65 results on '"Leora Witkowski"'
Search Results
2. P587: Leveraging extensive datasets to better classify SMARCA4 variants
- Author
-
Leora Witkowski, Nadine Demko, Elodie Petrecca, Marie Loncol, and William Foulkes
- Subjects
Genetics ,QH426-470 ,Medicine - Published
- 2023
- Full Text
- View/download PDF
3. Next-generation sequencing of non-small cell lung cancer at a Quebec health care cancer centre
- Author
-
Mark Sorin, Sophie Camilleri-Broët, Emilie Pichette, Justin-Pierre Lorange, Nasim Haghandish, Laurie-Rose Dubé, André Lametti, Caroline Huynh, Leora Witkowski, George Zogopoulos, Yifan Wang, Hangjun Wang, Jonathan Spicer, Logan A. Walsh, Roni Rayes, Guy Rouleau, Alan Spatz, Andrea Liliam Gomez Corredor, and Pierre Olivier Fiset
- Subjects
Lung cancer ,Mutations ,Next-generation sequencing ,NGS ,NSCLC ,Canada ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Lung cancer is the leading cause of cancer death in both men and women. Quebec has the highest lung cancer mortality out of all provinces in Canada, believed to be caused by higher smoking rates. Molecular testing for lung cancer is standard of care due to the discovery of actionable driver mutations that can be targeted with tyrosine kinase inhibitors. To date, no detailed molecular testing characterization of Quebec patients with lung cancer using next generation sequencing (NGS) has been performed. Materials and methods: The aim of this study was to describe the genomic landscape of patients with lung cancer (n = 997) who underwent NGS molecular testing at a tertiary care center in Quebec and to correlate it with clinical and pathology variables. Results: Compared to 10 other NGS studies found through a structured search strategy, our cohort had a higher prevalence of KRAS mutations (39.2%) compared to most geographical locations. Additionally, we observed a significant positive association between decreasing age and a higher proportion of KRAS G12C mutations. Conclusion: Overall, it remains important to assess institutional rates of actionable driver mutations to help guide governing bodies, fuel clinical trials and create benchmarks for expected rates as quality metrics.
- Published
- 2023
- Full Text
- View/download PDF
4. SMARCA4/2 loss inhibits chemotherapy-induced apoptosis by restricting IP3R3-mediated Ca2+ flux to mitochondria
- Author
-
Yibo Xue, Jordan L. Morris, Kangning Yang, Zheng Fu, Xianbing Zhu, Fraser Johnson, Brian Meehan, Leora Witkowski, Amber Yasmeen, Tunde Golenar, Mackenzie Coatham, Geneviève Morin, Anie Monast, Virginie Pilon, Pierre Olivier Fiset, Sungmi Jung, Anne V. Gonzalez, Sophie Camilleri-Broet, Lili Fu, Lynne-Marie Postovit, Jonathan Spicer, Walter H. Gotlieb, Marie-Christine Guiot, Janusz Rak, Morag Park, William Lockwood, William D. Foulkes, Julien Prudent, and Sidong Huang
- Subjects
Science - Abstract
SMARCA4/2 loss in ovarian and lung cancers is associated with chemotherapy resistance. Here, the authors show that SMARCA4/2 deficiency in cancer cells reduces the expression of the ER-Ca2+ channel IP3R3 and subsequently calcium transfer to the mitochondria, which inhibits apoptotic cell death.
- Published
- 2021
- Full Text
- View/download PDF
5. Author Correction: SMARCA4/2 loss inhibits chemotherapy-induced apoptosis by restricting IP3R3-mediated Ca2+ flux to mitochondria
- Author
-
Yibo Xue, Jordan L. Morris, Kangning Yang, Zheng Fu, Xianbing Zhu, Fraser Johnson, Brian Meehan, Leora Witkowski, Amber Yasmeen, Tunde Golenar, Mackenzie Coatham, Geneviève Morin, Anie Monast, Virginie Pilon, Pierre Olivier Fiset, Sungmi Jung, Anne V. Gonzalez, Sophie Camilleri-Broet, Lili Fu, Lynne-Marie Postovit, Jonathan Spicer, Walter H. Gotlieb, Marie-Christine Guiot, Janusz Rak, Morag Park, William Lockwood, William D. Foulkes, Julien Prudent, and Sidong Huang
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
6. CDK4/6 inhibitors target SMARCA4-determined cyclin D1 deficiency in hypercalcemic small cell carcinoma of the ovary
- Author
-
Yibo Xue, Brian Meehan, Elizabeth Macdonald, Sriram Venneti, Xue Qing D. Wang, Leora Witkowski, Petar Jelinic, Tim Kong, Daniel Martinez, Geneviève Morin, Michelle Firlit, Atefeh Abedini, Radia M. Johnson, Regina Cencic, Jay Patibandla, Hongbo Chen, Andreas I. Papadakis, Aurelie Auguste, Iris de Rink, Ron M. Kerkhoven, Nicholas Bertos, Walter H. Gotlieb, Blaise A. Clarke, Alexandra Leary, Michael Witcher, Marie-Christine Guiot, Jerry Pelletier, Josée Dostie, Morag Park, Alexander R. Judkins, Ralf Hass, Douglas A. Levine, Janusz Rak, Barbara Vanderhyden, William D. Foulkes, and Sidong Huang
- Subjects
Science - Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is driven by SMARCA4 loss. Here the authors demonstrate that SCCOHT cells are highly sensitive to CDK4/6 inhibition and provide mechanistic insights, whereby this druggable vulnerability is driven by cyclin D1 deficiency induced by SMARCA4 loss.
- Published
- 2019
- Full Text
- View/download PDF
7. Retrospective Analysis of Clinical Genetic Testing in Pediatric Primary Dilated Cardiomyopathy: Testing Outcomes and the Effects of Variant Reclassification
- Author
-
Daniel Quiat, Leora Witkowski, Hana Zouk, Kevin P. Daly, and Amy E. Roberts
- Subjects
familial dilated cardiomyopathy ,genetic testing ,idiopathic dilated cardiomyopathy ,variant of uncertain significance ,variant reanalysis ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Genetic testing in pediatric primary dilated cardiomyopathy (DCM) patients has identified numerous disease‐causing variants, but few studies have evaluated genetic testing outcomes in this population in the context of patient and familial clinical data or assessed the clinical implications of temporal changes in genetic testing results. Methods and Results We performed a retrospective analysis of all patients with primary DCM who presented to our institution between 2008 and 2018. Variants identified by genetic testing were reevaluated for pathogenicity on the basis of current guidelines for variant classification. A total of 73 patients with primary DCM presented to our institution and 63 (86%) were probands that underwent cardiomyopathy‐specific gene testing. A disease‐causing variant was identified in 19 of 63 (30%) of cases, with at least 9/19 (47%) variants occurring de novo. Positive family history was not associated with identification of a causal variant. Reclassification of variants resulted in the downgrading of a large proportion of variants of uncertain significance and did not identify any new disease‐causing variants. Conclusions Clinical genetic testing identifies a causal variant in one third of pediatric patients with primary DCM. Variant reevaluation significantly decreased the number of variants of uncertain significance, but a large burden of variants of uncertain significance remain. These results highlight the need for periodic reanalysis of genetic testing results, additional investigation of genotype‐phenotype correlations in DCM through large, multicenter genetic studies, and development of improved tools for functional characterization of variants of uncertain significance.
- Published
- 2020
- Full Text
- View/download PDF
8. Expanding the Noonan spectrum/RASopathy NGS panel: Benefits of adding NF1 and SPRED1
- Author
-
Leora Witkowski, Mitchell W. Dillon, Elissa Murphy, Matthew SLebo, and Heather Mason‐Suares
- Subjects
Legius syndrome ,Neurofibromatosis type 1 ,Neurofibromatosis‐Noonan syndrome (NFNS) ,NF1 ,Noonan syndrome (NS) ,Noonan syndrome with multiple lentigines (NSML) ,Genetics ,QH426-470 - Abstract
Abstract Background RASopathies are a group of disorders caused by disruptions to the RAS‒MAPK pathway. Despite being in the same pathway, Neurofibromatosis Type 1 (NF1) and Legius syndrome (LS) typically present with phenotypes distinct from Noonan spectrum disorders (NSDs). However, some NF1/LS individuals also exhibit NSD phenotypes, often referred to as Neurofibromatosis‐Noonan syndrome (NFNS), and may be mistakenly evaluated for NSDs, delaying diagnosis, and affecting patient management. Methods A derivation cohort of 28 patients with a prior negative NSD panel and either NFNS or a suspicion of NSD and café‐au‐lait spots underwent NF1 and SPRED1 sequencing. To further determine the utility and burden of adding these genes, a validation cohort of 505 patients with a suspected RASopathy were tested on a 14‐gene RASopathy‐associated panel. Results In the derivation cohort, six (21%) patients had disease‐causing NF1 or SPRED1 variants. In the validation cohort, 11 (2%) patients had disease‐causing variants and 15 (3%) had variants of uncertain significance in NF1 or SPRED1. Of those with disease‐causing variants, 5/17 only had an NSD diagnosis. Conclusions Adding NF1 and SPRED1 to RASopathy panels can speed diagnosis and improve patient management, without significantly increasing the burden of inconclusive results.
- Published
- 2020
- Full Text
- View/download PDF
9. Primary rhabdoid tumor of the ovary: When large cells become small cells
- Author
-
Alex Rabinovich, Leora Witkowski, Ruthy Shaco-Levi, Mihai Meirovitz, Martin Hasselblatt, and William D. Foulkes
- Subjects
Rhabdoid ,Ovary ,Tumor ,SMARCA4 ,Sequencing ,Genetic testing ,Gynecology and obstetrics ,RG1-991 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2015
- Full Text
- View/download PDF
10. Prophylactic oophorectomy for hereditary small cell carcinoma of the ovary, hypercalcemic type
- Author
-
Andrew Berchuck, Leora Witkowski, Martin Hasselblatt, and William D. Foulkes
- Subjects
Ovarian cancer ,SMARCA4 mutation ,Prophylactic oophorectomy ,Gynecology and obstetrics ,RG1-991 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2015
- Full Text
- View/download PDF
11. Cellular context determines <scp>DNA</scp> methylation profiles in <scp>SWI</scp> / <scp>SNF</scp> ‐deficient cancers of the gynecologic tract
- Author
-
Felix KF Kommoss, Basile Tessier‐Cloutier, Leora Witkowski, Erna Forgo, Christian Koelsche, Martin Köbel, William D Foulkes, Cheng‐Han Lee, David L Kolin, Andreas Deimling, and Brooke E Howitt
- Subjects
Lung Neoplasms ,DNA Helicases ,Nuclear Proteins ,DNA Methylation ,Small Cell Lung Carcinoma ,United Kingdom ,Endometrial Neoplasms ,Pathology and Forensic Medicine ,Hypercalcemia ,Humans ,Female ,Carcinoma, Small Cell ,Carcinoma, Endometrioid ,Rhabdoid Tumor ,Transcription Factors - Abstract
SWI/SNF (SWItch/Sucrose Non-Fermentable) complex deficiency has been reported in a wide variety of cancers and is often associated with an undifferentiated phenotype. In the gynecologic tract SWI/SNF-deficient cancers are diagnostically challenging and little is known about their cellular origins. Here we show that undifferentiated endometrial carcinoma (UDEC), SMARCA4-deficient uterine sarcoma (SDUS), and ovarian small cell carcinoma, hypercalcemic type (SCCOHT) harbor distinct DNA methylation signatures despite shared morphology and SWI/SNF inactivation. Our results indicate that the cellular context is an important determinant of the epigenetic landscape, even in the setting of core SWI/SNF deficiency, and therefore methylation profiling may represent a useful diagnostic tool in undifferentiated, SWI/SNF-deficient cancers. Furthermore, applying copy number analyses and group-wise differential methylation analyses including endometrioid endometrial carcinomas and extracranial malignant rhabdoid tumors, we uncover analogous molecular features in SDUS and SCCOHT in contrast to UDEC. These results suggest that SDUS and SCCOHT represent chromosomally stable SWI/SNF-deficient cancers of the gynecologic tract, which are within the broader spectrum of malignant rhabdoid tumors. © 2022 The Authors. The Journal of Pathology published by John WileySons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2022
- Full Text
- View/download PDF
12. Supplementary Data from Germline Missense Variants in CDC20 Result in Aberrant Mitotic Progression and Familial Cancer
- Author
-
Jose G. Teodoro, William D. Foulkes, Jacek Majewski, Marc Tischkowitz, David Barford, Celia M.T. Greenwood, William C.H. Chao, Ziguo Zhang, Colin J.R. Stewart, Reiner Siebert, Susanne Bens, Morten Hillmer, Dominik Schneider, Stefan Schönberger, Albert M. Berghuis, Leora Witkowski, Jian Carrot-Zhang, Lai Jiang, Caterina Pacifico, Isabelle Gamache, Yilin Wang, Somayyeh Fahiminiya, Barbara Rivera, Javad Nadaf, Mohamed Moustafa-Kamal, Ester Castellsagué, and Owen J. Chen
- Abstract
Supplementary Data from Germline Missense Variants in CDC20 Result in Aberrant Mitotic Progression and Familial Cancer
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Germline Missense Variants in CDC20 Result in Aberrant Mitotic Progression and Familial Cancer
- Author
-
Jose G. Teodoro, William D. Foulkes, Jacek Majewski, Marc Tischkowitz, David Barford, Celia M.T. Greenwood, William C.H. Chao, Ziguo Zhang, Colin J.R. Stewart, Reiner Siebert, Susanne Bens, Morten Hillmer, Dominik Schneider, Stefan Schönberger, Albert M. Berghuis, Leora Witkowski, Jian Carrot-Zhang, Lai Jiang, Caterina Pacifico, Isabelle Gamache, Yilin Wang, Somayyeh Fahiminiya, Barbara Rivera, Javad Nadaf, Mohamed Moustafa-Kamal, Ester Castellsagué, and Owen J. Chen
- Abstract
CDC20 is a coactivator of the anaphase promoting complex/cyclosome (APC/C) and is essential for mitotic progression. APC/CCDC20 is inhibited by the spindle assembly checkpoint (SAC), which prevents premature separation of sister chromatids and aneuploidy in daughter cells. Although overexpression of CDC20 is common in many cancers, oncogenic mutations have never been identified in humans. Using whole-exome sequencing, we identified heterozygous missense CDC20 variants (L151R and N331K) that segregate with ovarian germ cell tumors in two families. Functional characterization showed these mutants retain APC/C activation activity but have impaired binding to BUBR1, a component of the SAC. Expression of L151R and N331K variants promoted mitotic slippage in HeLa cells and primary skin fibroblasts derived from carriers. Generation of mice carrying the N331K variant using CRISPR-Cas9 showed that, although homozygous N331K mice were nonviable, heterozygotes displayed accelerated oncogenicity of Myc-driven cancers. These findings highlight an unappreciated role for CDC20 variants as tumor-promoting genes.Significance:Two germline CDC20 missense variants that segregate with cancer in two families compromise the spindle assembly checkpoint and lead to aberrant mitotic progression, which could predispose cells to transformation.See related commentary by Villarroya-Beltri and Malumbres, p. 3432
- Published
- 2023
- Full Text
- View/download PDF
14. Specifications of the ACMG/AMP Variant Classification Guidelines for Germline DICER1 Variant Curation
- Author
-
Jessica N. Hatton, Megan N. Frone, Hannah C. Cox, Stephanie B. Crowley, Susan Hiraki, Noriko N. Yokoyama, Noura S. Abul-Husn, James F. Amatruda, Michael J. Anderson, Xavier Bofill-De Ros, Ann G. Carr, Elizabeth C. Chao, Kenneth S. Chen, Shuo Gu, Cecilia Higgs, Jerry Machado, Deborah Ritter, Kris Ann P. Schultz, Emily R. Soper, Mona K. Wu, Jessica L. Mester, Jung Kim, William D. Foulkes, Leora Witkowski, and Douglas R. Stewart
- Subjects
Article Subject ,Genetics ,Genetics (clinical) - Abstract
Germline pathogenic variants in DICER1 predispose individuals to develop a variety of benign and malignant tumors. Accurate variant curation and classification are essential for reliable diagnosis of DICER1-related tumor predisposition and the identification of individuals who may benefit from surveillance. Since 2015, most labs have followed the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP) sequence variant classification guidelines for DICER1 germline variant curation. However, these general guidelines lack gene-specific nuances and leave room for subjectivity. Consequently, a group of DICER1 experts joined ClinGen to form the DICER1 and miRNA-Processing Genes Variant Curation Expert Panel (VCEP) to create DICER1-specific ACMG/AMP guidelines for germline variant curation. The VCEP followed the FDA-approved ClinGen protocol for adapting and piloting these guidelines. A diverse set of 40 DICER1 variants were selected for piloting, including 14 known pathogenic/likely pathogenic (P/LP) variants, 12 known benign/likely benign (B/LB) variants, and 14 variants classified as variants of uncertain significance (VUS) or with conflicting interpretations in ClinVar. Clinically meaningful classifications (i.e., P, LP, LB, or B) were achieved for 82.5% (33/40) of the pilot variants, with 100% concordance among the known P/LP and known B/LB variants. Half of the VUS or conflicting variants were resolved with four variants classified as LB and three as LP. These results demonstrate that the DICER1-specific guidelines for germline variant curation effectively classify known pathogenic and benign variants while reducing the frequency of uncertain classifications. Individuals and labs curating DICER1 variants should consider adopting this classification framework to encourage consistency and improve objectivity.
