63 results on '"Ozge Ceyhan-Birsoy"'
Search Results
2. Expanded genetic testing of GIST patients identifies high proportion of non-syndromic patients with germline alterations
- Author
-
Diana Mandelker, Antonio Marra, Nikita Mehta, Pier Selenica, Zarina Yelskaya, Ciyu Yang, Joshua Somar, Miika Mehine, Maksym Misyura, Olca Basturk, Alicia Latham, Maria Carlo, Michael Walsh, Zsofia K. Stadler, Kenneth Offit, Chaitanya Bandlamudi, Meera Hameed, Ping Chi, Jorge S. Reis-Filho, and Ozge Ceyhan-Birsoy
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Traditional genetic testing for patients with gastrointestinal stromal tumors (GISTs) focus on those with syndromic features. To assess whether expanded genetic testing of GIST patients could identify hereditary cancer predisposition, we analyzed matched tumor-germline sequencing results from 103 patients with GISTs over a 6-year period. Germline pathogenic/likely pathogenic (P/LP) variants in GIST-associated genes (SDHA, SDHB, SDHC, NF1, KIT) were identified in 69% of patients with KIT/PDGFRA-wildtype GISTs, 63% of whom did not have any personal or family history of syndromic features. To evaluate the frequency of somatic versus germline variants identified in tumor-only sequencing of GISTs, we analyzed 499 de-identified tumor-normal pairs. P/LP variants in certain genes (e.g., BRCA1/2, SDHB) identified in tumor-only sequencing of GISTs were almost exclusively germline in origin. Our results provide guidance for genetic testing of GIST patients and indicate that germline testing should be offered to all patients with KIT/PDGFRA-wildtype GISTs regardless of their history of syndromic features.
- Published
- 2023
- Full Text
- View/download PDF
3. Diagnostic yield and clinical relevance of expanded genetic testing for cancer patients
- Author
-
Ozge Ceyhan-Birsoy, Gowtham Jayakumaran, Yelena Kemel, Maksym Misyura, Umut Aypar, Sowmya Jairam, Ciyu Yang, Yirong Li, Nikita Mehta, Anna Maio, Angela Arnold, Erin Salo-Mullen, Margaret Sheehan, Aijazuddin Syed, Michael Walsh, Maria Carlo, Mark Robson, Kenneth Offit, Marc Ladanyi, Jorge S. Reis-Filho, Zsofia K. Stadler, Liying Zhang, Alicia Latham, Ahmet Zehir, and Diana Mandelker
- Subjects
Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background Genetic testing (GT) for hereditary cancer predisposition is traditionally performed on selected genes based on established guidelines for each cancer type. Recently, expanded GT (eGT) using large hereditary cancer gene panels uncovered hereditary predisposition in a greater proportion of patients than previously anticipated. We sought to define the diagnostic yield of eGT and its clinical relevance in a broad cancer patient population over a 5-year period. Methods A total of 17,523 cancer patients with a broad range of solid tumors, who received eGT at Memorial Sloan Kettering Cancer Center between July 2015 to April 2020, were included in the study. The patients were unselected for current GT criteria such as cancer type, age of onset, and/or family history of disease. The diagnostic yield of eGT was determined for each cancer type. For 9187 patients with five common cancer types frequently interrogated for hereditary predisposition (breast, colorectal, ovarian, pancreatic, and prostate cancer), the rate of pathogenic/likely pathogenic (P/LP) variants in genes that have been associated with each cancer type was analyzed. The clinical implications of additional findings in genes not known to be associated with a patients’ cancer type were investigated. Results 16.7% of patients in a broad cancer cohort had P/LP variants in hereditary cancer predisposition genes identified by eGT. The diagnostic yield of eGT in patients with breast, colorectal, ovarian, pancreatic, and prostate cancer was 17.5%, 15.3%, 24.2%, 19.4%, and 15.9%, respectively. Additionally, 8% of the patients with five common cancers had P/LP variants in genes not known to be associated with the patient’s current cancer type, with 0.8% of them having such a variant that confers a high risk for another cancer type. Analysis of clinical and family histories revealed that 74% of patients with variants in genes not associated with their current cancer type but which conferred a high risk for another cancer did not meet the current GT criteria for the genes harboring these variants. One or more variants of uncertain significance were identified in 57% of the patients. Conclusions Compared to targeted testing approaches, eGT can increase the yield of detection of hereditary cancer predisposition in patients with a range of tumors, allowing opportunities for enhanced surveillance and intervention. The benefits of performing eGT should be weighed against the added number of VUSs identified with this approach.
- Published
- 2022
- Full Text
- View/download PDF
4. P452: Specifying the ACMG/AMP variant sequence interpretation guidelines for congenital myopathies
- Author
-
Marina DiStefano, Ryan Webb, Hannah McCurry, Shannon McNulty Gray, Swati Tomar, Prasad Kopparapu, Eleanor Broeren, Kezang Tshering, Alan Beggs, Enrico Silvio Bertini, Adele D'Amico, Sandra Donkervoort, James Dowling, Fabiana Fattori, Ana Ferreiro, Casie Genetti, Hernan Gonorazky, Svetlana Gorokhova, Amanda Lindy, Livija Medne, Sander Pajusalu, Katarina Pelin, John Rendu, Matteo Vatta, Tom Winder, Hui Yang, Grace Yoon, Ozge Ceyhan-Birsoy, and Carsten Bönnemann
- Subjects
Genetics ,QH426-470 ,Medicine - Published
- 2023
- Full Text
- View/download PDF
5. Germline RAD51B variants confer susceptibility to breast and ovarian cancers deficient in homologous recombination
- Author
-
Jeremy Setton, Pier Selenica, Semanti Mukherjee, Rachna Shah, Isabella Pecorari, Biko McMillan, Isaac X. Pei, Yelena Kemel, Ozge Ceyhan-Birsoy, Margaret Sheehan, Kaitlyn Tkachuk, David N. Brown, Liying Zhang, Karen Cadoo, Simon Powell, Britta Weigelt, Mark Robson, Nadeem Riaz, Kenneth Offit, Jorge S. Reis-Filho, and Diana Mandelker
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Pathogenic germline mutations in the RAD51 paralog genes RAD51C and RAD51D, are known to confer susceptibility to ovarian and triple-negative breast cancer. Here, we investigated whether germline loss-of-function variants affecting another RAD51 paralog gene, RAD51B, are also associated with breast and ovarian cancer. Among 3422 consecutively accrued breast and ovarian cancer patients consented to tumor/germline sequencing, the observed carrier frequency of loss-of-function germline RAD51B variants was significantly higher than control cases from the gnomAD population database (0.26% vs 0.09%), with an odds ratio of 2.69 (95% CI: 1.4–5.3). Furthermore, we demonstrate that tumors harboring biallelic RAD51B alteration are deficient in homologous recombination DNA repair deficiency (HRD), as evidenced by analysis of sequencing data and in vitro functional assays. Our findings suggest that RAD51B should be considered as an addition to clinical germline testing panels for breast and ovarian cancer susceptibility.
- Published
- 2021
- Full Text
- View/download PDF
6. Fumarate hydratase c.914T > C (p.Phe305Ser) is a pathogenic variant associated with hereditary leiomyomatosis and renal cell cancer syndrome
- Author
-
Kelsey E. Breen, Maria I. Carlo, Yelena Kemel, Anna Maio, Ying‐Bei Chen, Liying Zhang, Ozge Ceyhan‐Birsoy, and Diana Mandelker
- Subjects
carcinoma ,fumarate hydratase ,leiomyomatosis ,mutation ,renal cell ,Genetics ,QH426-470 - Abstract
Abstract Background Hereditary leiomyomatosis and renal cell cancer syndrome (HLRCC), caused by heterozygous germline pathogenic variants in the FH, confers an increased risk for cutaneous and uterine leiomyomas and renal cancer. Methods About 13,722 advanced cancer patients, including 560 with renal cell carcinoma, had germline analysis performed in the context of tumor‐normal sequencing under an IRB approved protocol. Results We report two unrelated individuals with early onset kidney cancer who both carried the c.914C > T (p.Phe305Ser) germline variant in the FH. Both tumors exhibited loss of FH staining by immunohistochemistry and/or positive 2SC staining. Subsequent familial testing discovered that a daughter of a proband who carried the variant had both cutaneous and uterine leiomyomas. Conclusion This combination of evidence suggests that the FH c.914C > T (p.Phe305Ser) is pathogenic for HLRCC.
- Published
- 2020
- Full Text
- View/download PDF
7. The BabySeq project: implementing genomic sequencing in newborns
- Author
-
Ingrid A. Holm, Pankaj B. Agrawal, Ozge Ceyhan-Birsoy, Kurt D. Christensen, Shawn Fayer, Leslie A. Frankel, Casie A. Genetti, Joel B. Krier, Rebecca C. LaMay, Harvey L. Levy, Amy L. McGuire, Richard B. Parad, Peter J. Park, Stacey Pereira, Heidi L. Rehm, Talia S. Schwartz, Susan E. Waisbren, Timothy W. Yu, The BabySeq Project Team, Robert C. Green, and Alan H. Beggs
- Subjects
Newborn screening ,Newborn sequencing ,Whole exome sequencing ,Methods ,Randomized trial ,Ethical, legal, social implications ,Pediatrics ,RJ1-570 - Abstract
Abstract Background The greatest opportunity for lifelong impact of genomic sequencing is during the newborn period. The “BabySeq Project” is a randomized trial that explores the medical, behavioral, and economic impacts of integrating genomic sequencing into the care of healthy and sick newborns. Methods Families of newborns are enrolled from Boston Children’s Hospital and Brigham and Women’s Hospital nurseries, and half are randomized to receive genomic sequencing and a report that includes monogenic disease variants, recessive carrier variants for childhood onset or actionable disorders, and pharmacogenomic variants. All families participate in a disclosure session, which includes the return of results for those in the sequencing arm. Outcomes are collected through review of medical records and surveys of parents and health care providers and include the rationale for choice of genes and variants to report; what genomic data adds to the medical management of sick and healthy babies; and the medical, behavioral, and economic impacts of integrating genomic sequencing into the care of healthy and sick newborns. Discussion The BabySeq Project will provide empirical data about the risks, benefits and costs of newborn genomic sequencing and will inform policy decisions related to universal genomic screening of newborns. Trial registration The study is registered in ClinicalTrials.gov Identifier: NCT02422511. Registration date: 10 April 2015.
- Published
- 2018
- Full Text
- View/download PDF
8. Comprehensive analysis of germline drivers in endometrial cancer
- Author
-
Sushmita Gordhandas, Eric Rios-Doria, Karen A Cadoo, Amanda Catchings, Anna Maio, Yelena Kemel, Margaret Sheehan, Megha Ranganathan, Dina Green, Anjali Aryamvally, Angela G Arnold, Erin Salo-Mullen, Beryl Manning-Geist, Tiffany Sia, Pier Selenica, Arnaud Da Cruz Paula, Chad Vanderbilt, Maksym Misyura, Mario M Leitao, Jennifer J Mueller, Vicky Makker, Maria Rubinstein, Claire F Friedman, Qin Zhou, Alexia Iasonos, Alicia Latham, Maria I Carlo, Yonina R Murciano-Goroff, Marie Will, Michael F Walsh, Shirin Issa Bhaloo, Lora H Ellenson, Ozge Ceyhan-Birsoy, Michael F Berger, Mark E Robson, Nadeem Abu-Rustum, Carol Aghajanian, Kenneth Offit, Zsofia Stadler, Britta Weigelt, Diana L Mandelker, and Ying L Liu
- Subjects
Cancer Research ,Oncology - Abstract
Background We sought to determine the prevalence of germline pathogenic variants (gPVs) in unselected patients with endometrial cancer (EC), define biallelic gPVs within tumors, and describe their associations with clinicopathologic features. Methods Germline assessment of at least 76 cancer predisposition genes was performed in patients with EC undergoing clinical tumor-normal Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) sequencing from January 1, 2015, to June 30, 2021. In patients with gPVs, biallelic alterations in ECs were identified through analysis of loss of heterozygosity and somatic PVs. Clinicopathologic variables were compared using nonparametric tests. Results Of 1625 patients with EC, 216 (13%) had gPVs, and 15 patients had 2 gPVs. There were 231 gPVs in 35 genes (75 [32%] high penetrance; 39 [17%] moderate penetrance; and 117 [51%] low, recessive, or uncertain penetrance). Compared with those without gPVs, patients with gPVs were younger (P = .002), more often White (P = .009), and less obese (P = .025) and had differences in distribution of tumor histology (P = .017) and molecular subtype (P Conclusions Of unselected patients with EC, 13% had gPVs, with 63% of gPVs in high-penetrance genes (MMR and homologous recombination) exhibiting biallelic inactivation, potentially driving cancer development. This supports germline assessment in EC given implications for treatment and cancer prevention.
- Published
- 2023
9. Germline Testing for the Evaluation of Hereditary Cancer Predisposition
- Author
-
Ozge, Ceyhan-Birsoy
- Subjects
Germ Cells ,Neoplasms ,Biochemistry (medical) ,Clinical Biochemistry ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Germ-Line Mutation - Abstract
Germline testing for hereditary cancer predisposition has become increasingly important in the management of patients with cancer. Recent studies have demonstrated that hereditary cancer predisposition is more common than previously recognized and germline pathogenic variants may be actionable for patient treatment strategies. This article reviews the significance of hereditary cancer predisposition assessment and highlights the current practices in germline genetic testing approaches.
