357 results on '"Mary-Claire King"'
Search Results
2. Cisplatin +/− rucaparib after preoperative chemotherapy in patients with triple-negative or BRCA mutated breast cancer
- Author
-
Maitri Kalra, Yan Tong, David R. Jones, Tom Walsh, Michael A. Danso, Cynthia X. Ma, Paula Silverman, Mary-Claire King, Sunil S. Badve, Susan M. Perkins, and Kathy D. Miller
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Patients with triple-negative breast cancer (TNBC) who have residual disease after neoadjuvant therapy have a high risk of recurrence. We tested the impact of DNA-damaging chemotherapy alone or with PARP inhibition in this high-risk population. Patients with TNBC or deleterious BRCA mutation (TNBC/BRCAmut) who had >2 cm of invasive disease in the breast or persistent lymph node (LN) involvement after neoadjuvant therapy were assigned 1:1 to cisplatin alone or with rucaparib. Germline mutations were identified with BROCA analysis. The primary endpoint was 2-year disease-free survival (DFS) with 80% power to detect an HR 0.5. From Feb 2010 to May 2013, 128 patients were enrolled. Median tumor size at surgery was 1.9 cm (0–11.5 cm) with 1 (0–38) involved LN; median Residual Cancer Burden (RCB) score was 2.6. Six patients had known deleterious BRCA1 or BRCA2 mutations at study entry, but BROCA identified deleterious mutations in 22% of patients with available samples. Toxicity was similar in both arms. Despite frequent dose reductions (21% of patients) and delays (43.8% of patients), 73% of patients completed planned cisplatin. Rucaparib exposure was limited with median concentration 275 (82–4694) ng/mL post-infusion on day 3. The addition of rucaparib to cisplatin did not increase 2-year DFS (54.2% cisplatin vs. 64.1% cisplatin + rucaparib; P = 0.29). In the high-risk post preoperative TNBC/BRCAmut setting, the addition of low-dose rucaparib did not improve 2-year DFS or increase the toxicity of cisplatin. Genetic testing was underutilized in this high-risk population.
- Published
- 2021
- Full Text
- View/download PDF
3. Inherited predisposition to breast cancer in the Carolina Breast Cancer Study
- Author
-
Tom Walsh, Suleyman Gulsuner, Ming K. Lee, Melissa A. Troester, Andrew F. Olshan, H. Shelton Earp, Charles M. Perou, and Mary-Claire King
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract The Carolina Breast Cancer Study (CBCS) phases I–II was a case-control study of biological and social risk factors for invasive breast cancer that enrolled cases and controls between 1993 and 1999. Case selection was population-based and stratified by ancestry and age at diagnosis. Controls were matched to cases by age, self-identified race, and neighborhood of residence. Sequencing genomic DNA from 1370 cases and 1635 controls yielded odds ratios (with 95% confidence limits) for breast cancer of all subtypes of 26.7 (3.59, 189.1) for BRCA1, 8.8 (3.44, 22.48) for BRCA2, and 9.0 (2.06, 39.60) for PALB2; and for triple-negative breast cancer (TNBC) of 55.0 (7.01, 431.4) for BRCA1, 12.1 (4.18, 35.12) for BRCA2, and 10.8 (1.97, 59.11) for PALB2. Overall, 5.6% of patients carried a pathogenic variant in BRCA1, BRCA2, PALB2, or TP53, the four most highly penetrant breast cancer genes. Analysis of cases by tumor subtype revealed the expected association of TNBC versus other tumor subtypes with BRCA1, and suggested a significant association between TNBC versus other tumor subtypes with BRCA2 or PALB2 among African-American (AA) patients [2.95 (1.18, 7.37)], but not among European-American (EA) patients [0.62 (0.18, 2.09)]. AA patients with pathogenic variants in BRCA2 or PALB2 were 11 times more likely to be diagnosed with TNBC versus another tumor subtype than were EA patients with pathogenic variants in either of these genes (P = 0.001). If this pattern is confirmed in other comparisons of similarly ascertained AA and EA breast cancer patients, it could in part explain the higher prevalence of TNBC among AA breast cancer patients.
- Published
- 2021
- Full Text
- View/download PDF
4. Effects of germline and somatic events in candidate BRCA-like genes on breast-tumor signatures.
- Author
-
Weston R Bodily, Brian H Shirts, Tom Walsh, Suleyman Gulsuner, Mary-Claire King, Alyssa Parker, Moom Roosan, and Stephen R Piccolo
- Subjects
Medicine ,Science - Abstract
Mutations in BRCA1 and BRCA2 cause deficiencies in homologous recombination repair (HR), resulting in repair of DNA double-strand breaks by the alternative non-homologous end-joining pathway, which is more error prone. HR deficiency of breast tumors is important because it is associated with better responses to platinum salt therapies and PARP inhibitors. Among other consequences of HR deficiency are characteristic somatic-mutation signatures and gene-expression patterns. The term "BRCA-like" (or "BRCAness") describes tumors that harbor an HR defect but have no detectable germline mutation in BRCA1 or BRCA2. A better understanding of the genes and molecular events associated with tumors being BRCA-like could provide mechanistic insights and guide development of targeted treatments. Using data from The Cancer Genome Atlas (TCGA) for 1101 breast-cancer patients, we identified individuals with a germline mutation, somatic mutation, homozygous deletion, and/or hypermethylation event in BRCA1, BRCA2, and 59 other cancer-predisposition genes. Based on the assumption that BRCA-like events would have similar downstream effects on tumor biology as BRCA1/BRCA2 germline mutations, we quantified these effects based on somatic-mutation signatures and gene-expression profiles. We reduced the dimensionality of the somatic-mutation signatures and expression data and used a statistical resampling approach to quantify similarities among patients who had a BRCA1/BRCA2 germline mutation, another type of aberration in BRCA1 or BRCA2, or any type of aberration in one of the other genes. Somatic-mutation signatures of tumors having a non-germline aberration in BRCA1/BRCA2 (n = 80) were generally similar to each other and to tumors from BRCA1/BRCA2 germline carriers (n = 44). Additionally, somatic-mutation signatures of tumors with germline or somatic events in ATR (n = 16) and BARD1 (n = 8) showed high similarity to tumors from BRCA1/BRCA2 carriers. Other genes (CDKN2A, CTNNA1, PALB2, PALLD, PRSS1, SDHC) also showed high similarity but only for a small number of events or for a single event type. Tumors with germline mutations or hypermethylation of BRCA1 had relatively similar gene-expression profiles and overlapped considerably with the Basal-like subtype; but the transcriptional effects of the other events lacked consistency. Our findings confirm previously known relationships between molecular signatures and germline or somatic events in BRCA1/BRCA2. Our methodology represents an objective way to identify genes that have similar downstream effects on molecular signatures when mutated, deleted, or hypermethylated.
- Published
- 2020
- Full Text
- View/download PDF
5. Genetic features of myelodysplastic syndrome and aplastic anemia in pediatric and young adult patients
- Author
-
Siobán B. Keel, Angela Scott, Marilyn Sanchez-Bonilla, Phoenix A. Ho, Suleyman Gulsuner, Colin C. Pritchard, Janis L. Abkowitz, Mary-Claire King, Tom Walsh, and Akiko Shimamura
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
The clinical and histopathological distinctions between inherited versus acquired bone marrow failure and myelodysplastic syndromes are challenging. The identification of inherited bone marrow failure/myelodysplastic syndromes is critical to inform appropriate clinical management. To investigate whether a subset of pediatric and young adults undergoing transplant for aplastic anemia or myelodysplastic syndrome have germline mutations in bone marrow failure/myelodysplastic syndrome genes, we performed a targeted genetic screen of samples obtained between 1990–2012 from children and young adults with aplastic anemia or myelodysplastic syndrome transplanted at the Fred Hutchinson Cancer Research Center. Mutations in inherited bone marrow failure/myelodysplastic syndrome genes were found in 5.1% (5/98) of aplastic anemia patients and 13.6% (15/110) of myelodysplastic syndrome patients. While the majority of mutations were constitutional, a RUNX1 mutation present in the peripheral blood at a 51% variant allele fraction was confirmed to be somatically acquired in one myelodysplastic syndrome patient. This highlights the importance of distinguishing germline versus somatic mutations by sequencing DNA from a second tissue or from parents. Pathological mutations were present in DKC1, MPL, and TP53 among the aplastic anemia cohort, and in FANCA, GATA2, MPL, RTEL1, RUNX1, SBDS, TERT, TINF2, and TP53 among the myelodysplastic syndrome cohort. Family history or physical examination failed to reliably predict the presence of germline mutations. This study shows that while any single specific bone marrow failure/myelodysplastic syndrome genetic disorder is rare, screening for these disorders in aggregate identifies a significant subset of patients with inherited bone marrow failure/myelodysplastic syndrome.
- Published
- 2016
- Full Text
- View/download PDF
6. Genomic analysis of bone marrow failure and myelodysplastic syndromes reveals phenotypic and diagnostic complexity
- Author
-
Michael Y. Zhang, Siobán B. Keel, Tom Walsh, Ming K. Lee, Suleyman Gulsuner, Amanda C. Watts, Colin C. Pritchard, Stephen J. Salipante, Michael R. Jeng, Inga Hofmann, David A. Williams, Mark D. Fleming, Janis L. Abkowitz, Mary-Claire King, and Akiko Shimamura
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Accurate and timely diagnosis of inherited bone marrow failure and inherited myelodysplastic syndromes is essential to guide clinical management. Distinguishing inherited from acquired bone marrow failure/myelodysplastic syndrome poses a significant clinical challenge. At present, diagnostic genetic testing for inherited bone marrow failure/myelodysplastic syndrome is performed gene-by-gene, guided by clinical and laboratory evaluation. We hypothesized that standard clinically-directed genetic testing misses patients with cryptic or atypical presentations of inherited bone marrow failure/myelodysplastic syndrome. In order to screen simultaneously for mutations of all classes in bone marrow failure/myelodysplastic syndrome genes, we developed and validated a panel of 85 genes for targeted capture and multiplexed massively parallel sequencing. In patients with clinical diagnoses of Fanconi anemia, genomic analysis resolved subtype assignment, including those of patients with inconclusive complementation test results. Eight out of 71 patients with idiopathic bone marrow failure or myelodysplastic syndrome were found to harbor damaging germline mutations in GATA2, RUNX1, DKC1, or LIG4. All 8 of these patients lacked classical clinical stigmata or laboratory findings of these syndromes and only 4 had a family history suggestive of inherited disease. These results reflect the extensive genetic heterogeneity and phenotypic complexity of bone marrow failure/myelodysplastic syndrome phenotypes. This study supports the integration of broad unbiased genetic screening into the diagnostic workup of children and young adults with bone marrow failure and myelodysplastic syndromes.
