in Europe and the USA. Study-specific, multi-adjusted odds ratios were pooled into summary odds ratios (SOR) and 95% confidence intervals (CI) using random-effects models. RESULTS: 6891 CM cases and 5555 controls were included. CM risk was increased among MC1R variant carriers vs. wild-type individuals. The increase in risk was comparable across histological subtypes (SOR for any variant vs. wild-type ranged between 1.57 and 1.70, always statistical significant) except acral lentiginous melanoma, for which no association emerged; and slightly greater on chronically (1.74, 95% CI 1.47–2.07) than intermittently (1.55, 95% CI 1.34–1.78) sun-exposed skin. CM risk was greater for those carrying ‘R’ vs. ‘r’ variants; correlated with the number of variants; and was more evident among individuals not showing the red hair colour phenotype. Breslow thickness was not associated with MC1R status. CONCLUSION: MC1R variants were associated with an increased risk of CM of any histological subtype (except ALM) and occurring on both chronically and intermittently sun-exposed skin.
- Published
- 2020
41. Insights Into Genetic Susceptibility to Melanoma by Gene Panel Testing: Potential Pathogenic Variants in ACD, ATM, BAP1, and POT1
- Author
-
Paola Queirolo, Federica Cecchi, Francesco Spagnolo, Italian Melanoma Intergroup, Rita Danesi, Virginia Andreotti, Enrica Teresa Tanda, Giovanni Ponti, Paola Ghiorzo, Roberta La Starza, Siranoush Manoukian, Federica Grillo, Bruna Dalmasso, Pietro Chiurazzi, William Bruno, Maurizio Genuardi, Alisa M. Goldstein, Elena Sala, Valentina Zampiga, Giuseppe Spadola, Ignazio Stanganelli, Gabriele Maccanti, Luca Mastracci, Serena Sestini, Irene Vanni, Maria Grazia Tibiletti, Giulia Ciccarese, Lorenza Pastorino, Mario Mandalà, Alberto Ballestrero, Maria Antonietta Pizzichetta, Stefania Sciallero, L., Pastorino, V., Andreotti, B., Dalmasso, I., Vanni, G., Ciccarese, M., Mandala, G., Spadola, Pizzichetta, MARIA ANTONIETTA, G., Ponti, M., Grazia Tibiletti, E., Sala, M., Genuardi, P., Chiurazzi, G., Maccanti, S., Manoukian, S., Sestini, R., Danesi, V., Zampiga, R., La Starza, I., Stanganelli, A., Ballestrero, L., Mastracci, F., Grillo, S., Sciallero, F., Cecchi, E., Teresa Tanda, F., Spagnolo, P., Queirolo, A. M., Goldstein, W., Bruno, and P., Ghiorzo
- Subjects
0301 basic medicine ,Cancer Research ,Candidate gene ,Population ,Biology ,Settore MED/03 - GENETICA MEDICA ,ATM ,BAP1 ,CDKN2A ,POT1 ,familial melanoma ,gene panel sequencing ,genetic susceptibility ,high-penetrance genes ,missing heritability ,variant interpretation ,lcsh:RC254-282 ,High-penetrance gene ,Article ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Missing heritability problem ,Genetic susceptibility ,melanoma ,Genetic predisposition ,education ,neoplasms ,Variant interpretation ,Genetics ,education.field_of_study ,Gene panel sequencing ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Missing heritability ,Familial melanoma ,High-penetrance genes ,Penetrance ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cutaneous melanoma - Abstract
The contribution of recently established or candidate susceptibility genes to melanoma missing heritability has yet to be determined. Multigene panel testing could increase diagnostic yield and better define the role of candidate genes. We characterized 273 CDKN2A/ARF and CDK4-negative probands through a custom-designed targeted gene panel that included CDKN2A/ARF, CDK4, ACD, BAP1, MITF, POT1, TERF2IP, ATM, and PALB2. Co-segregation, loss of heterozygosity (LOH)/protein expression analysis, and splicing characterization were performed to improve variant classification. We identified 16 (5.9%) pathogenic and likely pathogenic variants in established high/medium penetrance cutaneous melanoma susceptibility genes (BAP1, POT1, ACD, MITF, and TERF2IP), including two novel variants in BAP1 and 4 in POT1. We also found four deleterious and five likely deleterious variants in ATM (3.3%). Thus, including potentially deleterious variants in ATM increased the diagnostic yield to about 9%. Inclusion of rare variants of uncertain significance would increase the overall detection yield to 14%. At least 10% of melanoma missing heritability may be explained through panel testing in our population. To our knowledge, this is the highest frequency of putative ATM deleterious variants reported in melanoma families, suggesting a possible role in melanoma susceptibility, which needs further investigation.