- Published
- 2022
10. Data from Cancer-Causative Mutations Occurring in Early Embryogenesis
- Author
-
Diana Mandelker, Jorge S. Reis-Filho, Mark E. Robson, Zsofia K. Stadler, Kenneth Offit, Marc Ladanyi, Anna-Katerina Hadjantonakis, Michael F. Walsh, Britta Weigelt, David H. Abramson, Nadeem Riaz, Xin Pei, Laetitia Borsu, Elizabeth Comen, Mahsa Vahdatinia, Christopher J. Schwartz, Jacklyn Casanova-Murphy, Yelena Kemel, Utsav Patel, Margaret Sheehan, Sowmya Jairam, Ozge Ceyhan-Birsoy, Michael F. Berger, Ryma Benayed, Ahmet Zehir, Antonio Marra, Ronglai Shen, Kelsey Breen, Arnaud Da Cruz Paula, Andrea M. Gazzo, Edaise M. da Silva, Pier Selenica, Fatemeh Derakhshan, David N. Brown, Ryan N. Ptashkin, and Fresia Pareja
- Abstract
Mosaic mutations in normal tissues can occur early in embryogenesis and be associated with hereditary cancer syndromes when affecting cancer susceptibility genes (CSG). Their contribution to apparently sporadic cancers is currently unknown. Analysis of paired tumor/blood sequencing data of 35,310 patients with cancer revealed 36 pathogenic mosaic variants affecting CSGs, most of which were not detected by prior clinical genetic testing. These CSG mosaic variants were consistently detected at varying variant allelic fractions in microdissected normal tissues (n = 48) from distinct embryonic lineages in all individuals tested, indicating their early embryonic origin, likely prior to gastrulation, and likely asymmetrical propagation. Tumor-specific biallelic inactivation of the CSG affected by a mosaic variant was observed in 91.7% (33/36) of cases, and tumors displayed the hallmark pathologic and/or genomic features of inactivation of the respective CSGs, establishing a causal link between CSG mosaic variants arising in early embryogenesis and the development of apparently sporadic cancers.Significance:Here, we demonstrate that mosaic variants in CSGs arising in early embryogenesis contribute to the oncogenesis of seemingly sporadic cancers. These variants can be systematically detected through the analysis of tumor/normal sequencing data, and their detection may affect therapeutic decisions as well as prophylactic measures for patients and their offspring.See related commentary by Liggett and Sankaran, p. 889.This article is highlighted in the In This Issue feature, p. 873
- Published
- 2023
11. Supplementary Data from Cancer-Causative Mutations Occurring in Early Embryogenesis
- Author
-
Diana Mandelker, Jorge S. Reis-Filho, Mark E. Robson, Zsofia K. Stadler, Kenneth Offit, Marc Ladanyi, Anna-Katerina Hadjantonakis, Michael F. Walsh, Britta Weigelt, David H. Abramson, Nadeem Riaz, Xin Pei, Laetitia Borsu, Elizabeth Comen, Mahsa Vahdatinia, Christopher J. Schwartz, Jacklyn Casanova-Murphy, Yelena Kemel, Utsav Patel, Margaret Sheehan, Sowmya Jairam, Ozge Ceyhan-Birsoy, Michael F. Berger, Ryma Benayed, Ahmet Zehir, Antonio Marra, Ronglai Shen, Kelsey Breen, Arnaud Da Cruz Paula, Andrea M. Gazzo, Edaise M. da Silva, Pier Selenica, Fatemeh Derakhshan, David N. Brown, Ryan N. Ptashkin, and Fresia Pareja
- Abstract
Supplementary Data from Cancer-Causative Mutations Occurring in Early Embryogenesis
- Published
- 2023
12. Supplementary Data from Microsatellite Instability–High Endometrial Cancers with MLH1 Promoter Hypermethylation Have Distinct Molecular and Clinical Profiles
- Author
-
Britta Weigelt, Diana L. Mandelker, Yulia Lakhman, Lora H. Ellenson, Dmitriy Zamarin, Alexia Iasonos, Jorge S. Reis-Filho, Zsofia K. Stadler, Nadeem R. Abu-Rustum, Carol Aghajanian, Claire F. Friedman, Maria M. Rubinstein, Sushmita Gordhandas, Timothy Hoang, Lea A. Moukarzel, Ozge Ceyhan-Birsoy, Pier Selenica, Weining Ma, Qin C. Zhou, Arnaud Da Cruz Paula, Kelly A. Devereaux, Ying L. Liu, and Beryl L. Manning-Geist
- Abstract
Supplementary Data from Microsatellite Instability–High Endometrial Cancers with MLH1 Promoter Hypermethylation Have Distinct Molecular and Clinical Profiles
- Published
- 2023
13. Data from Microsatellite Instability–High Endometrial Cancers with MLH1 Promoter Hypermethylation Have Distinct Molecular and Clinical Profiles
- Author
-
Britta Weigelt, Diana L. Mandelker, Yulia Lakhman, Lora H. Ellenson, Dmitriy Zamarin, Alexia Iasonos, Jorge S. Reis-Filho, Zsofia K. Stadler, Nadeem R. Abu-Rustum, Carol Aghajanian, Claire F. Friedman, Maria M. Rubinstein, Sushmita Gordhandas, Timothy Hoang, Lea A. Moukarzel, Ozge Ceyhan-Birsoy, Pier Selenica, Weining Ma, Qin C. Zhou, Arnaud Da Cruz Paula, Kelly A. Devereaux, Ying L. Liu, and Beryl L. Manning-Geist
- Abstract
Purpose:Microsatellite instability–high (MSI-H) endometrial carcinomas are underpinned by distinct mechanisms of DNA mismatch repair deficiency (MMR-D). We sought to characterize the clinical and genetic features of MSI-H endometrial cancers harboring germline or somatic mutations in MMR genes or MLH1 promoter hypermethylation (MLH1ph).Experimental Design:Of > 1,100 patients with endometrial cancer that underwent clinical tumor-normal sequencing, 184 had MSI-H endometrial cancers due to somatic MMR mutations or MLH1ph, or harbored pathogenic germline MMR mutations. Clinicopathologic features, mutational landscape, and tumor-infiltrating lymphocyte (TIL) scores were compared among MMR-D groups using nonparametric tests. Log-rank tests were used for categorical associations; Kaplan–Meier method and Wald test based on Cox proportional hazards models were employed for continuous variables and survival analyses.Results:Compared with patients with germline (n = 25) and somatic (n = 39) mutations, patients with MLH1ph endometrial cancers (n = 120) were older (P < 0.001), more obese (P = 0.001) and had more advanced disease at diagnosis (P = 0.025). MLH1ph endometrial cancers were enriched for JAK1 somatic mutations as opposed to germline MMR-D endometrial cancers which showed enrichment for pathogenic ERBB2 mutations. MLH1ph endometrial cancers exhibited lower tumor mutational burden and TIL scores compared with endometrial cancers harboring germline or somatic MMR mutations (P < 0.01). MLH1ph endometrial cancer patients had shorter progression-free survival (PFS) on univariate analysis, but in multivariable models, stage at diagnosis remained the only predictor of survival. For stage I/II endometrial cancer, two-year PFS was inferior for patients with MLH1ph endometrial cancers compared with germline and somatic MMR groups (70% vs. 100%, respectively).Conclusions:MLH1ph endometrial cancers likely constitute a distinct clinicopathologic entity compared with germline and somatic MMR-D ECs with potential treatment implications.
- Published
- 2023
14. ATM Germline-Mutated Gastroesophageal Junction Adenocarcinomas: Clinical Descriptors, Molecular Characteristics, and Potential Therapeutic Implications
- Author
-
Tony El Jabbour, Maksym Misyura, Darren Cowzer, Michal Zimmermann, Victoria Rimkunas, Antonio Marra, Fatemeh Derakhshan, Pier Selenica, Megan Parilla, Jeremy S Setton, Ozge Ceyhan-Birsoy, Yelena Kemel, Amanda Catchings, Megha Ranganathan, Geoffrey Y Ku, Yelena Y Janjigian, Michael Zinda, Maria Koehler, Zsofia Stadler, Jinru Shia, Jorge S Reis-Filho, and Diana Mandelker
- Subjects
Cancer Research ,Germ Cells ,Esophageal Neoplasms ,Oncology ,Stomach Neoplasms ,Humans ,Ataxia Telangiectasia Mutated Proteins ,Esophagogastric Junction ,Adenocarcinoma - Abstract
Background Gastroesophageal junction (GEJ) adenocarcinoma is a rare cancer associated with poor prognosis. The genetic factors conferring predisposition to GEJ adenocarcinoma have yet to be identified. Methods We analyzed germline testing results from 23 381 cancer patients undergoing tumor-normal sequencing, of which 312 individuals had GEJ adenocarcinoma. Genomic profiles and clinico-pathologic features were analyzed for the GEJ adenocarcinomas. Silencing of ATM and ATR was performed using validated short-interfering RNA species in GEJ, esophageal, and gastric adenocarcinoma cell lines. All statistical tests were 2-sided. Results Pathogenic or likely pathogenic ATM variants were identified in 18 of 312 patients (5.8%), and bi-allelic inactivation of ATM through loss of heterozygosity of the wild-type allele was detected in all (16 of 16) samples with sufficient tumor content. Germline ATM-mutated GEJ adenocarcinomas largely lacked somatic mutations in TP53, were more likely to harbor MDM2 amplification, and harbored statistically significantly fewer somatic single nucleotide variants (2.0 mutations/Mb vs 7.9 mutations/Mb; P < .001). A statistically significantly higher proportion of germline ATM-mutated than ATM–wild-type GEJ adenocarcinoma patients underwent a curative resection (10 [100%] vs 92 [86.8%], P = .04; Fisher’s exact test.), A synthetic lethal interaction between short-interfering RNA silencing of ATM and ATR was observed in the models analyzed. Conclusions Our results indicate that germline pathogenic variants in ATM drive oncogenesis in GEJ adenocarcinoma and might result in a distinct clinical phenotype. Given the high prevalence of germline ATM-mutated GEJ adenocarcinomas, genetic testing for individuals with GEJ adenocarcinomas may be considered to better inform prognostication, treatment decisions, and future cancer risk.
- Published
- 2022
15. The context-specific role of germline pathogenicity in tumorigenesis
- Author
-
Marc Ladanyi, Gowtham Jayakumaran, Karen Cadoo, Ahmet Zehir, Philip Jonsson, Xiang Li, Jason Hwee, Meera Prasad, Michael Walsh, Jinru Shia, Allison Richards, Alexander V Penson, Aijazuddin Syed, Joseph Vijai, Roy Cambria, Michael F. Berger, Barry S. Taylor, Mark E. Robson, Semanti Mukherjee, Chaitanya Bandlamudi, Jianjiong Gao, Christopher J. Fong, Ozge Ceyhan-Birsoy, Maria I. Carlo, Liying Zhang, David M. Hyman, Preethi Srinivasan, Ino de Bruijn, Diana Mandelker, Jesse Galle, Selcuk Onur Sumer, Craig M. Bielski, Kenneth Offit, Shweta S. Chavan, Nikolaus Schultz, Yelena Kemel, David B. Solit, and Zsofia K. Stadler
- Subjects
Heterozygote ,DNA Copy Number Variations ,Carcinogenesis ,Somatic cell ,Biology ,medicine.disease_cause ,DNA Mismatch Repair ,Medical and Health Sciences ,Germline ,Rare Diseases ,Clinical Research ,Neoplasms ,Genetics ,medicine ,Humans ,2.1 Biological and endogenous factors ,Genetic Predisposition to Disease ,Aetiology ,Allele ,Gene ,Germ-Line Mutation ,Cancer ,Human Genome ,Biological Sciences ,medicine.disease ,Phenotype ,Penetrance ,Developmental Biology - Abstract
Human cancers arise from environmental, heritable and somatic factors, but how these mechanisms interact in tumorigenesis is poorly understood. Studying 17,152 prospectively sequenced patients with cancer, we identified pathogenic germline variants in cancer predisposition genes, and assessed their zygosity and co-occurring somatic alterations in the concomitant tumors. Two major routes to tumorigenesis were apparent. In carriers of pathogenic germline variants in high-penetrance genes (5.1% overall), lineage-dependent patterns of biallelic inactivation led to tumors exhibiting mechanism-specific somatic phenotypes and fewer additional somatic oncogenic drivers. Nevertheless, 27% of cancers in these patients, and most tumors in patients with pathogenic germline variants in lower-penetrance genes, lacked particular hallmarks of tumorigenesis associated with the germline allele. The dependence of tumors on pathogenic germline variants is variable and often dictated by both penetrance and lineage, a finding with implications for clinical management. A study of 17,152 patients with cancer identified pathogenic germline variants in cancer predisposition genes. Although tumors showed biallelic inactivation for high-penetrance genes, this was not the case in most patients with pathogenic variants in low-penetrance genes, suggesting alternative routes to tumorigenesis.
- Published
- 2021
16. Prevalence and Characterization of Biallelic and Monoallelic NTHL1 and MSH3 Variant Carriers From a Pan-Cancer Patient Population
- Author
-
David B. Solit, Angela G. Arnold, Mark E. Robson, Diana Mandelker, Michael F. Berger, Shweta S. Chavan, Maksym Misyura, Michael Walsh, Anna Maio, Sarah R. Kane, Karen Cadoo, Yelena Kemel, Preethi Srinivasan, Ying Liu, Jinru Shia, Kenneth Offit, Kelsey Breen, Jennifer Kennedy, Margaret Sheehan, Semanti Mukherjee, Caitlin Bourque, Chaitanya Bandlamudi, Zalak Patel, Maria I. Carlo, Zsofia K. Stadler, Amanda Catchings, Erin E. Salo-Mullen, Arnold J. Markowitz, Barry S. Taylor, Prince Rainier Tejada, Rosalba Sacca, Vanessa Marcell, Alicia Latham, Ozge Ceyhan-Birsoy, Mark T.A. Donoghue, Kimberly Amoroso, and Megha Ranganathan
- Subjects
0301 basic medicine ,Genetics ,Cancer Research ,Pan cancer ,business.industry ,Cancer predisposition ,Cancer ,medicine.disease ,03 medical and health sciences ,Patient population ,030104 developmental biology ,0302 clinical medicine ,Oncology ,MSH3 ,030220 oncology & carcinogenesis ,medicine ,business ,Gene - Abstract
PURPOSE NTHL1 and MSH3 have been implicated as autosomal recessive cancer predisposition genes. Although individuals with biallelic NTHL1 and MSH3 pathogenic variants (PVs) have increased cancer and polyposis risk, risks for monoallelic carriers are uncertain. We sought to assess the prevalence and characterize NTHL1 and MSH3 from a large pan-cancer patient population. MATERIALS AND METHODS Patients with pan-cancer (n = 11,081) underwent matched tumor-normal sequencing with consent for germline analysis. Medical records and tumors were reviewed and analyzed. Prevalence of PVs was compared with reference controls (Genome Aggregation Database). RESULTS NTHL1-PVs were identified in 40 patients including 39 monoallelic carriers (39/11,081 = 0.35%) and one with biallelic variants (1/11,081 = 0.009%) and a diagnosis of isolated early-onset breast cancer. NTHL1-associated mutational signature 30 was identified in the tumors of the biallelic patient and two carriers. Colonic polyposis was not identified in any NTHL1 patient. MSH3-PVs were identified in 13 patients, including 12 monoallelic carriers (12/11,081 = 0.11%) and one with biallelic MSH3 variants (1/11,081 = 0.009%) and diagnoses of later-onset cancers, attenuated polyposis, and abnormal MSH3-protein expression. Of the 12 MSH3 carriers, two had early-onset cancer diagnoses with tumor loss of heterozygosity of the wild-type MSH3 allele. Ancestry-specific burden tests demonstrated that NTHL1 and MSH3 prevalence was not significantly different in this pan-cancer population versus controls. CONCLUSION NTHL1 and MSH3 germline alterations were not enriched in this pan-cancer patient population. However, tumor-specific findings, such as mutational signature 30 and loss of heterozygosity of the wild-type allele, suggest the potential contribution of monoallelic variants to tumorigenesis in a subset of patients.