- Published
- 2023
- Full Text
- View/download PDF
15. Neptune: an environment for the delivery of genomic medicine
- Author
-
Venner Eric, Victoria Yi, David Murdock, Sara E. Kalla, Tsung-Jung Wu, Aniko Sabo, Shoudong Li, Qingchang Meng, Xia Tian, Mullai Murugan, Michelle Cohen, Christie Kovar, Wei-Qi Wei, Wendy K. Chung, Chunhua Weng, Georgia L. Wiesner, Gail P. Jarvik, Donna Muzny, Richard A. Gibbs, Debra Abrams, Samuel E. Adunyah, Ladia Albertson-Junkans, Berta Almoguera, Darren C. Ames, Paul Appelbaum, Samuel Aronson, Sharon Aufox, Lawrence J. Babb, Adithya Balasubramanian, Hana Bangash, Melissa Basford, Lisa Bastarache, Samantha Baxter, Meckenzie Behr, Barbara Benoit, Elizabeth Bhoj, Suzette J. Bielinski, Sarah T. Bland, Carrie Blout, Kenneth Borthwick, Erwin P. Bottinger, Mark Bowser, Harrison Brand, Murray Brilliant, Wendy Brodeur, Pedro Caraballo, David Carrell, Andrew Carroll, Lisa Castillo, Victor Castro, Gauthami Chandanavelli, Theodore Chiang, Rex L. Chisholm, Kurt D. Christensen, Wendy Chung, Christopher G. Chute, Brittany City, Beth L. Cobb, John J. Connolly, Paul Crane, Katherine Crew, David R. Crosslin, Jyoti Dayal, Mariza De Andrade, Jessica De la Cruz, Josh C. Denny, Shawn Denson, Tim DeSmet, Ozan Dikilitas, Michael J. Dinsmore, Sheila Dodge, Phil Dunlea, Todd L. Edwards, Christine M. Eng, David Fasel, Alex Fedotov, Qiping Feng, Mark Fleharty, Andrea Foster, Robert Freimuth, Christopher Friedrich, Stephanie M. Fullerton, Birgit Funke, Stacey Gabriel, Vivian Gainer, Ali Gharavi, Andrew M. Glazer, Joseph T. Glessner, Jessica Goehringer, Adam S. Gordon, Chet Graham, Robert C. Green, Justin H. Gundelach, Heather S. Hain, Hakon Hakonarson, Maegan V. Harden, John Harley, Margaret Harr, Andrea Hartzler, M. Geoffrey Hayes, Scott Hebbring, Nora Henrikson, Andrew Hershey, Christin Hoell, Ingrid Holm, Kayla M. Howell, George Hripcsak, Jianhong Hu, Elizabeth Duffy Hynes, Joy C. Jayaseelan, Yunyun Jiang, Yoonjung Yoonie Joo, Sheethal Jose, Navya Shilpa Josyula, Anne E. Justice, Divya Kalra, Elizabeth W. Karlson, Brendan J. Keating, Melissa A. Kelly, Eimear E. Kenny, Dustin Key, Krzysztof Kiryluk, Terrie Kitchner, Barbara Klanderman, Eric Klee, David C. Kochan, Viktoriya Korchina, Leah Kottyan, Emily Kudalkar, Alanna Kulchak Rahm, Iftikhar J. Kullo, Philip Lammers, Eric B. Larson, Matthew S. Lebo, Magalie Leduc, Ming Ta (Michael) Lee, Niall J. Lennon, Kathleen A. Leppig, Nancy D. Leslie, Rongling Li, Wayne H. Liang, Chiao-Feng Lin, Jodell E. Linder, Noralane M. Lindor, Todd Lingren, James G. Linneman, Cong Liu, Wen Liu, Xiuping Liu, John Lynch, Hayley Lyon, Alyssa Macbeth, Harshad Mahadeshwar, Lisa Mahanta, Bradley Malin, Teri Manolio, Maddalena Marasa, Keith Marsolo, Michelle L. McGowan, Elizabeth McNally, Jim Meldrim, Frank Mentch, Hila Milo Rasouly, Jonathan Mosley, Shubhabrata Mukherjee, Thomas E. Mullen, Jesse Muniz, David R. Murdock, Shawn Murphy, Melanie F. Myers, Bahram Namjou, Yizhao Ni, Robert C. Onofrio, Aniwaa Owusu Obeng, Thomas N. Person, Josh F. Peterson, Lynn Petukhova, Cassandra J. Pisieczko, Siddharth Pratap, Cynthia A. Prows, Megan J. Puckelwartz, Ritika Raj, James D. Ralston, Arvind Ramaprasan, Andrea Ramirez, Luke Rasmussen, Laura Rasmussen-Torvik, Soumya Raychaudhuri, Heidi L. Rehm, Marylyn D. Ritchie, Catherine Rives, Beenish Riza, Dan M. Roden, Elisabeth A. Rosenthal, Avni Santani, Schaid Dan, Steven Scherer, Stuart Scott, Aaron Scrol, Soumitra Sengupta, Ning Shang, Himanshu Sharma, Richard R. Sharp, Rajbir Singh, Patrick M.A. Sleiman, Kara Slowik, Joshua C. Smith, Maureen E. Smith, Duane T. Smoot, Jordan W. Smoller, Sunghwan Sohn, Ian B. Stanaway, Justin Starren, Mary Stroud, Jessica Su, Casey Overby Taylor, Kasia Tolwinski, Sara L. Van Driest, Sean M. Vargas, Matthew Varugheese, David Veenstra, Eric Venner, Miguel Verbitsky, Gina Vicente, Michael Wagner, Kimberly Walker, Theresa Walunas, Liwen Wang, Qiaoyan Wang, Scott T. Weiss, Quinn S. Wells, Peter S. White, Ken L. Wiley, Janet L. Williams, Marc S. Williams, Michael W. Wilson, Leora Witkowski, Laura Allison Woods, Betty Woolf, Julia Wynn, Yaping Yang, Ge Zhang, Lan Zhang, and Hana Zouk
- Subjects
Computer science ,business.industry ,Process (engineering) ,MEDLINE ,High-Throughput Nucleotide Sequencing ,Genomics ,Data science ,Article ,Personalization ,Variety (cybernetics) ,Workflow ,Neptune ,Pharmacogenomics ,Health care ,Electronic Health Records ,Humans ,business ,Software ,Genetics (clinical) - Abstract
Genomic medicine holds great promise for improving health care, but integrating searchable and actionable genetic data into electronic health records (EHRs) remains a challenge. Here we describe Neptune, a system for managing the interaction between a clinical laboratory and an EHR system during the clinical reporting process. We developed Neptune and applied it to two clinical sequencing projects that required report customization, variant reanalysis, and EHR integration. Neptune has been applied for the generation and delivery of over 15,000 clinical genomic reports. This work spans two clinical tests based on targeted gene panels that contain 68 and 153 genes respectively. These projects demanded customizable clinical reports that contained a variety of genetic data types including single-nucleotide variants (SNVs), copy-number variants (CNVs), pharmacogenomics, and polygenic risk scores. Two variant reanalysis activities were also supported, highlighting this important workflow. Methods are needed for delivering structured genetic data to EHRs. This need extends beyond developing data formats to providing infrastructure that manages the reporting process itself. Neptune was successfully applied on two high-throughput clinical sequencing projects to build and deliver clinical reports to EHR systems. The software is open source and available at https://gitlab.com/bcm-hgsc/neptune .
- Published
- 2021
- Full Text
- View/download PDF
16. Small-Cell Carcinoma of the Ovary, Hypercalcemic Type–Genetics, New Treatment Targets, and Current Management Guidelines
- Author
-
Susana Banerjee, Patricia Pautier, Bernard E. Weissman, W. Glenn McCluggage, Krystal A. Orlando, David G. Huntsman, Sidong Huang, William P.D. Hendricks, Isabelle Ray-Coquard, Douglas A. Levine, Amit M. Oza, Leora Witkowski, Jeffrey M. Trent, William D. Foulkes, Jessica D. Lang, Marc Tischkowitz, Michael Witcher, and Jennifer Hague
- Subjects
0301 basic medicine ,Cancer Research ,DNA repair ,Ovariectomy ,Medical Oncology ,medicine.disease_cause ,Small-cell carcinoma ,Article ,Chromatin remodeling ,Germline ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Carcinoma ,Humans ,Carcinoma, Small Cell ,Ovarian Neoplasms ,Genetics ,Mutation ,business.industry ,Ovary ,DNA Helicases ,Nuclear Proteins ,Cancer ,Chromatin Assembly and Disassembly ,medicine.disease ,3. Good health ,Survival Rate ,Treatment Outcome ,030104 developmental biology ,Oncology ,Chemotherapy, Adjuvant ,Gynecology ,030220 oncology & carcinogenesis ,Practice Guidelines as Topic ,Hypercalcemia ,SMARCA4 ,Female ,Radiotherapy, Adjuvant ,business ,Stem Cell Transplantation ,Transcription Factors - Abstract
Small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare and highly aggressive ovarian malignancy. In almost all cases, it is associated with somatic and often germline pathogenic variants in SMARCA4, which encodes for the SMARCA4 protein (BRG1), a subunit of the SWI/SNF chromatin remodeling complex. Approximately 20% of human cancers possess pathogenic variants in at least one SWI/SNF subunit. Because of their role in regulating many important cellular processes including transcriptional control, DNA repair, differentiation, cell division, and DNA replication, SWI/SNF complexes with mutant subunits are thought to contribute to cancer initiation and progression. Fewer than 500 cases of SCCOHT have been reported in the literature and approximately 60% are associated with hypercalcemia. SCCOHT primarily affects females under 40 years of age who usually present with symptoms related to a pelvic mass. SCCOHT is an aggressive cancer, with long-term survival rates of 30% in early-stage cases. Although various treatment approaches have been proposed, there is no consensus on surveillance and therapeutic strategy. An international group of multidisciplinary clinicians and researchers recently formed the International SCCOHT Consortium to evaluate current knowledge and propose consensus surveillance and therapeutic recommendations, with the aim of improving outcomes. Here, we present an overview of the genetics of this cancer, provide updates on new treatment targets, and propose management guidelines for this challenging cancer.
- Published
- 2020
- Full Text
- View/download PDF
17. Malignant teratoid tumor of the thyroid gland: an aggressive primitive multiphenotypic malignancy showing organotypical elements and frequent DICER1 alterations—is the term 'thyroblastoma' more appropriate?
- Author
-
Manuel Sobrinho-Simões, Alfred Brütting, Randa M S Amin, Leora Witkowski, Abbas Agaimy, Markus Bährle, Robert Stoehr, Samir S. Amr, Joseph Christopher Castillo Cuenca, Arndt Hartmann, Catarina Eloy, William D. Foulkes, Markus Metzler, Konstantinos Mantsopoulos, and Carlos Alberto González-Muller
- Subjects
Male ,Ribonuclease III ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Thyroblastoma ,Adolescent ,DICER1 ,Teratocarcinosarcoma ,Biology ,Malignant teratoma ,Pathology and Forensic Medicine ,DEAD-box RNA Helicases ,Head and neck ,03 medical and health sciences ,0302 clinical medicine ,Terminology as Topic ,Rhabdomyosarcoma ,Germ cell tumor ,medicine ,Humans ,Thyroid Neoplasms ,ddc:610 ,Molecular Biology ,Aged ,DICER1 Syndrome ,Thyroid ,Teratoma ,Wilms' tumor ,Cell Biology ,General Medicine ,Middle Aged ,medicine.disease ,Teratoid tumor ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Blastoma ,Female ,Original Article ,PAX8 ,Germ cell - Abstract
Primary thyroid teratomas are exceedingly rare. Mature and immature variants recapitulate their gonadal counterparts (predilection for infants/children, triphasic germ layer differentiation, and favorable outcome). On the other hand, the so-called malignant teratomas affect predominantly adults and elderly, are highly aggressive, and, according to a few published cases, harbor DICER1 mutations. We describe three highly aggressive sporadic malignant teratoid thyroid tumors in 2 females (17 and 45 years) and one male (17 years). Histology showed triphasic neoplasms composed of solid nests of small primitive monomorphic cells embedded in a cellular stroma with primitive immature rhabdomyosarcoma-like (2) or pleomorphic sarcoma-like (1) phenotype. The third component was represented by TTF1+/PAX8+ primitive teratoid epithelial tubules reminiscent of primitive thyroid follicles and/or Wilms tumor, admixed with scattered respiratory- or enteric-type tubules, neuroepithelial rosettes, and fetal-type squamoid nests. Foci of cartilage were seen in two cases, but none contained mature organoid adult-type tissue or skin adnexa. SALL4 was expressed in the small cell (2) and stromal (1) component. Other germ cell markers were negative. Molecular testing revealed a known “hotspot” pathogenic DICER1 mutation in two cases. In addition, case 1 had a missense TP53 variant. This type of thyroid malignancy is distinct from genuine teratomas. The immunoprofile suggests primitive thyroid- or branchial cleft-like differentiation. Given that “blastoma” is a well-accepted terminology in the spectrum of DICER1-associated malignancies, the term “thyroblastoma” might be more convenient for these malignant teratoid tumors of the thyroid gland. Relationship of thyroblastoma to the DICER1 syndrome remains to be addressed. Electronic supplementary material The online version of this article (10.1007/s00428-020-02853-1) contains supplementary material, which is available to authorized users.