- Published
- 2020
42. Evolution of approaches to identify melanoma missing heritability
- Author
-
Bruna Dalmasso and Paola Ghiorzo
- Subjects
0301 basic medicine ,Candidate gene ,Skin Neoplasms ,familial cancer ,genetics ,germline ,Melanoma ,missing heritability ,Inheritance Patterns ,Biology ,Germline ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Missing heritability problem ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Family history ,Molecular Biology ,Gene ,Genetic Association Studies ,Germ-Line Mutation ,Genetics ,High-Throughput Nucleotide Sequencing ,Heritability ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Familial Cancer - Abstract
Introduction: Around 10% of melanoma patients have a positive family history of melanoma and/or related cancers. Although a germline pathogenic variant in a high-risk gene can be identified in up to 40% of these patients, the remaining part of melanoma heritability remains largely unexplained.Areas covered: The aim of this review is to provide an overview of the impact that new technologies and new research approaches had and are having on finding more efficient ways to unravel the missing heritability in melanoma.Expert opinion: High-throughput sequencing technologies have been crucial in increasing the number of genes/loci that might be implicated in melanoma predisposition. However, results from these approaches may have been inferior to the expectations, due to an increase in quantitative information which hasn't been followed at the same speed by an improvement of the methods to correctly interpret these data. Optimal approaches for improving our knowledge on melanoma heritability are currently based on segregation analysis coupled with functional assessment of candidate genes. An improvement of computational methods to infer genotype-phenotype correlations could help address the issue of missing heritability.
- Published
- 2020
43. MC1R variants as melanoma risk factors independent of at-risk phenotypic characteristics: a pooled analysis from the M-SKIP project
- Author
-
Chiara Menin, David Polsky, DeAnn Lazovich, Elena Tagliabue, Julia Newton-Bishop, Maria Concetta Fargnoli, Sara Raimondi, Rino Bellocco, Nelleke A. Gruis, José C. García-Borrón, Paola Ghiorzo, Julian Little, Jiali Han, Patrick Maisonneuve, Maria Teresa Landi, Peter A. Kanetsky, Sara Gandini, and Francesco Sera
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Melanoma ,Case-control study ,Subgroup analysis ,medicine.disease ,Phototype ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Genetic epidemiology ,030220 oncology & carcinogenesis ,Internal medicine ,Genotype ,Cutaneous melanoma ,medicine ,Genetic variability ,business - Abstract
Melanoma represents an important public health problem, due to its high case-fatality rate. Identification of individuals at high risk would be of major interest to improve early diagnosis and ultimately survival. The aim of this study was to evaluate whether MC1R variants predicted melanoma risk independently of at-risk phenotypic characteristics.Data were collected within an international collaboration - the M-SKIP project. The present pooled analysis included data on 3,830 single, primary, sporadic, cutaneous melanoma cases and 2,619 controls from seven previously published case-control studies. All the studies had information on MC1R gene variants by sequencing analysis and on hair color, skin phototype, and freckles, ie, the phenotypic characteristics used to define the red hair phenotype.The presence of any MC1R variant was associated with melanoma risk independently of phenotypic characteristics (OR 1.60; 95% CI 1.36-1.88). Inclusion of MC1R variants in a risk prediction model increased melanoma predictive accuracy (area under the receiver-operating characteristic curve) by 0.7% over a base clinical model (P=0.002), and 24% of participants were better assessed (net reclassification index 95% CI 20%-30%). Subgroup analysis suggested a possibly stronger role of MC1R in melanoma prediction for participants without the red hair phenotype (net reclassification index: 28%) compared to paler skinned participants (15%).The authors suggest that measuring the MC1R genotype might result in a benefit for melanoma prediction. The results could be a valid starting point to guide the development of scientific protocols assessing melanoma risk prediction tools incorporating the MC1R genotype.