- Published
- 2021
17. Microsatellite instability-high endometrial cancers with MLH1 promoter hypermethylation have distinct molecular and clinical profiles
- Author
-
Beryl L. Manning-Geist, Ying L. Liu, Kelly A. Devereaux, Arnaud Da Cruz Paula, Qin C. Zhou, Weining Ma, Pier Selenica, Ozge Ceyhan-Birsoy, Lea A. Moukarzel, Timothy Hoang, Sushmita Gordhandas, Maria M. Rubinstein, Claire F. Friedman, Carol Aghajanian, Nadeem R. Abu-Rustum, Zsofia K. Stadler, Jorge S. Reis-Filho, Alexia Iasonos, Dmitriy Zamarin, Lora H. Ellenson, Yulia Lakhman, Diana L. Mandelker, and Britta Weigelt
- Subjects
Cancer Research ,Brain Neoplasms ,DNA ,DNA Mismatch Repair ,Article ,Endometrial Neoplasms ,Oncology ,Neoplastic Syndromes, Hereditary ,Humans ,Female ,Microsatellite Instability ,Colorectal Neoplasms ,MutL Protein Homolog 1 ,Germ-Line Mutation - Abstract
Purpose: Microsatellite instability–high (MSI-H) endometrial carcinomas are underpinned by distinct mechanisms of DNA mismatch repair deficiency (MMR-D). We sought to characterize the clinical and genetic features of MSI-H endometrial cancers harboring germline or somatic mutations in MMR genes or MLH1 promoter hypermethylation (MLH1ph). Experimental Design: Of > 1,100 patients with endometrial cancer that underwent clinical tumor-normal sequencing, 184 had MSI-H endometrial cancers due to somatic MMR mutations or MLH1ph, or harbored pathogenic germline MMR mutations. Clinicopathologic features, mutational landscape, and tumor-infiltrating lymphocyte (TIL) scores were compared among MMR-D groups using nonparametric tests. Log-rank tests were used for categorical associations; Kaplan–Meier method and Wald test based on Cox proportional hazards models were employed for continuous variables and survival analyses. Results: Compared with patients with germline (n = 25) and somatic (n = 39) mutations, patients with MLH1ph endometrial cancers (n = 120) were older (P < 0.001), more obese (P = 0.001) and had more advanced disease at diagnosis (P = 0.025). MLH1ph endometrial cancers were enriched for JAK1 somatic mutations as opposed to germline MMR-D endometrial cancers which showed enrichment for pathogenic ERBB2 mutations. MLH1ph endometrial cancers exhibited lower tumor mutational burden and TIL scores compared with endometrial cancers harboring germline or somatic MMR mutations (P < 0.01). MLH1ph endometrial cancer patients had shorter progression-free survival (PFS) on univariate analysis, but in multivariable models, stage at diagnosis remained the only predictor of survival. For stage I/II endometrial cancer, two-year PFS was inferior for patients with MLH1ph endometrial cancers compared with germline and somatic MMR groups (70% vs. 100%, respectively). Conclusions: MLH1ph endometrial cancers likely constitute a distinct clinicopathologic entity compared with germline and somatic MMR-D ECs with potential treatment implications.
- Published
- 2022
18. Expanded genetic testing of GIST patients identifies high proportion of non-syndromic patients with germline alterations
- Author
-
Diana Mandelker, Antonio Marra, Nikita Mehta, Pier Selenica, Zarina Yelskaya, Ciyu Yang, Joshua Somar, Miika Mehine, Maksym Misyura, Olca Basturk, Alicia Latham, Maria Carlo, Michael Walsh, Zsofia K. Stadler, Kenneth Offit, Chaitanya Bandlamudi, Meera Hameed, Ping Chi, Jorge S. Reis-Filho, and Ozge Ceyhan-Birsoy
- Subjects
Cancer Research ,Oncology - Abstract
Traditional genetic testing for patients with gastrointestinal stromal tumors (GISTs) focus on those with syndromic features. To assess whether expanded genetic testing of GIST patients could identify hereditary cancer predisposition, we analyzed matched tumor-germline sequencing results from 103 patients with GISTs over a 6-year period. Germline pathogenic/likely pathogenic (P/LP) variants in GIST-associated genes (SDHA, SDHB, SDHC, NF1, KIT) were identified in 69% of patients with KIT/PDGFRA-wildtype GISTs, 63% of whom did not have any personal or family history of syndromic features. To evaluate the frequency of somatic versus germline variants identified in tumor-only sequencing of GISTs, we analyzed 499 de-identified tumor-normal pairs. P/LP variants in certain genes (e.g., BRCA1/2, SDHB) identified in tumor-only sequencing of GISTs were almost exclusively germline in origin. Our results provide guidance for genetic testing of GIST patients and indicate that germline testing should be offered to all patients with KIT/PDGFRA-wildtype GISTs regardless of their history of syndromic features.
- Published
- 2022
19. Paired Tumor-Normal Sequencing Provides Insights Into the TP53-Related Cancer Spectrum in Patients With Li-Fraumeni Syndrome
- Author
-
M. Herman Chui, Ciyu Yang, Maksym Misyura, Zsofia K. Stadler, Jorge S. Reis-Filho, Gowtham Jayakumaran, Sowmya Jairam, Ryan Ptashkin, Nikita Mehta, Marc Ladanyi, Ozge Ceyhan-Birsoy, Kenneth Offit, Anna Maio, Yirong Li, Yelena Kemel, Mark E. Robson, Maria I. Carlo, Margaret Sheehan, Alicia Latham, Pier Selenica, Ahmet Zehir, Erin E. Salo-Mullen, Michael Walsh, Umut Aypar, and Diana Mandelker
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Somatic cell ,Cancer ,medicine.disease ,DNA sequencing ,Germline ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Immunohistochemistry ,Li fraumeni ,Allele ,business ,Genetic testing - Abstract
Background Genetic testing for Li-Fraumeni syndrome (LFS) is performed by using blood specimens from patients selected based on phenotype-dependent guidelines. This approach is problematic for understanding the LFS clinical spectrum because patients with nonclassical presentations are missed, clonal hematopoiesis–related somatic blood alterations cannot be distinguished from germline variants, and unrelated tumors cannot be differentiated from those driven by germline TP53 defects. Methods To provide insights into the LFS-related cancer spectrum, we analyzed paired tumor-blood DNA sequencing results in 17 922 patients with cancer and distinguished clonal hematopoiesis–related, mosaic, and germline TP53 variants. Loss of heterozygosity and TP53 mutational status were assessed in tumors, followed by immunohistochemistry for p53 expression on a subset to identify those lacking biallelic TP53 inactivation. Results Pathogenic/likely pathogenic TP53 variants were identified in 50 patients, 12 (24.0%) of which were clonal hematopoiesis related and 4 (8.0%) of which were mosaic. Twelve (35.3%) of 34 patients with germline TP53 variants did not meet LFS testing criteria. Loss of heterozygosity of germline TP53 variant was observed in 96.0% (95% confidence interval [CI] = 79.7% to 99.9%) of core LFS spectrum–type tumors vs 45.5% (95% CI = 16.8% to 76.6%) of other tumors and 91.3% (95% CI = 72.0% to 98.9%) of tumors from patients who met LFS testing criteria vs 61.5% (95% CI = 31.6% to 86.1%) of tumors from patients who did not. Tumors retaining the wild-type TP53 allele exhibited wild-type p53 expression. Conclusions Our results indicate that some TP53 variants identified in blood-only sequencing are not germline and a substantial proportion of patients with LFS are missed based on current testing guidelines. Additionally, a subset of tumors from patients with LFS do not have biallelic TP53 inactivation and may represent cancers unrelated to their germline TP53 defect.
- Published
- 2021
20. Concurrent Germline BRCA1/2 and Mismatch Repair Mutations in Young-Onset Pancreatic and Colorectal Cancer: The Importance of Comprehensive Germline and Somatic Characterization to Inform Therapeutic Options
- Author
-
Muhammet Ozer, Megha Ranganathan, Nicolas Lecomte, Juan M. Schvartzman, Henry S. Walch, Walid K. Chatila, Jungeui Hong, Maria I. Carlo, Michael F. Walsh, Margaret Sheehan, Diana Mandelker, Ozge Ceyhan-Birsoy, Anna Maio, Yelena Kemel, Christine A. Iacobuzio-Donahue, Eileen M. O'Reilly, and Kenneth H. Yu
- Subjects
Cancer Research ,Germ Cells ,Oncology ,BRCA1 Protein ,Mutation ,Humans ,Case Reports ,Colorectal Neoplasms ,DNA Mismatch Repair - Published
- 2022
21. Abstract 5214: Expanding the spectrum of germline-driven cancers by leveraging population-scale targeted tumor and normal sequencing
- Author
-
Miika Mehine, Rebecca Caeser, Yelena Kemel, Daniel Muldoon, Sebastià Franch-Expósito, A. Rose Brannon, Aijazuddin Syed, Ozge Ceyhan-Birsoy, Maksym Misyura, Panieh Terraf, David B. Solit, Marc Ladanyi, Kenneth Offit, Zsofia K. Stadler, Diana L. Mandelker, Yonina R. Murciano-Goroff, Charles M. Rudin, Michael F. Berger, and Chaitanya Bandlamudi
- Subjects
Cancer Research ,Oncology - Abstract
Large case-control and familial studies have established clear cancer-specific risk profiles for several key cancer predisposition genes (CPGs). For example, germline pathogenic variants (PVs) in BRCA1/2 (gBRCA) are associated with increased risk for developing breast, ovarian, pancreatic, and prostate cancers. However, the extent to which gBRCA mutations are involved in mediating the tumorigenesis of other cancer types remains challenging to characterize. We hypothesized that integrating orthogonal features such as selection for biallelic inactivation of the PVs and depletion of canonical somatic drivers among the carriers can enrich the signal for identifying novel gene and cancer type associations. We then extend this framework to identify novel CPGs as well as to understand how tumors arise in patients with PVs in oncogenes. To study this, we leveraged the prospective MSK-IMPACT matched tumor-normal sequencing cohort of 49,291 patients across 77 major cancer types. We study 90 well-known CPGs as well as >300 cancer genes not previously associated with cancer predisposition. Overall, 8% (N=3,964) of patients harbored a PV in high or moderate penetrance CPGs. We identified 90 gene and cancer type associations with enrichment for biallelic inactivation (q Among carriers of PVs in oncogenes, we observe two possible mechanisms of first somatic hit towards malignant transformation. We find enrichment for copy number gain or copy neutral loss of heterozygosity of the germline PV in thyroid cancers with a PV in RET. We also find that lung cancers with a germline PV in EGFR frequently developed additional somatic point mutations located in cis with the PV. Investigating genes with no prior association with germline predisposition to cancer, we find evidence for KEAP1 and CIC as likely novel CPGs. Lung (n=8) and thyroid (n=4) cancers with deleterious germline variants in KEAP1 were characterized by loss of the wild-type allele, co-occurring somatic STK11 mutations, and depletion of canonical drivers such as EGFR. We also found biallelic loss of CIC in two patients with Neuroblastoma, each carrying a different germline loss-of-function mutation in CIC. Both tumors were also negative for MYCN and ALK defects. Collectively, our findings expand our understanding of cancer predisposition in cancer, shed new insights into how tumors arise in germline carriers, and provide a framework for identifying new CPGs using population scale tumor-normal paired clinical sequencing data. Citation Format: Miika Mehine, Rebecca Caeser, Yelena Kemel, Daniel Muldoon, Sebastià Franch-Expósito, A. Rose Brannon, Aijazuddin Syed, Ozge Ceyhan-Birsoy, Maksym Misyura, Panieh Terraf, David B. Solit, Marc Ladanyi, Kenneth Offit, Zsofia K. Stadler, Diana L. Mandelker, Yonina R. Murciano-Goroff, Charles M. Rudin, Michael F. Berger, Chaitanya Bandlamudi. Expanding the spectrum of germline-driven cancers by leveraging population-scale targeted tumor and normal sequencing. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5214.
- Published
- 2023
22. Quantifying Downstream Healthcare Utilization in Studies of Genomic Testing
- Author
-
Zoë P. Mackay, Dmitry Dukhovny, Kathryn A. Phillips, Alan H. Beggs, Robert C. Green, Richard B. Parad, Kurt D. Christensen, Pankaj B. Agrawal, Ozge Ceyhan-Birsoy, Shawn Fayer, Leslie A. Frankel, Casie A. Genetti, Amanda M. Gutierrez, Maegan Harden, Ingrid A. Holm, Joel B. Krier, Matthew S. Lebo, Kalotina Machini, Amy L. McGuire, Medha Naik, Tiffany T. Nguyen, Stacey Pereira, Vivek Ramanathan, Heidi L. Rehm, Amy Roberts, Jill O. Robinson, Sergei Roumiantsev, Talia S. Schwartz, Tina K. Truong, Grace E. VanNoy, Susan E. Waisbren, and Timothy W. Yu
- Subjects
Male ,Parents ,Psychological intervention ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Surveys and Questionnaires ,Humans ,Medicine ,Genetic Testing ,Longitudinal Studies ,030212 general & internal medicine ,Medical diagnosis ,Genetic testing ,Downstream (petroleum industry) ,medicine.diagnostic_test ,business.industry ,030503 health policy & services ,Health Policy ,Medical record ,Public Health, Environmental and Occupational Health ,Infant ,Genomics ,Patient Acceptance of Health Care ,medicine.disease ,Telephone ,Healthcare utilization ,Female ,Personalized medicine ,Medical emergency ,0305 other medical science ,business - Abstract
Objectives The challenges of understanding how interventions influence follow-up medical care are magnified during genomic testing because few patients have received it to date and because the scope of information it provides is complex and often unexpected. We tested a novel strategy for quantifying downstream healthcare utilization after genomic testing to more comprehensively and efficiently identify related services. We also evaluated the effectiveness of different methods for collecting these data. Methods We developed a risk-based approach for a trial of newborn genomic sequencing in which we defined primary conditions based on existing diagnoses and family histories of disease and defined secondary conditions based on unexpected findings. We then created patient-specific lists of services associated with managing primary and secondary conditions. Services were quantified based on medical record reviews, surveys, and telephone check-ins with parents. Results By focusing on services that genomic testing would most likely influence in the short-term, we reduced the number of services in our analyses by more than 90% compared with analyses of all observed services. We also identified the same services that were ordered in response to unexpected findings as were identified during expert review and by confirming whether recommendations were completed. Data also showed that quantifying healthcare utilization with surveys and telephone check-ins alone would have missed the majority of attributable services. Conclusions Our risk-based strategy provides an improved approach for assessing the short-term impact of genomic testing and other interventions on healthcare utilization while conforming as much as possible to existing best-practice recommendations.