- Published
- 2015
- Full Text
- View/download PDF
7. Evidence is evidence: an interview with Mary-Claire King. Interviewed by Jane Gitschier.
- Author
-
Mary-Claire King
- Subjects
Genetics ,QH426-470 - Published
- 2013
- Full Text
- View/download PDF
8. A genomewide screen for suppressors of Alu-mediated rearrangements reveals a role for PIF1.
- Author
-
Karen M Chisholm, Sarah D Aubert, Krister P Freese, Virginia A Zakian, Mary-Claire King, and Piri L Welcsh
- Subjects
Medicine ,Science - Abstract
Alu-mediated rearrangement of tumor suppressor genes occurs frequently during carcinogenesis. In breast cancer, this mechanism contributes to loss of the wild-type BRCA1 allele in inherited disease and to loss of heterozygosity in sporadic cancer. To identify genes required for suppression of Alu-mediated recombination we performed a genomewide screen of a collection of 4672 yeast gene deletion mutants using a direct repeat recombination assay. The primary screen and subsequent analysis identified 12 candidate genes including TSA, ELG1, and RRM3, which are known to play a significant role in maintaining genomic stability. Genetic analysis of the corresponding human homologs was performed in sporadic breast tumors and in inherited BRCA1-associated carcinomas. Sequencing of these genes in high risk breast cancer families revealed a potential role for the helicase PIF1 in cancer predisposition. PIF1 variant L319P was identified in three breast cancer families; importantly, this variant, which is predicted to be functionally damaging, was not identified in a large series of controls nor has it been reported in either dbSNP or the 1000 Genomes Project. In Schizosaccharomyces pombe, Pfh1 is required to maintain both mitochondrial and nuclear genomic integrity. Functional studies in yeast of human PIF1 L319P revealed that this variant cannot complement the essential functions of Pfh1 in either the nucleus or mitochondria. Our results provide a global view of nonessential genes involved in suppressing Alu-mediated recombination and implicate variation in PIF1 in breast cancer predisposition.
- Published
- 2012
- Full Text
- View/download PDF
9. Clinically Complex LRBA Deficiency Due to a Founder Allele in the Georgian Jewish Population
- Author
-
Tal Freund, Sarah K. Baxter, Tom Walsh, Hana Golan, Joseph Kapelushnik, Michal Abramsohn-Goldenberg, Shira Benor, Nadav Sarid, Ron Ram, Yifat Alcalay, Reeval Segel, Paul Renbaum, Polina Stepensky, Mary-Claire King, Troy R. Torgerson, and David Hagin
- Subjects
Immunology ,Immunology and Allergy - Abstract
Pathogenic variants in LRBA, encoding the LPS Responsive Beige-Like Anchor (LRBA) protein, are responsible for recessive, early-onset hypogammaglobulinemia, severe multi-organ autoimmunity, and lymphoproliferation, with increased risk for malignancy. LRBA deficiency has a wide clinical spectrum with variable age of onset and disease severity. Three apparently unrelated patients with LRBA deficiency, of Georgian Jewish descent, were homozygous for LRBA c.6640C T, p.R2214*, leading to a stop upstream of the LRBA BEACH domain. Despite carrying the same LRBA genotype, the three patients differed in clinical course: the first patient was asymptomatic until age 25 years; the second presented with failure to thrive at age 3 months; and the third presented at age 7 years with immune cytopenias and severe infections. Two of the patients developed malignancies: the first patient was diagnosed with recurrent Hodgkin's disease at age 36 years, and the second patient developed aggressive gastric cancer at age 15 years. Among Georgian Jews, the carrier frequency of the LRBA p.R2214* allele was 1.6% (4 of 236 Georgian Jewish controls). The allele was absent from other populations. Haplotype analysis showed a shared origin of the mutation. These three patients revealed a pathogenic LRBA founder allele in the Georgian Jewish population, support the diverse and complex clinical spectrum of LRBA deficiency, and support the possibility that LRBA deficiency predisposes to malignancy.
- Published
- 2022
10. A paradoxical genotype-phenotype relationship: Low level of GOSR2 translation from a non-AUG start codon in a family with profound hearing loss
- Author
-
Amal Aburayyan, Ryan J Carlson, Grace N Rabie, Ming K Lee, Suleyman Gulsuner, Tom Walsh, Karen B Avraham, Moien N Kanaan, and Mary-Claire King
- Subjects
Genetics ,General Medicine ,Molecular Biology ,Genetics (clinical) - Published
- 2023
11. Table S1 from Mutations in Homologous Recombination Genes and Outcomes in Ovarian Carcinoma Patients in GOG 218: An NRG Oncology/Gynecologic Oncology Group Study
- Author
-
Elizabeth M. Swisher, Michael J. Birrer, Mary Claire King, Nilsa C. Ramirez, Heather A. Lankes, John Soper, Stephen Rubin, Cheryl Bailey, Gretchen Glaser, Robert S. Mannel, Floor Backes, Krishnansu S. Tewari, Robert A. Burger, Silvia Casadei, Sarah S. Bernards, Suleyman Gulsuner, Ming K. Lee, Tom Walsh, Maria I. Harrell, Mark F. Brady, and Barbara M. Norquist
- Abstract
Table of non-BRCA mutations with clinical characteristics
- Published
- 2023
12. Supplementary Table 4 from Germline and Somatic Mutations in Homologous Recombination Genes Predict Platinum Response and Survival in Ovarian, Fallopian Tube, and Peritoneal Carcinomas
- Author
-
Elizabeth M. Swisher, Mary-Claire King, Rochelle L. Garcia, Sheena Scroggins, Colin C. Pritchard, Kathy J. Agnew, Alexander S. Nord, Silvia Casadei, Barbara M. Norquist, Anne Thornton, Mara H. Rendi, Christopher C. Pennil, Ming K. Lee, Maria I. Harrell, Tom Walsh, and Kathryn P. Pennington
- Abstract
PDF file 48K, Genomic regions for targeted capture
- Published
- 2023
13. Figure S1, Table S2 from Mutations in Homologous Recombination Genes and Outcomes in Ovarian Carcinoma Patients in GOG 218: An NRG Oncology/Gynecologic Oncology Group Study
- Author
-
Elizabeth M. Swisher, Michael J. Birrer, Mary Claire King, Nilsa C. Ramirez, Heather A. Lankes, John Soper, Stephen Rubin, Cheryl Bailey, Gretchen Glaser, Robert S. Mannel, Floor Backes, Krishnansu S. Tewari, Robert A. Burger, Silvia Casadei, Sarah S. Bernards, Suleyman Gulsuner, Ming K. Lee, Tom Walsh, Maria I. Harrell, Mark F. Brady, and Barbara M. Norquist
- Abstract
Survival by mutation type, and mutation data by the source of sequenced DNA
- Published
- 2023
14. Table S1, Table S2, Table S3; source file format .docx from Intensive Surveillance with Biannual Dynamic Contrast-Enhanced Magnetic Resonance Imaging Downstages Breast Cancer in BRCA1 Mutation Carriers
- Author
-
Olufunmilayo I. Olopade, Dezheng Huo, Gillian M. Newstead, Gregory Karczmar, Mary-Claire King, Colin C. Pritchard, Marcio Debiasi, Xiaoming Wang, Nora Jaskowiak, Robert Livingston, Susan Hong, Iris L. Romero, Hongyuan Cao, Akila Raoul, Fang Liu, Galina Khramtsova, Jeffrey Mueller, Elias Obeid, Jane Churpek, Angela R. Bradbury, Marion S. Verp, Deepa Sheth, Kirti Kulkarni, David Schacht, Tom Walsh, Toshio F. Yoshimatsu, Kristen Whitaker, Hiroyuki Abe, Yonglan Zheng, and Rodrigo Santa Cruz Guindalini
- Abstract
Table S1. Specifics of DCE-MRI Techniques Used; Table S2. Pathogenic mutations identified in study participants; Table S3. Patients' reasons of withdrawls from the study within 5 years
- Published
- 2023
15. Supplementary Table 3 from Germline and Somatic Mutations in Homologous Recombination Genes Predict Platinum Response and Survival in Ovarian, Fallopian Tube, and Peritoneal Carcinomas
- Author
-
Elizabeth M. Swisher, Mary-Claire King, Rochelle L. Garcia, Sheena Scroggins, Colin C. Pritchard, Kathy J. Agnew, Alexander S. Nord, Silvia Casadei, Barbara M. Norquist, Anne Thornton, Mara H. Rendi, Christopher C. Pennil, Ming K. Lee, Maria I. Harrell, Tom Walsh, and Kathryn P. Pennington
- Abstract
PDF file 69K, Paired primary and recurrent carcinomas
- Published
- 2023
16. Data from Germline and Somatic Mutations in Homologous Recombination Genes Predict Platinum Response and Survival in Ovarian, Fallopian Tube, and Peritoneal Carcinomas
- Author
-
Elizabeth M. Swisher, Mary-Claire King, Rochelle L. Garcia, Sheena Scroggins, Colin C. Pritchard, Kathy J. Agnew, Alexander S. Nord, Silvia Casadei, Barbara M. Norquist, Anne Thornton, Mara H. Rendi, Christopher C. Pennil, Ming K. Lee, Maria I. Harrell, Tom Walsh, and Kathryn P. Pennington
- Abstract
Purpose: Hallmarks of germline BRCA1/2-associated ovarian carcinomas include chemosensitivity and improved survival. The therapeutic impact of somatic BRCA1/2 mutations and mutations in other homologous recombination DNA repair genes is uncertain.Experimental Design: Using targeted capture and massively parallel genomic sequencing, we assessed 390 ovarian carcinomas for germline and somatic loss-of-function mutations in 30 genes, including BRCA1, BRCA2, and 11 other genes in the homologous recombination pathway.Results: Thirty-one percent of ovarian carcinomas had a deleterious germline (24%) and/or somatic (9%) mutation in one or more of the 13 homologous recombination genes: BRCA1, BRCA2, ATM, BARD1, BRIP1, CHEK1, CHEK2, FAM175A, MRE11A, NBN, PALB2, RAD51C, and RAD51D. Nonserous ovarian carcinomas had similar rates of homologous recombination mutations to serous carcinomas (28% vs. 31%, P = 0.6), including clear cell, endometrioid, and carcinosarcoma. The presence of germline and somatic homologous recombination mutations was highly predictive of primary platinum sensitivity (P = 0.0002) and improved overall survival (P = 0.0006), with a median overall survival of 66 months in germline homologous recombination mutation carriers, 59 months in cases with a somatic homologous recombination mutation, and 41 months for cases without a homologous recombination mutation.Conclusions: Germline or somatic mutations in homologous recombination genes are present in almost one third of ovarian carcinomas, including both serous and nonserous histologies. Somatic BRCA1/2 mutations and mutations in other homologous recombination genes have a similar positive impact on overall survival and platinum responsiveness as germline BRCA1/2 mutations. The similar rate of homologous recombination mutations in nonserous carcinomas supports their inclusion in PARP inhibitor clinical trials. Clin Cancer Res; 20(3); 764–75. ©2013 AACR.