- Published
- 2018
44. 1482P Landscape of germline pathogenic variants beyond BRCA in pancreatic cancer patients
- Author
-
R. Borea, S. Puglisi, Lorenza Pastorino, Alberto Puccini, F. Catalano, Paola Ghiorzo, Giuseppe Fornarini, Stefania Sciallero, Virginia Andreotti, Irene Vanni, Alessio Signori, William Bruno, and Bruna Dalmasso
- Subjects
Oncology ,business.industry ,Pancreatic cancer ,Cancer research ,Medicine ,Hematology ,business ,medicine.disease ,Germline - Published
- 2021
45. TP63 mutations are frequent in cutaneous melanoma, support UV etiology, but their role in melanomagenesis is unclear
- Author
-
Paola Monti, Giorgia Foggetti, Paola Menichini, Gilberto Fronza, Alberto Izzotti, Paola Queirolo, and Paola Ghiorzo
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Neoplasms, Radiation-Induced ,Skin Neoplasms ,Carcinogenesis ,Ultraviolet Rays ,medicine.disease_cause ,functional analysis ,03 medical and health sciences ,0302 clinical medicine ,TP63 ,Humans ,Medicine ,Melanoma ,Molecular Epidemiology ,Mutation ,Oncogene ,business.industry ,Tumor Suppressor Proteins ,Cancer ,Tumor Protein p73 ,Articles ,General Medicine ,mutations ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Etiology ,Tumor Suppressor Protein p53 ,Skin cancer ,cutaneous melanomas ,business ,yeast assay ,Transcription Factors - Abstract
In contrast to TP53, cancer development is rarely associated with mutations in the TP63 and TP73 genes. Recently, next generation sequencing analysis revealed that TP63 mutations are frequent, specifically in cutaneous melanomas. Cutaneous melanoma represents 4% of skin cancers but it is responsible for 80% of skin cancer related deaths. In the present study, we first determined whether all three members of the P53 family of transcription factors were found mutated in cutaneous melanomas by retrieving all TP53, TP63 and TP73 mutations from cBioPortal (http://www.cbioportal.org/). TP53 and TP63 were frequently mutated [15.0% (91/605) and 14.7% (89/605), respectively], while TP73 [1.5% (9/605)] was more rarely mutated (p70%). This result does not support a major role of the mutant P63 protein in melanomagenesis while it is still consistent with the TP63 gene being a recorder of UV exposure. The TP63 mutation spectrum from cutaneous melanomas, when compared with that observed at the germinal level in patients affected by P63-associated diseases [ectodermal dysplasia syndromes, (EDs)], revealed significant differences. The TP63 mutations were more frequent at CpGs sites (p
- Published
- 2017
46. Heterogeneity and frequency of BRAF mutations in primary melanoma: Comparison between molecular methods and immunohistochemistry
- Author
-
Federica Grillo, Virginia Andreotti, Paola Ghiorzo, Luca Mastracci, Lorenza Pastorino, Claudia Martinuzzi, Paola Queirolo, Francesco Spagnolo, Marina Gualco, Giovanna Bianchi-Scarrà, Francesco Cabiddu, Alberto Ballestrero, Bruna Dalmasso, and William Bruno
- Subjects
Male ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Skin Neoplasms ,Metastatic lesions ,Concordance ,DNA Mutational Analysis ,Real-Time Polymerase Chain Reaction ,Metastatic tumor ,BRAF ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Predictive Value of Tests ,Biomarkers, Tumor ,Humans ,Medicine ,Genetic Predisposition to Disease ,PNA-clamping ,heterogeneity ,immunohistochemistry ,primary melanoma ,Mutation frequency ,Melanoma ,Observer Variation ,business.industry ,High-Throughput Nucleotide Sequencing ,Reproducibility of Results ,Middle Aged ,Prognosis ,medicine.disease ,Primary tumor ,Phenotype ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Mutation testing ,Immunohistochemistry ,Female ,business ,Research Paper - Abstract
// William Bruno 1 , Claudia Martinuzzi 1 , Virginia Andreotti 1 , Lorenza Pastorino 1 , Francesco Spagnolo 2 , Bruna Dalmasso 1 , Francesco Cabiddu 3 , Marina Gualco 3 , Alberto Ballestrero 1 , Giovanna Bianchi-Scarra 1 , Paola Queirolo 2 , Federica Grillo 4, * , Luca Mastracci 4, * , Paola Ghiorzo 1, * on behalf of the Italian Melanoma Intergroup (IMI) 1 Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy 2 Department of Medical Oncology, IRCCS AOU San Martino-IST, Genoa, Italy 3 Department of Pathology, IRCCS AOU San Martino-IST, Genoa, Italy 4 Department of Surgical and Diagnostic Sciences, Pathology Unit, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy * These authors have contributed equally to this work Correspondence to: Martinuzzi Claudia, email: claudia.martinuzzi@edu.unige.it Keywords: BRAF, primary melanoma, PNA-clamping, immunohistochemistry, heterogeneity Abbreviations: PNA: peptide