- Published
- 2020
23. Abstract P6-09-01: RAD51B loss-of-function variants confer susceptibility to hereditary breast and ovarian cancers and result in tumors with genomic features of homologous recombination repair defects
- Author
-
Mark E. Robson, Yelena Kemel, Pier Selenica, Britta Weigelt, David N Brown, Semanti Mukherjee, Diana SReis-FilhoReis-Filho Mandelker, Margaret Sheehan, Simon N. Powell, Jorge S. Reis-Filho, Kenneth Offit, Ozge Ceyhan-Birsoy, Jeremy Setton, Nadeem Riaz, and Kaitlyn Tkachuk
- Subjects
Cancer Research ,PALB2 ,Cancer ,Biology ,medicine.disease ,Germline ,Loss of heterozygosity ,Breast cancer ,Germline mutation ,Oncology ,PARP inhibitor ,medicine ,Cancer research ,Ovarian cancer - Abstract
Introduction: Germline pathogenic variants in genes in the homologous recombination (HR) pathway such as BRCA1, BRCA2, ATM, PALB2, RAD51C and RAD51D confer an increased risk of breast and ovarian cancers. Screening for germline variants in these cancer susceptibility genes is critical as prophylactic measures and monitoring can be performed in these individuals to minimize their risk of developing breast and/or ovarian cancers. More recently, it has been recognized that cancers arising in carriers of germline pathogenic (P)/ likely pathogenic (LP) variants in HR genes often show a homologous recombination (HR) deficient (HRD) phenotype and can be targeted with platinum agents or PARP inhibitors. RAD51B is a RAD51 paralog that binds to RAD51C and functions as a heterodimer in the HR pathway. Whilst RAD51B germline variants have been reported in isolated cases of breast and ovarian cancers, it is unclear as to whether RAD51B P/LP germline variants would confer predisposition to these cancer types. Materials and Methods: We screened 9,287 consecutive unselected cancer patients who consented for both tumor and germline testing using the MSK-IMPACT platform for the presence of RAD51B germline truncating variants. Selected RAD51B germline mutant breast cancers identified by MSK-IMPACT were subjected to whole-exome sequencing (WES) analysis to determine the dominant mutational signatures using DeconstructSigs. Functional assays were performed to ascertain the impact of RAD51B loss on HR DNA repair and PARP inhibitor sensitivity using genome editing methods in non-malignant breast epithelial cell lines. Results: Out of the 9,287 cancer patients, we detected likely pathogenic loss of function RAD51B germline variants in 11 patients (0.12%), which was similar to the frequency detected in the gnomAD database (0.09%). All female carriers of RAD51B loss of function variants (n=8) had breast or ovarian cancers (8/1,619 breast or ovarian cancers, 0.5%). The observed carrier frequencies of germline truncating variants in RAD51B was statistically enriched in breast and ovarian cancer patients compared to individuals in the gnomAD database (0.5% vs 0.09% P=0.0003; odds ratio = 5.06 (95% CI: 2.1-10.3)). Although segregation studies were not available, 9/11 of the RAD51B germline loss of function variant carriers had a personal or family history of breast or ovarian cancers. All five breast and ovarian cancers from RAD51B mutation carriers investigated by WES were found to harbor RAD51B bi-allelic inactivation through loss of heterozygosity of the RAD51B wild-type allele. In addition, these five cases were found to display genomic features of HRD including high large-scale state transition scores and a dominant mutational signature 3. CRISPR/Cas9 genome editing of RAD51B in a non-malignant breast epithelial cell model revealed that RAD51B deficient cells display PARP inhibitor sensitivity similar to that reported in BRCA1 and BRCA2 deficient cell lines. Conclusion: RAD51B loss-of-function germline variants confer susceptibility to breast and ovarian cancer development. Breast and ovarian cancers occurring in the context of RAD51B germline mutations harbor bi-allelic inactivation of RAD51B through loss-of-heterozygosity of the wild-type allele, genomic features of HRD and are likely sensitive to PARP inhibition. Albeit rarely germline mutated, RAD51B should be considered as an addition to clinical germline testing panels for hereditary breast and ovarian cancer syndrome patients. Citation Format: Diana SReis-FilhoReis-Filho Mandelker, Semanti Mukherjee, Jeremy Setton, Pier Selenica, Yelena Kemel, Ozge Ceyhan-Birsoy, Margaret Sheehan, Kaitlyn Tkachuk, David N Brown, Simon Powell, Britta Weigelt, Mark E Robson, Nadeem Riaz, Kenneth Offit, Jorge S Reis-Filho. RAD51B loss-of-function variants confer susceptibility to hereditary breast and ovarian cancers and result in tumors with genomic features of homologous recombination repair defects [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P6-09-01.
- Published
- 2020
24. Evolving Significance of Tumor-Normal Sequencing in Cancer Care
- Author
-
Ozge Ceyhan-Birsoy and Diana Mandelker
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Somatic cell ,DNA Mutational Analysis ,Antineoplastic Agents ,Article ,Germline ,Circulating Tumor DNA ,03 medical and health sciences ,0302 clinical medicine ,Neoplastic Syndromes, Hereditary ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Molecular Targeted Therapy ,Precision Medicine ,Clinical care ,Germ-Line Mutation ,business.industry ,Tumor biology ,Liquid Biopsy ,Prognosis ,Advanced cancer ,Patient management ,030104 developmental biology ,030220 oncology & carcinogenesis ,Hereditary Cancer ,Treatment decision making ,business - Abstract
Molecular tests assist at various stages of cancer patient management, including providing diagnosis, predicting prognosis, identifying therapeutic targets, and determining hereditary cancer risk. The current testing paradigm involves germline testing in a subset of patients determined to be at high risk for having a hereditary cancer syndrome, and tumor-only sequencing for treatment decisions in advanced cancer patients. A major limitation of tumor-only sequencing is its inability to distinguish germline versus somatic mutations. Tumor-normal sequencing has emerged as a comprehensive analysis for both hereditary cancer predisposition and somatic profiling. Here, we review recent studies involving tumor-normal sequencing, discuss its benefits in clinical care, challenges for its implementation, and novel insights it has provided regarding tumor biology and germline contribution to cancer.
- Published
- 2020
25. Germline Variants Identified in Patients with Early-onset Renal Cell Carcinoma Referred for Germline Genetic Testing
- Author
-
Peter Reisz, Hong Truong, Kenneth Offit, Andrew T. Lenis, A. Ari Hakimi, Sujata Patil, Nikita Mehta, Neil J. Shah, Marc Ladanyi, Chung-Han Lee, Robert J. Motzer, Ozge Ceyhan-Birsoy, Aliya Khurram, Michael Walsh, Paul Russo, Ritesh Kotecha, Yelena Kemel, Martin H. Voss, Diana Mandelker, Jonathan A. Coleman, Zsofia K. Stadler, Vijai Joseph, Maria I. Carlo, Darren R. Feldman, Alicia Latham, Rania Sheikh, and Mark E. Robson
- Subjects
Oncology ,medicine.medical_specialty ,Genetic counseling ,Urology ,Gene mutation ,urologic and male genital diseases ,Kidney ,Medical Oncology ,Article ,Germline mutation ,Renal cell carcinoma ,Internal medicine ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Genetic Testing ,Retroperitoneal Neoplasms ,neoplasms ,CHEK2 ,Carcinoma, Renal Cell ,Genetic testing ,Retrospective Studies ,medicine.diagnostic_test ,business.industry ,Cancer ,medicine.disease ,female genital diseases and pregnancy complications ,Kidney Neoplasms ,Germ Cells ,Surgery ,Ureter ,business ,Kidney cancer - Abstract
Background Despite guidelines recommending genetic counseling for patients with early-onset renal cell carcinoma (RCC), studies interrogating the spectrum of germline mutations and clinical associations in patients with early-onset RCC are lacking. Objective To define the germline genetic spectrum and clinical associations for patients with early-onset RCC diagnosed at age ≤46 yr who underwent genetic testing. Design, setting, and participants We retrospectively identified patients with early-onset RCC who underwent germline testing at our institution from February 2003 to June 2020. Outcome measurement and statistical analysis The frequency and spectrum of pathogenic/likely pathogenic (P/LP) variants were determined. Clinical characteristics associated with mutation status were analyzed using two-sample comparison (Fisher’s exact or χ2 test). Results and limitations Of 232 patients with early-onset RCC, 50% had non–clear-cell histology, including unclassified RCC (12.1%), chromophobe RCC (9.7%), FH-deficient RCC (7.0%), papillary RCC (6.6%), and translocation-associated RCC (4.3%). Overall, 43.5% had metastatic disease. Germline P/LP variants were identified in 41 patients (17.7%), of which 21 (9.1%) were in an RCC-associated gene and 20 (8.6%) in a non–RCC-associated gene, including 17 (7.3%) in DNA damage repair genes such as BRCA1/2, ATM, and CHEK2. Factors associated with RCC P/LP variants include bilateral/multifocal renal tumors, non–clear-cell histology, and additional extrarenal primary malignancies. In patients with only a solitary clear-cell RCC, the prevalence of P/LP variants in RCC-associated and non–RCC-associated genes was 0% and 9.9%, respectively. Conclusions Patients with early-onset RCC had high frequencies of germline P/LP variants in genes associated with both hereditary RCC and other cancer predispositions. Germline RCC panel testing has the highest yield when patients have clinical phenotypes suggestive of underlying RCC gene mutations. Patients with early-onset RCC should undergo comprehensive assessment of personal and family history to guide appropriate genetic testing. Patient summary In this study of 232 patients with early-onset kidney cancer who underwent genetic testing, we found a high prevalence of mutations in genes that increase the risk of cancer in both kidneys and other organs for patients and their at-risk family members. Our study suggests that patients with early-onset kidney cancer should undergo comprehensive genetic risk assessment.
- Published
- 2021
26. Germline RAD51B variants confer susceptibility to breast and ovarian cancers deficient in homologous recombination
- Author
-
Kenneth Offit, Karen Cadoo, Diana Mandelker, Britta Weigelt, Isabella Pecorari, S.N. Powell, Liying Zhang, Semanti Mukherjee, Isaac X Pei, Mark E. Robson, Nadeem Riaz, Yelena Kemel, Rachna Shah, David N Brown, Jorge S. Reis-Filho, Ozge Ceyhan-Birsoy, Pier Selenica, Jeremy Setton, Kaitlyn Tkachuk, Margaret Sheehan, Biko McMillan, RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, and School Office GROW
- Subjects
GENES ,RAD51 ,Biology ,Article ,Germline ,Rare Diseases ,Breast cancer ,Germline mutation ,Breast Cancer ,Genetics ,medicine ,2.1 Biological and endogenous factors ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,Cancer genetics ,Gene ,RC254-282 ,Cancer ,RISK ,LONG-RANGE INTERACTION ,LANDSCAPE ,MUTATIONS ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,DNA ,ASSOCIATION ,BRCA1 ,medicine.disease ,Ovarian Cancer ,4.1 Discovery and preclinical testing of markers and technologies ,Oncology ,Cancer research ,RAD51C ,COMPLEXES ,Homologous recombination ,Ovarian cancer ,GENOMICS ,Biotechnology - Abstract
Pathogenic germline mutations in the RAD51 paralog genes RAD51C and RAD51D, are known to confer susceptibility to ovarian and triple-negative breast cancer. Here, we investigated whether germline loss-of-function variants affecting another RAD51 paralog gene, RAD51B, are also associated with breast and ovarian cancer. Among 3422 consecutively accrued breast and ovarian cancer patients consented to tumor/germline sequencing, the observed carrier frequency of loss-of-function germline RAD51B variants was significantly higher than control cases from the gnomAD population database (0.26% vs 0.09%), with an odds ratio of 2.69 (95% CI: 1.4–5.3). Furthermore, we demonstrate that tumors harboring biallelic RAD51B alteration are deficient in homologous recombination DNA repair deficiency (HRD), as evidenced by analysis of sequencing data and in vitro functional assays. Our findings suggest that RAD51B should be considered as an addition to clinical germline testing panels for breast and ovarian cancer susceptibility.