- Published
- 2020
- Full Text
- View/download PDF
18. Frequency of genomic incidental findings among 21,915 eMERGE network participants
- Author
-
Ian B. Stanaway, Dan M. Roden, Hakon Hakonarson, Maureen E. Smith, Laura J. Rasmussen-Torvik, Marc S. Williams, Magalie S. Leduc, Jodell E. Linder, Donna M. Muzny, Cynthia A. Prows, Georgia L. Wiesner, Hana Zouk, Iftikhar J. Kullo, Leora Witkowski, Heidi L. Rehm, Sara L. Van Driest, Hila Milo Rasouly, Birgit Funke, Gail P. Jarvik, David Carrell, Eric B. Larson, Wendy K. Chung, Richard A. Gibbs, Christine M. Eng, Joshua C. Denny, Alexander Fedotov, Niall J. Lennon, Kimberly Walker, Elisabeth A. Rosenthal, David R. Crosslin, Eric Venner, Adam S. Gordon, Kathleen A. Leppig, Laura M. Amendola, and Rex L. Chisholm
- Subjects
Incidental Findings ,business.industry ,Genome, Human ,Personal Genomics ,Genomics ,Article ,Clinical Sequencing ,eMERGE ,Neoplasms ,Medicine ,Humans ,Exome ,Genetic Testing ,business ,Genetics (clinical) - Abstract
Discovering an incidental finding (IF) or secondary finding (SF) is a potential result of genomic testing, but few data exist describing types and frequencies of SFs likely to appear in broader clinical populations.The Electronic Medical Records and Genomics Network Phase III (eMERGE III) developed a CLIA-compliant sequencing panel of 109 genes and 1551 variants of clinical relevance or research interest and deployed this panel at ten clinical sites. We evaluated medically actionable SFs across 67 genes and 14 single-nucleotide variants (SNVs) in a diverse cohort of 21,915 participants drawn from a variety of settings (e.g., primary care, biobanks, specialty clinics).Correcting for testing indication, we found a 3.02% overall frequency of SFs; 2.54% from 59 genes the American College of Medical Genetics and Genomics recommends for SF return, and 0.48% in other genes, primarily HFE and PALB2. SFs associated with cancer susceptibility were most frequent (1.38%), followed by cardiovascular diseases (0.87%), and lipid disorders (0.50%). After removing HFE, the frequency of SFs and proportion of pathogenic versus likely pathogenic SFs did not differ in those self-identifying as White versus others.Here we present frequencies and types of medically actionable secondary findings to support informed decision making by patients, participants, and practitioners engaged in genomic medicine.
- Published
- 2020
19. Association of Pathogenic Variants in Hereditary Cancer Genes With Multiple Diseases
- Author
-
Chenjie Zeng, Lisa A. Bastarache, Ran Tao, Eric Venner, Scott Hebbring, Justin D. Andujar, Sarah T. Bland, David R. Crosslin, Siddharth Pratap, Ayorinde Cooley, Jennifer A. Pacheco, Kurt D. Christensen, Emma Perez, Carrie L. Blout Zawatsky, Leora Witkowski, Hana Zouk, Chunhua Weng, Kathleen A. Leppig, Patrick M. A. Sleiman, Hakon Hakonarson, Marc. S. Williams, Yuan Luo, Gail P. Jarvik, Robert C. Green, Wendy K. Chung, Ali G. Gharavi, Niall J. Lennon, Heidi L. Rehm, Richard A. Gibbs, Josh F. Peterson, Dan M. Roden, Georgia L. Wiesner, and Joshua C. Denny
- Subjects
Male ,Cancer Research ,Oncology ,Pancreatitis ,Neoplastic Syndromes, Hereditary ,Gastritis ,Acute Disease ,Humans ,Female ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Original Investigation - Abstract
IMPORTANCE: Knowledge about the spectrum of diseases associated with hereditary cancer syndromes may improve disease diagnosis and management for patients and help to identify high-risk individuals. OBJECTIVE: To identify phenotypes associated with hereditary cancer genes through a phenome-wide association study. DESIGN, SETTING, AND PARTICIPANTS: This phenome-wide association study used health data from participants in 3 cohorts. The Electronic Medical Records and Genomics Sequencing (eMERGEseq) data set recruited predominantly healthy individuals from 10 US medical centers from July 16, 2016, through February 18, 2018, with a mean follow-up through electronic health records (EHRs) of 12.7 (7.4) years. The UK Biobank (UKB) cohort recruited participants from March 15, 2006, through August 1, 2010, with a mean (SD) follow-up of 12.4 (1.0) years. The Hereditary Cancer Registry (HCR) recruited patients undergoing clinical genetic testing at Vanderbilt University Medical Center from May 1, 2012, through December 31, 2019, with a mean (SD) follow-up through EHRs of 8.8 (6.5) years. EXPOSURES: Germline variants in 23 hereditary cancer genes. Pathogenic and likely pathogenic variants for each gene were aggregated for association analyses. MAIN OUTCOMES AND MEASURES: Phenotypes in the eMERGEseq and HCR cohorts were derived from the linked EHRs. Phenotypes in UKB were from multiple sources of health-related data. RESULTS: A total of 214 020 participants were identified, including 23 544 in eMERGEseq cohort (mean [SD] age, 47.8 [23.7] years; 12 611 women [53.6%]), 187 234 in the UKB cohort (mean [SD] age, 56.7 [8.1] years; 104 055 [55.6%] women), and 3242 in the HCR cohort (mean [SD] age, 52.5 [15.5] years; 2851 [87.9%] women). All 38 established gene-cancer associations were replicated, and 19 new associations were identified. These included the following 7 associations with neoplasms: CHEK2 with leukemia (odds ratio [OR], 3.81 [95% CI, 2.64-5.48]) and plasma cell neoplasms (OR, 3.12 [95% CI, 1.84-5.28]), ATM with gastric cancer (OR, 4.27 [95% CI, 2.35-7.44]) and pancreatic cancer (OR, 4.44 [95% CI, 2.66-7.40]), MUTYH (biallelic) with kidney cancer (OR, 32.28 [95% CI, 6.40-162.73]), MSH6 with bladder cancer (OR, 5.63 [95% CI, 2.75-11.49]), and APC with benign liver/intrahepatic bile duct tumors (OR, 52.01 [95% CI, 14.29-189.29]). The remaining 12 associations with nonneoplastic diseases included BRCA1/2 with ovarian cysts (OR, 3.15 [95% CI, 2.22-4.46] and 3.12 [95% CI, 2.36-4.12], respectively), MEN1 with acute pancreatitis (OR, 33.45 [95% CI, 9.25-121.02]), APC with gastritis and duodenitis (OR, 4.66 [95% CI, 2.61-8.33]), and PTEN with chronic gastritis (OR, 15.68 [95% CI, 6.01-40.92]). CONCLUSIONS AND RELEVANCE: The findings of this genetic association study analyzing the EHRs of 3 large cohorts suggest that these new phenotypes associated with hereditary cancer genes may facilitate early detection and better management of cancers. This study highlights the potential benefits of using EHR data in genomic medicine.
- Published
- 2022
20. Germline pathogenicSMARCA4variants in neuroblastoma
- Author
-
Leora Witkowski, Kim E. Nichols, Marjolijn Jongmans, Nienke van Engelen, Ronald R de Krijger, Jennifer Herrera-Mullar, Lieve Tytgat, Armita Bahrami, Helen Mar Fan, Aimee L Davidson, Thomas Robertson, Michael Anderson, Martin Hasselblatt, Sharon E. Plon, and William D Foulkes
- Subjects
Genetics ,Genetics (clinical) - Abstract
Heterozygous germline pathogenic variants (GPVs) inSMARCA4, the gene encoding the ATP-dependent chromatin remodelling protein SMARCA4 (previously known as BRG1), predispose to several rare tumour types, including small cell carcinoma of the ovary, hypercalcaemic type, atypical teratoid and malignant rhabdoid tumour, and uterine sarcoma. The increase in germline testing ofSMARCA4in recent years has revealed putative GPVs affectingSMARCA4in patients with other cancer types. Here we describe 11 patients with neuroblastoma (NBL), including 4 previously unreported cases, all of whom were found to harbour heterozygous germline variants inSMARCA4. Median age at diagnosis was 5 years (range 2 months–26 years); nine were male; and eight of nine cases had tumour location information in the adrenal gland. Eight of the germline variants were expected to result in loss of function ofSMARCA4(large deletion, truncating and canonical splice variants), while the remaining four were missense variants. Loss of heterozygosity of the wild-typeSMARCA4allele was found in all eight cases where somatic testing was performed, supporting the notion that SMARCA4 functions as a classic tumour suppressor. Altogether, these findings strongly suggest that NBL should be included in the spectrum ofSMARCA4-associated tumours.
- Published
- 2023
- Full Text
- View/download PDF
21. Germline Missense Variants in CDC20 Result in Aberrant Mitotic Progression and Familial Cancer
- Author
-
Owen J. Chen, Ester Castellsagué, Mohamed Moustafa-Kamal, Javad Nadaf, Barbara Rivera, Somayyeh Fahiminiya, Yilin Wang, Isabelle Gamache, Caterina Pacifico, Lai Jiang, Jian Carrot-Zhang, Leora Witkowski, Albert M. Berghuis, Stefan Schönberger, Dominik Schneider, Morten Hillmer, Susanne Bens, Reiner Siebert, Colin J.R. Stewart, Ziguo Zhang, William C.H. Chao, Celia M.T. Greenwood, David Barford, Marc Tischkowitz, Jacek Majewski, William D. Foulkes, and Jose G. Teodoro
- Subjects
Cancer Research ,Cdc20 Proteins ,Medizin ,Mitosis ,Cell Cycle Proteins ,Spindle Apparatus ,Anaphase-Promoting Complex-Cyclosome ,Mice ,Germ Cells ,Oncology ,Neoplasms ,Animals ,Humans ,HeLa Cells ,Protein Binding - Abstract
CDC20 is a coactivator of the anaphase promoting complex/cyclosome (APC/C) and is essential for mitotic progression. APC/CCDC20 is inhibited by the spindle assembly checkpoint (SAC), which prevents premature separation of sister chromatids and aneuploidy in daughter cells. Although overexpression of CDC20 is common in many cancers, oncogenic mutations have never been identified in humans. Using whole-exome sequencing, we identified heterozygous missense CDC20 variants (L151R and N331K) that segregate with ovarian germ cell tumors in two families. Functional characterization showed these mutants retain APC/C activation activity but have impaired binding to BUBR1, a component of the SAC. Expression of L151R and N331K variants promoted mitotic slippage in HeLa cells and primary skin fibroblasts derived from carriers. Generation of mice carrying the N331K variant using CRISPR-Cas9 showed that, although homozygous N331K mice were nonviable, heterozygotes displayed accelerated oncogenicity of Myc-driven cancers. These findings highlight an unappreciated role for CDC20 variants as tumor-promoting genes. Significance: Two germline CDC20 missense variants that segregate with cancer in two families compromise the spindle assembly checkpoint and lead to aberrant mitotic progression, which could predispose cells to transformation. See related commentary by Villarroya-Beltri and Malumbres, p. 3432
- Published
- 2021
22. Performance of the McGill Interactive Pediatric OncoGenetic Guidelines for Identifying Cancer Predisposition Syndromes
- Author
-
Josée Brossard, Kathleen Felton, Jemma Say, Adam Fleming, Valerie Larouche, Ronald Grant, Katherine A Blood, Uri Tabori, Melissa Tachdjian, Chantel Cacciotti, Rawan Hammad, Noelle Cullinan, Catherine Goudie, Vijay Ramaswamy, Lucie Lafay-Cousin, Kalene van Engelen, Kim E. Nichols, Ledia Brunga, Linlea Armstrong, Kimberly Caswell, Stephanie Vairy, Jonathan D. Wasserman, Mary Egan Clark, Conrad V. Fernandez, Katherine M. Tucker, Nandini Dendukuri, James A. Whitlock, Ian Schiller, David Malkin, Gino Somers, Annie-Kim Toupin, Nada Jabado, M. Stephen Meyn, Nicolas Waespe, Sonia Cellot, Daniel Sinnett, Paul Gibson, My Linh Thibodeau, Donna L. Johnston, William D. Foulkes, Rebecca J. Deyell, Angela Punnett, David S. Ziegler, Irene Lara-Corrales, Junne Kamihara, Sarah R. Kane, Meghan Pike, Natalie Mathews, Catherine Clinton, Renee Perrier, Lynn W. Harrison, Meredith S. Irwin, Noemi Fuentes-Bolanos, Orli Michaeli, Meera Warby, Adam Shlien, Leora Witkowski, Anita Villani, Hallie Coltin, Paul C. Nathan, and Lara Reichman
- Subjects
Cancer Research ,medicine.medical_specialty ,Referral ,Genetic counseling ,Internal medicine ,Neoplasms ,Medicine ,Humans ,In patient ,Genetic Predisposition to Disease ,Genetic Testing ,Medical diagnosis ,610 Medicine & health ,Child ,Early Detection of Cancer ,Genetic testing ,Original Investigation ,medicine.diagnostic_test ,business.industry ,Cancer predisposition ,Cancer ,Syndrome ,medicine.disease ,Oncology ,Child, Preschool ,business ,Risk assessment ,360 Social problems & social services - Abstract
Importance Prompt recognition of a child with a cancer predisposition syndrome (CPS) has implications for cancer management, surveillance, genetic counseling, and cascade testing of relatives. Diagnosis of CPS requires practitioner expertise, access to genetic testing, and test result interpretation. This diagnostic process is not accessible in all institutions worldwide, leading to missed CPS diagnoses. Advances in electronic health technology can facilitate CPS risk assessment. Objective To evaluate the diagnostic accuracy of a CPS prediction tool (McGill Interactive Pediatric OncoGenetic Guidelines [MIPOGG]) in identifying children with cancer who have a low or high likelihood of having a CPS. Design, Setting, and Participants In this international, multicenter diagnostic accuracy study, 1071 pediatric (
- Published
- 2021
23. Germline Missense Variants in CDC20 Result in Aberrant Mitotic Progression and Familial Cancer
- Author
-
Jacek Majewski, Ziguo Zhang, Marc Tischkowitz, Celia M. T. Greenwood, Dominik Schneider, Colin J.R. Stewart, Stefan Schoenberger, Owen J. Chen, William Chong Hang Chao, Jian Carrot-Zhang, Barbara Rivera, Susanne Bens, Ester Castellsagué, Caterina Pacifico, Yilin Wang, Mohamed Moustafa-Kamal, Albert M. Berghuis, Lai Jiang, Leora Witkowski, Jose G. Teodoro, Reiner Siebert, Javad Nadaf, Somayyeh Fahiminiya, Isabelle Gamache, William D. Foulkes, and David Barford
- Subjects
Spindle checkpoint ,Cancer research ,medicine ,Sister chromatids ,Missense mutation ,Aneuploidy ,CDC20 ,Anaphase-promoting complex ,Biology ,medicine.disease ,Mitosis ,Germline - Abstract
SUMMARYCDC20 is a co-activator of the anaphase promoting complex/cyclosome (APC/C) and is essential for mitotic progression. APC/CCDC20 is inhibited by the spindle assembly checkpoint (SAC), which prevents premature separation of sister chromatids and aneuploidy in daughter cells. Although overexpression of CDC20 is common in many cancers, oncogenic mutations have never been identified in humans. Using whole exome sequencing, we identified heterozygous missense CDC20 variants (L151R and N331K) that segregate with cancer in two families. Characterization of these mutants showed they retain APC/C activation activity but show reduced binding to BUBR1, a component of the SAC. Expression of L151R and N331K promoted mitotic slippage in HeLa cells and primary skin fibroblasts derived from carriers. CRISPR/Cas9 was used to generate mice carrying N331K. Homozygous mice carrying N331K were non-viable, however, heterozygotes displayed accelerated oncogenicity in Myc-driven cancers. These findings highlight an unappreciated role for CDC20 variants as tumor promoting genes in humans.