nucleic acid – PNA-clamping real-time PCR; IHC: immunohistochemistry; NGS: next generation sequencing; FFPE: formalin-fixed, paraffin embedded Received: September 20, 2016 Accepted: November 24, 2016 Published: December 22, 2016 ABSTRACT Finding the best technique to identify BRAF mutations with a high sensitivity and specificity is mandatory for accurate patient selection for target therapy. BRAF mutation frequency ranges from 40 to 60% depending on melanoma clinical characteristics and detection technique used. Intertumoral heterogeneity could lead to misinterpretation of BRAF mutational status; this is especially important if testing is performed on primary specimens, when metastatic lesions are unavailable. Aim of this study was to identify the best combination of methods for detecting BRAF mutations (among peptide nucleic acid – PNA-clamping real-time PCR, immunohistochemistry and capillary sequencing) and investigate BRAF mutation heterogeneity in a series of 100 primary melanomas and a subset of 25 matched metastatic samples. Overall, we obtained a BRAF mutation frequency of 62%, based on the combination of at least two techniques. Concordance between mutation status in primary and metastatic tumor was good but not complete (67%), when agreement of at least two techniques were considered. Next generation sequencing was used to quantify the threshold of detected mutant alleles in discordant samples. Combining different methods excludes that the observed heterogeneity is technique-based. We propose an algorithm for BRAF mutation testing based on agreement between immunohistochemistry and PNA; a third molecular method could be added in case of discordance of the results. Testing the primary tumor when the metastatic sample is unavailable is a good option if at least two methods of detection are used, however the presence of intertumoral heterogeneity or the occurrence of additional primaries should be carefully considered.
- Published
- 2016
47. Estimating CDKN2A mutation carrier probability among global familial melanoma cases using GenoMELPREDICT
- Author
-
Alisa M. Goldstein, Helen Schmid, Peter A. Kanetsky, Eduardo Nagore, Nicholas J. Taylor, Johan Hansson, Anne E. Cust, William Bruno, Veronica Höiom, Maria Teresa Landi, David E. Elder, Alejandra Larre-Borges, Nicholas K. Hayward, Christian Ingvar, Graham J. Mann, Marko Hočevar, Nienke van der Stoep, Marie-Françoise Avril, Nandita Mitra, Antonia L. Pritchard, D. Timothy Bishop, Julia Newton-Bishop, Donato Calista, Florence Demenais, Francisco Cuellar, Margaret A. Tucker, Mark Harland, Lu Qian, Karin Wadt, Susana Puig, Elizabeth A. Holland, Barbara Perić, Brigitte Bressac-de Paillerets, Gilles Landman, Paola Ghiorzo, Dace Pjanova, Nelleke A. Gruis, Thaís Corsetti Grazziotin, Jane M. Palmer, Xiaohong R. Yang, Anne-Marie Gerdes, and Håkan Olsson
- Subjects
Oncology ,Internationality ,Skin Neoplasms ,CDKN2A, familial melanoma, GenoMEL, GenoMELPREDICT, mutation prediction ,030207 dermatology & venereal diseases ,0302 clinical medicine ,Risk Factors ,CDKN2A ,mutation prediction ,Child ,Melanoma ,familial melanoma ,Aged, 80 and over ,education.field_of_study ,medicine.diagnostic_test ,Middle Aged ,Phenotype ,Area Under Curve ,030220 oncology & carcinogenesis ,Adult ,Heterozygote ,medicine.medical_specialty ,Adolescent ,Population ,Dermatology ,Article ,Young Adult ,03 medical and health sciences ,Germline mutation ,Predictive Value of Tests ,Internal medicine ,Pancreatic cancer ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,education ,Cyclin-Dependent Kinase Inhibitor p16 ,Germ-Line Mutation ,Aged ,Probability ,Genetic testing ,Receiver operating characteristic ,business.industry ,GenoMEL ,medicine.disease ,Confidence interval ,Pancreatic Neoplasms ,Logistic Models ,ROC Curve ,business ,GenoMELPREDICT - Abstract
Background: Although rare in the general population, highly penetrant germline mutations in CDKN2A are responsible for 5%-40% of melanoma cases reported in melanoma-prone families. We sought to determine whether MELPREDICT was generalizable to a global series of families with melanoma and whether performance improvements can be achieved.Methods: In total, 2116 familial melanoma cases were ascertained by the international GenoMEL Consortium. We recapitulated the MELPREDICT model within our data (GenoMELPREDICT) to assess performance improvements by adding phenotypic risk factors and history of pancreatic cancer. We report areas under the curve (AUC) with 95% confidence intervals (CIs) along with net reclassification indices (NRIs) as performance metrics.Results: MELPREDICT performed