- Published
- 2021
27. Psychosocial Effect of Newborn Genomic Sequencing on Families in the BabySeq Project: A Randomized Clinical Trial
- Author
-
Heidi L. Rehm, Bethany Zettler, Amy L. McGuire, Sergei Roumiantsev, Dmitry Dukhovny, Kalotina Machini, Talia S. Schwartz, Ozge Ceyhan-Birsoy, Hadley Stevens Smith, Timothy W. Yu, Jill O. Robinson, Devan Petersen, Pankaj B. Agrawal, Chet Graham, Amy E. Roberts, Tiffany T. Nguyen Dolphyn, Tina K. Truong, Maegan Harden, Carrie L. Blout Zawatsky, Casie A. Genetti, Ingrid A. Holm, Shawn Fayer, Xingquan Lu, Harvey L. Levy, Vivek Ramanathan, Richard B. Parad, Leslie A. Frankel, Jaclyn B. Murry, Amanda M. Gutierrez, Wendi N. Betting, Kaitlyn B. Lee, Grace E. VanNoy, Susan E. Waisbren, Robert C. Green, Stacey Pereira, Alan H. Beggs, Matthew S. Lebo, Kurt D. Christensen, Medha Naik, Hayley A. Peoples, Rubaiya Islam, Uma Ramamurthy, and Joel B. Krier
- Subjects
Adult ,Male ,Parents ,Pediatrics ,medicine.medical_specialty ,law.invention ,Neonatal Screening ,Randomized controlled trial ,law ,Intensive care ,Exome Sequencing ,Medicine ,Humans ,Genetic Predisposition to Disease ,Parent-Child Relations ,Family history ,Child ,Generalized estimating equation ,Original Investigation ,Newborn screening ,business.industry ,Infant, Newborn ,Genomics ,Edinburgh Postnatal Depression Scale ,Pediatrics, Perinatology and Child Health ,Anxiety ,Female ,medicine.symptom ,business ,Psychosocial - Abstract
Importance Newborn genomic sequencing (nGS) may provide health benefits throughout the life span, but there are concerns that it could also have an unfavorable (ie, negative) psychosocial effect on families. Objective To assess the psychosocial effect of nGS on families from the BabySeq Project, a randomized clinical trial evaluating the effect of nGS on the clinical care of newborns from well-baby nurseries and intensive care units. Design, Setting, and Participants In this randomized clinical trial conducted from May 14, 2015, to May 21, 2019, at well-baby nurseries and intensive care units at 3 Boston, Massachusetts, area hospitals, 519 parents of 325 infants completed surveys at enrollment, immediately after disclosure of nGS results, and 3 and 10 months after results disclosure. Statistical analysis was performed on a per-protocol basis from January 16, 2019, to December 1, 2019. Intervention Newborns were randomized to receive either standard newborn screening and a family history report (control group) or the same plus an nGS report of childhood-onset conditions and highly actionable adult-onset conditions (nGS group). Main Outcomes and Measures Mean responses were compared between groups and, within the nGS group, between parents of children who received a monogenic disease risk finding and those who did not in 3 domains of psychosocial impact: parent-child relationship (Mother-to-Infant Bonding Scale), parents’ relationship (Kansas Marital Satisfaction Scale), and parents’ psychological distress (Edinburgh Postnatal Depression Scale anxiety subscale). Results A total of 519 parents (275 women [53.0%]; mean [SD] age, 35.1 [4.5] years) were included in this study. Although mean scores differed for some outcomes at singular time points, generalized estimating equations models did not show meaningful differences in parent-child relationship (between-group difference in adjusted mean [SE] Mother-to-Infant Bonding Scale scores: postdisclosure, 0.04 [0.15]; 3 months, –0.18 [0.18]; 10 months, –0.07 [0.20]; jointP = .57) or parents’ psychological distress (between-group ratio of adjusted mean [SE] Edinburgh Postnatal Depression Scale anxiety subscale scores: postdisclosure, 1.04 [0.08]; 3 months, 1.07 [0.11]; jointP = .80) response patterns between study groups over time for any measures analyzed in these 2 domains. Response patterns on one parents’ relationship measure differed between groups over time (between-group difference in adjusted mean [SE] Kansas Marital Satisfaction Scale scores: postdisclosure, –0.19 [0.07]; 3 months, –0.04 [0.07]; and 10 months, –0.01 [0.08]; jointP = .02), but the effect decreased over time and no difference was observed on the conflict measure responses over time. We found no evidence of persistent negative psychosocial effect in any domain. Conclusions and Relevance In this randomized clinical trial of nGS, there was no persistent negative psychosocial effect on families among those who received nGS nor among those who received a monogenic disease risk finding for their infant. Trial Registration ClinicalTrials.gov Identifier:NCT02422511
- Published
- 2021
28. Cancer-Causative Mutations Occurring in Early Embryogenesis
- Author
-
Fresia Pareja, Ryan N. Ptashkin, David N. Brown, Fatemeh Derakhshan, Pier Selenica, Edaise M. da Silva, Andrea M. Gazzo, Arnaud Da Cruz Paula, Kelsey Breen, Ronglai Shen, Antonio Marra, Ahmet Zehir, Ryma Benayed, Michael F. Berger, Ozge Ceyhan-Birsoy, Sowmya Jairam, Margaret Sheehan, Utsav Patel, Yelena Kemel, Jacklyn Casanova-Murphy, Christopher J. Schwartz, Mahsa Vahdatinia, Elizabeth Comen, Laetitia Borsu, Xin Pei, Nadeem Riaz, David H. Abramson, Britta Weigelt, Michael F. Walsh, Anna-Katerina Hadjantonakis, Marc Ladanyi, Kenneth Offit, Zsofia K. Stadler, Mark E. Robson, Jorge S. Reis-Filho, and Diana Mandelker
- Subjects
Oncology ,Neoplasms ,Mutation ,Embryonic Development ,Humans ,Genetic Testing ,Alleles ,Article - Abstract
Mosaic mutations in normal tissues can occur early in embryogenesis and be associated with hereditary cancer syndromes when affecting cancer susceptibility genes (CSG). Their contribution to apparently sporadic cancers is currently unknown. Analysis of paired tumor/blood sequencing data of 35,310 patients with cancer revealed 36 pathogenic mosaic variants affecting CSGs, most of which were not detected by prior clinical genetic testing. These CSG mosaic variants were consistently detected at varying variant allelic fractions in microdissected normal tissues (n = 48) from distinct embryonic lineages in all individuals tested, indicating their early embryonic origin, likely prior to gastrulation, and likely asymmetrical propagation. Tumor-specific biallelic inactivation of the CSG affected by a mosaic variant was observed in 91.7% (33/36) of cases, and tumors displayed the hallmark pathologic and/or genomic features of inactivation of the respective CSGs, establishing a causal link between CSG mosaic variants arising in early embryogenesis and the development of apparently sporadic cancers. Significance: Here, we demonstrate that mosaic variants in CSGs arising in early embryogenesis contribute to the oncogenesis of seemingly sporadic cancers. These variants can be systematically detected through the analysis of tumor/normal sequencing data, and their detection may affect therapeutic decisions as well as prophylactic measures for patients and their offspring. See related commentary by Liggett and Sankaran, p. 889. This article is highlighted in the In This Issue feature, p. 873
- Published
- 2021
29. Prospective pan-cancer germline testing using MSK-IMPACT informs clinical translation in 751 patients with pediatric solid tumors
- Author
-
Yasmin Khakoo, Maria I. Carlo, Elise Fiala, Ozge Ceyhan-Birsoy, Leonard H. Wexler, Nancy Bouvier, Diana Mandelker, Jasmine H. Francis, Alicia Latham, Sowmya Jairam, Marc Ladanyi, Gowtham Jayakumaran, Sameer Farouk Sait, Filemon S. Dela Cruz, Ira J. Dunkel, Michael Walsh, Michael F. Berger, Todd E. Heaton, Ellen M. Basu, Matthias A. Karajannis, Ciyu Yang, Christopher J. Forlenza, Neerav Shukla, Emily K. Slotkin, Julia Glade Bender, Tanya M. Trippett, Nai-Kong Cheung, Danielle Novetsky Friedman, Justin T. Gerstle, Semanti Murkherjee, Ahmet Zehir, Zsofia K. Stadler, Erin E. Salo-Mullen, Maria Luisa Sulis, Yelena Kemel, Karen Cadoo, Sabine Topka, Jennifer Kennedy, Megan Harlan Fleischut, Nikolaus Schultz, Richard J. O'Reilly, Anna Maio, Margaret Sheehan, Shakeel Modak, Michael V. Ortiz, Alex Kentsis, Andrew L. Kung, Paul A. Meyers, Stephen S. Roberts, Kenneth Offit, Angela G. Arnold, Mark E. Robson, David H. Abramson, Stephen Gilheeney, Liying Zhang, Audrey Mauguen, and Joseph Vijai
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Cascade testing ,Pediatric Cancer ,Germline ,Article ,Rare Diseases ,Clinical Research ,Internal medicine ,Neoplasms ,Genetics ,medicine ,2.1 Biological and endogenous factors ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Prospective Studies ,Aetiology ,Child ,Likely pathogenic ,Germ-Line Mutation ,Cancer ,Pediatric ,screening and diagnosis ,Pan cancer ,business.industry ,Human Genome ,medicine.disease ,Penetrance ,Pediatric cancer ,Detection ,Good Health and Well Being ,Germ Cells ,Medical genetics ,business ,4.2 Evaluation of markers and technologies - Abstract
The spectrum of germline predisposition in pediatric cancer continues to be realized. Here we report 751 solid tumor patients who underwent prospective matched tumor-normal DNA sequencing and downstream clinical use (clinicaltrials.gov NCT01775072). Germline pathogenic and likely pathogenic (P/LP) variants were reported. One or more P/LP variants were found in 18% (138/751) of individuals when including variants in low, moderate, and high penetrance dominant or recessive genes, or 13% (99/751) in moderate and high penetrance dominant genes. 34% of high or moderate penetrance variants were unexpected based on the patient's diagnosis and previous history. 76% of patients with positive results completed a clinical genetics visit, and 21% had at least one relative undergo cascade testing as a result of this testing. Clinical actionability additionally included screening, risk reduction in relatives, reproductive use, and use of targeted therapies. Germline testing should be considered for all children with cancer.
- Published
- 2021
30. Therapeutic Implications of Germline Testing in Patients With Advanced Cancers
- Author
-
Marc Ladanyi, Michael Walsh, Anna M. Varghese, Ahmet Zehir, Erin E. Salo-Mullen, Kaitlyn Tkachuk, Nikolaus Schultz, Michael F. Berger, Wassim Abida, Bastien Nguyen, Shaleigh A. Smith, Margaret Sheehan, Diana Mandelker, Michael J. Morris, Christopher J. Fong, Maria I. Carlo, Carol Aghajanian, Gowtham Jayakumaran, Karen Cadoo, Anna Maio, Mark E. Robson, Nadeem R. Abu-Rustum, Amanda Erakky, Kenneth Offit, Luis A. Diaz, Alexander Drilon, Hongxin Zhang, Eileen M. O'Reilly, Zsofia K. Stadler, Debyani Chakravarty, Alicia Latham, Ozge Ceyhan-Birsoy, Ritika Kundra, David B. Solit, Yelena Kemel, Jesse Galle, and Ying Liu
- Subjects
Male ,Cancer Research ,MEDLINE ,Bioinformatics ,Germline ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Neoplasms ,Profiling (information science) ,Medicine ,Humans ,In patient ,Genetic Predisposition to Disease ,030212 general & internal medicine ,Prospective Studies ,Selection (genetic algorithm) ,Germ-Line Mutation ,business.industry ,ORIGINAL REPORTS ,Middle Aged ,Advanced cancer ,Oncology ,030220 oncology & carcinogenesis ,Female ,business - Abstract
PURPOSE Tumor mutational profiling is increasingly performed in patients with advanced cancer. We determined the extent to which germline mutation profiling guides therapy selection in patients with advanced cancer. METHODS Patients with cancer undergoing tumor genomic profiling were prospectively consented for germline cancer predisposition gene analysis (2015-2019). In patients harboring germline likely pathogenic or pathogenic (LP/P) alterations, therapeutic actionability was classified using a precision oncology knowledge base. Patients with metastatic or recurrent cancer receiving germline genotype–directed therapy were determined. RESULTS Among 11,947 patients across > 50 malignancies, 17% (n = 2,037) harbored a germline LP/P variant. By oncology knowledge base classification, 9% (n = 1042) had an LP/P variant in a gene with therapeutic implications (4% level 1; 4% level 3B; < 1% level 4). BRCA1/2 variants accounted for 42% of therapeutically actionable findings, followed by CHEK2 (13%), ATM (12%), mismatch repair genes (11%), and PALB2 (5%). When limited to the 9,079 patients with metastatic or recurrent cancer, 8% (n = 710) harbored level 1 or 3B genetic findings and 3.2% (n = 289) received germline genotype–directed therapy. Germline genotype–directed therapy was received by 61% and 18% of metastatic cancer patients with level 1 and level 3B findings, respectively, and by 54% of BRCA1/2, 75% of mismatch repair, 43% of PALB2, 35% of RAD51C/D, 24% of BRIP1, and 19% of ATM carriers. Of BRCA1/2 patients receiving a poly(ADP-ribose) polymerase inhibitor, 45% (84 of 188) had tumors other than breast or ovarian cancer, wherein the drug, at time of delivery, was delivered in an investigational setting. CONCLUSION In a pan-cancer analysis, 8% of patients with advanced cancer harbored a germline variant with therapeutic actionability with 40% of these patients receiving germline genotype–directed treatment. Germline sequence analysis is additive to tumor sequence analysis for therapy selection and should be considered for all patients with advanced cancer.
- Published
- 2021
31. Interpretation of Genomic Sequencing Results in Healthy and Ill Newborns: Results from the BabySeq Project
- Author
-
Ozge Ceyhan-Birsoy, Jaclyn B. Murry, Kalotina Machini, Matthew S. Lebo, Timothy W. Yu, Shawn Fayer, Casie A. Genetti, Talia S. Schwartz, Pankaj B. Agrawal, Richard B. Parad, Ingrid A. Holm, Amy L. McGuire, Robert C. Green, Heidi L. Rehm, Alan H. Beggs, Wendi N. Betting, Kurt D. Christensen, Dmitry Dukhovny, Leslie A. Frankel, Chet Graham, Amanda M. Guiterrez, Maegan Harden, Joel B. Krier, Harvey L. Levy, Xingquan Lu, Medha Naik, Tiffany T. Nguyen, Hayley A. Peoples, Stacey Pereira, Devan Petersen, Uma Ramamurthy, Vivek Ramanathan, Amy Roberts, Jill O. Robinson, Serguei Roumiantsev, Tina K. Truong, Grace E. VanNoy, and Susan E. Waisbren
- Subjects
Male ,0301 basic medicine ,Heterozygote ,Pediatrics ,medicine.medical_specialty ,Neonatal intensive care unit ,Disease ,030105 genetics & heredity ,03 medical and health sciences ,Exome Sequencing ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Age of Onset ,Family history ,Genetics (clinical) ,Exome sequencing ,Genetic testing ,Newborn screening ,medicine.diagnostic_test ,Genome, Human ,business.industry ,Racial Groups ,Infant, Newborn ,Genetic Variation ,Genomics ,Sequence Analysis, DNA ,030104 developmental biology ,Health ,Pharmacogenetics ,Pharmacogenomics ,Female ,Age of onset ,business - Abstract
Genomic sequencing provides many opportunities in newborn clinical care, but the challenges of interpreting and reporting newborn genomic sequencing (nGS) results need to be addressed for its broader and effective application. The BabySeq Project is a pilot randomized clinical trial that explores the medical, behavioral, and economic impacts of nGS in well newborns and those admitted to a neonatal intensive care unit (NICU). Here we present childhood-onset and actionable adult-onset disease risk, carrier status, and pharmacogenomics findings from nGS of 159 newborns in the BabySeq Project. nGS revealed a risk of childhood-onset disease in 15/159 (9.4%) newborns; none of the disease risks were anticipated based on the infants' known clinical or family histories. nGS also revealed actionable adult-onset disease risk in 3/85 (3.5%) newborns whose parents consented to receive this information. Carrier status for recessive diseases and pharmacogenomics variants were reported in 88% and 5% of newborns, respectively. Additional indication-based analyses were performed in 29/32 (91%) NICU newborns and 6/127 (5%) healthy newborns who later had presentations that prompted a diagnostic analysis. No variants that sufficiently explained the reason for the indications were identified; however, suspicious but uncertain results were reported in five newborns. Testing parental samples contributed to the interpretation and reporting of results in 13/159 (8%) newborns. Our results suggest that nGS can effectively detect risk and carrier status for a wide range of disorders that are not detectable by current newborn screening assays or predicted based on the infant's known clinical or family history, and the interpretation of results can substantially benefit from parental testing.