- Published
- 2021
- Full Text
- View/download PDF
24. SMARCA4/2 loss inhibits chemotherapy-induced apoptosis by restricting IP3R3-mediated Ca 2+ flux to mitochondria
- Author
-
Sophie Camilleri-Broët, Fraser Johnson, Anie Monast, Janusz Rak, Walter H. Gotlieb, Jonathan Spicer, Zheng Fu, Virginie Pilon, Sidong Huang, William W. Lockwood, Jordan L. Morris, Yibo Xue, Pierre Fiset, William D. Foulkes, Tunde Golenar, Mackenzie Coatham, Julien Prudent, Brian Meehan, Marie-Christine Guiot, Lynne-Marie Postovit, Leora Witkowski, Geneviève Morin, Lili Fu, Sungmi Jung, Xianbing Zhu, Kangning Yang, Anne V. Gonzalez, Morag Park, Amber Yasmeen, Xue, Yibo [0000-0003-4252-4446], Morris, Jordan L. [0000-0002-4663-4776], Yang, Kangning [0000-0002-5211-913X], Witkowski, Leora [0000-0002-5736-2534], Camilleri-Broet, Sophie [0000-0002-3453-636X], Spicer, Jonathan [0000-0003-2708-1309], Rak, Janusz [0000-0002-2912-5566], Park, Morag [0000-0001-5400-606X], Lockwood, William [0000-0001-9831-3408], Foulkes, William D. [0000-0001-7427-4651], Prudent, Julien [0000-0003-3821-6088], Huang, Sidong [0000-0002-2838-4726], Apollo - University of Cambridge Repository, Morris, Jordan L [0000-0002-4663-4776], and Foulkes, William D [0000-0001-7427-4651]
- Subjects
Male ,49/47 ,General Physics and Astronomy ,Apoptosis ,Mice, SCID ,13/2 ,0302 clinical medicine ,13/44 ,Intracellular organelle ,Mice, Inbred NOD ,Neoplasms ,Inositol 1,4,5-Trisphosphate Receptors ,14/19 ,13/89 ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,Chemistry ,Histone deacetylase inhibitor ,article ,Nuclear Proteins ,3. Good health ,Chromatin ,Mitochondria ,631/67/68/2486 ,Gene Expression Regulation, Neoplastic ,13/31 ,030220 oncology & carcinogenesis ,631/67/1244 ,SMARCA4 ,38/39 ,medicine.drug ,631/80/82/23 ,medicine.drug_class ,631/67/1059/2326 ,Science ,Antineoplastic Agents ,13/106 ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,82/80 ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,030304 developmental biology ,Cisplatin ,Ion Transport ,Gene Expression Profiling ,DNA Helicases ,General Chemistry ,Xenograft Model Antitumor Assays ,HEK293 Cells ,Cancer cell ,Mutation ,13/95 ,13/51 ,Cancer research ,Calcium ,38/15 ,Transcription Factors - Abstract
Acknowledgements: The authors thank A. Leary for sharing RNA-seq data and R. Hass for sharing the SCCOHT-1 cells. The authors thank members of the Pelletier Laboratory for assistance with flow cytometry studies. This work was supported by Canadian Institutes of Health Research (CIHR) grants MOP-130540, PJT-156233, and PJT-438303 to S.H. and grant FDN-148390 to W.D.F. and a core grant from Medical Research Council (MRC) MC_UU_00015/7 to J.P. Author Y.X. was supported by CIHR Fellowship (MFE-171249), J.L.M. was supported by a MRC-funded graduate student fellowship, and S.H. was supported by a CRC Chair in Functional Genomics., Inactivating mutations in SMARCA4 and concurrent epigenetic silencing of SMARCA2 characterize subsets of ovarian and lung cancers. Concomitant loss of these key subunits of SWI/SNF chromatin remodeling complexes in both cancers is associated with chemotherapy resistance and poor prognosis. Here, we discover that SMARCA4/2 loss inhibits chemotherapy-induced apoptosis through disrupting intracellular organelle calcium ion (Ca2+) release in these cancers. By restricting chromatin accessibility to ITPR3, encoding Ca2+ channel IP3R3, SMARCA4/2 deficiency causes reduced IP3R3 expression leading to impaired Ca2+ transfer from the endoplasmic reticulum to mitochondria required for apoptosis induction. Reactivation of SMARCA2 by a histone deacetylase inhibitor rescues IP3R3 expression and enhances cisplatin response in SMARCA4/2-deficient cancer cells both in vitro and in vivo. Our findings elucidate the contribution of SMARCA4/2 to Ca2+-dependent apoptosis induction, which may be exploited to enhance chemotherapy response in SMARCA4/2-deficient cancers.
- Published
- 2021
25. Harmonizing Clinical Sequencing and Interpretation for the eMERGE III Network
- Author
-
Ian B. Stanaway, Dan M. Roden, Divya Kalra, Dustin Key, Debra J. Abrams, David Fasel, Victor Castro, Brad Malin, Berta Almoguera, Beenish Riza, Meckenzie A. Behr, Eric Venner, Christine M. Eng, Joy Jayaseelan, Scott J. Hebbring, Michelle L. McGowan, Steven E. Scherer, Theresa L. Walunas, Mark Bowser, James D. Ralston, Wei-Qi Wei, Liwen Wang, David R. Murdock, Wayne H. Liang, Julia Wynn, Nancy D. Leslie, Laura J. Rasmussen-Torvik, Ming Ta (Michael) Lee, Frank D. Mentch, Lan Zhang, Alanna Kulchak Rahm, Josh F. Peterson, Jodell E. Linder, Joshua C. Smith, Soumitra Sengupta, Brendan J. Keating, Gina Vicente, Andrew Carroll, Nora B. Henrikson, Anne E. Justice, Heather S. Hain, Wen Liu, Andrea H. Ramirez, Matthew S. Lebo, Hana Zouk, Georgia L. Wiesner, Andrea L. Hartzler, Cassandra J. Pisieczko, Catherine M. Rives, Jessica Goehringer, Maegan V. Harden, John Lynch, Chiao-Feng Lin, Peter White, Phil Dunlea, Shawn N. Murphy, Mullai Murugan, Harshad Mahadeshwar, Mark Fleharty, Andrea Foster, Arvind Ramaprasan, Christopher A. Friedrich, Justin H. Gundelach, Hayley Lyon, Niall J. Lennon, Eric W. Klee, David R. Crosslin, Ge Zhang, Rongling Li, Ozan Dikilitas, Xiuping Liu, Christin Hoell, Aniwaa Owusu Obeng, Katherine D. Crew, Lisa M. Castillo, Justin Starren, Jonathan D. Mosley, Carrie L. Blout, Himanshu Sharma, Elizabeth M. McNally, Sarah T. Bland, Megan J. Puckelwartz, Matthew Varugheese, Keith Marsolo, Betty Woolf, Sharon Aufox, Janet L. Williams, Kimberly Walker, Murray H. Brilliant, Birgit Funke, Laura Allison Woods, Marylyn D. Ritchie, Brittany City, Todd Lingren, Hila Milo Rasouly, Lawrence J. Babb, Alex Fedotov, Robert C. Onofrio, Margaret Harr, Suzette J. Bielinski, Michael W. Wilson, Shubhabrata Mukherjee, Robert R. Freimuth, Chet Graham, Todd L. Edwards, Quinn S. Wells, Marc S. Williams, Jordan W. Smoller, Wendy K. Chung, Avni Santani, Paul K. Crane, George Hripcsak, QiPing Feng, Ali G. Gharavi, Yizhao Ni, Iftikhar J. Kullo, Michael Wagner, Philip E. Lammers, Michael J. Dinsmore, Thomas N. Person, Victoria Yi, Samuel E. Adunyah, Tim DeSmet, Eric B. Larson, Elizabeth Hynes, David C. Kochan, Eimear E. Kenny, Magalie S. Leduc, Lisa Mahanta, David Carrell, Paul S. Appelbaum, Viktoriya Korchina, Beth L. Cobb, Lynn Petukhova, Jessica De la Cruz, Patrick M. A. Sleiman, Stuart A. Scott, Tsung-Jung Wu, Gail P. Jarvik, Erwin P. Bottinger, Ken Wiley, Josh C. Denny, Melissa A. Basford, Samuel J. Aronson, David L. Veenstra, Yaping Yang, Kayla Marie Howell, John J. Connolly, Jessica Su, Yoonjung Yoonie Joo, Miguel Verbitsky, Sean M. Vargas, Cong Liu, Barbara Benoit, Andrew Hershey, Richard A. Gibbs, Cynthia A. Prows, Hana Bangash, Wendy Brodeur, Gauthami Chandanavelli, Sara L. Van Driest, Kurt D. Christensen, Elizabeth J. Bhoj, Vivian S. Gainer, Adam S. Gordon, Robert C. Green, Hakon Hakonarson, Krzysztof Kiryluk, Elisabeth A. Rosenthal, Rajbir Singh, James G. Linneman, Harrison Brand, Theodore Chiang, Sheila Dodge, Ingrid A. Holm, M. Geoffrey Hayes, Yunyun Jiang, Ning Shang, Samantha Baxter, Noralane M. Lindor, Kathleen A. Leppig, Teri A. Manolio, Sara E. Kalla, Pedro J. Caraballo, Ritika Raj, Aaron Scrol, Jyoti G. Dayal, Richard R. Sharp, Christie Kovar, Soumya Raychaudhuri, Sunghwan Sohn, Emily Kudalkar, Maddalena Marasa, Stacey Gabriel, Dan Schaid, Ladia Albertson-Junkans, Rex L. Chisholm, Maureen E. Smith, Donna M. Muzny, Casey Overby Taylor, Jianhong Hu, Elizabeth W. Karlson, Lisa Bastarache, Darren C. Ames, Joseph T. Glessner, Leora Witkowski, Siddharth Pratap, Qiaoyan Wang, Melissa A. Kelly, Adithya Balasubramanian, Kara Slowik, Terrie Kitchner, Barbara J. Klanderman, Shawn Denson, Mary Stroud, Alyssa Macbeth, Melanie F. Myers, Jesse Muniz, Kasia Tolwinski, Scott T. Weiss, Chunhua Weng, Stephanie M. Fullerton, John B. Harley, Christopher G. Chute, Heidi L. Rehm, Sheethal Jose, Andrew M. Glazer, Navya Shilpa Josyula, Kenneth M. Borthwick, Thomas E. Mullen, Mariza de Andrade, Leah C. Kottyan, Luke V. Rasmussen, James Meldrim, and Bahram Namjou
- Subjects
0301 basic medicine ,Standardization ,Test data generation ,business.industry ,Computer science ,Sequence Analysis, DNA ,030105 genetics & heredity ,Precision medicine ,Data science ,Clinical decision support system ,Biobank ,Article ,3. Good health ,Data sharing ,03 medical and health sciences ,030104 developmental biology ,Genetics ,Humans ,Genetic Testing ,Prospective Studies ,Sample collection ,Personalized medicine ,Precision Medicine ,business ,Genetics (clinical) - Abstract
The advancement of precision medicine requires new methods to coordinate and deliver genetic data from heterogeneous sources to physicians and patients. The eMERGE III Network enrolled >25,000 participants from biobank and prospective cohorts of predominantly healthy individuals for clinical genetic testing to determine clinically actionable findings. The network developed protocols linking together the 11 participant collection sites and 2 clinical genetic testing laboratories. DNA capture panels targeting 109 genes were used for testing of DNA and sample collection, data generation, interpretation, reporting, delivery, and storage were each harmonized. A compliant and secure network enabled ongoing review and reconciliation of clinical interpretations, while maintaining communication and data sharing between clinicians and investigators. A total of 202 individuals had positive diagnostic findings relevant to the indication for testing and 1,294 had additional/secondary findings of medical significance deemed to be returnable, establishing data return rates for other testing endeavors. This study accomplished integration of structured genomic results into multiple electronic health record (EHR) systems, setting the stage for clinical decision support to enable genomic medicine. Further, the established processes enable different sequencing sites to harmonize technical and interpretive aspects of sequencing tests, a critical achievement toward global standardization of genomic testing. The eMERGE protocols and tools are available for widespread dissemination.