well (AUC 0.752, 95% CI 0.730-0.775), and GenoMELPREDICT performance was similar (AUC 0.748, 95% CI 0.726-0.771). Adding a reported history of pancreatic cancer yielded discriminatory improvement (P < .0001) in GenoMELPREDICT (AUC 0.772, 95% CI 0.750-0.793, NRI 0.40). Including phenotypic risk factors did not improve performance.Conclusion: The MELPREDICT model functioned well in a global data set of familial melanoma cases. Adding pancreatic cancer history improved model prediction. GenoMELPREDICT is a simple tool for predicting CDKN2A mutational status among melanoma patients from melanoma-prone families and can aid in directing these patients to receive genetic testing or cancer risk counseling.
- Published
- 2019
48. Contribution of Common Genetic Variants to Familial Aggregation of Disease and Implications for Sequencing Studies
- Author
-
Ruth M. Pfeiffer, Susana Puig, Maria Teresa Landi, Donato Calista, Ketty Peris, Joshua N. Sampson, Maria Concetta Fargnoli, Eduardo Nagore, Paola Ghiorzo, Andrew Schlafly, Tongwu Zhang, Lei Song, Jianxin Shi, and Chiara Menin
- Subjects
Melanomas ,Cancer Research ,Multifactorial Inheritance ,Heredity ,Epidemiology ,Test Statistics ,Social Sciences ,Genome-wide association study ,Disease ,QH426-470 ,medicine.disease_cause ,Epidemiologia genètica ,0302 clinical medicine ,Mathematical and Statistical Techniques ,Sociology ,Risk Factors ,Consortia ,Medicine and Health Sciences ,Genetic epidemiology ,Càncer ,Melanoma ,Genetics (clinical) ,Cancer ,Genetics ,0303 health sciences ,Factors de risc en les malalties ,Statistics ,Family aggregation ,Single Nucleotide ,Pedigree ,Phenotype ,Oncology ,Genetic Diseases ,Physical Sciences ,symbols ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,Sequence Analysis ,Research Article ,Genotyping ,Genotype ,Risk factors in diseases ,Biology ,Research and Analysis Methods ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,symbols.namesake ,medicine ,SNP ,Humans ,Genetic Predisposition to Disease ,Allele ,Polymorphism ,Statistical Methods ,Molecular Biology Techniques ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Alleles ,030304 developmental biology ,Genetic Diseases, Inborn ,Genetic Variation ,Biology and Life Sciences ,Cancers and Neoplasms ,DNA ,Sequence Analysis, DNA ,Heritability ,Inborn ,Genetic Loci ,Genetics of Disease ,Mendelian inheritance ,030217 neurology & neurosurgery ,Mathematics ,Genome-Wide Association Study - Abstract
Despite genetics being accepted as the primary cause of familial aggregation for most diseases, it is still unclear whether afflicted families are likely to share a single highly penetrant rare variant, many minimally penetrant common variants, or a combination of the two types of variants. We therefore use recent estimates of SNP heritability and the liability threshold model to estimate the proportion of afflicted families likely to carry a rare, causal variant. We then show that Polygenic Risk Scores (PRS) may be useful for identifying families likely to carry such a rare variant and therefore for prioritizing families to include in sequencing studies with that aim. Specifically, we introduce a new statistic that estimates the proportion of individuals carrying causal rare variants based on the family structure, disease pattern, and PRS of genotyped individuals. Finally, we consider data from the MelaNostrum consortium and show that, despite an estimated PRS heritability of only 0.05 for melanoma, families carrying putative causal variants had a statistically significantly lower PRS, supporting the idea that PRS prioritization may be a useful future tool. However, it will be important to evaluate whether the presence of rare mendelian variants are generally associated with the proposed test statistic or lower PRS in future and larger studies., Author summary Multiple members in a family can be diagnosed with the same disease. In such families, genetics may be a significant factor in disease risk. However, it remains unclear whether such familial aggregation of disease is likely due to a single highly penetrant rare variant (HPRV), many minimally penetrant common variants, or a combination of the two types of variants. We therefore use recent estimates of SNP heritability and the liability threshold model to estimate the proportion of afflicted families likely to carry a rare, causal variant. We then show that Polygenic Risk Scores (PRS) may be useful for identifying families likely to carry such a rare variant and introduce a related statistic that can be used to select families for sequencing studies trying to identify HPRV.