- Published
- 2023
17. Supplementary Table 2 from Germline and Somatic Mutations in Homologous Recombination Genes Predict Platinum Response and Survival in Ovarian, Fallopian Tube, and Peritoneal Carcinomas
- Author
-
Elizabeth M. Swisher, Mary-Claire King, Rochelle L. Garcia, Sheena Scroggins, Colin C. Pritchard, Kathy J. Agnew, Alexander S. Nord, Silvia Casadei, Barbara M. Norquist, Anne Thornton, Mara H. Rendi, Christopher C. Pennil, Ming K. Lee, Maria I. Harrell, Tom Walsh, and Kathryn P. Pennington
- Abstract
PDF file 51K, Cases with both germline and somatic HR mutations
- Published
- 2023
18. Supplementary Table 1 from Germline and Somatic Mutations in Homologous Recombination Genes Predict Platinum Response and Survival in Ovarian, Fallopian Tube, and Peritoneal Carcinomas
- Author
-
Elizabeth M. Swisher, Mary-Claire King, Rochelle L. Garcia, Sheena Scroggins, Colin C. Pritchard, Kathy J. Agnew, Alexander S. Nord, Silvia Casadei, Barbara M. Norquist, Anne Thornton, Mara H. Rendi, Christopher C. Pennil, Ming K. Lee, Maria I. Harrell, Tom Walsh, and Kathryn P. Pennington
- Abstract
PDF file 80K, Cases with deleterious germline mutations, somatic HR mutations, and somatic PTEN mutations
- Published
- 2023
19. Data from Intensive Surveillance with Biannual Dynamic Contrast-Enhanced Magnetic Resonance Imaging Downstages Breast Cancer in BRCA1 Mutation Carriers
- Author
-
Olufunmilayo I. Olopade, Dezheng Huo, Gillian M. Newstead, Gregory Karczmar, Mary-Claire King, Colin C. Pritchard, Marcio Debiasi, Xiaoming Wang, Nora Jaskowiak, Robert Livingston, Susan Hong, Iris L. Romero, Hongyuan Cao, Akila Raoul, Fang Liu, Galina Khramtsova, Jeffrey Mueller, Elias Obeid, Jane Churpek, Angela R. Bradbury, Marion S. Verp, Deepa Sheth, Kirti Kulkarni, David Schacht, Tom Walsh, Toshio F. Yoshimatsu, Kristen Whitaker, Hiroyuki Abe, Yonglan Zheng, and Rodrigo Santa Cruz Guindalini
- Abstract
Purpose:To establish a cohort of high-risk women undergoing intensive surveillance for breast cancer.Experimental Design: We performed dynamic contrast-enhanced MRI every 6 months in conjunction with annual mammography (MG). Eligible participants had a cumulative lifetime breast cancer risk ≥20% and/or tested positive for a pathogenic mutation in a known breast cancer susceptibility gene.Results:Between 2004 and 2016, we prospectively enrolled 295 women, including 157 mutation carriers (75 BRCA1, 61 BRCA2); participants' mean age at entry was 43.3 years. Seventeen cancers were later diagnosed: 4 ductal carcinoma in situ (DCIS) and 13 early-stage invasive breast cancers. Fifteen cancers occurred in mutation carriers (11 BRCA1, 3 BRCA2, 1 CDH1). Median size of the invasive cancers was 0.61 cm. No patients had lymph node metastasis at time of diagnosis, and no interval invasive cancers occurred. The sensitivity of biannual MRI alone was 88.2% and annual MG plus biannual MRI was 94.1%. The cancer detection rate of biannual MRI alone was 0.7% per 100 screening episodes, which is similar to the cancer detection rate of 0.7% per 100 screening episodes for annual MG plus biannual MRI. The number of recalls and biopsies needed to detect one cancer by biannual MRI were 2.8 and 1.7 in BRCA1 carriers, 12.0 and 8.0 in BRCA2 carriers, and 11.7 and 5.0 in non-BRCA1/2 carriers, respectively.Conclusions:Biannual MRI performed well for early detection of invasive breast cancer in genomically stratified high-risk women. No benefit was associated with annual MG screening plus biannual MRI screening.See related commentary by Kuhl and Schrading, p. 1693
- Published
- 2023
20. Data from Mutations in Homologous Recombination Genes and Outcomes in Ovarian Carcinoma Patients in GOG 218: An NRG Oncology/Gynecologic Oncology Group Study
- Author
-
Elizabeth M. Swisher, Michael J. Birrer, Mary Claire King, Nilsa C. Ramirez, Heather A. Lankes, John Soper, Stephen Rubin, Cheryl Bailey, Gretchen Glaser, Robert S. Mannel, Floor Backes, Krishnansu S. Tewari, Robert A. Burger, Silvia Casadei, Sarah S. Bernards, Suleyman Gulsuner, Ming K. Lee, Tom Walsh, Maria I. Harrell, Mark F. Brady, and Barbara M. Norquist
- Abstract
Purpose: We hypothesized that mutations in homologous recombination repair (HRR) genes beyond BRCA1 and BRCA2 improve outcomes for ovarian carcinoma patients treated with platinum therapy and would impact the relative benefit of adding prolonged bevacizumab.Experimental Design: We sequenced DNA from blood and/or neoplasm from 1,195 women enrolled in GOG-0218, a randomized phase III trial in advanced ovarian carcinoma of bevacizumab added to carboplatin and paclitaxel. Defects in HRR were defined as damaging mutations in 16 genes. Proportional hazards models were used to estimate relative hazards for progression-free survival (PFS) and overall survival (OS).Results: Of 1,195 women with ovarian carcinoma, HRR mutations were identified in 307 (25.7%). Adjusted hazards for progression and death compared with those without mutations were lower for women with non-BRCA HRR mutations [HR = 0.73; 95% confidence interval (CI), 0.57–0.94; P = 0.01 for PFS; HR = 0.67; 95% CI, 0.50–0.90; P = 0.007 for OS] and BRCA1 mutations (HR = 0.80; 95% CI, 0.66–0.97; P = 0.02 for PFS; HR = 0.74; 95% CI, 0.59–0.94; P = 0.01 for OS) and were lowest for BRCA2 mutations (HR = 0.52; 95% CI, 0.40–0.67; P < 0.0001 for PFS; HR = 0.36; 95% CI, 0.25–0.53; P < 0.0001 for OS). A test of interaction showed no difference in the effect of bevacizumab on PFS between cases with and without mutations.Conclusions: HRR mutations, including non-BRCA genes, significantly prolong PFS and OS in ovarian carcinoma and should be stratified for in clinical trials. The benefit of adding bevacizumab was not significantly modified by mutation status. Clin Cancer Res; 24(4); 777–83. ©2017 AACR.
- Published
- 2023
21. Figure S1, Figure S2, Figure S3; source file format .docx from Intensive Surveillance with Biannual Dynamic Contrast-Enhanced Magnetic Resonance Imaging Downstages Breast Cancer in BRCA1 Mutation Carriers
- Author
-
Olufunmilayo I. Olopade, Dezheng Huo, Gillian M. Newstead, Gregory Karczmar, Mary-Claire King, Colin C. Pritchard, Marcio Debiasi, Xiaoming Wang, Nora Jaskowiak, Robert Livingston, Susan Hong, Iris L. Romero, Hongyuan Cao, Akila Raoul, Fang Liu, Galina Khramtsova, Jeffrey Mueller, Elias Obeid, Jane Churpek, Angela R. Bradbury, Marion S. Verp, Deepa Sheth, Kirti Kulkarni, David Schacht, Tom Walsh, Toshio F. Yoshimatsu, Kristen Whitaker, Hiroyuki Abe, Yonglan Zheng, and Rodrigo Santa Cruz Guindalini
- Abstract
Figure S1. Study protocol design; Figure S2. MRI compliance during the first 5 years of study; Figure S3. Recall and biopsy rates by imaging modality
- Published
- 2023
22. Supplementary Tables 1-4, Figure 1 from Contribution of Inherited Mutations in the BRCA2-Interacting Protein PALB2 to Familial Breast Cancer
- Author
-
Mary-Claire King, John A. Stamatoyannopoulos, Ming K. Lee, Jessica B. Mandell, Sunday Stray, Tom Walsh, Barbara M. Norquist, and Silvia Casadei
- Abstract
Supplementary Tables 1-4, Figure 1 from Contribution of Inherited Mutations in the BRCA2-Interacting Protein PALB2 to Familial Breast Cancer
- Published
- 2023
23. Data from Contribution of Inherited Mutations in the BRCA2-Interacting Protein PALB2 to Familial Breast Cancer
- Author
-
Mary-Claire King, John A. Stamatoyannopoulos, Ming K. Lee, Jessica B. Mandell, Sunday Stray, Tom Walsh, Barbara M. Norquist, and Silvia Casadei
- Abstract
Inherited mutations in the BRCA2-interacting protein PALB2 are known to be associated with increased risks of developing breast cancer. To evaluate the contribution of PALB2 to familial breast cancer in the United States, we sequenced the coding sequences and flanking regulatory regions of the gene from constitutional genomic DNA of 1,144 familial breast cancer patients with wild-type sequences at BRCA1 and BRCA2. Overall, 3.4% (33/972) of patients not selected by ancestry and 0% (0/172) of patients specifically of Ashkenazi Jewish ancestry were heterozygous for a nonsense, frameshift, or frameshift-associated splice mutation in PALB2. Mutations were detected in both male and female breast cancer patients. All mutations were individually rare: the 33 heterozygotes harbored 13 different mutations, 5 previously reported and 8 novel mutations. PALB2 heterozygotes were 4-fold more likely to have a male relative with breast cancer (P = 0.0003), 6-fold more likely to have a relative with pancreatic cancer (P = 0.002), and 1.3-fold more likely to have a relative with ovarian cancer (P = 0.18). Compared with their female relatives without mutations, increased risk of developing breast cancer for female PALB2 heterozygotes was 2.3-fold (95% CI: 1.5–4.2) by age 55 and 3.4-fold (95% CI: 2.4–5.9) by age 85. Loss of the wild-type PALB2 allele was observed in laser-dissected tumor specimens from heterozygous patients. Given this mutation prevalence and risk, consideration might be given to clinical testing of PALB2 by complete genomic sequencing for familial breast cancer patients with wild-type sequences at BRCA1 and BRCA2. Cancer Res; 71(6); 2222–9. ©2011 AACR.
- Published
- 2023
24. Mutational spectrum of breast cancer susceptibility genes among women ascertained in a cancer risk clinic in Northeast Brazil
- Author
-
Gabriela E. S. Felix, Rodrigo Santa Cruz Guindalini, Yonglan Zheng, Tom Walsh, Elisabeth Sveen, Taisa Manuela Machado Lopes, Juliana Côrtes, Jing Zhang, Polyanna Carôzo, Irlânia Santos, Thaís Ferreira Bonfim, Bernardo Garicochea, Maria Betânia Pereira Toralles, Roberto Meyer, Eduardo Martins Netto, Kiyoko Abe-Sandes, Mary-Claire King, Ivana Lucia de Oliveira Nascimento, and Olufunmilayo I. Olopade
- Subjects
BRCA2 Protein ,Cancer Research ,Oncology ,BRCA1 Protein ,Genes, BRCA2 ,Mutation ,DNA Helicases ,Humans ,Breast Neoplasms ,Female ,Genetic Predisposition to Disease ,Brazil ,Germ-Line Mutation - Abstract
Purpose There is a paucity of data on the spectrum and prevalence of pathogenic variants among women of African ancestry in the Northeast region of Brazil. Methods We performed BROCA panel sequencing to identify inherited loss-of-function variants in breast cancer susceptibility genes among 292 Brazilian women referred to a single institution cancer risk assessment program. Results The study included a convenient cohort of 173 women with invasive breast cancer (cases) and 119 women who were cancer-free at the time of ascertainment. The majority of the women self-reported as African-descended (67% for cases and 90.8% for unaffected volunteers). Thirty-seven pathogenic variants were found in 36 (20.8%) patients. While the spectrum of pathogenic variants was heterogeneous, the majority (70.3%) of the pathogenic variants were detected in high-risk genes BRCA1, BRCA2, PALB2, and TP53. Pathogenic variants were also found in the ATM, BARD1, BRIP1, FAM175A, FANCM, NBN, and SLX4 genes in 6.4% of the affected women. Four recurrent pathogenic variants were detected in 11 patients of African ancestry. Only one unaffected woman had a pathogenic variant in the RAD51C gene. Different risk assessment models examined performed well in predicting risk of carrying germline loss-of-function variants in BRCA1 and/or BRCA2 in breast cancer cases. Conclusion The high prevalence and heterogenous spectrum of pathogenic variants identified among self-reported African descendants in Northeast Brazil is consistent with studies in other African ancestry populations with a high burden of aggressive young onset breast cancer. It underscores the need to integrate comprehensive cancer risk assessment and genomic testing in the management of newly diagnosed Black women with breast cancer across the African Diaspora, enabling improved cancer control in admixed underserved and understudied populations.