- Published
- 2019
- Full Text
- View/download PDF
26. A scalable EHR-based approach for phenotype discovery and variant interpretation for hereditary cancer genes
- Author
-
Ali G. Gharavi, Josh F. Peterson, Yuan Luo, Joshua C. Denny, Scott J. Hebbring, Blout Zawatsky Cl, Patrick M. A. Sleiman, David R. Crosslin, Dan M. Roden, Hakon Hakonarson, Sarah Bland, Eric Venner, Andujar Jd, Hana Zouk, Leora Witkowski, Ayorinde Cooley, Marc S. Williams, Jennifer A. Pacheco, Emma Perez, Kurt D. Christensen, Zeng C, Kathleen A. Leppig, Gail P. Jarvik, Ran Tao, Chunhua Weng, Georgia L. Wiesner, Robert C. Green, Richard A. Gibbs, Niall J. Lennon, Wendy K. Chung, Heidi L. Rehm, Lisa Bastarache, and Siddharth Pratap
- Subjects
Genetics ,medicine.diagnostic_test ,Electronic health record ,Cohort ,medicine ,Genomic medicine ,Hereditary Cancer ,Biology ,Pathogenicity ,Gene ,Phenotype ,Genetic testing - Abstract
Knowledge of the clinical spectrum of rare genetic disorders helps in disease management and variant pathogenicity interpretation. Leveraging electronic health record (EHR)-linked genetic testing data from the eMERGE network, we determined the associations between a set of 23 hereditary cancer genes and 3017 phenotypes in 23544 individuals. This phenome-wide association study replicated 45% (184/406) of known gene-phenotype associations (P = 5.1×10−125). Meta-analysis with an independent EHR-derived cohort of 3242 patients confirmed 14 novel associations with phenotypes in the neoplastic, genitourinary, digestive, congenital, metabolic, mental and neurologic categories. Phenotype risk scores (PheRS) based on weighted aggregations of EHR phenotypes accurately predicted variant pathogenicity for at least 50% of pathogenic variants for 8/23 genes. We generated a catalog of PheRS for 7800 variants, including 5217 variants of uncertain significance, to provide empirical evidence of potential pathogenicity. This study highlights the potential of EHR data in genomic medicine.
- Published
- 2021
- Full Text
- View/download PDF
27. SMARCA4/2 loss inhibits chemotherapy-induced apoptosis by restricting IP3R3-mediated Ca
- Author
-
Yibo, Xue, Jordan L, Morris, Kangning, Yang, Zheng, Fu, Xianbing, Zhu, Fraser, Johnson, Brian, Meehan, Leora, Witkowski, Amber, Yasmeen, Tunde, Golenar, Mackenzie, Coatham, Geneviève, Morin, Anie, Monast, Virginie, Pilon, Pierre Olivier, Fiset, Sungmi, Jung, Anne V, Gonzalez, Sophie, Camilleri-Broet, Lili, Fu, Lynne-Marie, Postovit, Jonathan, Spicer, Walter H, Gotlieb, Marie-Christine, Guiot, Janusz, Rak, Morag, Park, William, Lockwood, William D, Foulkes, Julien, Prudent, and Sidong, Huang
- Subjects
Male ,Antineoplastic Agents ,Apoptosis ,Mice, SCID ,Article ,Cancer epigenetics ,Mice, Inbred NOD ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Inositol 1,4,5-Trisphosphate Receptors ,Tumour-suppressor proteins ,Mice, Knockout ,Ion Transport ,Gene Expression Profiling ,DNA Helicases ,Nuclear Proteins ,Xenograft Model Antitumor Assays ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Cancer therapeutic resistance ,HEK293 Cells ,Mutation ,Calcium ,Transcription Factors - Abstract
Inactivating mutations in SMARCA4 and concurrent epigenetic silencing of SMARCA2 characterize subsets of ovarian and lung cancers. Concomitant loss of these key subunits of SWI/SNF chromatin remodeling complexes in both cancers is associated with chemotherapy resistance and poor prognosis. Here, we discover that SMARCA4/2 loss inhibits chemotherapy-induced apoptosis through disrupting intracellular organelle calcium ion (Ca2+) release in these cancers. By restricting chromatin accessibility to ITPR3, encoding Ca2+ channel IP3R3, SMARCA4/2 deficiency causes reduced IP3R3 expression leading to impaired Ca2+ transfer from the endoplasmic reticulum to mitochondria required for apoptosis induction. Reactivation of SMARCA2 by a histone deacetylase inhibitor rescues IP3R3 expression and enhances cisplatin response in SMARCA4/2-deficient cancer cells both in vitro and in vivo. Our findings elucidate the contribution of SMARCA4/2 to Ca2+-dependent apoptosis induction, which may be exploited to enhance chemotherapy response in SMARCA4/2-deficient cancers., SMARCA4/2 loss in ovarian and lung cancers is associated with chemotherapy resistance. Here, the authors show that SMARCA4/2 deficiency in cancer cells reduces the expression of the ER-Ca2+ channel IP3R3 and subsequently calcium transfer to the mitochondria, which inhibits apoptotic cell death.
- Published
- 2020
28. Retrospective Analysis of Clinical Genetic Testing in Pediatric Primary Dilated Cardiomyopathy: Testing Outcomes and the Effects of Variant Reclassification
- Author
-
Kevin P. Daly, Daniel Quiat, Hana Zouk, Leora Witkowski, and Amy E. Roberts
- Subjects
Cardiomyopathy, Dilated ,Pediatrics ,medicine.medical_specialty ,Cardiomyopathy ,Familial dilated cardiomyopathy ,familial dilated cardiomyopathy ,variant reanalysis ,030204 cardiovascular system & hematology ,genetic testing ,03 medical and health sciences ,0302 clinical medicine ,Idiopathic dilated cardiomyopathy ,Genetics ,medicine ,Clinical genetic ,Retrospective analysis ,Humans ,Child ,Original Research ,Retrospective Studies ,030304 developmental biology ,Genetic testing ,0303 health sciences ,medicine.diagnostic_test ,business.industry ,variant of uncertain significance ,Primary dilated cardiomyopathy ,Pedigree ,idiopathic dilated cardiomyopathy ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background Genetic testing in pediatric primary dilated cardiomyopathy (DCM) patients has identified numerous disease‐causing variants, but few studies have evaluated genetic testing outcomes in this population in the context of patient and familial clinical data or assessed the clinical implications of temporal changes in genetic testing results. Methods and Results We performed a retrospective analysis of all patients with primary DCM who presented to our institution between 2008 and 2018. Variants identified by genetic testing were reevaluated for pathogenicity on the basis of current guidelines for variant classification. A total of 73 patients with primary DCM presented to our institution and 63 (86%) were probands that underwent cardiomyopathy‐specific gene testing. A disease‐causing variant was identified in 19 of 63 (30%) of cases, with at least 9/19 (47%) variants occurring de novo. Positive family history was not associated with identification of a causal variant. Reclassification of variants resulted in the downgrading of a large proportion of variants of uncertain significance and did not identify any new disease‐causing variants. Conclusions Clinical genetic testing identifies a causal variant in one third of pediatric patients with primary DCM. Variant reevaluation significantly decreased the number of variants of uncertain significance, but a large burden of variants of uncertain significance remain. These results highlight the need for periodic reanalysis of genetic testing results, additional investigation of genotype‐phenotype correlations in DCM through large, multicenter genetic studies, and development of improved tools for functional characterization of variants of uncertain significance.
- Published
- 2020
- Full Text
- View/download PDF
29. Association of Rare Pathogenic DNA Variants for Familial Hypercholesterolemia, Hereditary Breast and Ovarian Cancer Syndrome, and Lynch Syndrome With Disease Risk in Adults According to Family History
- Author
-
Megan H. Hawley, Kenney Ng, Kalotina Machini, Christopher A. Cassa, Aniruddh P. Patel, Renee C. Pelletier, Leora Witkowski, Akl C. Fahed, Patrick T. Ellinor, Minxian Wang, Christopher Koch, Sami S. Amr, Sekar Kathiresan, Matthew S. Lebo, Deanna Brockman, Amit Khera, Anthony A. Philippakis, and Heather Mason-Suares
- Subjects
Male ,medicine.medical_specialty ,Heterozygote ,Context (language use) ,Familial hypercholesterolemia ,Disease ,Article ,Cohort Studies ,Hyperlipoproteinemia Type II ,Uterine cancer ,Internal medicine ,Exome Sequencing ,Medicine ,Humans ,Genetic Predisposition to Disease ,Family history ,Aged ,Proportional Hazards Models ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Colorectal Neoplasms, Hereditary Nonpolyposis ,Lynch syndrome ,United Kingdom ,Cancer registry ,Pedigree ,Hereditary Breast and Ovarian Cancer Syndrome ,Female ,business ,Ovarian cancer - Abstract
Importance Pathogenic DNA variants associated with familial hypercholesterolemia, hereditary breast and ovarian cancer syndrome, and Lynch syndrome are widely recognized as clinically important and actionable when identified, leading some clinicians to recommend population-wide genomic screening. Objectives To assess the prevalence and clinical importance of pathogenic or likely pathogenic variants associated with each of 3 genomic conditions (familial hypercholesterolemia, hereditary breast and ovarian cancer syndrome, and Lynch syndrome) within the context of contemporary clinical care. Design, Setting, and Participants This cohort study used gene-sequencing data from 49 738 participants in the UK Biobank who were recruited from 22 sites across the UK between March 21, 2006, and October 1, 2010. Inpatient hospital data date back to 1977; cancer registry data, to 1957; and death registry data, to 2006. Statistical analysis was performed from July 22, 2019, to November 15, 2019. Exposures Pathogenic or likely pathogenic DNA variants classified by a clinical laboratory geneticist. Main Outcomes and Measures Composite end point specific to each genomic condition based on atherosclerotic cardiovascular disease events for familial hypercholesterolemia, breast or ovarian cancer for hereditary breast and ovarian cancer syndrome, and colorectal or uterine cancer for Lynch syndrome. Results Among 49 738 participants (mean [SD] age, 57 [8] years; 27 144 female [55%]), 441 (0.9%) harbored a pathogenic or likely pathogenic variant associated with any of 3 genomic conditions, including 131 (0.3%) for familial hypercholesterolemia, 235 (0.5%) for hereditary breast and ovarian cancer syndrome, and 76 (0.2%) for Lynch syndrome. Presence of these variants was associated with increased risk of disease: for familial hypercholesterolemia, 28 of 131 carriers (21.4%) vs 4663 of 49 607 noncarriers (9.4%) developed atherosclerotic cardiovascular disease; for hereditary breast and ovarian cancer syndrome, 32 of 116 female carriers (27.6%) vs 2080 of 27 028 female noncarriers (7.7%) developed associated cancers; and for Lynch syndrome, 17 of 76 carriers (22.4%) vs 929 of 49 662 noncarriers (1.9%) developed colorectal or uterine cancer. The predicted probability of disease at age 75 years despite contemporary clinical care was 45.3% for carriers of familial hypercholesterolemia, 41.1% for hereditary breast and ovarian cancer syndrome, and 38.3% for Lynch syndrome. Across the 3 conditions, 39.7% (175 of 441) of the carriers reported a family history of disease vs 23.2% (34 517 of 148 772) of noncarriers. Conclusions and Relevance The findings suggest that approximately 1% of the middle-aged adult population in the UK Biobank harbored a pathogenic variant associated with any of 3 genomic conditions. These variants were associated with an increased risk of disease despite contemporary clinical care and were not reliably detected by family history.
- Published
- 2020
30. Expanding the Noonan spectrum/RASopathy NGS panel: Benefits of adding NF1 and SPRED1
- Author
-
Matthew S. Lebo, Mitchell W Dillon, Leora Witkowski, Elissa Murphy, and Heather Mason-Suares
- Subjects
0301 basic medicine ,Male ,Pediatrics ,Watson syndrome ,030105 genetics & heredity ,Child ,Uncertain significance ,Genetics (clinical) ,Neurofibromin 1 ,Noonan Syndrome ,High-Throughput Nucleotide Sequencing ,Derivation cohort ,Patient management ,Legius syndrome ,Neurofibromatosis‐Noonan syndrome (NFNS) ,Child, Preschool ,Female ,Original Article ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,SPRED1 ,Neurofibromatosis 1 ,lcsh:QH426-470 ,RASopathy ,03 medical and health sciences ,Genetics ,medicine ,Humans ,Genetic Testing ,Neurofibromatosis ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,business.industry ,Infant ,Sequence Analysis, DNA ,Original Articles ,Noonan syndrome (NS) ,medicine.disease ,lcsh:Genetics ,030104 developmental biology ,NF1 ,Mutation ,ras Proteins ,business ,Validation cohort ,Noonan syndrome with multiple lentigines (NSML) ,Neurofibromatosis type 1 - Abstract
Background RASopathies are a group of disorders caused by disruptions to the RAS‒MAPK pathway. Despite being in the same pathway, Neurofibromatosis Type 1 (NF1) and Legius syndrome (LS) typically present with phenotypes distinct from Noonan spectrum disorders (NSDs). However, some NF1/LS individuals also exhibit NSD phenotypes, often referred to as Neurofibromatosis‐Noonan syndrome (NFNS), and may be mistakenly evaluated for NSDs, delaying diagnosis, and affecting patient management. Methods A derivation cohort of 28 patients with a prior negative NSD panel and either NFNS or a suspicion of NSD and café‐au‐lait spots underwent NF1 and SPRED1 sequencing. To further determine the utility and burden of adding these genes, a validation cohort of 505 patients with a suspected RASopathy were tested on a 14‐gene RASopathy‐associated panel. Results In the derivation cohort, six (21%) patients had disease‐causing NF1 or SPRED1 variants. In the validation cohort, 11 (2%) patients had disease‐causing variants and 15 (3%) had variants of uncertain significance in NF1 or SPRED1. Of those with disease‐causing variants, 5/17 only had an NSD diagnosis. Conclusions Adding NF1 and SPRED1 to RASopathy panels can speed diagnosis and improve patient management, without significantly increasing the burden of inconclusive results., Adding the NF1 and SPRED1 genes to Noonan spectrum disorder/RASopathy NGS gene panels modestly increases clinical diagnoses without significantly increasing the VUS burden. Since a diagnosis of NF1 or LS would change patient management, the NF1 and SPRED1 genes should be included on all to Noonan spectrum disorder/RASopathy NGS gene panels, including those used prenatally.
- Published
- 2019
31. Case 35-2018: A Woman with Back Pain and a Remote History of Breast Cancer. Reply
- Author
-
William D. Foulkes and Leora Witkowski
- Subjects
medicine.medical_specialty ,business.industry ,General surgery ,MEDLINE ,Breast Neoplasms ,General Medicine ,medicine.disease ,Breast cancer ,Text mining ,Back Pain ,medicine ,Back pain ,Humans ,Female ,medicine.symptom ,business ,Aged - Published
- 2019
32. The hereditary nature of small cell carcinoma of the ovary, hypercalcemic type: two new familial cases
- Author
-
Nancy Donini, William D. Foulkes, Rebecca Byler-Dann, Steffen Albrecht, James A. Knost, Andrew Berchuck, Martin Hasselblatt, W. Glenn McCluggage, and Leora Witkowski
- Subjects
0301 basic medicine ,Adult ,Cancer Research ,Pathology ,medicine.medical_specialty ,Somatic cell ,Short Communication ,Biology ,medicine.disease_cause ,Small-cell carcinoma ,Germline ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Germline mutation ,Ovarian cancer ,SMARCA4 ,Rhabdoid ,Genetics ,medicine ,SCCOHT ,Humans ,Genetics(clinical) ,Genetic Predisposition to Disease ,SMARCB1 ,Carcinoma, Small Cell ,Genetics (clinical) ,Ovarian Neoplasms ,Mutation ,DNA Helicases ,Nuclear Proteins ,medicine.disease ,Pedigree ,030104 developmental biology ,Hereditary ,Oncology ,030220 oncology & carcinogenesis ,Hypercalcemia ,Female ,Transcription Factors - Abstract
Small cell carcinoma of the ovary, hypercalcemic type, (SCCOHT) is the most common undifferentiated ovarian cancer in women aged under 40 years. SCCOHT is a monogenic disease, characterized by germline and somatic SMARCA4 mutations. Recent studies have stressed its morphological and clinical similarity to malignant rhabdoid tumours, which are usually caused by mutations in the related gene, SMARCB1. While familial tumours are rare, the incidence of germline mutations is relatively high, with up to 43% of SCCOHTs and 35% of rhabdoid tumours caused by germline mutations in SMARCA4 and SMARCB1, respectively. We report two new familial cases of SCCOHT. Affected members in both families and the associated tumours were found to carry SMARCA4 germline and somatic mutations, respectively, leading to loss of SMARCA4 protein expression in the tumours. Despite the rarity of familial SCCOHT, the high incidence of germline mutations is important to note, as without a family history of the disease, the hereditary nature of SCCOHT may be missed, especially if the mutation was inherited from the father or acquired de novo. The similarity between SCCOHT and rhabdoid tumours should be recognized, as infant carriers of SMARCA4 mutations may be at risk for these tumours in addition to SCCOHT.