- Published
- 2019
32. Discordant results between conventional newborn screening and genomic sequencing in the BabySeq Project
- Author
-
Monica H. Wojcik, Tian Zhang, Ozge Ceyhan-Birsoy, Casie A. Genetti, Matthew S. Lebo, Timothy W. Yu, Richard B. Parad, Ingrid A. Holm, Heidi L. Rehm, Alan H. Beggs, Robert C. Green, Pankaj B. Agrawal, Wendi N. Betting, Kurt D. Christensen, Dmitry Dukhovny, Shawn Fayer, Leslie A. Frankel, Chet Graham, Amanda M. Guiterrez, Maegan Harden, Joel B. Krier, Harvey L. Levy, Xingquan Lu, Kalotina Machini, Amy L. McGuire, Jaclyn B. Murry, Medha Naik, Tiffany T. Nguyen, Hayley A. Peoples, Stacey Pereira, Devan Petersen, Uma Ramamurthy, Vivek Ramanathan, Amy Roberts, Jill O. Robinson, Serguei Roumiantsev, Talia S. Schwartz, Tina K. Truong, Grace E. VanNoy, and Susan E. Waisbren
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,Population ,030105 genetics & heredity ,Monogenic disease ,Article ,03 medical and health sciences ,Neonatal Screening ,Risk Factors ,Exome Sequencing ,False positive paradox ,Medicine ,Humans ,education ,Genetics (clinical) ,Exome sequencing ,Newborn screening ,education.field_of_study ,business.industry ,Genomic sequencing ,Infant, Newborn ,food and beverages ,Chromosome Mapping ,Infant ,Hearing screen ,Genomics ,030104 developmental biology ,embryonic structures ,business ,Genetic diagnosis - Abstract
Purpose Newborn screening (NBS) is performed to identify neonates at risk for actionable, severe, early-onset disorders, many of which are genetic. The BabySeq Project randomized neonates to receive conventional NBS or NBS plus exome sequencing (ES) capable of detecting sequence variants that may also diagnose monogenic disease or indicate genetic disease risk. We therefore evaluated how ES and conventional NBS results differ in this population. Methods We compared results of NBS (including hearing screens) and ES for 159 infants in the BabySeq Project. Infants were considered "NBS positive" if any abnormal result was found indicating disease risk and "ES positive" if ES identified a monogenic disease risk or a genetic diagnosis. Results Most infants (132/159, 84%) were NBS and ES negative. Only one infant was positive for the same disorder by both modalities. Nine infants were NBS positive/ES negative, though seven of these were subsequently determined to be false positives. Fifteen infants were ES positive/NBS negative, all of which represented risk of genetic conditions that are not included in NBS programs. No genetic explanation was identified for eight infants referred on the hearing screen. Conclusion These differences highlight the complementarity of information that may be gleaned from NBS and ES in the newborn period.
- Published
- 2021
33. Contributors
- Author
-
Ana Catarina Alves, Christina Anne Austin-Tse, Mafalda Bourbon, Marcelo A. Carvalho, Ozge Ceyhan-Birsoy, George S. Charames, Joana Rita Chora, Mara Colombo, Xavier de la Cruz, Johan T. den Dunnen, Niels de Wind, Orland Diez, Anna B.R. Elias, D Gareth Evans, Lidia Feliubadaló, Vanessa C. Fernandes, Ivo F.A.C. Fokkema, Cristina Fortuno, Alice Garrett, Paolo Gasparini, Giorgia Girotto, Anna González-Neira, Karen W. Gripp, Sara Gutiérrez-Enríquez, Steven M. Harrison, Miguel de la Hoya, Jodie Ingles, Renee Johnson, Jordan Lerner-Ellis, Harvey Levy, Conxi Lázaro, Heather Mason-Suares, Ana Margarida Medeiros, Jessica L. Mester, Alejandro Moles-Fernández, Alvaro N.A. Monteiro, Anna Morgan, Thales C. Nepomuceno, Rocío Núñez-Torres, Selen Özkan, Natàlia Padilla, Michael T. Parsons, Tina F. Pesaran, Marta Pineda, Paolo Radice, Farrah Rajabi, Ebony Richardson, Peter Sabatini, Stephanie Sacharow, Amanda Spurdle, Bryony A. Thompson, Emma Tudini, Clare Turnbull, Lisa M. Vincent, Michael F. Walsh, and Nicholas Watkins
- Published
- 2021
34. Approaches to the comprehensive interpretation of genome-scale sequencing
- Author
-
Christina Austin-Tse and Ozge Ceyhan-Birsoy
- Subjects
Interpretation (philosophy) ,Genomic data ,Carrier status ,Genome scale ,Disease ,Computational biology ,Biology ,Return of results ,Gene - Abstract
Genome-scale sequencing (GS) provides unparalleled opportunities to screen thousands of genes simultaneously and reveal a wide range of health-related information including cause of genetic disease in affected individuals, risk of disease in apparently unaffected individuals, carrier status for recessive disease, and pharmacogenetic variants that implicate drug sensitivity or response. Comprehensive interpretation of GS relies on selecting the right approaches for variant detection, filtration, and return of results depending on the purpose of testing. This chapter will review contexts in which GS is applied, important concepts in the analysis of genomic data, criteria used in selecting returnable results, and the types of results that may be returned.
- Published
- 2021
35. Prevalence and Characterization of Biallelic and Monoallelic
- Author
-
Erin E, Salo-Mullen, Anna, Maio, Semanti, Mukherjee, Chaitanya, Bandlamudi, Jinru, Shia, Yelena, Kemel, Karen A, Cadoo, Ying, Liu, Maria, Carlo, Megha, Ranganathan, Sarah, Kane, Preethi, Srinivasan, Shweta S, Chavan, Mark T A, Donoghue, Caitlin, Bourque, Margaret, Sheehan, Prince Rainier, Tejada, Zalak, Patel, Angela G, Arnold, Jennifer A, Kennedy, Kimberly, Amoroso, Kelsey, Breen, Amanda, Catchings, Rosalba, Sacca, Vanessa, Marcell, Arnold J, Markowitz, Alicia, Latham, Michael, Walsh, Maksym, Misyura, Ozge, Ceyhan-Birsoy, David B, Solit, Michael F, Berger, Mark E, Robson, Barry S, Taylor, Kenneth, Offit, Diana, Mandelker, and Zsofia K, Stadler
- Subjects
Adult ,Male ,Heterozygote ,Adolescent ,Colonic Polyps ,Genetic Variation ,Infant ,ORIGINAL REPORTS ,Middle Aged ,Deoxyribonuclease (Pyrimidine Dimer) ,Young Adult ,Child, Preschool ,MutS Homolog 3 Protein ,Humans ,Female ,Child ,Colorectal Neoplasms ,Alleles ,Aged - Abstract
NTHL1 and MSH3 have been implicated as autosomal recessive cancer predisposition genes. Although individuals with biallelic NTHL1 and MSH3 pathogenic variants (PVs) have increased cancer and polyposis risk, risks for monoallelic carriers are uncertain. We sought to assess the prevalence and characterize NTHL1 and MSH3 from a large pan-cancer patient population. MATERIALS AND METHODS: Patients with pan-cancer (n = 11,081) underwent matched tumor-normal sequencing with consent for germline analysis. Medical records and tumors were reviewed and analyzed. Prevalence of PVs was compared with reference controls (Genome Aggregation Database). RESULTS: NTHL1-PVs were identified in 40 patients including 39 monoallelic carriers (39/11,081 = 0.35%) and one with biallelic variants (1/11,081 = 0.009%) and a diagnosis of isolated early-onset breast cancer. NTHL1-associated mutational signature 30 was identified in the tumors of the biallelic patient and two carriers. Colonic polyposis was not identified in any NTHL1 patient. MSH3-PVs were identified in 13 patients, including 12 monoallelic carriers (12/11,081 = 0.11%) and one with biallelic MSH3 variants (1/11,081 = 0.009%) and diagnoses of later-onset cancers, attenuated polyposis, and abnormal MSH3-protein expression. Of the 12 MSH3 carriers, two had early-onset cancer diagnoses with tumor loss of heterozygosity of the wild-type MSH3 allele. Ancestry-specific burden tests demonstrated that NTHL1 and MSH3 prevalence was not significantly different in this pan-cancer population versus controls. CONCLUSION: NTHL1 and MSH3 germline alterations were not enriched in this pan-cancer patient population. However, tumor-specific findings, such as mutational signature 30 and loss of heterozygosity of the wild-type allele, suggest the potential contribution of monoallelic variants to tumorigenesis in a subset of patients.
- Published
- 2020
36. Fumarate hydratase c.914T > C (p.Phe305Ser) is a pathogenic variant associated with hereditary leiomyomatosis and renal cell cancer syndrome
- Author
-
Diana Mandelker, Kelsey Breen, Yelena Kemel, Ozge Ceyhan-Birsoy, Maria I. Carlo, Anna Maio, Ying-Bei Chen, and Liying Zhang
- Subjects
0301 basic medicine ,Proband ,Male ,fumarate hydratase ,Skin Neoplasms ,lcsh:QH426-470 ,Mutation, Missense ,Context (language use) ,030105 genetics & heredity ,carcinoma ,urologic and male genital diseases ,leiomyomatosis ,Germline ,Clinical Reports ,03 medical and health sciences ,Leiomyomatosis ,Renal cell carcinoma ,Neoplastic Syndromes, Hereditary ,Genetics ,medicine ,Humans ,Molecular Biology ,Genetics (clinical) ,Germ-Line Mutation ,Clinical Report ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,renal cell ,Pedigree ,lcsh:Genetics ,030104 developmental biology ,Hereditary leiomyomatosis and renal cell cancer syndrome ,Uterine Neoplasms ,Cancer research ,Female ,mutation ,business ,Kidney cancer - Abstract
Background Hereditary leiomyomatosis and renal cell cancer syndrome (HLRCC), caused by heterozygous germline pathogenic variants in the FH, confers an increased risk for cutaneous and uterine leiomyomas and renal cancer. Methods About 13,722 advanced cancer patients, including 560 with renal cell carcinoma, had germline analysis performed in the context of tumor‐normal sequencing under an IRB approved protocol. Results We report two unrelated individuals with early onset kidney cancer who both carried the c.914C > T (p.Phe305Ser) germline variant in the FH. Both tumors exhibited loss of FH staining by immunohistochemistry and/or positive 2SC staining. Subsequent familial testing discovered that a daughter of a proband who carried the variant had both cutaneous and uterine leiomyomas. Conclusion This combination of evidence suggests that the FH c.914C > T (p.Phe305Ser) is pathogenic for HLRCC., Immunohistochemical analysis of kidney tumors from two unrelated patients with the germline FH c.914T > C (p.Phe305Ser) variant revealed a loss of fumarate hydratase (FH) staining and positive S‐(2‐succino)‐cysteine (2SC) staining. We provide evidence to suggest the FH variant c.914C > T (p.Phe305Ser) is pathogenic for HLRCC.
- Published
- 2020
37. A Clinical Approach to Detecting Germline Pathogenic Variants From Tumor-Only Sequencing
- Author
-
Maksym Misyura, Ozge Ceyhan-Birsoy, and Diana Mandelker
- Subjects
Cancer Research ,Text mining ,Solicited Editorial ,Oncology ,business.industry ,Medicine ,Computational biology ,business ,Germline - Published
- 2020
38. Integrating somatic variant data and biomarkers for germline variant classification in cancer predisposition genes
- Author
-
Michael Walsh, Michael W. Drazer, Deborah I. Ritter, Kenneth Offit, Minjie Luo, Elizabeth C. Chao, Diana Mandelker, Dmitriy Sonkin, Liying Zhang, Yelena Kemel, Dale Hedges, Gang Wu, Sharon E. Plon, Shashikant Kulkarni, Debyani Chakravarty, Kristy Lee, Rajarshi Ghosh, Chimene Kesserwan, and Ozge Ceyhan-Birsoy
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Somatic cell ,Genomics ,Computational biology ,Biology ,Genome ,Article ,Germline ,Loss of heterozygosity ,03 medical and health sciences ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Allele ,Alleles ,Germ-Line Mutation ,Genetics (clinical) ,Genome, Human ,Molecular pathology ,Computational Biology ,Genetic Variation ,030104 developmental biology ,Mutation ,Medical genetics - Abstract
In its landmark paper about Standards and Guidelines for the Interpretation of Sequence Variants, the American College of Medical Genetics and Genomics (ACMG), and Association for Molecular Pathology (AMP) did not address how to use tumor data when assessing the pathogenicity of germline variants. The Clinical Genome Resource (ClinGen) established a multidisciplinary working group, the Germline/Somatic Variant Subcommittee (GSVS) with this focus. The GSVS implemented a survey to determine current practices of integrating somatic data when classifying germline variants in cancer predisposition genes. The GSVS then reviewed and analyzed available resources of relevant somatic data, and performed integrative germline variant curation exercises. The committee determined that somatic hotspots could be systematically integrated into moderate evidence of pathogenicity (PM1). Tumor RNA sequencing data showing altered splicing may be considered as strong evidence in support of germline pathogenicity (PVS1) and tumor phenotypic features such as mutational signatures be considered supporting evidence of pathogenicity (PP4). However, at present, somatic data such as focal loss of heterozygosity and mutations occurring on the alternative allele are not recommended to be systematically integrated, instead, incorporation of this type of data should take place under the advisement of multidisciplinary cancer center tumor-normal sequencing boards.
- Published
- 2018
39. A synonymous germline variant PALB2 c.18G>T (p.Gly6=) disrupts normal splicing in a family with pancreatic and breast cancers
- Author
-
Ozge Ceyhan-Birsoy, Michael Walsh, Ciyu Yang, Diana Mandelker, Sowmya Jairam, Amanda Catchings, Eileen M. O'Reilly, and Liying Zhang
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,PALB2 ,Breast Neoplasms ,Biology ,Article ,Germline ,Frameshift mutation ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Breast cancer ,medicine ,Humans ,Protein Isoforms ,Genetic Predisposition to Disease ,Germ-Line Mutation ,Ovarian Neoplasms ,Cloning ,Cancer ,Exons ,Middle Aged ,medicine.disease ,Pedigree ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,RNA splicing ,Cancer research ,Female ,Fanconi Anemia Complementation Group N Protein - Abstract
Mutations in PALB2 have been associated with a predisposition to breast and pancreatic cancers. This study aims to characterize a novel PALB2 synonymous variant c.18G>T (p.Gly6=) identified in a family with pancreatic and breast cancers. The PALB2 c.18G>T (p.Gly6=) variant in this family was identified using Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT™). RT-PCR and subsequent cloning were performed to investigate whether this variant affects normal splicing. This variant completely disrupts normal splicing and leads to several abnormal transcripts, which presumably leads to premature protein truncation. The major abnormal transcript resulted in a deletion of 32 base pairs in exon 1 and frameshift. Our results indicate that the PALB2 c.18G>T (p.Gly6=) variant is likely pathogenic. This study provided important laboratory evidence for classification of this variant and guided improved patient management.