- Published
- 2022
25. 906 De novomutations in childhood-onset systemic lupus erythematosus
- Author
-
Sarah K Baxter, Suleyman Gulsuner, Mary M Eckert, Ming K Lee, Tom Walsh, Anne Stevens, and Mary-Claire King
- Published
- 2022
26. Adaptation and validation of a computerized neurocognitive battery in the Xhosa of South Africa
- Author
-
Mary Claire King, Jon McClellan, Megan M. Campbell, J. Cobb Scott, Kosha Ruparel, Adele Pretorius, Allison M. Port, Zukiswa Zingela, Ezra Susser, Chad T. Jackson, Goodman Sibeko, Tyler M. Moore, Dan J. Stein, Ruben C. Gur, Linda Ngqengelele, and Mohammed Nagdee
- Subjects
education.field_of_study ,Concurrent validity ,Population ,Neuropsychology ,Reproducibility of Results ,Cognition ,Test validity ,Neuropsychological Tests ,Executive functions ,Article ,Executive Function ,South Africa ,Neuropsychology and Physiological Psychology ,Social cognition ,Humans ,education ,Psychology ,Neurocognitive ,Clinical psychology - Abstract
Objective Large-scale studies have revolutionized biomedical research, and neurocognitive tests can help elucidate the biological basis of neuropsychiatric diseases. However, studies have predominantly been conducted in Western settings. We describe the development and validation of a computerized battery (PennCNB) with the Xhosa population of South Africa. Method Individuals with schizophrenia (n = 525) and a normative comparison group (n = 744) were balanced on age, sex, education, and region. Participants provided blood samples, were assessed psychiatrically, and were administered a PennCNB translation to isiXhosa, including measures of executive functions, episodic memory, complex cognition, social cognition, and sensorimotor speed. Feasibility was examined with test completion rates and input from administrators, and psychometric structural validity and associations with clinical and demographic characteristics were examined. Results Tests were well tolerated by participants, as >87% had one (or fewer) test missing. Results suggested a similar factor structure to prior PennCNB studies in Western contexts, and expected age and sex effects were apparent. Furthermore, a similar profile of schizophrenia was observed, with neurocognitive deficits most pronounced for executive functions, especially attention, as well as memory, social cognition, and motor speed relative to complex cognition and sensorimotor speed. Conclusions Results support the feasibility of implementing a culturally adapted computerized neurocognitive battery in sub-Saharan African settings and provide evidence supporting the concurrent validity of the translated instrument. Thus, the PennCNB is implementable on a large scale in non-Western contexts, shows expected factor structure, and can detect cognitive deficits associated with neuropsychiatric disorders. Obtaining valid measures of cognition by nonspecialized proctors is especially suitable in resource-limited settings, where traditional testing is prohibitive. Future work should establish normative standards, test-retest reliability, and sensitivity to treatment. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
- Published
- 2021
27. Diagnostic yield of chromosomal microarray and trio whole exome sequencing in cryptogenic cerebral palsy
- Author
-
Alla Kuzminsky, Liora Sagi, Adi Aran, Luba Blumkin, Tehila Klopstock, Dorit Lev, Dafna Guttman, Lilach Shemer Meiri, Reeval Segel, Suleyman Gulsuner, Michal Yechieli, Mary Claire King, Varda Gross-Tsur, Aviva Fattal, Paul Renbaum, Hilla Ben-Pazi, Ephrat Lahad Levy, Sharon Zeligson, Nira Schneebaum Sender, Dorit Shmueli, Thomas J. Walsh, and Amnon Lahad
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Candidate gene ,Movement disorders ,DNA Copy Number Variations ,Microarray ,medicine.disease_cause ,Cerebral palsy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Exome Sequencing ,Genetics ,medicine ,Humans ,Genetics (clinical) ,Exome sequencing ,Genetic testing ,Mutation ,medicine.diagnostic_test ,business.industry ,Cerebral Palsy ,Point mutation ,Microarray Analysis ,medicine.disease ,030104 developmental biology ,Child, Preschool ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
ObjectiveTo determine the yield of genetic diagnoses using chromosomal microarray (CMA) and trio whole exome sequencing (WES), separately and combined, among patients with cryptogenic cerebral palsy (CP).MethodsTrio WES of patients with prior CMA analysis for cryptogenic CP, defined as disabling, non-progressive motor symptoms beginning before the age of 3 years without known cause.ResultsGiven both CMA analysis and trio WES, clinically significant genetic findings were identified for 58% of patients (26 of 45). Diagnoses were eight large CNVs detected by CMA and 18 point mutations detected by trio WES. None had more than one severe mutation. Approximately half of events (14 of 26) were de novo. Yield was significantly higher in patients with CP with comorbidities (69%, 22 of 32) than in those with pure motor CP (31%, 4 of 13; p=0.02). Among patients with genetic diagnoses, CNVs were more frequent than point mutations among patients with congenital anomalies (OR 7.8, 95% CI 1.2 to 52.4) or major dysmorphic features (OR 10.5, 95% CI 1.4 to 73.7). Clinically significant mutations were identified in 18 different genes: 14 with known involvement in CP-related disorders and 4 responsible for other neurodevelopmental conditions. Three possible new candidate genes for CP were ARGEF10, RTF1 and TAOK3.ConclusionsCryptogenic CP is genetically highly heterogeneous. Genomic analysis has a high yield and is warranted in all these patients. Trio WES has higher yield than CMA, except in patients with congenital anomalies or major dysmorphic features, but these methods are complementary. Patients with negative results with one approach should also be tested by the other.
- Published
- 2021
28. Genetic Heterogeneity and Core Clinical Features of NOG-Related-Symphalangism Spectrum Disorder
- Author
-
Kathleen A. Leppig, Thomas J. Walsh, Dawson Wells, Dror Gilony, Karen B. Avraham, Mary Claire King, Ryan J. Carlson, Jay T. Rubinstein, Suleyman Gulsuner, Zippora Brownstein, and Alicia M. Quesnel
- Subjects
Foot Deformities, Congenital ,business.industry ,Genetic heterogeneity ,Point mutation ,Tarsal Bones ,medicine.disease ,Bioinformatics ,Article ,Stapes ,Sensory Systems ,Conductive hearing loss ,Genetic Heterogeneity ,Synostosis ,Otorhinolaryngology ,Temporal bone ,medicine ,Humans ,Otosclerosis ,Missense mutation ,Neurology (clinical) ,business ,Hand Deformities, Congenital ,Carpal Bones ,Chromosomal Deletion - Abstract
Objectives To better distinguish NOG-related-symphalangism spectrum disorder (NOG-SSD) from chromosomal 17q22 microdeletion syndromes and to inform surgical considerations in stapes surgery for patients with NOG-SSD. Background Mutations in NOG cause a variety of skeletal syndromes that often include conductive hearing loss. Several microdeletions of chromosome 17q22 lead to severe syndromes with clinical characteristics that overlap NOG-SSD. Isolated deletion of NOG has not been described, and therefore the contribution of NOG deletion in these syndromes is unknown. Methods Two families with autosomal dominant NOG-SSD exhibited stapes ankylosis, facial dysmorphisms, and skeletal and joint anomalies. In each family, NOG was evaluated by genomic sequencing and candidate mutations confirmed as damaging by in vitro assays. Temporal bone histology of a patient with NOG-SSD was compared with temporal bones of 40 patients diagnosed with otosclerosis. Results Family 1 harbors a 555 kb chromosomal deletion encompassing only NOG and ANKFN1. Family 2 harbors a missense mutation in NOG leading to absence of noggin protein. The incus-footplate distance of the temporal bone was significantly longer in a patient with NOG-SSD than in patients with otosclerosis. Conclusion The chromosomal microdeletion of family 1 led to a phenotype comparable to that due to a NOG point mutation and much milder than the phenotypes due to other chromosome 17q22 microdeletions. Severe clinical findings in other microdeletion cases are likely due to deletion of genes other than NOG. Based on temporal bone findings, we recommend that surgeons obtain longer stapes prostheses before stapes surgery in individuals with NOG-SSD stapes ankylosis.
- Published
- 2021
29. A novel founder MSH2 deletion in Ethiopian Jews is mainly associated with early-onset colorectal cancer
- Author
-
Zohar Levi, Sari Lieberman, G Reznick Levi, M Goldenberg, Lior H. Katz, Elizabeth E. Half, Yael Goldberg, L Walsh, D Rothstein, Thomas J. Walsh, T Yablonski Peretz, Ayala Hubert, Ido Laish, Inbal Kedar, Colin C. Pritchard, Lina Basel-Salmon, S Naftaly Nathan, Mary Claire King, R Tomashov-Matar, A Abu Shtaya, Ola Aleme, I Lagovsky, and Sergi Castellví-Bel
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Colorectal cancer ,Genetic counseling ,030105 genetics & heredity ,medicine.disease ,digestive system diseases ,Lynch syndrome ,Human genetics ,03 medical and health sciences ,0302 clinical medicine ,MSH2 ,030220 oncology & carcinogenesis ,Internal medicine ,Epidemiology ,Anticipation (genetics) ,Genetics ,Medicine ,DNA mismatch repair ,business ,Genetics (clinical) - Abstract
Lynch syndrome is an inherited cancer predisposition syndrome caused by germline defects in any of the mismatch repair (MMR) genes. Diagnosis of carriers makes precision prevention, early detection, and tailored treatment possible. Herein we report a novel founder deletion of 18,758 bp, mediated by Alu repeats on both sides, detected in Ethiopian Jews. The deletion, which encompasses exon 9–10 of the MSH2 coding sequence, is associated mainly with early-onset MSH2/MSH6-deficient colorectal cancer (CRC) and liposarcoma. Testing of 35 members of 5 seemingly unrelated families of Ethiopian origin yielded 10/21 (48%) carriers, of whom 9 had CRC. Age at first tumor diagnosis ranged from 16 to 89 years. Carriers from the oldest generations were diagnosed after age 45 years (mean 57), and carriers from the younger generation were diagnosed before age 45 years (mean 30). Awareness of this founder deletion is important to improve patient diagnosis, institute surveillance from an early age, and refer patients for genetic counseling addressing the risk of bi-allelic constitutional MMR deficiency syndrome.