- Published
- 2016
33. Loss of SMARCA4 (BRG1) protein expression as determined by immunohistochemistry in small-cell carcinoma of the ovary, hypercalcaemic type distinguishes these tumours from their mimics
- Author
-
Anthony N. Karnezis, W. Glenn McCluggage, Janez Lamovec, Neil J. Sebire, Tuyet Nhung Ton Nu, C. Blake Gilks, Patricia Shaw, Leora Witkowski, David G. Huntsman, Martin Hasselblatt, Blaise A. Clarke, William D. Foulkes, Colin J.R. Stewart, and Lawrence M. Roth
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Histology ,Ovary ,Carcinoma, Ovarian Epithelial ,Biology ,Sensitivity and Specificity ,Small-cell carcinoma ,Pathology and Forensic Medicine ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Biomarkers, Tumor ,medicine ,Carcinoma ,Humans ,Neoplasms, Glandular and Epithelial ,Carcinoma, Small Cell ,SMARCB1 ,Nuclear protein ,Ovarian Neoplasms ,DNA Helicases ,Nuclear Proteins ,General Medicine ,medicine.disease ,Immunohistochemistry ,Staining ,030104 developmental biology ,medicine.anatomical_structure ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,SMARCA4 ,Cancer research ,Female ,Transcription Factors - Abstract
Aims Molecular investigation of small-cell carcinoma of the ovary, hypercalcaemic type (SCCOHT) has revealed that it is a monogenetic tumour characterized by alteration of SMARCA4 (BRG1), encoding a member of the switch/sucrose non-fermentable (SWI/SNF) chromatin remodelling complex. A large majority of cases show loss of expression of the corresponding SMARCA4/BRG1 protein. Furthermore, three cases of SCCOHT with retained SMARCA4 protein expression showed loss of SMARCB1/INI1 expression. The aim of this study was to assess the sensitivity and specificity of loss of SMARCA4 expression as a diagnostic test for SCCOHT. Methods and results We performed SMARCA4 and SMARCB1 staining in 245 tumours, many of which were potentially in the differential diagnosis of SCCOHT. We also stained 56 cases of SCCOHT for SMARCA4 and 37 of these for SMARCB1. Fifty-four of the SCCOHT cases showed complete absence of SMARCA4 expression. The two cases with retained expression showed molecular alteration of SMARCA4. Of the 217 other neoplasms with interpretable staining, all retained SMARCA4 expression. Although the majority showed diffuse, strong nuclear expression, a heterogeneous, typically weak staining pattern was present in 13% of cases. All 37 cases of SCCOHT tested and all other neoplasms, apart from three malignant rhabdoid tumours, showed retained nuclear SMARCB1 expression. Loss of SMARCA4 expression had a sensitivity of 96.55% and specificity of 100%. Conclusions Loss of SMARCA4 expression is sensitive and specific for SCCOHT. Although some mimics show heterogeneous expression, there is retention of nuclear staining in at least a part of the tumour; therefore, only complete loss of staining should be regarded as being supportive of SCCOHT.
- Published
- 2016
- Full Text
- View/download PDF
34. Small-Cell Carcinoma of the Ovary of Hypercalcemic Type (Malignant Rhabdoid Tumor of the Ovary)
- Author
-
Leora Witkowski, W. Glenn McCluggage, William D. Foulkes, and Catherine Goudie
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Somatic cell ,Ovary ,Biology ,medicine.disease ,Small-cell carcinoma ,Germline ,Pathology and Forensic Medicine ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Carcinoma ,medicine ,SMARCA4 ,Immunohistochemistry ,Surgery - Abstract
Small-cell carcinoma of the ovary of hypercalcemic type (SCCOHT) is a highly malignant and aggressive tumor and is the most common undifferentiated ovarian malignancy to occur in women younger than 40. SCCOHT is characterized by deleterious germline or somatic mutations in SMARCA4. Given the striking morphologic and molecular similarities between SCCOHT and atypical teratoid/malignant rhabdoid tumor, we propose this should be reflected in a nomenclature change and that SCCOHT be renamed malignant rhabdoid tumor of the ovary. SMARCA4 (BRG1) immunohistochemistry is useful in diagnosis because there is loss of nuclear immunoreactivity in SCCOHT but retention of staining in mimics.
- Published
- 2016
- Full Text
- View/download PDF
35. Detection of Pathogenic Variants With Germline Genetic Testing Using Deep Learning vs Standard Methods in Patients With Prostate Cancer and Melanoma
- Author
-
Haitham Elmarakeby, Eric Kofman, Nicholas M. Moore, Brendan Reardon, Abdulsalam A. Al-Sulaiman, Hani Choudhry, Sabrina Y. Camp, Keyan Salari, Eliezer M. Van Allen, Saud H. AlDubayan, Jake Conway, Amaro Taylor-Weiner, Seunghun Han, Leora Witkowski, Abdullah M. Al-Rubaish, and Amein K. Al-Ali
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,Cross-sectional study ,business.industry ,Melanoma ,010102 general mathematics ,Cancer ,General Medicine ,medicine.disease ,01 natural sciences ,Gastroenterology ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Germline mutation ,Predictive value of tests ,Internal medicine ,Cohort ,medicine ,030212 general & internal medicine ,0101 mathematics ,business ,Genetic testing - Abstract
Importance Less than 10% of patients with cancer have detectable pathogenic germline alterations, which may be partially due to incomplete pathogenic variant detection. Objective To evaluate if deep learning approaches identify more germline pathogenic variants in patients with cancer. Design, setting, and participants A cross-sectional study of a standard germline detection method and a deep learning method in 2 convenience cohorts with prostate cancer and melanoma enrolled in the US and Europe between 2010 and 2017. The final date of clinical data collection was December 2017. Exposures Germline variant detection using standard or deep learning methods. Main outcomes and measures The primary outcomes included pathogenic variant detection performance in 118 cancer-predisposition genes estimated as sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV). The secondary outcomes were pathogenic variant detection performance in 59 genes deemed actionable by the American College of Medical Genetics and Genomics (ACMG) and 5197 clinically relevant mendelian genes. True sensitivity and true specificity could not be calculated due to lack of a criterion reference standard, but were estimated as the proportion of true-positive variants and true-negative variants, respectively, identified by each method in a reference variant set that consisted of all variants judged to be valid from either approach. Results The prostate cancer cohort included 1072 men (mean [SD] age at diagnosis, 63.7 [7.9] years; 857 [79.9%] with European ancestry) and the melanoma cohort included 1295 patients (mean [SD] age at diagnosis, 59.8 [15.6] years; 488 [37.7%] women; 1060 [81.9%] with European ancestry). The deep learning method identified more patients with pathogenic variants in cancer-predisposition genes than the standard method (prostate cancer: 198 vs 182; melanoma: 93 vs 74); sensitivity (prostate cancer: 94.7% vs 87.1% [difference, 7.6%; 95% CI, 2.2% to 13.1%]; melanoma: 74.4% vs 59.2% [difference, 15.2%; 95% CI, 3.7% to 26.7%]), specificity (prostate cancer: 64.0% vs 36.0% [difference, 28.0%; 95% CI, 1.4% to 54.6%]; melanoma: 63.4% vs 36.6% [difference, 26.8%; 95% CI, 17.6% to 35.9%]), PPV (prostate cancer: 95.7% vs 91.9% [difference, 3.8%; 95% CI, -1.0% to 8.4%]; melanoma: 54.4% vs 35.4% [difference, 19.0%; 95% CI, 9.1% to 28.9%]), and NPV (prostate cancer: 59.3% vs 25.0% [difference, 34.3%; 95% CI, 10.9% to 57.6%]; melanoma: 80.8% vs 60.5% [difference, 20.3%; 95% CI, 10.0% to 30.7%]). For the ACMG genes, the sensitivity of the 2 methods was not significantly different in the prostate cancer cohort (94.9% vs 90.6% [difference, 4.3%; 95% CI, -2.3% to 10.9%]), but the deep learning method had a higher sensitivity in the melanoma cohort (71.6% vs 53.7% [difference, 17.9%; 95% CI, 1.82% to 34.0%]). The deep learning method had higher sensitivity in the mendelian genes (prostate cancer: 99.7% vs 95.1% [difference, 4.6%; 95% CI, 3.0% to 6.3%]; melanoma: 91.7% vs 86.2% [difference, 5.5%; 95% CI, 2.2% to 8.8%]). Conclusions and relevance Among a convenience sample of 2 independent cohorts of patients with prostate cancer and melanoma, germline genetic testing using deep learning, compared with the current standard genetic testing method, was associated with higher sensitivity and specificity for detection of pathogenic variants. Further research is needed to understand the relevance of these findings with regard to clinical outcomes.
- Published
- 2020
- Full Text
- View/download PDF
36. Expanding the diagnostic yield of germline genetic testing in cancer patients using deep learning
- Author
-
Jake Conway, Amaro Taylor-Weiner, Abdulsalam A. Al-Sulaiman, Seunghun Han, Sabrina Y. Camp, Eliezer M. Van Allen, Saud H. AlDubayan, Haitham Elmarakeby, Brendan Reardon, Eric Kofman, Amein K. Al-Ali, Leora Witkowski, and Abdullah M. Al-Rubaish
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Cancer prevention ,medicine.diagnostic_test ,business.industry ,Deep learning ,Cancer ,medicine.disease ,Germline ,Feature (computer vision) ,Internal medicine ,medicine ,Artificial intelligence ,business ,Genetic testing - Abstract
1518 Background: Germline genetic analysis is an essential tool for implementing precision cancer prevention and treatment. However, only a small fraction of cancer patients, even those with features suggestive of a cancer-predisposition syndrome, have detectable pathogenic germline events, which may in part reflect incomplete pathogenic variant detection by current gold-standard methods. Here, we leveraged deep learning approaches to expand the diagnostic utility of genetic analysis in cancer patients. Methods: Systematic analysis of the detection rate of pathogenic cancer-predisposition variants using the standard clinical variant detection method and a deep learning approach in germline whole-exome sequencing data of 2367 cancer patients (n = 1072 prostate cancer, 1295 melanoma). Results: Of 1072 prostate cancer patients, deep learning variant detection identified 16 additional prostate cancer patients with clinically actionable pathogenic cancer-predisposition variants that went undetected by the gold-standard method (198 vs. 182), yielding higher sensitivity (94.7% vs. 87.1%), specificity (64.0% vs. 36.0%), positive predictive value (95.7% vs. 91.9%), and negative predictive value (59.3% vs. 25.0%). Similarly, germline genetic analysis of 1295 melanoma patients showed that, compared with the standard method, deep learning detected 19 additional patients with validated pathogenic variants (93 vs. 74) with fewer false-positive calls (78 vs. 135) leading to a higher diagnostic yield. Collectively, deep learning identified one additional patient with a pathogenic cancer-risk variant, that went undetected by the standard method, for every 52 to 67 cancer patients undergoing germline analysis. Superior performance of deep learning, for detecting putative loss-of-function variants, was also seen across 5197 clinically relevant Mendelian genes in these cohorts. Conclusions: The gold-standard germline variant detection method, universally used in clinical and research settings, has significant limitations for identifying clinically relevant pathogenic disease-causing variants. We determined that deep learning approaches have a clinically significant increase in the diagnostic yield across commonly examined Mendelian gene sets.
- Published
- 2020
- Full Text
- View/download PDF
37. CDK4/6 inhibitors target SMARCA4-determined cyclin D1 deficiency in hypercalcemic small cell carcinoma of the ovary
- Author
-
Alexander R. Judkins, Jerry Pelletier, Sidong Huang, Regina Cencic, Ron M. Kerkhoven, Douglas A. Levine, Yibo Xue, William D. Foulkes, Morag Park, Barbara C. Vanderhyden, Radia M. Johnson, Nicholas Bertos, Janusz Rak, Alexandra Leary, Aurélie Auguste, Jay R. Patibandla, Leora Witkowski, Marie-Christine Guiot, Hongbo Chen, Michelle Firlit, Brian Meehan, Daniel Martinez, Xue Qing D. Wang, Tim Kong, Petar Jelinic, Blaise A. Clarke, Michael Witcher, Geneviève Morin, Josée Dostie, Andreas I. Papadakis, Sriram Venneti, Elizabeth Macdonald, Walter H. Gotlieb, Ralf Hass, Iris de Rink, and Atefeh Abedini
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,Cell Survival ,Pyridines ,Science ,General Physics and Astronomy ,Aminopyridines ,02 engineering and technology ,Mice, SCID ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Piperazines ,Article ,03 medical and health sciences ,Mice ,Cyclin D1 ,Downregulation and upregulation ,RNA interference ,Cell Line, Tumor ,Animals ,Humans ,Kinase activity ,Carcinoma, Small Cell ,RNA, Small Interfering ,lcsh:Science ,Protein Kinase Inhibitors ,Ovarian Neoplasms ,Multidisciplinary ,Oncogene ,Kinase ,DNA Helicases ,Nuclear Proteins ,General Chemistry ,021001 nanoscience & nanotechnology ,3. Good health ,030104 developmental biology ,Purines ,SMARCA4 ,Cancer research ,Hypercalcemia ,lcsh:Q ,Benzimidazoles ,Female ,biological phenomena, cell phenomena, and immunity ,0210 nano-technology ,Chromatin immunoprecipitation ,Transcription Factors - Abstract
Inactivating mutations in SMARCA4 (BRG1), a key SWI/SNF chromatin remodelling gene, underlie small cell carcinoma of the ovary, hypercalcemic type (SCCOHT). To reveal its druggable vulnerabilities, we perform kinase-focused RNAi screens and uncover that SMARCA4-deficient SCCOHT cells are highly sensitive to the inhibition of cyclin-dependent kinase 4/6 (CDK4/6). SMARCA4 loss causes profound downregulation of cyclin D1, which limits CDK4/6 kinase activity in SCCOHT cells and leads to in vitro and in vivo susceptibility to CDK4/6 inhibitors. SCCOHT patient tumors are deficient in cyclin D1 yet retain the retinoblastoma-proficient/p16INK4a-deficient profile associated with positive responses to CDK4/6 inhibitors. Thus, our findings indicate that CDK4/6 inhibitors, approved for a breast cancer subtype addicted to CDK4/6 activation, could be repurposed to treat SCCOHT. Moreover, our study suggests a novel paradigm whereby critically low oncogene levels, caused by loss of a driver tumor suppressor, may also be exploited therapeutically., Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is driven by SMARCA4 loss. Here the authors demonstrate that SCCOHT cells are highly sensitive to CDK4/6 inhibition and provide mechanistic insights, whereby this druggable vulnerability is driven by cyclin D1 deficiency induced by SMARCA4 loss.