- Published
- 2018
40. Parental Interest in Genomic Sequencing of Newborns: Enrollment Experience from the BabySeq Project
- Author
-
Casie A. Genetti, Talia S. Schwartz, Jill O. Robinson, Grace E. VanNoy, Devan Petersen, Stacey Pereira, Shawn Fayer, Hayley A. Peoples, Pankaj B. Agrawal, Wendi N. Betting, Ingrid A. Holm, Amy L. McGuire, Susan E. Waisbren, Timothy W. Yu, Robert C. Green, Alan H. Beggs, Richard B. Parad, Ozge Ceyhan-Birsoy, Kurt D. Christensen, Dmitry Dukhovny, Leslie A. Frankel, Chet Graham, Amanda M. Gutierrez, Maegan Harden, Joel B. Krier, Matthew S. Lebo, Harvey L. Levy, Xingquan Lu, Kalotina Machini, Jaclyn B. Murry, Medha Naik, Tiffany T. Nguyen, Uma Ramamurthy, Vivek Ramanathan, Heidi L. Rehm, Amy Roberts, Serguei Roumiantsev, and Tina K. Truong
- Subjects
0301 basic medicine ,Adult ,Male ,Parents ,medicine.medical_specialty ,Parental Concerns ,media_common.quotation_subject ,Genetic counseling ,Consent for Genomic Testing ,030105 genetics & heredity ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Neonatal Screening ,Randomized controlled trial ,law ,Informed consent ,030225 pediatrics ,medicine ,Humans ,Genetic Testing ,Predictive testing ,Insurability ,Genetics (clinical) ,media_common ,Genetic testing ,Newborn screening ,Genomic Sequencing ,Informed Consent ,medicine.diagnostic_test ,business.industry ,Patient Selection ,Infant, Newborn ,Sequence Analysis, DNA ,Newborn ,3. Good health ,Feeling ,Family medicine ,Female ,business ,Attitude to Health ,Newborn Screening - Abstract
Purpose: Newborn genomic sequencing (nGS) has great potential to improve pediatric care. Parental interest and concerns about genomics are relatively unexplored. Understanding why parents decline research consent for nGS may reveal implementation barriers. Methods: We evaluated parental interest in a randomized trial of nGS in well-baby and ICU nursery settings. Interested families attended an informational enrollment session (ES) with a genetic counselor prior to consenting. Reason(s) for declining participation and sociodemographic associations were analyzed. Results: Of 3,860 eligible approached families, 10% attended ES, 67% of whom enrolled. Of 1,760 families queried for decline reasons, 58% were uninterested in research. Among 499 families considering research, principal reasons for decline prior to ES included burdensome study logistics (48%), feeling overwhelmed postpartum (17%), and lack of interest/discomfort with genetic testing (17%). Decliners after ES more often cited concerns about privacy/insurability (41%) and uncertain/unfavorable results (23%). Conclusions: Low interest in research and study logistics were major initial barriers to postpartum enrollment and are likely generic to many postpartum research efforts. Concerns over privacy and result implications were most commonly cited in decliners after ES. Understanding parental concerns around research nGS may inform future integration of nGS into newborn screening, predictive testing, and pediatric diagnostics.
- Published
- 2018
41. Automated typing of red blood cell and platelet antigens: a whole-genome sequencing study
- Author
-
Kalotina Machini, Nicole Soranzo, Maria Aguad, Christine E. Seidman, Leslie E. Silberstein, Erica F. Schonman, Michael F. Murray, David W. Bates, Denise L. Perry, Heather M. McLaughlin, John Danesh, Sek Won Kong, Klaudia Walter, Tina Hambuch, Isaac S. Kohane, Judy Garber, William J. Lane, Allison L. Cirino, Carrie L. Blout, Connie M. Westhoff, Matthew S. Lebo, Ellen A. Tsai, Pamela M. Diamond, Eleanor Steffens, Lindsay Z. Feuerman, Heidi L. Rehm, Helen Mah, Cynthia C. Morton, Wendi N. Betting, David J. Roberts, Sunitha Vege, Richard M. Kaufman, Lisa Soleymani Lehmann, Jill O. Robinson, Daimon P. Simmons, Amy L. McGuire, Peter Kraft, Nicholas A. Watkins, Kaitlyn B. Lee, Adam S. Butterworth, Emanuele Di Angelantonio, Willem H. Ouwehand, J. Scott Roberts, Shamil R. Sunyaev, Calum A. MacRae, Nicholas Gleadall, Joel B. Krier, Robin Smeland-Wagman, Jason L. Vassy, Ozge Ceyhan-Birsoy, Danielle R. Azzariti, Samuel J. Aronson, Tiffany T. Nguyen, Robert C. Green, Kurt D. Christensen, Carolyn Y. Ho, Kelly Davis, Peter A. Ubel, Melody J. Slashinski, and Jennifer Blumenthal-Barby
- Subjects
0301 basic medicine ,Whole genome sequencing ,medicine.medical_specialty ,Blood transfusion ,business.industry ,medicine.medical_treatment ,Transfusion medicine ,Hematology ,Computational biology ,030204 cardiovascular system & hematology ,Genome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,ABO blood group system ,medicine ,SNP ,Human genome ,Typing ,business - Abstract
Summary Background There are more than 300 known red blood cell (RBC) antigens and 33 platelet antigens that differ between individuals. Sensitisation to antigens is a serious complication that can occur in prenatal medicine and after blood transfusion, particularly for patients who require multiple transfusions. Although pre-transfusion compatibility testing largely relies on serological methods, reagents are not available for many antigens. Methods based on single-nucleotide polymorphism (SNP) arrays have been used, but typing for ABO and Rh—the most important blood groups—cannot be done with SNP typing alone. We aimed to develop a novel method based on whole-genome sequencing to identify RBC and platelet antigens. Methods This whole-genome sequencing study is a subanalysis of data from patients in the whole-genome sequencing arm of the MedSeq Project randomised controlled trial (NCT01736566) with no measured patient outcomes. We created a database of molecular changes in RBC and platelet antigens and developed an automated antigen-typing algorithm based on whole-genome sequencing (bloodTyper). This algorithm was iteratively improved to address cis–trans haplotype ambiguities and homologous gene alignments. Whole-genome sequencing data from 110 MedSeq participants (30 × depth) were used to initially validate bloodTyper through comparison with conventional serology and SNP methods for typing of 38 RBC antigens in 12 blood-group systems and 22 human platelet antigens. bloodTyper was further validated with whole-genome sequencing data from 200 INTERVAL trial participants (15 × depth) with serological comparisons. Findings We iteratively improved bloodTyper by comparing its typing results with conventional serological and SNP typing in three rounds of testing. The initial whole-genome sequencing typing algorithm was 99·5% concordant across the first 20 MedSeq genomes. Addressing discordances led to development of an improved algorithm that was 99·8% concordant for the remaining 90 MedSeq genomes. Additional modifications led to the final algorithm, which was 99·2% concordant across 200 INTERVAL genomes (or 99·9% after adjustment for the lower depth of coverage). Interpretation By enabling more precise antigen-matching of patients with blood donors, antigen typing based on whole-genome sequencing provides a novel approach to improve transfusion outcomes with the potential to transform the practice of transfusion medicine. Funding National Human Genome Research Institute, Doris Duke Charitable Foundation, National Health Service Blood and Transplant, National Institute for Health Research, and Wellcome Trust.
- Published
- 2018
42. A Novel Missense Variant in the AGRN Gene; Congenital Myasthenic Syndrome Presenting With Head Drop
- Author
-
Mert Karakaya, Haluk Topaloglu, Alan H. Beggs, and Ozge Ceyhan-Birsoy
- Subjects
Male ,0301 basic medicine ,Weakness ,Pathology ,medicine.medical_specialty ,DNA Mutational Analysis ,Mutation, Missense ,Article ,LMNA ,03 medical and health sciences ,0302 clinical medicine ,Ptosis ,medicine ,Humans ,Missense mutation ,Exome ,Agrin ,Exome sequencing ,Myasthenic Syndromes, Congenital ,business.industry ,Infant ,General Medicine ,Congenital myasthenic syndrome ,medicine.disease ,Hypotonia ,030104 developmental biology ,Neurology ,Child, Preschool ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Congenital myasthenic syndromes (CMS) are a heterogeneous group of diseases of the neuromuscular junction caused by compromised synaptic transmission. Clinical features include early-onset weakness of limbs and oculobulbar muscles resulting in hypotonia, bulbar paresis, ptosis, and hypoventilation. The first dropped head syndrome in children were detected in 2 patients with LMNA and SEPN1 mutations. We report a 17-month-old boy with dropped head and limb-girdle weakness, who had no ptosis or ophthalmoplegia at presentation. We performed whole exome sequencing, which revealed a homozygous missense variant in the AGRN gene c.5023G>A, p.Gly1675Ser in the LG2 domain, which is predicted to be likely disease causing by in silico tools. Agrin is known to play a critical role in the development and maintenance of the neuromuscular junction. Agrin-related CMS is one of the rarest subtypes. Of note, our patient is the first described patient with agrin-related CMS with dropped head phenotype.
- Published
- 2017
43. Analyzing and Reanalyzing the Genome: Findings from the MedSeq Project
- Author
-
Lisa Mahanta, Matthew S. Lebo, Peter Rossetti, Himanshu Sharma, Laura Hutchinson, Danielle R. Azzariti, Ozge Ceyhan-Birsoy, Robert C. Green, Kalotina Machini, Heidi L. Rehm, and Heather M. McLaughlin
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Multifactorial Inheritance ,Cardiomyopathy ,Disease ,030105 genetics & heredity ,Genome ,DNA sequencing ,Article ,03 medical and health sciences ,Internal medicine ,Genotype ,Genetics ,Medicine ,Humans ,Family ,Genetic Predisposition to Disease ,Gene ,Genetics (clinical) ,Randomized Controlled Trials as Topic ,Whole Genome Sequencing ,business.industry ,Genome, Human ,Computational Biology ,Genetic Variation ,Sequence Analysis, DNA ,medicine.disease ,030104 developmental biology ,Pharmacogenomics ,Case-Control Studies ,Data Interpretation, Statistical ,Cohort ,Female ,business ,Cardiomyopathies - Abstract
Although genome sequencing is increasingly available in clinical and research settings, many questions remain about the interpretation of sequencing data. In the MedSeq Project, we explored how much effort is required to evaluate and report on more than 4,500 genes reportedly associated with monogenic conditions, as well as pharmacogenomic (PGx) markers, blood antigen serotyping, and polygenic risk scores in 100 individuals (50 with cardiomyopathy and 50 healthy) randomized to the sequencing arm. We defined the quality thresholds for determining the need for Sanger confirmation. Finally, we examined the effort needed and new findings revealed by reanalyzing each genome (6–23 months after initial analysis; mean 13 months). Monogenic disease risk and carrier status were reported in 21% and 94% of participants, respectively. Only two participants had no monogenic disease risk or carrier status identified. For the PGx results (18 genotypes in six genes for five drugs), the identified diplotypes prompted recommendation for non-standard dosing of at least one of the analyzed drugs in 95% of participants. For blood antigen studies, we found that 31% of participants had a rare blood antigen genotype. In the cardiomyopathy cohort, an explanation for disease was identified in 48% of individuals. Over the course of the study, 14 variants were reclassified and, upon reanalysis, 18 new variants met criteria for reporting. These findings highlight the quantity of medically relevant findings from a broad analysis of genomic sequencing data as well as the need for periodic reinterpretation and reanalysis of data for both diagnostic indications and secondary findings.
- Published
- 2019
44. The Landscape of Somatic Genetic Alterations in Breast Cancers from CHEK2 Germline Mutation Carriers
- Author
-
Jorge S. Reis-Filho, Rajesh Kumar, David N Brown, Ozge Ceyhan-Birsoy, Jeremy Setton, Xin Pei, Pier Selenica, Diana Mandelker, Nadeem Riaz, Mark E. Robson, Larry Norton, Simon N. Powell, Britta Weigelt, Hannah Y Wen, and Sasi Arunachalam
- Subjects
Genetics ,0303 health sciences ,Cancer Research ,Somatic cell ,business.industry ,Cell cycle ,Brief Communication ,Phenotype ,Germline ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Oncology ,030220 oncology & carcinogenesis ,Medicine ,Allele ,business ,skin and connective tissue diseases ,CHEK2 ,030304 developmental biology - Abstract
Pathogenic germline variants in checkpoint kinase 2 (CHEK2), which plays pivotal roles in DNA damage response and cell cycle regulation, confer an increased breast cancer (BC) risk. Here, we investigated the phenotypic and genomic characteristics of 33 BCs from CHEK2 germline mutation carriers (16 high-risk variants and 17 low-risk p.Ile157Thr variants). CHEK2-associated BCs from patients with high-risk germline variants were largely hormone receptor-positive (87%, 13/15), and 81% (13/16) exhibited loss of heterozygosity (LOH) of the CHEK2 wild-type allele. Conversely, CHEK2-associated BCs from patients with the low-risk p.Ile157Thr variant displayed less-frequent loss of heterozygosity (5/17, 29%) and higher levels of CHEK2 protein expression than those with high-risk germline variants. CHEK2-associated BCs lacked a dominant mutational signature 3, a genomics feature of homologous recombination DNA repair deficiency (HRD). Our findings indicate that CHEK2-associated BCs are generally hormone receptor-positive and lack HRD-related mutational signatures, recapitulating the features of ATM-associated BCs. Specific CHEK2 germline variants may have a distinct impact on tumor biology.