- Published
- 2019
49. Comprehensive Study of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families Worldwide
- Author
-
Carla Daniela Robles-Espinoza, Hilary Racher, Luca Mastracci, Robert Pilarski, Frederick H. Davidorf, Saleem Taibjee, William Glasson, Martine J. Jager, Jo Burke, Ivana K. Kim, Lisa Golmard, Rajmohan Murali, Hayley Hamilton, Giulia Ciccarese, Jane M. Palmer, Madeleine Howlie, Arnaud de la Fouchardière, David J. Adams, Tero Kivelä, Sonika Dahiya, Peter Johansson, Lorenza Pastorino, Sebastian Walpole, Annelies de Klein, Bruna Dalmasso, Anne Marie Lane, Marina Marinkovic, William Bruno, Judith Symmons, Sonja Klebe, Julia Newton-Bishop, Marc-Henri Stern, Michael R. Speicher, Joni A. Turunen, Colleen M. Cebulla, Dominique Stoppa-Lyonnet, Carsten Bergmann, Hildur Helgadottir, Mohamed H. Abdel-Rahman, S.J. O’Shea, Meredith Stautberg, Ching-Ni Njauw, Jens Folke Kiilgaard, Pauliina Repo, Erin M. Garfield, Paola Queirolo, Rana'a T. Al-Jamal, Remco van Doorn, Antonia L. Pritchard, Gregorius P M Luyten, Michael L. Nickerson, Mitchell Cheung, Pedram Gerami, Miriam Potrony, Paola Ghiorzo, Odile Cabaret, Julian Adlard, Veronica Höiom, Hensin Tsao, Cindy Chau, J. William Harbour, Irma Dianzani, Joseph R. Testa, Brigitte Bressac-de Paillerets, Sunil K Warrier, Maartje Nielsen, Marta Betti, Susana Puig, Emine Kilic, Sandro Santagata, Karin Wadt, Nicholas K. Hayward, Virginia Andreotti, Raul Ossio, Natasha M. van Poppelen, Nicola K. Poplawski, Reetta Stiina Järvinen, Clinical Genetics, and Ophthalmology
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Genotype ,Reviews ,Biology ,Risk Assessment ,Germline ,Age of Onset ,Alleles ,Female ,Gene Frequency ,Humans ,Neoplastic Syndromes, Hereditary ,Phenotype ,Tumor Suppressor Proteins ,Ubiquitin Thiolesterase ,Genetic Association Studies ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Genotype-phenotype distinction ,Germline mutation ,Missense mutation ,Neoplastic Syndromes ,Allele ,BAP1 ,3. Good health ,Hereditary ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Cancer research - Abstract
BACKGROUND: The BRCA1-associated protein-1 (BAP1) tumor predisposition syndrome (BAP1-TPDS) is a hereditary tumor syndrome caused by germline pathogenic variants in BAP1 encoding a tumor suppressor associated with uveal melanoma, mesothelioma, cutaneous melanoma, renal cell carcinoma, and cutaneous BAP1-inactivated melanocytic tumors. However, the full spectrum of tumors associated with the syndrome is yet to be determined. Improved understanding of the BAP1-TPDS is crucial for appropriate clinical management of BAP1 germline variant carriers and their families, including genetic counseling and surveillance for new tumors. METHODS: We collated germline variant status, tumor diagnoses, and information on BAP1 immunohistochemistry or loss of somatic heterozygosity on 106 published and 75 unpublished BAP1 germline variant-positive families worldwide to better characterize the genotypes and phenotypes associated with the BAP1-TPDS. Tumor spectrum and ages of onset were compared between missense and null variants. All statistical tests were two-sided. RESULTS: The 181 families carried 140 unique BAP1 germline variants. The collated data confirmed the core tumor spectrum associated with the BAP1-TPDS and showed that some families carrying missense variants can exhibit this phenotype. A variety of noncore BAP1-TPDS -associated tumors were found in families of variant carriers. Median ages of onset of core tumor types were lower in null than missense variant carriers for all tumors combined (P
- Published
- 2018