- Published
- 2018
38. Molecular analyses reveal close similarities between small cell carcinoma of the ovary, hypercalcemic type and atypical teratoid/rhabdoid tumor
- Author
-
Catherine Goudie, Marcel Kool, Ryan S. Lee, Jaclyn A. Biegel, Martin Hasselblatt, William D. Foulkes, Charles W. M. Roberts, Nada Jabado, Jacek Majewski, Tenzin Gayden, Pascal Johann, Jian Carrot-Zhang, Javad Nadaf, Somayyeh Fahiminiya, and Leora Witkowski
- Subjects
Epigenomics ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Biology ,ATRT ,Germline ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,SCCOHT ,Exome ,Genetic Predisposition to Disease ,Epigenetics ,Carcinoma, Small Cell ,SMARCB1 ,Rhabdoid Tumor ,Exome sequencing ,Ovarian Neoplasms ,DNA Helicases ,Teratoma ,Nuclear Proteins ,Sequence Analysis, DNA ,DNA Methylation ,medicine.disease ,Cystadenocarcinoma, Serous ,SWI/SNF ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,DNA methylation ,Atypical teratoid rhabdoid tumor ,Hypercalcemia ,SMARCA4 ,Female ,methylation ,exome sequencing ,Genome-Wide Association Study ,Transcription Factors ,Research Paper - Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is the most common undifferentiated ovarian malignancy diagnosed in women under age 40. We and others recently determined that germline and/or somatic deleterious mutations in SMARCA4 characterize SCCOHT. Alterations in this gene, or the related SWI/SNF chromatin remodeling gene SMARCB1, have been previously reported in atypical teratoid/rhabdoid tumors (ATRTs) and malignant rhabdoid tumors (MRTs). To further describe the somatic landscape of SCCOHT, we performed whole exome sequencing on 14 tumors and their matched normal tissues and compared their genomic alterations with those in ATRT and ovarian high grade serous carcinoma (HGSC). We confirmed that SMARCA4 is the only recurrently mutated gene in SCCOHT, and show that recurrent allelic imbalance is observed exclusively on chromosome 19p, where SMARCA4 resides. By comparing genomic alterations between SCCOHT, ATRT and HGSC, we demonstrate that SCCOHTs, like ATRTs, have a remarkably simple genome and harbor significantly fewer somatic protein-coding mutations and chromosomal alterations than HGSC. Furthermore, a comparison of global DNA methylation profiles of 45 SCCOHTs, 65 ATRTs, and 92 HGSCs demonstrates a strong epigenetic correlation between SCCOHT and ATRT. Our results further confirm that the genomic and epigenomic signatures of SCCOHT are more similar to those of ATRT than HGSC, supporting our previous hypothesis that SCCOHT is a rhabdoid tumor and should be renamed MRT of the ovary. Furthermore, we conclude that SMARCA4 inactivation is the main cause of SCCOHT, and that new distinct therapeutic approaches should be developed to specifically target this devastating tumor.
- Published
- 2015
- Full Text
- View/download PDF
39. In Brief: Picturing the complex world of chromatin remodelling families
- Author
-
Leora Witkowski and William D. Foulkes
- Subjects
Genetics ,Evolutionary biology ,medicine ,Cancer ,Epigenetics ,Biology ,medicine.disease ,Chromatin remodelling ,SWI/SNF ,Pathology and Forensic Medicine ,Chromatin - Abstract
Over the past decade, chromatin remodelling emerged as one of the most important causes of both abnormal development and cancer. Although much has been written about one or another of the complexes, no recent concise summary of the chromatin remodelling families as a whole is available. In this short review, we introduce the family members, briefly summarize their role in developmental abnormalities and neoplasia, and outline the different ways in which these families remodel chromatin.
- Published
- 2015
- Full Text
- View/download PDF
40. Ovarian Sex Cord-Stromal Tumors in Patients With Probable or Confirmed Germline DICER1 Mutations
- Author
-
Jason Tan, Sharron Townshend, Adrian Charles, Hein Sonnendecker, William D. Foulkes, Stuart G. Salfinger, Colin J.R. Stewart, Yee Leung, E. Ebo Oost, Catherine S. Choong, and Leora Witkowski
- Subjects
Adult ,Ribonuclease III ,endocrine system ,Pathology ,medicine.medical_specialty ,Adolescent ,Somatic cell ,Granulosa cell ,CD99 ,Sex Cord-Gonadal Stromal Tumors ,Biology ,Neuroendocrine differentiation ,Germline ,Pathology and Forensic Medicine ,DEAD-box RNA Helicases ,Germline mutation ,Biomarkers, Tumor ,medicine ,Humans ,Germ-Line Mutation ,Ovarian Neoplasms ,Obstetrics and Gynecology ,Sertoli cell ,Immunohistochemistry ,medicine.anatomical_structure ,Female - Abstract
The DICER1 gene encodes an endoribonuclease involved in the production of mature microRNAs which regulates gene expression through several mechanisms. Recent studies have demonstrated somatic mutations in DICER1 in approximately 60% of ovarian Sertoli-Leydig cell tumors. Furthermore, patients with germline mutations in DICER1 are predisposed to developing a range of rare neoplasms including ovarian sex cord-stromal tumors most of which have been classified as Sertoli-Leydig cell tumor. However, the histologic features of these tumors have not been reported in detail. We describe the morphologic and immunophenotypic findings of 4 sex cord-stromal tumors arising in patients with proven or likely germline DICER1 mutations including 3 individuals from 1 family. Three tumors showed similar appearances characterized by marked architectural and cytologic heterogeneity including sertoliform, juvenile granulosa cell tumor-like, and unclassifiable elements. The remaining case mainly showed heterologous mucinous epithelial and neuroendocrine differentiation with only a minor intermediate-grade Sertoli cell component. This tumor and one of the 3 former cases arose in related patients with identical germline DICER1 mutations indicating that additional factors influence tumor morphology. All tumors were positive for steroidogenic factor-1 and FOXL2 on immunohistochemical analysis, whereas there was more variable expression of inhibin, calretinin, CD56, CD99, and hormone receptors. The present small series suggests that some ovarian Sertoli-Leydig cell tumor associated with germline DICER1 mutations may show distinctive histologic features in particular admixed Sertoli cell and juvenile granulosa cell tumor-like features. Larger studies are required to establish whether heterologous elements are also a more common feature of these tumors.
- Published
- 2015
- Full Text
- View/download PDF
41. Specifications of the ACMG/AMP variant curation guidelines for the analysis of germline CDH1 sequence variants
- Author
-
Elizabeth C. Chao, Sharon E. Plon, Arjen R. Mensenkamp, Rajarshi Ghosh, Liying Zhang, Tyler Landrith, Shuwei Li, Leora Witkowski, Charles S. Yi, Carla Oliveira, Kristy Lee, Amanda B. Spurdle, Rachid Karam, Mackenzie Trapp, Joana Figueiredo, Kasmintan A. Schrader, Fátima Carneiro, David G. Huntsman, Carolina Pardo, Tina Pesaran, Katherine Dixon, Michael J. Anderson, Pardeep Kaurah, Kate Krempely, Maegan E. Roberts, Matthew Richardson, Chimene Kesserwan, and Thomas P. Slavin
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Genomics ,Computational biology ,Biology ,Genome ,Article ,Germline ,03 medical and health sciences ,Breast cancer ,All institutes and research themes of the Radboud University Medical Center ,Genetics ,medicine ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Humans ,Genetic Testing ,Allele ,Alleles ,Societies, Medical ,Genetics (clinical) ,Genome, Human ,Molecular pathology ,Computational Biology ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Sequence Analysis, DNA ,medicine.disease ,United States ,3. Good health ,030104 developmental biology ,Mutation ,Medical genetics ,Hereditary diffuse gastric cancer - Abstract
The variant curation guidelines published in 2015 by the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP) provided the genetics community with a framework to assess variant pathogenicity; however, these rules are not gene-specific. Germline pathogenic variants in the CDH1 gene cause hereditary diffuse gastric cancer and lobular breast cancer, a clinically challenging cancer predisposition syndrome that often requires a multidisciplinary team of experts to be properly managed. Given this challenge, the Clinical Genome Resource (ClinGen) Hereditary Cancer Domain prioritized the development of the CDH1 Variant Curation Expert Panel (VCEP) to develop and implement rules for CDH1 variant classifications. Here we describe the CDH1 specifications of the ACMG/AMP guidelines, which were developed and validated after a systematic evaluation of variants obtained from a cohort of clinical laboratory data encompassing ~827,000 CDH1 sequenced alleles. Comparing previously reported germline variants that were classified using the 2015 ACMG/AMP guidelines to the CDH1 VCEP recommendations resulted in reduced variants of uncertain significance and facilitated resolution of variants with conflicted assertions in ClinVar. Overall, the ClinGen CDH1 VCEP recommends the use of these CDH1-specific guidelines for the assessment and classification of variants identified in this clinically actionable gene.
- Published
- 2018
42. Paediatric ovarian tumours and their associated cancer susceptibility syndromes
- Author
-
W. Glenn McCluggage, Stephanie Vairy, Leora Witkowski, William D. Foulkes, and Catherine Goudie
- Subjects
0301 basic medicine ,endocrine system ,medicine.medical_specialty ,Pathology ,genetic epidemiology ,Granulosa cell ,cancer: colon ,Ovary ,Biology ,Small-cell carcinoma ,Germline ,cancer: endocrine ,cancer: breast ,Diagnosis, Differential ,genome-wide ,diagnostics tests ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Genetic predisposition ,Cancer Genetics ,Humans ,Genetic Predisposition to Disease ,genetics ,Child ,Position Statement ,Pathological ,Genetics (clinical) ,Ovarian Neoplasms ,Complex Traits ,Syndrome ,genetic screening/counselling ,medicine.disease ,Sertoli cell ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,030220 oncology & carcinogenesis ,molecular genetics ,oncology ,Female ,Germ cell ,clinical genetics ,reproductive medicine - Abstract
Background Germline pathogenic variants in SDHB/SDHC/SDHD are the most frequent causes of inherited phaeochromocytomas/paragangliomas. Insufficient information regarding penetrance and phenotypic variability hinders optimum management of mutation carriers. We estimate penetrance for symptomatic tumours and elucidate genotype–phenotype correlations in a large cohort of SDHB/SDHC/SDHD mutation carriers. Methods A retrospective survey of 1832 individuals referred for genetic testing due to a personal or family history of phaeochromocytoma/paraganglioma. 876 patients (401 previously reported) had a germline mutation in SDHB/SDHC/SDHD (n=673/43/160). Tumour risks were correlated with in silico structural prediction analyses. Results Tumour risks analysis provided novel penetrance estimates and genotype–phenotype correlations. In addition to tumour type susceptibility differences for individual genes, we confirmed that the SDHD:p.Pro81Leu mutation has a distinct phenotype and identified increased age-related tumour risks with highly destabilising SDHB missense mutations. By Kaplan-Meier analysis, the penetrance (cumulative risk of clinically apparent tumours) in SDHB and (paternally inherited) SDHD mutation-positive non-probands (n=371/67 with detailed clinical information) by age 60 years was 21.8% (95% CI 15.2% to 27.9%) and 43.2% (95% CI 25.4% to 56.7%), respectively. Risk of malignant disease at age 60 years in non-proband SDHB mutation carriers was 4.2%(95% CI 1.1% to 7.2%). With retrospective cohort analysis to adjust for ascertainment, cumulative tumour risks for SDHB mutation carriers at ages 60 years and 80 years were 23.9% (95% CI 20.9% to 27.4%) and 30.6% (95% CI 26.8% to 34.7%). Conclusions Overall risks of clinically apparent tumours for SDHB mutation carriers are substantially lower than initially estimated and will improve counselling of affected families. Specific genotype–tumour risk associations provides a basis for novel investigative strategies into succinate dehydrogenase-related mechanisms of tumourigenesis and the development of personalised management for SDHB/SDHC/SDHD mutation carriers.
- Published
- 2017
43. Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type
- Author
-
Avi Saskin, Jocelyne Arseneau, Lawrence M. Roth, Anna Margiolaki, François Plourde, Janez Lamovec, Barbara Rivera, Rachel Silva-Smith, Steffen Albrecht, Reiner Siebert, Vassilis Georgoulias, Helen J. Mackay, Andrew Berchuck, William D. Foulkes, Emmanouil Saloustros, Jacek Majewski, W Glenn McCluggage, Sharon M. Castellino, Madeleine Arseneault, Kitty Pavlakis, Ester Castellsagué, Yasser Riazalhosseini, Somayyeh Fahiminiya, Nancy Hamel, Thomas M. Ulbright, Inga Nagel, Elizabeth A. Reilly, Frederick R. Ueland, Tracey A Bender, Mona Wu, Javad Nadaf, Blaise A. Clarke, David Grynspan, Eva Tomiak, Catherine Gilpin, Leora Witkowski, Colin J.R. Stewart, Michel Longy, Marc Tischkowitz, Rouzan G. Karabakhtsian, Martin Hasselblatt, and Jian Carrot-Zhang
- Subjects
Genetics ,Mutation ,Somatic cell ,Biology ,medicine.disease ,medicine.disease_cause ,Germline ,Germline mutation ,Carcinoma ,medicine ,Allele ,Ovarian cancer ,Exome sequencing - Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is the most common undifferentiated ovarian malignancy in women under 40 years of age. We sequenced the exomes of six individuals from three families with SCCOHT. After discovering segregating deleterious germline mutations in SMARCA4 in all three families, we tested DNA from a fourth affected family, which also carried a segregating SMARCA4 germline mutation. All the familial tumors sequenced harbored either a somatic mutation or loss of the wild-type allele. Immunohistochemical analysis of these cases and additional familial and non-familial cases showed loss of SMARCA4 (BRG1) protein in 38 of 40 tumors overall. Sequencing of cases with available DNA identified at least one germline or somatic deleterious SMARCA4 mutation in 30 of 32 cases. Additionally, the SCCOHT cell line BIN-67 had biallelic deleterious mutations in SMARCA4. Our findings identify alterations in SMARCA4 as the major cause of SCCOHT, which could lead to improvements in genetic counseling and new treatment approaches.