- Published
- 2019
45. Activating mutations in CSF1R and additional receptor tyrosine kinases in histiocytic neoplasms
- Author
-
Sydney X. Lu, Benjamin H. Durham, Frederic Geissmann, Maria E. Arcila, Eli L. Diamond, Mariko Yabe, Jennifer Picarsic, Julien Haroche, David H. Abramson, Jaume Mora, Alexander Drilon, David M. Hyman, David B. Solit, Michelle Ki, Jean-François Emile, Caroline Erickson, Kseniya Petrova-Drus, Vicente Santa-María López, Veronica Rotemberg, Steven De Munck, Gary A. Ulaner, Erwin Pannecoucke, Omar Abdel-Wahab, Ozge Ceyhan-Birsoy, Estibaliz Lopez Rodrigo, Ruth Saganty, Akihide Yoshimi, Ira J. Dunkel, Mario E. Lacouture, Olivier Decaux, Marc Ladanyi, Michael Walsh, Alessandro Pastore, Diana Mandelker, Michael F. Berger, and Savvas N. Savvides
- Subjects
0301 basic medicine ,Male ,endocrine system diseases ,Pyridines ,Aminopyridines ,medicine.disease_cause ,Receptor tyrosine kinase ,0302 clinical medicine ,Anaplastic Lymphoma Kinase ,Receptor ,Child ,Picolinic Acids ,Mutation ,biology ,Kinase ,General Medicine ,Histiocytosis ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,030220 oncology & carcinogenesis ,Histiocytoses ,Child, Preschool ,Hematologic Neoplasms ,Female ,medicine.drug ,Adult ,Adolescent ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Young Adult ,Exome Sequencing ,medicine ,Humans ,Pyrroles ,Benzothiazoles ,Protein Kinase Inhibitors ,neoplasms ,Histiocyte ,Crizotinib ,Genome, Human ,Proto-Oncogene Proteins c-ret ,Infant ,Receptor Protein-Tyrosine Kinases ,Twins, Monozygotic ,medicine.disease ,030104 developmental biology ,biology.protein ,Cancer research ,Pyrazoles - Abstract
Histiocytoses are clonal hematopoietic disorders frequently driven by mutations mapping to the BRAF and MEK1 and MEK2 kinases. Currently, however, the developmental origins of histiocytoses in patients are not well understood, and clinically meaningful therapeutic targets outside of BRAF and MEK are undefined. In this study, we uncovered activating mutations in CSF1R and rearrangements in RET and ALK that conferred dramatic responses to selective inhibition of RET (selpercatinib) and crizotinib, respectively, in patients with histiocytosis.
- Published
- 2019
46. Next generation sequencing‐based copy number analysis reveals low prevalence of deletions and duplications in 46 genes associated with genetic cardiomyopathies
- Author
-
Ozge Ceyhan-Birsoy, Mark Bowser, Trevor J. Pugh, Lisa Mahanta, Ashley L. Frisella, Matthew S. Lebo, Sami S. Amr, Elizabeth Hynes, and Birgit Funke
- Subjects
0301 basic medicine ,Genetics ,Genetic heterogeneity ,Copy number analysis ,next‐generation sequencing ,Original Articles ,Biology ,Molecular diagnostics ,DNA sequencing ,genetic heterogeneity ,molecular diagnostics ,03 medical and health sciences ,Exon ,030104 developmental biology ,DNA copy number variants ,Original Article ,Copy-number variation ,False positive rate ,Cardiomyopathies ,Molecular Biology ,Gene ,Genetics (clinical) - Abstract
Background Diagnostic testing for genetic cardiomyopathies has undergone dramatic changes in the last decade with next generation sequencing (NGS) expanding the number of genes that can be interrogated simultaneously. Exon resolution copy number analysis is increasingly incorporated into routine diagnostic testing via cytogenomic arrays and more recently via NGS. While NGS is an attractive option for laboratories that have no access to array platforms, its higher false positive rate requires weighing the added cost incurred by orthogonal confirmation against the magnitude of the increase in diagnostic yield. Although copy number variants (CNVs) have been reported in various cardiomyopathy genes, their contribution has not been systematically studied. Methods We performed single exon resolution NGS‐based deletion/duplication analysis for up to 46 cardiomyopathy genes in >1400 individuals with cardiomyopathies including HCM, DCM, ARVC, RCM, and LVNC. Results and Conclusion Clinically significant deletions and duplications were identified in only 9 of 1425 (0.63%) individuals. The majority of those (6/9) represented intragenic events. We conclude that the added benefit of exon level deletion/duplication analysis is low for currently known cardiomyopathy genes and may not outweigh the increased cost and complexity of incorporating it into routine diagnostic testing for these disorders.
- Published
- 2015
47. Prevalence of Germline Mutations in Cancer Susceptibility Genes in Patients With Advanced Renal Cell Carcinoma
- Author
-
Carolyn Stewart, Kuo-Cheng Huang, Angela G. Arnold, Mark E. Robson, Darren R. Feldman, Devyn Taylor Coskey, Ying-Bei Chen, Nisha Pradhan, Liying Zhang, Andrea Knezevic, Robert J. Motzer, Maria I. Carlo, Yelena Kemel, Joseph Vijai, Martin H. Voss, Almedina Redzematovic, A. Ari Hakimi, Semanti Mukherjee, Sujata Patil, Diana Mandelker, Zsofia K. Stadler, David M. Hyman, Chung-Han Lee, Marc Ladanyi, Michael Walsh, Karen Cadoo, Kenneth Offit, Jonathan A. Coleman, and Ozge Ceyhan-Birsoy
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Population ,Gene mutation ,urologic and male genital diseases ,medicine.disease_cause ,Germline ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Germline mutation ,Internal medicine ,medicine ,Carcinoma ,Prevalence ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,education ,Carcinoma, Renal Cell ,Genetic Association Studies ,Germ-Line Mutation ,Genetic testing ,Aged ,Mutation ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Cancer ,Middle Aged ,medicine.disease ,Kidney Neoplasms ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,business - Abstract
Importance Identification of patients with hereditary renal cell carcinoma (RCC) is important for cancer screening and, in patients with advanced disease, for guiding treatment. The prevalence of cancer-related germline mutations in patients with advanced RCC and the phenotypes associated with some rare mutations are unknown. Objectives To examine the prevalence of germline mutations in both known RCC predisposition genes and other cancer-associated genes and to identify clinical and pathologic factors associated with germline mutations. Design, Setting, and Participants In this cohort study conducted from October 1, 2015, to July 31, 2017, 254 of 267 patients with advanced (American Joint Committee on Cancer stage III or IV) RCC who were seen in medical oncology or urology clinics agreed to germline sequencing and disclosure of results under an institutional protocol of matched tumor-germline DNA sequencing. Main Outcomes and Measures Mutation prevalence and spectrum in patients with advanced RCC were determined. Clinical characteristics were assessed by mutation status. Results Of the 254 patients (median age [range], 56 [13-79] years; 179 [70.5%] male; 211 [83.1%] non-Hispanic white), germline mutations were identified in 41 (16.1%); 14 (5.5%) had mutations in syndromic RCC-associated genes (7 inFH, 3 inBAP1, and 1 each inVHL,MET,SDHA,andSDHB). The most frequent mutations wereCHEK2(n = 9) andFH(n = 7). Of genes not previously associated with RCC risk,CHEK2was overrepresented in patients compared with the general population, with an odds ratio of RCC of 3.0 (95% CI, 1.3-5.8;P = .003). Patients with non–clear cell RCC were significantly more likely to have an RCC-associated gene mutation (9 [11.7%] of 74 vs 3 [1.7%] of 177;P = .001), and 8 (10.0%) had a mutation in a gene that could guide therapy. Of patients with mutations in RCC-associated genes, 5 (35.7%) failed to meet current clinical guidelines for genetic testing. Conclusions and Relevance Of patients with non–clear cell RCC, more than 20% had a germline mutation, of which half had the potential to direct systemic therapy. Current referral criteria for genetic testing did not identify a substantial portion of patients with mutations, supporting the role of a more inclusive sequencing approach.
- Published
- 2018
48. The BabySeq project: implementing genomic sequencing in newborns
- Author
-
Talia S. Schwartz, Peter J. Park, Kurt D. Christensen, Harvey L. Levy, Ozge Ceyhan-Birsoy, Timothy W. Yu, Richard B. Parad, Stacey Pereira, Alan H. Beggs, Leslie A. Frankel, Ingrid A. Holm, Amy L. McGuire, Pankaj B. Agrawal, Joel B. Krier, Rebecca C. LaMay, Robert C. Green, Shawn Fayer, Susan E. Waisbren, Casie A. Genetti, and Heidi L. Rehm
- Subjects
0301 basic medicine ,Newborn screening ,medicine.medical_specialty ,Ethical, legal, social implications ,Genetic Counseling ,030105 genetics & heredity ,Risk Assessment ,Pharmacogenomic Variants ,law.invention ,03 medical and health sciences ,Study Protocol ,Neonatal Screening ,Randomized controlled trial ,law ,Health care ,Exome Sequencing ,Methods ,Medicine ,Humans ,Family ,Genetic Predisposition to Disease ,Exome sequencing ,Newborn sequencing ,business.industry ,Genomic sequencing ,Medical record ,lcsh:RJ1-570 ,Whole exome sequencing ,Infant, Newborn ,lcsh:Pediatrics ,Health Care Costs ,3. Good health ,030104 developmental biology ,Family medicine ,Pediatrics, Perinatology and Child Health ,Randomized trial ,business ,Return of results - Abstract
The greatest opportunity for lifelong impact of genomic sequencing is during the newborn period. The “BabySeq Project” is a randomized trial that explores the medical, behavioral, and economic impacts of integrating genomic sequencing into the care of healthy and sick newborns. Families of newborns are enrolled from Boston Children’s Hospital and Brigham and Women’s Hospital nurseries, and half are randomized to receive genomic sequencing and a report that includes monogenic disease variants, recessive carrier variants for childhood onset or actionable disorders, and pharmacogenomic variants. All families participate in a disclosure session, which includes the return of results for those in the sequencing arm. Outcomes are collected through review of medical records and surveys of parents and health care providers and include the rationale for choice of genes and variants to report; what genomic data adds to the medical management of sick and healthy babies; and the medical, behavioral, and economic impacts of integrating genomic sequencing into the care of healthy and sick newborns. The BabySeq Project will provide empirical data about the risks, benefits and costs of newborn genomic sequencing and will inform policy decisions related to universal genomic screening of newborns. The study is registered in ClinicalTrials.gov Identifier: NCT02422511 . Registration date: 10 April 2015.
- Published
- 2018
49. NGS testing for cardiomyopathy: Utility of adding RASopathy-associated genes
- Author
-
Elizabeth Hynes, Maya M Miatkowski, Ozge Ceyhan-Birsoy, Birgit Funke, and Heather Mason-Suares
- Subjects
0301 basic medicine ,Cardiomyopathy, Dilated ,Male ,Proto-Oncogene Proteins B-raf ,Adolescent ,DNA Copy Number Variations ,Cardiomyopathy ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,RASopathy ,Biology ,Article ,03 medical and health sciences ,Genetics ,medicine ,Missense mutation ,Humans ,Genetic Predisposition to Disease ,Copy-number variation ,Genetics (clinical) ,Germ-Line Mutation ,Genetic testing ,Aged, 80 and over ,Mitogen-Activated Protein Kinase Kinases ,medicine.diagnostic_test ,Noonan Syndrome ,Hypertrophic cardiomyopathy ,High-Throughput Nucleotide Sequencing ,Infant ,Dilated cardiomyopathy ,Cardiomyopathy, Hypertrophic ,Middle Aged ,medicine.disease ,030104 developmental biology ,Noonan syndrome ,Female ,SOS1 Protein ,Signal Transduction - Abstract
RASopathies include a group of syndromes caused by pathogenic germline variants in RAS-MAPK pathway genes and typically present with facial dysmorphology, cardiovascular disease, and musculoskeletal anomalies. Recently, variants in RASopathy-associated genes have been reported in individuals with apparently nonsyndromic cardiomyopathy, suggesting that subtle features may be overlooked. To determine the utility and burden of adding RASopathy-associated genes to cardiomyopathy panels, we tested 11 RASopathy-associated genes by next-generation sequencing (NGS), including NGS-based copy number variant assessment, in 1,111 individuals referred for genetic testing for hypertrophic cardiomyopathy (HCM) or dilated cardiomyopathy (DCM). Disease-causing variants were identified in 0.6% (four of 692) of individuals with HCM, including three missense variants in the PTPN11, SOS1, and BRAF genes. Overall, 36 variants of uncertain significance (VUSs) were identified, averaging ∼3VUSs/100 cases. This study demonstrates that adding a subset of the RASopathy-associated genes to cardiomyopathy panels will increase clinical diagnoses without significantly increasing the number of VUSs/case.
- Published
- 2018
50. Germline SDHA mutations in children and adults with cancer
- Author
-
Diana Mandelker, Karen Cadoo, Sabine Topka, Yirong Li, Marianne Dubard Gault, Kıvanç Birsoy, Jennifer Kennedy, Neerav Shukla, Andrew L. Kung, Liying Zhang, Sowmaya Jairam, Jaclyn F. Hechtman, Shakeel Modak, Margaret Sheehan, Ozge Ceyhan-Birsoy, Wendy Kohlman, Michael Walsh, Kenneth Offit, Alicia Latham Schwark, Hikmat Al-Ahmadie, Alex Kentsis, Joseph Vijai, Erol C. Bayraktar, Ciyu Yang, Joshua D. Schiffman, Yelena Kemel, Vanessa Marcell, Ed Reznik, Renzo G. DiNatale, Zsofia K. Stadler, Ester Borras Flores, Carolyn Stewart, Stephen S. Roberts, Angela G. Arnold, Mark E. Robson, Deborah DeLair, Maria I. Carlo, and Beth Siegel
- Subjects
0301 basic medicine ,Research Report ,Adult ,Male ,Adolescent ,SDHA ,clear cell renal cell carcinoma ,medicine.disease_cause ,Germline ,Loss of heterozygosity ,03 medical and health sciences ,neuroblastoma ,0302 clinical medicine ,Germline mutation ,Gene Frequency ,neoplasm of the breast ,Cell Line, Tumor ,Neoplasms ,medicine ,Humans ,Stromal tumor ,Child ,neoplasms ,Germ-Line Mutation ,GiST ,gastrointestinal stroma tumor ,business.industry ,Electron Transport Complex II ,Cancer ,General Medicine ,Middle Aged ,medicine.disease ,3. Good health ,030104 developmental biology ,HEK293 Cells ,030220 oncology & carcinogenesis ,Child, Preschool ,Cancer research ,Female ,business ,Carcinogenesis - Abstract
Mutations in succinate dehydrogenase complex genes predispose to familial paraganglioma-pheochromocytoma syndrome (FPG) and gastrointestinal stromal tumors (GIST). Here we describe cancer patients undergoing agnostic germline testing at Memorial Sloan Kettering Cancer Center and found to harbor germline SDHA mutations. Using targeted sequencing covering the cancer census genes, we identified 10 patients with SDHA germline mutations. Cancer diagnoses for these patients carrying SDHA germline mutations included neuroblastoma (n = 1), breast (n = 1), colon (n = 1), renal (n = 1), melanoma and uterine (n = 1), prostate (n = 1), endometrial (n = 1), bladder (n = 1), and gastrointestinal stromal tumor (GIST) (n = 2). Immunohistochemical staining and assessment of patient tumors for second hits and loss of heterozygosity in SDHA confirmed GIST as an SDHA-associated tumor and suggests SDHA germline mutations may be a driver in neuroblastoma tumorigenesis.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.