- Published
- 2021
30. 439. Functional Characterization of Damaging Mutations in Patients With Sporadic Schizophrenia Using CRISPRi in iPSCS
- Author
-
Anna Sunshine, Suleyman Gulsuner, Tom Walsh, Mary-Claire King, and Jon McClellan
- Subjects
Biological Psychiatry - Published
- 2023
31. Germline variants drive myelodysplastic syndrome in young adults
- Author
-
Ulla Wartiovaara-Kautto, Ulrich Germing, Hari Prasanna Subramanian, Tara Cronin, Gudrun Goehring, Elizabeth A. Griffiths, Guimin Gao, Eunice S. Wang, Mary Claire King, Simone Feurstein, Carolyn Owen, Thomas Schroeder, Brigitte Schlegelberger, Outi Kilpivaara, Marja Hakkarainen, Torsten Haferlach, Divij Verma, Felicitas Thol, Stefanie Geyh, Hartmut Döhner, Colin C. Pritchard, Sioban Keel, Juehua Gao, Zejuan Li, Tom Walsh, Daniela S. Krause, Suleyman Gulsuner, Michael Heuser, Lucy A. Godley, Daniela del Gaudio, Ming Lee, Jane E. Churpek, Konstanze Döhner, Soma Das, and Christian Pohlkamp
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Article ,Germline ,Young Adult ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Aplastic anemia ,Young adult ,Germ-Line Mutation ,business.industry ,Anemia, Aplastic ,Hematology ,Prognosis ,medicine.disease ,Myelodysplastic Syndromes ,Female ,Line (text file) ,business ,Biomarkers - Published
- 2021
32. Spectrum of genes for inherited hearing loss in the Israeli Jewish population, including the novel human deafness gene<scp>ATOH1</scp>
- Author
-
Tom Walsh, Silvia Casadei, Ofer Isakov, Mary Claire King, Matthew W. Kelley, Noa Ruhrman-Shahar, Eiríkur Steingrímsson, Maria Birkan, Mor Bordeynik-Cohen, Ronna Hertzano, Nadra Samra, Morad Khayat, Nada Danial-Farran, Naama Zvi, Zippora Brownstein, Moshe Frydman, Elon Pras, Ophir Handzel, Moien Kanaan, Fabio Tadeu Arrojo Martins, Michal Macarov, Noam Shomron, Asgeir O. Arnthorsson, Bella Davidov, Doaa Ali-Naffaa, Michal Sagi, Lara Kamal, Reuven Sharony, Lina Basel-Salmon, Ming K. Lee, Meirav Sokolov, Weise Chang, Ory Madgar, Michael Wolf, Dorit Lev, Karen B. Avraham, Hagit Baris-Feldman, Dror Gilony, Ryan J. Carlson, Hana Poran, Noga Lipschitz, Shahar Taiber, Suleyman Gulsuner, Amal Abu-Rayyan, Stavit Allon-Shalev, Chana Vinkler, Amihood Singer, Amir Peleg, Efrat Sofrin‐Drucker, and Mordechai Shohat
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,Hearing loss ,Genetic counseling ,Population ,Genomics ,Deafness ,030105 genetics & heredity ,Biology ,Article ,Young Adult ,03 medical and health sciences ,Genotype ,Basic Helix-Loop-Helix Transcription Factors ,otorhinolaryngologic diseases ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Israel ,Allele ,Child ,Hearing Loss ,education ,Genetic Association Studies ,Genetics (clinical) ,Newborn screening ,education.field_of_study ,Massive parallel sequencing ,Pedigree ,030104 developmental biology ,Child, Preschool ,Jews ,Female ,medicine.symptom - Abstract
Mutations in more than 150 genes are responsible for inherited hearing loss, with thousands of different, severe causal alleles that vary among populations. The Israeli Jewish population includes communities of diverse geographic origins, revealing a wide range of deafness-associated variants and enabling clinical characterization of the associated phenotypes. Our goal was to identify the genetic causes of inherited hearing loss in this population, and to determine relationships among genotype, phenotype, and ethnicity. Genomic DNA samples from informative relatives of 88 multiplex families, all of self-identified Jewish ancestry, with either non-syndromic or syndromic hearing loss, were sequenced for known and candidate deafness genes using the HEar-Seq gene panel. The genetic causes of hearing loss were identified for 60% of the families. One gene was encountered for the first time in human hearing loss: ATOH1 (Atonal), a basic helix-loop-helix transcription factor responsible for autosomal dominant progressive hearing loss in a five-generation family. Our results demonstrate that genomic sequencing with a gene panel dedicated to hearing loss is effective for genetic diagnoses in a diverse population. Comprehensive sequencing enables well-informed genetic counseling and clinical management by medical geneticists, otolaryngologists, audiologists, and speech therapists and can be integrated into newborn screening for deafness.
- Published
- 2020
33. NKX2-2 Mutation Causes Congenital Diabetes and Infantile Obesity With Paradoxical Glucose-Induced Ghrelin Secretion
- Author
-
Ariella Weinberg-Shukron, Adi Auerbach, Suleyman Gulsuner, Abdulsalam Abu-Libdeh, Amitay Cohen, David Zangen, Mary Claire King, Rina Hemi, Noa Ofek Shlomai, and Ephrat Levy-Lahad
- Subjects
0301 basic medicine ,Pediatric Obesity ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Clinical Biochemistry ,030209 endocrinology & metabolism ,Context (language use) ,Gene mutation ,Biochemistry ,ABCC8 ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Neonatal diabetes mellitus ,Internal medicine ,Diabetes mellitus ,Exome Sequencing ,Diabetes Mellitus ,medicine ,Humans ,Infant, Very Low Birth Weight ,Homeodomain Proteins ,biology ,business.industry ,Insulin ,Biochemistry (medical) ,Infant ,Nuclear Proteins ,Infantile Obesity ,Zebrafish Proteins ,medicine.disease ,Ghrelin ,Homeobox Protein Nkx-2.2 ,030104 developmental biology ,Child, Preschool ,Mutation ,Mutation (genetic algorithm) ,biology.protein ,Female ,business ,Ghrelin secretion ,Transcription Factors - Abstract
Context NKX2-2 is a crucial transcription factor that enables specific β-cell gene expression. Nkx2-2(–/–) mice manifest with severe neonatal diabetes and changes in β-cell progenitor fate into ghrelin-producing cells. In humans, recessive NKX2-2 gene mutations have been recently reported as a novel etiology for neonatal diabetes, with only 3 cases known worldwide. This study describes the genetic analysis, distinctive clinical features, the therapeutic challenges, and the unique pathophysiology causing neonatal diabetes in human NKX2-2 dysfunction. Case Description An infant with very low birth weight (VLBW) and severe neonatal diabetes (NDM) presented with severe obesity and developmental delay already at age 1 year. The challenge of achieving glycemic control in a VLBW infant was unexpectedly met by a regimen of 3 daily doses of long-acting insulin analogues. Sanger sequencing of known NDM genes (such as ABCC8 and EIF2AK3) was followed by whole-exome sequencing that revealed homozygosity of a pathogenic frameshift variant, c.356delG, p.P119fs64*, in the islet cells transcription factor, NKX2-2. To elucidate the cause for the severe obesity, an oral glucose tolerance test was conducted at age 3.5 years and revealed undetectable C-peptide levels with a paradoxically unexpected 30% increase in ghrelin levels. Conclusion Recessive NKX2-2 loss of function causes severe NDM associated with VLBW, childhood obesity, and developmental delay. The severe obesity phenotype is associated with postprandial paradoxical ghrelin secretion, which may be related to human β-cell fate change to ghrelin-secreting cells, recapitulating the finding in Nkx2-2(–/–) mice islet cells.
- Published
- 2020
34. A defect in GPI synthesis as a suggested mechanism for the role of ARV1 in intellectual disability and seizures
- Author
-
Mary Claire King, Adi Aran, Tom Walsh, Kouichi Funato, Ephrat Levy-Lahad, Haruka Okai, Tzvia Rosen, Hiroto Denda, Hiroki Nakamura, Paul Renbaum, Sharon Zeligson, Katsuki Eto, Suleyman Gulsuner, Reeval Segel, and Rachel Beeri
- Subjects
Male ,0301 basic medicine ,Adolescent ,Glycosylphosphatidylinositols ,Mutant ,Golgi Apparatus ,Endoplasmic Reticulum ,medicine.disease_cause ,Mannosyltransferases ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,symbols.namesake ,0302 clinical medicine ,Glycolipid ,Protein Domains ,Seizures ,Intellectual Disability ,Genetics ,medicine ,Humans ,Child ,Genetics (clinical) ,Brain Diseases ,Mutation ,Chemistry ,Endoplasmic reticulum ,Genetic Complementation Test ,Homozygote ,Temperature ,Membrane Proteins ,Fibroblasts ,Golgi apparatus ,Lipids ,Sphingolipid ,Transmembrane protein ,Pedigree ,Cell biology ,carbohydrates (lipids) ,Complementation ,030104 developmental biology ,symbols ,lipids (amino acids, peptides, and proteins) ,Carrier Proteins ,030217 neurology & neurosurgery - Abstract
Deficiency of the endoplasmic reticulum transmembrane protein ARV1 leads to epileptic encephalopathy in humans and in mice. ARV1 is highly conserved, but its function in human cells is unknown. Studies of yeast arv1 null mutants indicate that it is involved in a number of biochemical processes including the synthesis of sphingolipids and glycosylphosphatidylinositol (GPI), a glycolipid anchor that is attached to the C-termini of many membrane bound proteins. GPI anchors are post-translational modifications, enabling proteins to travel from the endoplasmic reticulum (ER) through the Golgi and to attach to plasma membranes. We identified a homozygous pathogenic mutation in ARV1, p.Gly189Arg, in two brothers with infantile encephalopathy, and characterized the biochemical defect caused by this mutation. In addition to reduced expression of ARV1 transcript and protein in patients' fibroblasts, complementation tests in yeast showed that the ARV1 p.Gly189Arg mutation leads to deficient maturation of Gas1, a GPI-anchored protein, but does not affect sphingolipid synthesis. Our results suggest, that similar to mutations in other proteins in the GPI-anchoring pathway, including PIGM, PIGA, and PIGQ, ARV1 p.Gly189Arg causes a GPI anchoring defect and leads to early onset epileptic encephalopathy.