- Published
- 2014
- Full Text
- View/download PDF
44. Primary rhabdoid tumor of the ovary: When large cells become small cells
- Author
-
Ruthy Shaco-Levi, Leora Witkowski, Martin Hasselblatt, William D. Foulkes, Mihai Meirovitz, and Alex Rabinovich
- Subjects
Pathology ,medicine.medical_specialty ,Tumor ,Genetic testing ,Malignant rhabdoid tumor ,Ovary ,Obstetrics and Gynecology ,Case Report ,Biology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Small-cell carcinoma ,lcsh:Gynecology and obstetrics ,lcsh:RC254-282 ,medicine.anatomical_structure ,Oncology ,SMARCA4 ,Rhabdoid ,medicine ,Immunohistochemistry ,Sequencing ,SMARCB1 ,lcsh:RG1-991 - Abstract
Highlights • The third case of pure primary malignant rhabdoid tumor of the ovary (MRTO) is described • SMARCA4 and SMARCB1 genetic analysis and immunohistochemistry are necessary for correct diagnosis of MRTO • MRTO and small cell carcinoma of the ovary, hypercalcemic type are essentially the same and should be treated as such
- Published
- 2015
- Full Text
- View/download PDF
45. DICER1 hotspot mutations in non-epithelial gonadal tumours
- Author
-
Nicholas Coleman, Jocelyne Arseneau, Dominik T. Schneider, Gabriele Calaminus, Jorge S. Reis-Filho, David G. Huntsman, Colin J.R. Stewart, Matthew J. Murray, James Nicholson, W G McCluggage, Leora Witkowski, Stefan Schönberger, Catherine S. Choong, William D. Foulkes, and J. Mattina
- Subjects
Male ,Ribonuclease III ,Cancer Research ,Pathology ,Somatic cell ,DNA Mutational Analysis ,Sex Cord-Gonadal Stromal Tumors ,DICER1 ,DEAD-box RNA Helicases ,0302 clinical medicine ,Gene Frequency ,Child ,Ovarian Neoplasms ,Sanger sequencing ,0303 health sciences ,microRNA ,Middle Aged ,Neoplasms, Germ Cell and Embryonal ,medicine.anatomical_structure ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,symbols ,Female ,Germ cell ,Adult ,endocrine system ,medicine.medical_specialty ,Adolescent ,RNase P ,germ cell tumours ,Biology ,Young Adult ,03 medical and health sciences ,symbols.namesake ,Testicular Neoplasms ,medicine ,ovarian ,Humans ,Gene ,Allele frequency ,Aged ,030304 developmental biology ,DICER1 Syndrome ,Infant, Newborn ,Infant ,Genetics and Genomics ,testicular ,Mutation ,Cancer research ,sex cord-stromal tumours - Abstract
Background: Non-epithelial gonadal tumours largely comprise sex cord-stromal tumours (SCSTs) and germ cell tumours (GCTs). Specific somatic mutations in DICER1, a microRNA maturation pathway gene, have been identified in these tumours. We conducted a study that aimed to confirm, refine and extend the previous observations. Methods: We used Sanger sequencing to sequence the RNase IIIa and IIIb domains of DICER1 in 154 gonadal tumours from 135 females and 19 males, as well as 43 extra-gonadal GCTs from 26 females and 17 males. Results: We identified heterozygous non-synonymous mutations in the RNase IIIb domain of DICER1 in 14/197 non-epithelial tumours (7.1%). Mutations were found in 9/28 SCSTs (32%), 5/118 gonadal GCTs (4.2%), 0/43 extra-gonadal GCTs and 0/8 miscellaneous tumours. The 14 mutations affected only five residues: E1705, D1709, E1788, D1810 and E1813. In all five patients where matched and constitutional DNA was available, the mutations were only somatic. There were no mutations found in the RNase IIIa domain. Conclusion: More than half (8/15) of Sertoli–Leydig cell tumours (SLCTs) harbour DICER1 mutations in the RNase IIIb domain, while mutations are rarely found in GCTs. Genetic alterations in SLCTs may aid in classification and provide new approaches to therapy.
- Published
- 2013
- Full Text
- View/download PDF
46. Familial rhabdoid tumour 'avant la lettre '-from pathology review to exome sequencing and back again
- Author
-
Sandra Thompson May, Steffen Albrecht, Emilie Lalonde, Nancy Hamel, Martin Hasselblatt, Jian Zhang, Matthew J. Murray, William D. Foulkes, James Nicholson, Nicholas Coleman, Stefan Schönberger, David G. Huntsman, Marc Tischkowitz, Jacek Majewski, Leora Witkowski, Peter F. Tauber, Luca Cavallone, and David W. Yandell
- Subjects
Sanger sequencing ,Daughter ,Pathology ,medicine.medical_specialty ,media_common.quotation_subject ,Nonsense mutation ,Context (language use) ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,Malignant rhabdoid tumour ,symbols.namesake ,SMARCA4 ,medicine ,symbols ,Immature teratoma ,Exome sequencing ,media_common - Abstract
Here we provide compelling evidence that next-generation sequencing will revolutionize diagnostics. We reappraised a case from 1991, published in 1993, describing the unique occurrence of an ovarian immature teratoma arising in a young woman and a clonally distinct intracerebral immature teratoma developing in her daughter. We conducted whole-exome sequencing on constitutional DNA from the mother and her daughter and identified a previously unreported nonsense mutation (c.3533G>A; p.Trp1178*) in the chromatin remodelling gene, SMARCA4, that was present in both individuals and was subject to nonsense-mediated decay. Tumour analysis by Sanger sequencing revealed a somatic SMARCA4 mutation in both the mother (c.2438+1G>T) and her daughter (c.3229C>T; p.Arg1077*), which are predicted to be truncating. As immature teratomas are classified as germ cell tumours, we performed a comprehensive mutation survey of 106 apparently sporadic germ cell tumours, but did not find any other clearly deleterious SMARCA4 mutations. Recently, inactivating mutations in SMARCA4 have been found in two cases of rhabdoid tumour predisposition syndrome type 2. In the light of these findings, renewed efforts to locate previously unobtainable tumour samples were successfully undertaken. Histopathological and immunohistochemical re-analysis of the daughter's tumour revealed that it was indeed a rhabdoid tumour (atypical teratoid/rhabdoid tumour). In this context, the original pathology report of the mother's ovarian tumour was re-interpreted as describing a malignant rhabdoid tumour of the ovary. This report raises the question as to whether molecular genetic analysis should be included in tumour classification, alongside more traditional microscopy-based methods. The use of new sequencing technologies, particularly when applied to archived samples, will lead to many more 'molecular rediagnoses'. This is the earliest known case of rhabdoid tumour predisposition syndrome type 2 and the first described case with an autosomal dominant pattern of inheritance, only discovered through an exome sequencing project. Copyright © 2013 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
- Published
- 2013
- Full Text
- View/download PDF
47. Clinical, morphological and immunohistochemical evidence that small-cell carcinoma of the ovary of hypercalcaemic type (SCCOHT) may be a primitive germ-cell neoplasm
- Author
-
William D. Foulkes, W. Glenn McCluggage, Blaise A. Clarke, and Leora Witkowski
- Subjects
0301 basic medicine ,Adult ,Pathology ,medicine.medical_specialty ,Histology ,Biology ,Histogenesis ,Small-cell carcinoma ,Glypican 3 ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,SALL4 ,medicine ,Biomarkers, Tumor ,Humans ,Carcinoma, Small Cell ,Ovarian Neoplasms ,General Medicine ,Neoplasms, Germ Cell and Embryonal ,medicine.disease ,Immunohistochemistry ,030104 developmental biology ,medicine.anatomical_structure ,Dermoid cyst ,030220 oncology & carcinogenesis ,Hypercalcemia ,Immature teratoma ,Female ,Teratoma ,Germ cell - Abstract
Aims The histogenesis and cell lineage of small cell carcinoma of the ovary of hypercalcaemic type (SCCOHT) is unknown. We aim to provide evidence that this may be a primitive germ cell neoplasm arising from a teratoma. Methods/ Results Following the identification of two cases of SCCOHT associated with germ cell tumours (one dermoid cyst, one immature teratoma with a focus of yolk sac tumour), we undertook a literature review to look for any prior reports of SCCOHT in association with other neoplasms or elements. This revealed two cases associated with immature teratomas, one arising in an ovary where a cystectomy had previously been undertaken for a teratoma and another arising in association with a mucinous borderline tumour. Mucinous elements have also been reported in SCCOHT, this type of epithelium potentially being of teratomatous derivation. We stained whole tissue sections of 9 cases of SCCOHT and a tissue microarray (TMA) containing 34 different SCCOHT with germ cell markers SALL4, OCT3/4, alpha fetoprotein (AFP) and glypican 3. All except one of the whole tissue sections and approximately half of the TMA cases were positive with SALL4 while all cases were OCT3/4, AFP and glypican 3 negative, except for focal glypican 3 staining in an occasional case. Conclusions Our findings provide additional evidence to that proposed by others that SCCOHT is a primitive germ cell neoplasm arising from a teratoma. This article is protected by copyright. All rights reserved.
- Published
- 2016
48. The McGill Interactive Pediatric OncoGenetic Guidelines: An approach to identifying pediatric oncology patients most likely to benefit from a genetic evaluation
- Author
-
Stephanie Mourad, William D. Foulkes, Leora Witkowski, Hallie Coltin, Catherine Goudie, and David Malkin
- Subjects
0301 basic medicine ,medicine.medical_specialty ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Pediatric oncology ,Medicine ,Humans ,Medical physics ,Genetic Predisposition to Disease ,Genetic Testing ,business.industry ,Cancer predisposition ,Hematology ,Guideline ,Mobile Applications ,3. Good health ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer genetics ,Pediatrics, Perinatology and Child Health ,Physical therapy ,Familial Cancer ,business ,Algorithms - Abstract
Identifying cancer predisposition syndromes in children with tumors is crucial, yet few clinical guidelines exist to identify children at high risk of having germline mutations. The McGill Interactive Pediatric OncoGenetic Guidelines project aims to create a validated pediatric guideline in the form of a smartphone/tablet application using algorithms to process clinical data and help determine whether to refer a child for genetic assessment. This paper discusses the initial stages of the project, focusing on its overall structure, the methodology underpinning the algorithms, and the upcoming algorithm validation process.
- Published
- 2016
49. Recently characterized molecular events in uncommon gynaecological neoplasms and their clinical importance
- Author
-
William D. Foulkes, Leora Witkowski, and W. Glenn McCluggage
- Subjects
0301 basic medicine ,Identification methods ,Pathology ,medicine.medical_specialty ,Histology ,Somatic cell ,Genital Neoplasms, Female ,Biology ,Bioinformatics ,Germline ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Carcinoma ,Humans ,Pathological ,Gene ,High-Throughput Nucleotide Sequencing ,General Medicine ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,SMARCA4 ,Female ,Embryonal rhabdomyosarcoma - Abstract
The introduction of new sequencing technologies has resulted in the discovery of commonly mutated genes in uncommon cancers, including non-epithelial ovarian neoplasms and other rare gynaecological tumours, such as cervical embryonal rhabdomyosarcoma. In some of these neoplasms, mutations in certain genes are both frequent and specific enough for the genomic mutations and sometimes their associated protein loss or overexpression to be used as an aid to diagnosis. In this review, we contrast previous gene identification methods with newer ones, and discuss how the new sequencing technologies (collectively referred to as 'next-generation sequencing') have permitted the identification of specific molecular events that characterize several rare gynaecological neoplasms. We highlight the value of using sequencing to complement traditional pathological methods when diagnosing certain tumours, and provide practical advice to pathologists dealing with these neoplasms. We focus on adult granulosa cell tumours (somatic monoallelic mutations in FOXL2), Sertoli-Leydig cell tumours, gynaecological embryonal rhabdomyosarcomas (germline and somatic mutations in DICER1), and small-cell carcinoma of the ovary, hypercalcaemic type (biallelic mutations in SMARCA4). The new genetic findings provided by next-generation sequencing in these uncommon neoplasms have brought these disorders back into focus, and point the way towards new diagnostic, preventive and therapeutic avenues.
- Published
- 2016
50. The influence of clinical and genetic factors on patient outcome in small cell carcinoma of the ovary, hypercalcemic type
- Author
-
W. Glenn McCluggage, Heather E. Cunliffe, Michel Longy, Andrew Berchuck, Anthony N. Karnezis, Catherine Goudie, Jeffrey M. Trent, Talia Boshari, Emmanouil Saloustros, Jean Sébastien Brunet, Douglas A. Levine, Leora Witkowski, David G. Huntsman, William D. Foulkes, William P.D. Hendricks, Patricia Pautier, Colin J.R. Stewart, James A. Knost, Martin Hasselblatt, and Pilar Ramos
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_treatment ,Stem cell rescue ,Kaplan-Meier Estimate ,Bioinformatics ,Germline ,Cohort Studies ,0302 clinical medicine ,SMARCA4 ,SCCOHT ,Young adult ,Carcinoma, Small Cell ,Child ,Cancer ,Ovarian Neoplasms ,Age Factors ,Obstetrics and Gynecology ,Nuclear Proteins ,Prognosis ,030220 oncology & carcinogenesis ,Female ,Adult ,medicine.medical_specialty ,Adolescent ,Genetic counseling ,Oncology and Carcinogenesis ,Small-cell carcinoma ,Article ,Paediatrics and Reproductive Medicine ,03 medical and health sciences ,Young Adult ,Germline mutation ,Ovarian cancer ,Clinical Research ,Internal medicine ,medicine ,Chemotherapy ,Humans ,Oncology & Carcinogenesis ,Germ-Line Mutation ,Neoplasm Staging ,business.industry ,Carcinoma ,DNA Helicases ,Small Cell ,medicine.disease ,Stem Cell Research ,Radiation therapy ,030104 developmental biology ,Mutation ,Hypercalcemia ,business ,Transcription Factors - Abstract
OBJECTIVE: Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is an aggressive tumor, with long term survival at ~30% in early stage disease. SCCOHT is caused by germline and somatic SMARCA4 mutations, but the effect of the mutation type on patients remains unknown. Furthermore, the rarity of SCCOHT has resulted in varied treatment, with no standardized protocols. We analyzed 293 cases to determine the effect of treatment modalities and SMARCA4 mutations on patient diagnosis and outcome. METHODS: In 293 SCCOHT patients we collected information on age and stage at diagnosis, treatment modality (surgery, chemotherapy, radiotherapy, and/or high-dose chemotherapy with autologous stem cell rescue (HDC-aSCR)), SMARCA4 mutation origin (germline/somatic), and overall survival. Cox analysis and log-rank tests were performed on 257 cases with available survival data. RESULTS: The strongest prognostic factors were stage at diagnosis (p = 2.72e-15) and treatment modality (p = 3.87e-13). For FIGO stages II-IV, 5-year survival was 71% for patients who received HDC-aSCR, compared to 25% in patients who received conventional chemotherapy alone following surgery (p = 0.002). Patients aged ≥40 had a worse outcome than younger patients (p = 0.04). Twenty-six of 60 tested patients carried a germline SMARCA4 mutation, including all patients diagnosed
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.