50. Prevalence of pathogenic/likely pathogenic variants in the 24 cancer genes of the ACMG Secondary Findings v2.0 list in a large cancer cohort and ethnicity-matched controls
- Author
-
Ketty Peris, Jennifer T. Loud, Maisa Pinheiro, Alexander Pemov, Stephen J. Chanock, Seth A. Brodie, Susana Puig, Jennifer J. Johnston, Maria Concetta Fargnoli, Joshua N. Sampson, Xiaohong R. Yang, Payal P. Khincha, Lindsay M. Morton, Sharon A. Savage, Talia Wegman-Ostrosky, Megan N. Frone, Neil E. Caporaso, Eduardo Nagore, Lisa J. McReynolds, Shahinaz M. Gadalla, Mingyi Wang, Michael L. Nickerson, Paola Ghiorzo, Mark H. Greene, Mary L. McMaster, Kelvin C. de Andrade, Mia M. Gaudet, Michael Dean, Bin Zhu, Leslie G. Biesecker, Donato Calista, Belynda Hicks, Margaret A. Tucker, Jia Liu, Wen Luo, Maria Isabel Achatz, Matthew Gianferante, Lisa Mirabello, Neal D. Freedman, Melissa Rotunno, Lynn R. Goldin, Jung Kim, Alisa M. Goldstein, Shengchao A. Li, Blanche P. Alter, Fernanda P. Fortes, Douglas R. Stewart, Maria Teresa Landi, Susan M. Gapstur, and Karina Miranda Santiago
- Subjects
0301 basic medicine ,Oncology ,Male ,DNA Mutational Analysis ,Prevalence ,lcsh:Medicine ,Cohort Studies ,Familial cancer exome ,Neoplasms ,Ethnicity ,Genetics (clinical) ,Single Nucleotide ,3. Good health ,Variant classification ,Cohort ,Molecular Medicine ,Medical genetics ,Population study ,Female ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,medicine.medical_specialty ,lcsh:QH426-470 ,Concordance ,Ethnic Groups ,ACMG secondary findings ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Aged ,Genes, Neoplasm ,Humans ,Genetic Predisposition to Disease ,Mutation ,Internal medicine ,Genetics ,medicine ,Polymorphism ,Molecular Biology ,Gene ,business.industry ,Research ,lcsh:R ,Cancer ,medicine.disease ,Human genetics ,lcsh:Genetics ,030104 developmental biology ,Genes ,ACMG secondary findings, Familial cancer exome, Population study, Variant classification ,Neoplasm ,business - Abstract
Background Prior research has established that the prevalence of pathogenic/likely pathogenic (P/LP) variants across all of the American College of Medical Genetics (ACMG) Secondary Findings (SF) genes is approximately 0.8–5%. We investigated the prevalence of P/LP variants in the 24 ACMG SF v2.0 cancer genes in a family-based cancer research cohort (n = 1173) and in cancer-free ethnicity-matched controls (n = 982). Methods We used InterVar to classify variants and subsequently conducted a manual review to further examine variants of unknown significance (VUS). Results In the 24 genes on the ACMG SF v2.0 list associated with a cancer phenotype, we observed 8 P/LP unique variants (8 individuals; 0.8%) in controls and 11 P/LP unique variants (14 individuals; 1.2%) in cases, a non-significant difference. We reviewed 115 VUS. The median estimated per-variant review time required was 30 min; the first variant within a gene took significantly (p = 0.0009) longer to review (median = 60 min) compared with subsequent variants (median = 30 min). The concordance rate was 83.3% for the variants examined by two reviewers. Conclusion The 115 VUS required database and literature review, a time- and labor-intensive process hampered by the difficulty in interpreting conflicting P/LP determinations. By rigorously investigating the 24 ACMG SF v2.0 cancer genes, our work establishes a benchmark P/LP variant prevalence rate in a familial cancer cohort and controls. Electronic supplementary material The online version of this article (10.1186/s13073-018-0607-5) contains supplementary material, which is available to authorized users.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.