- Published
- 2020
35. Whole Exome Sequencing Identifies Candidate Genes Associated with Hereditary Predisposition to Uveal Melanoma
- Author
-
Lynn Schoenfield, Silvia Casadei, Meghan J DeBenedictis, Mohamed H. Abdel-Rahman, Ben Kelly, Peter White, Frederick H. Davidorf, Andrew W. Stacey, Daniel D. Kinnamon, Mary Claire King, David M Gordon, Arun D. Singh, James B. Massengill, T. Walsh, Nicholas K. Hayward, Getachew Boru, Klarke M. Sample, James Scarth, Timothy W. Grosel, Ellie Fewings, Colleen M. Cebulla, Peter Johansson, Marc Tischkowitz, and Robert Pilarski
- Subjects
Adult ,Male ,Uveal Neoplasms ,Oncology ,Candidate gene ,medicine.medical_specialty ,Adolescent ,MLH1 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Internal medicine ,Exome Sequencing ,medicine ,Humans ,Genetic Predisposition to Disease ,Child ,Melanoma ,Germ-Line Mutation ,Exome sequencing ,Aged ,Retrospective Studies ,030304 developmental biology ,Genetic testing ,Aged, 80 and over ,0303 health sciences ,medicine.diagnostic_test ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Tumor Suppressor Proteins ,Infant ,Cancer ,DNA, Neoplasm ,Odds ratio ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Neoplasm Proteins ,3. Good health ,MSH6 ,Ophthalmology ,Child, Preschool ,030221 ophthalmology & optometry ,Female ,business ,Ubiquitin Thiolesterase ,Genes, Neoplasm - Abstract
To identify susceptibility genes associated with hereditary predisposition to uveal melanoma (UM) in patients with no detectable germline BAP1 alterations.Retrospective case series from academic referral centers.Cohort of 154 UM patients with high risk of hereditary cancer defined as patients with 1 or more of the following: (1) familial UM, (2) young age (35 years) at diagnosis, (3) personal history of other primary cancers, and (4) family history of 2 or more primary cancers with no detectable mutation or deletion in BAP1 gene.Whole exome sequencing, a cancer gene panel, or both were carried out. Probands included 27 patients with familial UM, 1 patient with bilateral UM, 1 patient with congenital UM, and 125 UM patients with strong personal or family histories, or both, of cancer. Functional validation of variants was carried out by immunohistochemistry, reverse-transcriptase polymerase chain reaction, and genotyping.Clinical characterization of UM patients with germline alterations in known cancer genes.We identified actionable pathogenic variants in 8 known hereditary cancer predisposition genes (PALB2, MLH1, MSH6, CHEK2, SMARCE1, ATM, BRCA1, and CTNNA1) in 9 patients, including 3 of 27 patients (11%) with familial UM and 6 of 127 patients (4.7%) with a high risk for cancer. Two patients showed pathogenic variants in CHEK2 and PALB2, whereas variants in the other genes each occurred in 1 patient. Biallelic inactivation of PALB2 and MLH1 was observed in tumors from the respective patients. The frequencies of pathogenic variants in PALB2, MLH1, and SMARCE1 in UM patients were significantly higher than the observed frequencies in noncancer controls (PALB2: P = 0.02; odds ratio, 8.9; 95% confidence interval, 1.5-30.6; MLH1: P = 0.04; odds ratio, 25.4; 95% confidence interval, 1.2-143; SMARCE1: P = 0.001; odds ratio, 2047; 95% confidence interval, 52-4.5e15, respectively).The study provided moderate evidence of gene and disease association of germline mutations in PALB2 and MLH1 with hereditary predisposition to UM. It also identified several other candidate susceptibility genes. The results suggest locus heterogeneity in predisposition to UM. Genetic testing for hereditary predisposition to cancer is warranted in UM patients with strong personal or family history of cancers, or both.
- Published
- 2020
36. Systematic misclassification of missense variants in BRCA1 and BRCA2 'coldspots'
- Author
-
Tom Walsh, Colin C. Pritchard, Thomas P. Slavin, Douglas M. Fowler, Jennifer N. Dines, Mary Claire King, and Brian H. Shirts
- Subjects
0301 basic medicine ,endocrine system diseases ,Mutation, Missense ,Breast Neoplasms ,Classification scheme ,Biology ,Article ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Humans ,Missense mutation ,Coding region ,Genetic Predisposition to Disease ,skin and connective tissue diseases ,variant classification ,Genetics (clinical) ,Likely pathogenic ,BRCA2 Protein ,Ovarian Neoplasms ,Genetics ,BRCA1 Protein ,Bayes Theorem ,Exons ,Pathogenicity ,BRCA1 ,Human genetics ,Confidence interval ,VUS ,030104 developmental biology ,coldspot ,030220 oncology & carcinogenesis ,ACMG ,Female - Abstract
Purpose Guidelines for variant interpretation incorporate variant hotspots in critical functional domains as evidence for pathogenicity (e.g., PM1 and PP2), but do not use “coldspots,” that is, regions without essential functions that tolerate variation, as evidence a variant is benign. To improve variant classification we evaluated BRCA1 and BRCA2 missense variants reported in ClinVar to identify regions where pathogenic missenses are extremely infrequent, defined as coldspots. Methods We used Bayesian approaches to model variant classification in these regions. Results BRCA1 exon 11 (~60% of the coding sequence), and BRCA2 exons 10 and 11 (~65% of the coding sequence), are coldspots. Of 89 pathogenic (P) or likely pathogenic (LP) missense variants in BRCA1, none are in exon 11 (odds
- Published
- 2020
37. Characterization of splice-altering mutations in inherited predisposition to cancer
- Author
-
Yael Goldberg, Colin C. Pritchard, Hannah M. Kortbawi, Jessica B. Mandell, Brian H. Shirts, Zheng Tan, Ming K. Lee, Elizabeth M. Swisher, Mary Claire King, Suleyman Gulsuner, Robert O’Connor, Tom Walsh, Silvia Casadei, and B. Norquist
- Subjects
Genetics ,Mutation ,Multidisciplinary ,Intron ,Biological Sciences ,Biology ,medicine.disease_cause ,Exon skipping ,Exon ,genomic DNA ,RNA splicing ,medicine ,splice ,Gene - Abstract
Mutations responsible for inherited disease may act by disrupting normal transcriptional splicing. Such mutations can be difficult to detect, and their effects difficult to characterize, because many lie deep within exons or introns where they may alter splice enhancers or silencers or introduce new splice acceptors or donors. Multiple mutation-specific and genome-wide approaches have been developed to evaluate these classes of mutations. We introduce a complementary experimental approach, cBROCA, which yields qualitative and quantitative assessments of the effects of genomic mutations on transcriptional splicing of tumor suppressor genes. cBROCA analysis is undertaken by deriving complementary DNA (cDNA) from puromycin-treated patient lymphoblasts, hybridizing the cDNA to the BROCA panel of tumor suppressor genes, and then multiplex sequencing to very high coverage. At each splice junction suggested by split sequencing reads, read depths of test and control samples are compared. Significant Z scores indicate altered transcripts, over and above naturally occurring minor transcripts, and comparisons of read depths indicate relative abundances of mutant and normal transcripts. BROCA analysis of genomic DNA suggested 120 rare mutations from 150 families with cancers of the breast, ovary, uterus, or colon, in >600 informative genotyped relatives. cBROCA analysis of their transcripts revealed a wide variety of consequences of abnormal splicing in tumor suppressor genes, including whole or partial exon skipping, exonification of intronic sequence, loss or gain of exonic and intronic splicing enhancers and silencers, complete intron retention, hypomorphic alleles, and combinations of these alterations. Combined with pedigree analysis, cBROCA sequencing contributes to understanding the clinical consequences of rare inherited mutations.
- Published
- 2019
38. A tipping point in neuropsychiatric genetics
- Author
-
Jon McClellan and Mary Claire King
- Subjects
0301 basic medicine ,Genetics ,Mechanism (biology) ,Genetic heterogeneity ,General Neuroscience ,Protocadherin ,Biology ,Cadherins ,medicine.disease ,Article ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Gene Frequency ,Schizophrenia ,Jews ,medicine ,Humans ,Ashkenazi Jewish ,Allele ,Alleles ,030217 neurology & neurosurgery ,Founder effect - Abstract
Identification of rare variants associated with schizophrenia has proven challenging due to genetic heterogeneity, which is reduced in founder populations. In samples from the Ashkenazi Jewish population, we report that schizophrenia cases had greater frequency of novel missense or loss of function (MisLoF) ultra-rare variants (URVs) compared to controls, and MisLoF URV burden was inversely correlated with polygenic risk scores in cases. Characterizing 141 “case-only” genes (MisLoF URVs in ≥ 3 cases with none in controls), the cadherin gene set was associated with schizophrenia. We report a recurrent case mutation in PCDHA3 which results in formation of cytoplasmic aggregates and failure to engage in homophilic interactions on the plasma membrane in cultured cells. Modeling purifying selection, we demonstrate that deleterious URVs are greatly over-represented in the Ashkenazi population, yielding enhanced power for association studies. Identification of the cadherin/protocadherin family as risk genes helps specify the synaptic abnormalities central to schizophrenia.
- Published
- 2021
39. Mutational Spectrum of Breast Cancer Susceptibility Genes among Women in Northeast Brazil
- Author
-
Gabriela ES Felix, Rodrigo Santa Cruz Guindalini, Yonglan Zheng, Tom Walsh, Elisabeth Sveen, Taisa Manuela Machado Lopes, Juliana Côrtes, Jing Zhang, Polyanna Carôzo, Irlânia Santos, Thaís Ferreira Bonfim, Bernardo Garicochea, Betânia Pereira Toralles, Roberto Meyer, Eduardo Martins Netto, Kiyoko Abe-Sandes, Mary-Claire King, Ivana Lucia de Oliveira Nascimento, and Olufunmilayo I. Olopade
- Abstract
Purpose: There is a paucity of data on the spectrum and prevalence of pathogenic variants among women of African ancestry in the Northeast region of Brazil. Methods: We performed BROCA panel sequencing to identify inherited loss-of-function variants in breast cancer susceptibility genes among 292 Brazilian women referred to a single institution cancer risk assessment program. Results: The study included a convenient cohort of 173 women with invasive breast cancer and 119 women who were cancer-free at the time of ascertainment. The majority of the women self-reported as African-descended (67% for cases and 90.8% for controls). Thirty-seven pathogenic variants were found in 36 (20.8%) patients. While the spectrum of pathogenic variants was heterogeneous, the majority (70.3%) of the pathogenic variants were detected in high-risk genes BRCA1 , BRCA2 , PALB2 , and TP53 . Pathogenic variants were also found in the ATM , BARD1 , BRIP1 , FAM175A , FANCM , NBN , and SLX4 genes in 6.4% of the affected women. Four recurrent pathogenic variants were detected in 11 patients of African ancestry. Only one unaffected woman had a pathogenic variant in the RAD51C gene. Conclusion: The high prevalence and heterogenous spectrum of pathogenic variants identified among self-reported African descendants in Northeast Brazil is consistent with studies in other African populations with a high burden of aggressive young onset breast cancer. It underscores the need to integrate comprehensive cancer risk assessment and genomic testing in the management of newly diagnosed Black women with breast cancer across the African Diaspora, enabling improved cancer control in admixed underserved and understudied populations.
- Published
- 2021
40. Hiding in Plain Sight — Somatic Mutation in Human Disease
- Author
-
Ephrat Levy-Lahad and Mary Claire King
- Subjects
03 medical and health sciences ,medicine.medical_specialty ,0302 clinical medicine ,Human disease ,Germline mutation ,business.industry ,Medicine ,030212 general & internal medicine ,General Medicine ,030204 cardiovascular system & hematology ,business ,Dermatology ,Inflammatory disorder - Abstract
Beck et al. define an adult-onset inflammatory disorder that unites multiple clinical conditions with previously different diagnoses or with no diagnosis at all; their findings are now reported in ...
- Published
- 2020
41. CRISPR–Cas9/long-read sequencing approach to identify cryptic mutations in BRCA1 and other tumour suppressor genes
- Author
-
Tom Walsh, Silvia Casadei, Katherine M. Munson, Mary Eng, Jessica B. Mandell, Mary Claire King, and Suleyman Gulsuner
- Subjects
0301 basic medicine ,Genetics ,Mutation ,Sequence analysis ,Biology ,medicine.disease_cause ,Structural variation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Germline mutation ,medicine ,CRISPR ,Indel ,Gene ,030217 neurology & neurosurgery ,Genetics (clinical) ,Exome sequencing - Abstract
Current clinical approaches for mutation discovery are based on short sequence reads (100–300 bp) of exons and flanking splice sites targeted by multigene panels or whole exomes. Short-read sequencing is highly accurate for detection of single nucleotide variants, small indels and simple copy number differences but is of limited use for identifying complex insertions and deletions and other structural rearrangements. We used CRISPR-Cas9 to excise complete BRCA1 and BRCA2 genomic regions from lymphoblast cells of patients with breast cancer, then sequenced these regions with long reads (>10 000 bp) to fully characterise all non-coding regions for structural variation. In a family severely affected with early-onset bilateral breast cancer and with negative (normal) results by gene panel and exome sequencing, we identified an intronic SINE-VNTR-Alu retrotransposon insertion that led to the creation of a pseudoexon in the BRCA1 message and introduced a premature truncation. This combination of CRISPR–Cas9 excision and long-read sequencing reveals a class of complex, damaging and otherwise cryptic mutations that may be particularly frequent in tumour suppressor genes replete with intronic repeats.
- Published
- 2020
42. Inherited predisposition to malignant mesothelioma and overall survival following platinum chemotherapy
- Author
-
Betsy Morrow, Ming K. Lee, Jun Wei, Anish Thomas, Liqiang Xi, Raffit Hassan, Idrees Mian, Mary Claire King, Tom Walsh, Jingli Zhang, Snehal Patel, Vasiliki Panou, David S. Schrump, Jane E. Churpek, Arpita Desai, Mark Raffeld, Emerson Padiernos, Javed Khan, Meghana Gadiraju, Shaojian Gao, Kathleen A. Calzone, Mary Hesdorffer, Suleyman Gulsuner, Christine Alewine, Seth M. Steinberg, and Hedy L. Kindler
- Subjects
Adult ,Male ,Mesothelioma ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,DNA Repair ,DNA repair ,Pleural Neoplasms ,medicine.disease_cause ,Young Adult ,Survival ,Prostate ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Genotype ,medicine ,Humans ,BAP1 ,Genetic Predisposition to Disease ,Gene ,Germ-Line Mutation ,Aged ,Platinum ,Aged, 80 and over ,Mutation ,Multidisciplinary ,DNA repair genes ,business.industry ,Tumor Suppressor Proteins ,Nherited genetics ,Mesothelioma, Malignant ,Biological Sciences ,Middle Aged ,medicine.disease ,Survival Analysis ,respiratory tract diseases ,medicine.anatomical_structure ,Peritoneal mesothelioma ,Female ,business ,Ubiquitin Thiolesterase - Abstract
Survival from malignant mesothelioma, particularly pleural mesothelioma, is very poor. For patients with breast, ovarian, or prostate cancers, overall survival is associated with increased sensitivity to platinum chemotherapy due to loss-of-function mutations in DNA repair genes. The goal of this project was to evaluate, in patients with malignant mesothelioma, the relationship between inherited loss-of-function mutations in DNA repair and other tumor suppressor genes and overall survival following platinum chemotherapy. Patients with histologically confirmed malignant mesothelioma were evaluated for inherited mutations in tumor suppressor genes. Survival was evaluated with respect to genotype and site of mesothelioma. Among 385 patients treated with platinum chemotherapy, median overall survival was significantly longer for patients with loss-of-function mutations in any of the targeted genes compared with patients with no such mutation (P = 0.0006). The effect of genotype was highly significant for patients with pleural mesothelioma (median survival 7.9 y versus 2.4 y, P = 0.0012), but not for patients with peritoneal mesothelioma (median survival 8.2 y versus 5.4 y, P = 0.47). Effect of patient genotype on overall survival, measured at 3 y, remained independently significant after adjusting for gender and age at diagnosis, two other known prognostic factors. Patients with pleural mesothelioma with inherited mutations in DNA repair and other tumor suppressor genes appear to particularly benefit from platinum chemotherapy compared with patients without inherited mutations. These patients may also benefit from other DNA repair targeted therapies such as poly-ADP ribose polymerase (PARP) inhibitors.
- Published
- 2019
43. Intensive Surveillance with Biannual Dynamic Contrast-Enhanced Magnetic Resonance Imaging Downstages Breast Cancer in BRCA1 Mutation Carriers
- Author
-
Akila Raoul, Elias Obeid, Olufunmilayo I. Olopade, Mary Claire King, Marcio Debiasi, Marion S. Verp, Kristen Danielle Whitaker, Jeffrey Mueller, Xiaoming Wang, Yonglan Zheng, Fang Liu, Iris L. Romero, Angela R. Bradbury, Galina Khramtsova, Jane E. Churpek, Robert B. Livingston, Kirti Kulkarni, David V Schacht, Colin C. Pritchard, Susan Hong, Hongyuan Cao, Tom Walsh, Rodrigo Santa Cruz Guindalini, Nora Jaskowiak, Gregory S. Karczmar, Hiroyuki Abe, Deepa Sheth, Dezheng Huo, Toshio F. Yoshimatsu, and Gillian M. Newstead
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Biopsy ,Genes, BRCA1 ,Breast Neoplasms ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,medicine ,Mass Screening ,Humans ,Mammography ,Genetic Predisposition to Disease ,Prospective Studies ,skin and connective tissue diseases ,Early Detection of Cancer ,Brca1 gene ,Neoplasm Staging ,medicine.diagnostic_test ,BRCA1 Protein ,business.industry ,Cancer ,Magnetic resonance imaging ,Middle Aged ,Ductal carcinoma ,medicine.disease ,Magnetic Resonance Imaging ,Dynamic contrast ,030220 oncology & carcinogenesis ,Mutation ,Cohort ,Female ,business - Abstract
Purpose: To establish a cohort of high-risk women undergoing intensive surveillance for breast cancer. Experimental Design: We performed dynamic contrast-enhanced MRI every 6 months in conjunction with annual mammography (MG). Eligible participants had a cumulative lifetime breast cancer risk ≥20% and/or tested positive for a pathogenic mutation in a known breast cancer susceptibility gene. Results: Between 2004 and 2016, we prospectively enrolled 295 women, including 157 mutation carriers (75 BRCA1, 61 BRCA2); participants' mean age at entry was 43.3 years. Seventeen cancers were later diagnosed: 4 ductal carcinoma in situ (DCIS) and 13 early-stage invasive breast cancers. Fifteen cancers occurred in mutation carriers (11 BRCA1, 3 BRCA2, 1 CDH1). Median size of the invasive cancers was 0.61 cm. No patients had lymph node metastasis at time of diagnosis, and no interval invasive cancers occurred. The sensitivity of biannual MRI alone was 88.2% and annual MG plus biannual MRI was 94.1%. The cancer detection rate of biannual MRI alone was 0.7% per 100 screening episodes, which is similar to the cancer detection rate of 0.7% per 100 screening episodes for annual MG plus biannual MRI. The number of recalls and biopsies needed to detect one cancer by biannual MRI were 2.8 and 1.7 in BRCA1 carriers, 12.0 and 8.0 in BRCA2 carriers, and 11.7 and 5.0 in non-BRCA1/2 carriers, respectively. Conclusions: Biannual MRI performed well for early detection of invasive breast cancer in genomically stratified high-risk women. No benefit was associated with annual MG screening plus biannual MRI screening. See related commentary by Kuhl and Schrading, p. 1693
- Published
- 2019
44. Targeted long-read sequencing identifies missing disease-causing variation
- Author
-
Tim Cherry, Seth J. Perlman, Rando Allikmets, Christina Lam, Katrina M Dipple, Alexias Safi, Hailey Loucks, Penny M Chow, Ian A. Glass, Xue Zou, Heather C Mefford, Angela Sun, Deborah A. Nickerson, Danny E. Miller, Dawn L. Earl, James T. Bennett, Alexandra P. Lewis, Stephanie Austin, Margaret P Adam, Apoorva K Iyengar, Arvis Sulovari, Edith P Almanza Fuerte, Andrew S. Allen, Audrey Squire, Karynne E. Patterson, Erin Huggins, Winston Lee, William H. Majoros, Emily S Bonkowski, Tianyun Wang, Priya S. Kishnani, Robin L. Bennett, Mary Claire King, Tara L. Wenger, Erika Beckman, Kendra Hoekzema, Gregory E. Crawford, Timothy E. Reddy, Evan E. Eichler, Irene Chang, Anne V. Hing, Zoe Nelson, Thomas J. Walsh, Dan Doherty, Megan C. Sikes, Michael J. Bamshad, Catherine R Paschal, Jessica X. Chong, Jenny Thies, and Katherine M. Munson
- Subjects
Male ,DNA Copy Number Variations ,Computational biology ,Disease ,Biology ,Article ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Genetics ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Gene ,Genetics (clinical) ,030304 developmental biology ,Sequence (medicine) ,Data source ,Chromosome Aberrations ,0303 health sciences ,Genome, Human ,Genetic Diseases, Inborn ,High-Throughput Nucleotide Sequencing ,Sequence Analysis, DNA ,Phenotype ,Karyotyping ,Mutation (genetic algorithm) ,Cytogenetic Analysis ,Mutation ,Mendelian inheritance ,symbols ,Female ,Nanopore sequencing ,030217 neurology & neurosurgery - Abstract
Despite widespread clinical genetic testing, many individuals with suspected genetic conditions lack a precise diagnosis, limiting their opportunity to take advantage of state-of-the-art treatments. In some cases, testing reveals difficult-to-evaluate structural differences, candidate variants that do not fully explain the phenotype, single pathogenic variants in recessive disorders, or no variants in genes of interest. Thus, there is a need for better tools to identify a precise genetic diagnosis in individuals when conventional testing approaches have been exhausted. We performed targeted long-read sequencing (T-LRS) using adaptive sampling on the Oxford Nanopore platform on 40 individuals, 10 of whom lacked a complete molecular diagnosis. We computationally targeted up to 151 Mbp of sequence per individual and searched for pathogenic substitutions, structural variants, and methylation differences using a single data source. We detected all genomic aberrations-including single-nucleotide variants, copy number changes, repeat expansions, and methylation differences-identified by prior clinical testing. In 8/8 individuals with complex structural rearrangements, T-LRS enabled more precise resolution of the mutation, leading to changes in clinical management in one case. In ten individuals with suspected Mendelian conditions lacking a precise genetic diagnosis, T-LRS identified pathogenic or likely pathogenic variants in six and variants of uncertain significance in two others. T-LRS accurately identifies pathogenic structural variants, resolves complex rearrangements, and identifies Mendelian variants not detected by other technologies. T-LRS represents an efficient and cost-effective strategy to evaluate high-priority genes and regions or complex clinical testing results.
- Published
- 2021
45. Cisplatin +/− rucaparib after preoperative chemotherapy in patients with triple-negative or BRCA mutated breast cancer
- Author
-
Susan M. Perkins, Cynthia X. Ma, Thomas J. Walsh, Michael A. Danso, Kathy D. Miller, Maitri Kalra, Paula Silverman, Yan Tong, David R. Jones, Sunil Badve, and Mary Claire King
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Population ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Internal medicine ,medicine ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,education ,Rucaparib ,Lymph node ,Cancer genetics ,RC254-282 ,Neoadjuvant therapy ,Cisplatin ,education.field_of_study ,Chemotherapy ,business.industry ,BRCA mutation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Translational research ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,business ,medicine.drug - Abstract
Patients with triple-negative breast cancer (TNBC) who have residual disease after neoadjuvant therapy have a high risk of recurrence. We tested the impact of DNA-damaging chemotherapy alone or with PARP inhibition in this high-risk population. Patients with TNBC or deleterious BRCA mutation (TNBC/BRCAmut) who had >2 cm of invasive disease in the breast or persistent lymph node (LN) involvement after neoadjuvant therapy were assigned 1:1 to cisplatin alone or with rucaparib. Germline mutations were identified with BROCA analysis. The primary endpoint was 2-year disease-free survival (DFS) with 80% power to detect an HR 0.5. From Feb 2010 to May 2013, 128 patients were enrolled. Median tumor size at surgery was 1.9 cm (0–11.5 cm) with 1 (0–38) involved LN; median Residual Cancer Burden (RCB) score was 2.6. Six patients had known deleterious BRCA1 or BRCA2 mutations at study entry, but BROCA identified deleterious mutations in 22% of patients with available samples. Toxicity was similar in both arms. Despite frequent dose reductions (21% of patients) and delays (43.8% of patients), 73% of patients completed planned cisplatin. Rucaparib exposure was limited with median concentration 275 (82–4694) ng/mL post-infusion on day 3. The addition of rucaparib to cisplatin did not increase 2-year DFS (54.2% cisplatin vs. 64.1% cisplatin + rucaparib; P = 0.29). In the high-risk post preoperative TNBC/BRCAmut setting, the addition of low-dose rucaparib did not improve 2-year DFS or increase the toxicity of cisplatin. Genetic testing was underutilized in this high-risk population.
- Published
- 2021
46. Inherited predisposition to breast cancer in the Carolina Breast Cancer Study
- Author
-
Mary Claire King, Andrew F. Olshan, Ming K. Lee, Suleyman Gulsuner, H. Shelton Earp, Tom Walsh, Charles M. Perou, and Melissa A. Troester
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,PALB2 ,Population ,Article ,Inherited Predisposition ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,medicine ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,education ,skin and connective tissue diseases ,Cancer genetics ,RC254-282 ,education.field_of_study ,Carolina Breast Cancer Study ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Odds ratio ,medicine.disease ,Confidence interval ,Tumor Subtype ,030104 developmental biology ,030220 oncology & carcinogenesis ,business - Abstract
The Carolina Breast Cancer Study (CBCS) phases I–II was a case-control study of biological and social risk factors for invasive breast cancer that enrolled cases and controls between 1993 and 1999. Case selection was population-based and stratified by ancestry and age at diagnosis. Controls were matched to cases by age, self-identified race, and neighborhood of residence. Sequencing genomic DNA from 1370 cases and 1635 controls yielded odds ratios (with 95% confidence limits) for breast cancer of all subtypes of 26.7 (3.59, 189.1) for BRCA1, 8.8 (3.44, 22.48) for BRCA2, and 9.0 (2.06, 39.60) for PALB2; and for triple-negative breast cancer (TNBC) of 55.0 (7.01, 431.4) for BRCA1, 12.1 (4.18, 35.12) for BRCA2, and 10.8 (1.97, 59.11) for PALB2. Overall, 5.6% of patients carried a pathogenic variant in BRCA1, BRCA2, PALB2, or TP53, the four most highly penetrant breast cancer genes. Analysis of cases by tumor subtype revealed the expected association of TNBC versus other tumor subtypes with BRCA1, and suggested a significant association between TNBC versus other tumor subtypes with BRCA2 or PALB2 among African-American (AA) patients [2.95 (1.18, 7.37)], but not among European-American (EA) patients [0.62 (0.18, 2.09)]. AA patients with pathogenic variants in BRCA2 or PALB2 were 11 times more likely to be diagnosed with TNBC versus another tumor subtype than were EA patients with pathogenic variants in either of these genes (P = 0.001). If this pattern is confirmed in other comparisons of similarly ascertained AA and EA breast cancer patients, it could in part explain the higher prevalence of TNBC among AA breast cancer patients.
- Published
- 2021
47. P478. Functional Characterization of Rare and Protein-Damaging Mutations in Patients With Sporadic Schizophrenia Using CRISPRi in iPSCs
- Author
-
Anna Sunshine, Suleyman Gulsuner, Tom Walsh, Mary-Claire King, and Jon McClellan
- Subjects
Biological Psychiatry - Published
- 2022
48. Telomere biology disorder prevalence and phenotypes in adults with familial hematologic and/or pulmonary presentations
- Author
-
Aliya N. Husain, Mary Claire King, Sandeep Gurbuxani, Padma Sheila Rajagopal, Hari Prasanna Subramanian, Robert H. Collins, Mary E. Strek, Ayodeji Adegunsoye, Soma Das, Daniela del Gaudio, Danijela Mojsilovic, Rekha Vij, Jane E. Churpek, Lucy A. Godley, Zejuan Li, Peter L. Greenberg, Raymond H. Kim, Jeremy P. Segal, Suleyman Gulsuner, Simone Feurstein, Steven D. Gore, Afaf Osman, Allison H. West DePersia, and Tom Walsh
- Subjects
0301 basic medicine ,Proband ,medicine.medical_specialty ,Population ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Prevalence ,Copy-number variation ,Family history ,education ,Biology ,Telomerase ,In Situ Hybridization, Fluorescence ,Genetic testing ,education.field_of_study ,Hematology ,Myeloid Neoplasia ,medicine.diagnostic_test ,business.industry ,Bone marrow failure ,Telomere ,medicine.disease ,Penetrance ,030104 developmental biology ,Phenotype ,Immunology ,Mutation ,business ,030215 immunology - Abstract
Telomere biology disorders (TBDs) present heterogeneously, ranging from infantile bone marrow failure associated with very short telomeres to adult-onset interstitial lung disease (ILD) with normal telomere length. Yield of genetic testing and phenotypic spectra for TBDs caused by the expanding list of telomere genes in adults remain understudied. Thus, we screened adults aged ≥18 years with a personal and/or family history clustering hematologic disorders and/or ILD enrolled on The University of Chicago Inherited Hematologic Disorders Registry for causative variants in 13 TBD genes. Sixteen (10%) of 153 probands carried causative variants distributed among TERT (n = 6), TERC (n = 4), PARN (n = 5), or RTEL1 (n = 1), of which 19% were copy number variants. The highest yield (9 of 22 [41%]) was in families with mixed hematologic and ILD presentations, suggesting that ILD in hematology populations and hematologic abnormalities in ILD populations warrant TBD genetic testing. Four (3%) of 117 familial hematologic disorder families without ILD carried TBD variants, making TBD second to only DDX41 in frequency for genetic diagnoses in this population. Phenotypes of 17 carriers with heterozygous PARN variants included 4 (24%) with hematologic abnormalities, 67% with lymphocyte telomere lengths measured by flow cytometry and fluorescence in situ hybridization at or above the 10th percentile, and a high penetrance for ILD. Alternative etiologies for cytopenias and/or ILD such as autoimmune features were noted in multiple TBD families, emphasizing the need to maintain clinical suspicion for a TBD despite the presence of alternative explanations.
- Published
- 2020
49. Genomic analysis of inherited hearing loss in the Palestinian population
- Author
-
Hashem Shahin, Christina Canavati, Fouad Zahdeh, Karen B. Avraham, Tamara Jaraysa, Grace Rabie, Amal Abu Rayyan, Ming K. Lee, Suleyman Gulsuner, Tom Walsh, Silvia Casadei, Mary Claire King, Lara Kamal, Dima Dweik, Zippora Brownstein, Ryan J. Carlson, and Moien Kanaan
- Subjects
0301 basic medicine ,Adult ,Male ,Adolescent ,Hearing loss ,Population ,Consanguinity ,030105 genetics & heredity ,Biology ,medicine.disease_cause ,03 medical and health sciences ,Middle East ,Young Adult ,medicine ,Humans ,Allele ,education ,Child ,Hearing Loss ,Gene ,Alleles ,Genetics ,Mutation ,education.field_of_study ,Multidisciplinary ,Genetic heterogeneity ,Exons ,Genomics ,Middle Aged ,Biological Sciences ,Human genetics ,Pedigree ,030104 developmental biology ,Child, Preschool ,Female ,medicine.symptom - Abstract
The genetic characterization of a common phenotype for an entire population reveals both the causes of that phenotype for that place and the power of family-based, population-wide genomic analysis for gene and mutation discovery. We characterized the genetics of hearing loss throughout the Palestinian population, enrolling 2,198 participants from 491 families from all parts of the West Bank and Gaza. In Palestinian families with no prior history of hearing loss, we estimate that 56% of hearing loss is genetic and 44% is not genetic. For the great majority (87%) of families with inherited hearing loss, panel-based genomic DNA sequencing, followed by segregation analysis of large kindreds and transcriptional analysis of participant RNA, enabled identification of the causal genes and mutations, including at distant noncoding sites. Genetic heterogeneity of hearing loss was striking with respect to both genes and alleles: The 337 solved families harbored 143 different mutations in 48 different genes. For one in four solved families, a transcription-altering mutation was the responsible allele. Many of these mutations were cryptic, either exonic alterations of splice enhancers or silencers or deeply intronic events. Experimentally calibrated in silico analysis of transcriptional effects yielded inferences of high confidence for effects on splicing even of mutations in genes not expressed in accessible tissue. Most (58%) of all hearing loss in the population was attributable to consanguinity. Given the ongoing decline in consanguineous marriage, inherited hearing loss will likely be much rarer in the next generation.
- Published
- 2020
50. Spectrum of genes for inherited hearing loss in the Israeli Jewish population, including the novel human deafness geneATOH1
- Author
-
Noam Shomron, Mor Bordeynik-Cohen, Lara Kamal, Dror Gilony, Ryan J. Carlson, Morad Khayat, Asgeir Orn Arnporsson, Tom Walsh, Silvia Casadei, Naama Zvi, Noga Lipschitz, Hana Poran, Michal Sagi, Maria Birkan, Weise Chang, Ory Madgar, Amihood Singer, Shahar Taiber, Ronna Hertzano, Noa Ruhrman-Shahar, Ophir Handzel, Eiríkur Steingrímsson, Moien Kanaan, Michael Wolf, Hagit Baris-Feldman, Amir Peleg, Chana Vinkler, Bella Davidov, Michal Macarov, Stavit Allon-Shalev, Nadra Samara, Ming Lee, Reuven Sharony, Meirav Sokolov, Elon Pras, Karen B. Avraham, Zippora Brownstein, Fabio Tadeu Arrojo Martins, Efrat Sofrin, Matthew W. Kelley, Dorit Lev, Mordechai Shohat, Moshe Frydman, Lina Basel-Salmon, Ofer Isakov, Mary Claire King, Nada Danial-Farran, Amal Abu Rayyan, Suleyman Gulsuner, and Doaa Ali-Naffaa
- Subjects
Genetics ,Newborn screening ,education.field_of_study ,Hearing loss ,Genetic counseling ,Population ,Biology ,Phenotype ,Genotype ,otorhinolaryngologic diseases ,medicine ,Allele ,medicine.symptom ,education ,Gene - Abstract
Mutations in more than 150 genes are responsible for inherited hearing loss, with thousands of different, severe causal alleles that vary among populations. The Israeli Jewish population includes communities of diverse geographic origins, revealing a wide range of deafness-associated variants and enabling clinical characterization of the associated phenotypes. Our goal was to identify the genetic causes of inherited hearing loss in this population, and to determine relationships among genotype, phenotype, and ethnicity. Genomic DNA samples from informative relatives of 88 multiplex families, all of self-identified Jewish ancestry, with either non-syndromic or syndromic hearing loss, were sequenced for known and candidate deafness genes using the HEar-Seq gene panel. The genetic causes of hearing loss were identified for 60% of the families. One gene was encountered for the first time in human hearing loss:ATOH1(Atonal), a basic helix-loop-helix transcription factor responsible for autosomal dominant progressive hearing loss in a five-generation family. Our results demonstrate that genomic sequencing with a gene panel dedicated to hearing loss is effective for genetic diagnoses in a diverse population. Comprehensive sequencing enables well-informed genetic counseling and clinical management by medical geneticists, otolaryngologists, audiologists, and speech therapists and can be integrated into newborn screening for deafness.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.