395 results on '"Margaret A. Tucker"'
Search Results
2. Data from Use of Nonsteroidal Anti-Inflammatory Drugs and Incidence of Melanoma in the United States Radiologic Technologists Study
- Author
-
Elizabeth K. Cahoon, Margaret A. Tucker, Martha S. Linet, Bruce H. Alexander, Mark P. Little, Michael R. Sargen, Cari M. Kitahara, and Jim Z. Mai
- Abstract
Although NSAIDs have been associated with both reduced and increased cutaneous melanoma risk, few studies have examined these associations by ultraviolet radiation (UVR) or personal sun-sensitivity. We examined the associations between NSAID use and first primary invasive cutaneous melanoma among 58,227 non-Hispanic white participants in the United States Radiologic Technologists cohort study. Poisson regression was used to calculate rate ratios (RR) and 95% likelihood-based confidence intervals (CI), adjusting for attained age, birth cohort, and ambient UVR. No significant association of melanoma was observed for any use of NSAIDs (RR, 0.87; 95% CI, 0.71–1.09). The relative risks of melanoma for the highest categories of aspirin and other NSAID use (≥5 times per month vs. none) were 0.93 (95% CI, 0.74–1.16) and 1.02 (95% CI, 0.83–1.25), respectively. Further analyses did not reveal dose–response for trends in frequency of NSAID use or interactions with sex, UVR, eye and hair color, and skin complexion. In this large nationwide study, NSAID use was not associated with melanoma risk.Prevention Relevance:NSAIDs have been associated with both reduced and increased melanoma risk. However, few studies have examined the role of UVR or personal sun-sensitivity on these associations. Our findings strengthen the evidence that NSAID use is not associated with melanoma risk, even in sun-sensitive subgroups.
- Published
- 2023
3. Data from Pooling Prospective Studies to Investigate the Etiology of Second Cancers
- Author
-
Lindsay M. Morton, Amy Berrington de Gonzalez, Joshua N. Sampson, Joanne Elena, Rochelle E. Curtis, Anne Zeleniuch-Jacquotte, James R. Cerhan, Robert N. Hoover, Margaret A. Tucker, Patricia Hartge, Joseph F. Fraumeni, Mark P. Purdue, Gabriella Andreotti, Laura E. Beane Freeman, Stephanie J. Weinstein, Demetrius Albanes, Kim Robien, Yikyung Park, Meredith S. Shiels, Todd M. Gibson, and Amanda Black
- Abstract
Background: With over 13 million cancer survivors in the United States today, second cancers are of rapidly growing importance. However, data on nontreatment risk factors for second cancers are sparse. We explored the feasibility of pooling data from cohort studies of cancer incidence to investigate second cancer etiology.Methods: We combined data from five prospective studies including more than 800,000 individuals. We compared study designs and populations; evaluated availability of and ability to harmonize risk factor data; compared incidence and survival for common first primary malignancies and incidence of second primary malignancies; and estimated sample size requirements.Results: Overall, 96,513 incident, first primary malignancies were diagnosed during 1985 to 2009. Incidence rates and survival following the first primary varied among the cohorts, but most of the heterogeneity could be explained by characteristics of the study populations (age, sex, smoking, and screening rates). A total of 7,890 second primary cancers (excluding original primary site) were identified, yielding sufficient statistical power (≥80%) for detecting modest associations with risk of all second cancers among survivors of common first primary malignancies (e.g., colorectal cancer); however, there were insufficient events for studying survivors of rarer cancers or identifying risk factors for specific second cancers.Conclusions: Pooling data from cohort studies to investigate nontreatment risk factors for second primary cancers seems feasible but there are important methodologic issues—some of which are barriers to specific research questions—that require special attention.Impact: Increased understanding of nontreatment risk factors for second cancers will provide valuable prevention and surveillance information. Cancer Epidemiol Biomarkers Prev; 23(8); 1598–608. ©2014 AACR.
- Published
- 2023
4. Data from MicroRNA Expression Differentiates Histology and Predicts Survival of Lung Cancer
- Author
-
Ena Wang, Lisa M. McShane, Neil E. Caporaso, Angela C. Pesatori, Pier Alberto Bertazzi, Margaret A. Tucker, Francesco M. Marincola, Ilona Linnoila, Alisa M. Goldstein, Maurizia Rubagotti, Andrew W. Bergen, Hui Liu, Jill Koshiol, Melissa Rotunno, Yingdong Zhao, and Maria Teresa Landi
- Abstract
Purpose: The molecular drivers that determine histology in lung cancer are largely unknown. We investigated whether microRNA (miR) expression profiles can differentiate histologic subtypes and predict survival for non–small cell lung cancer.Experimental Design: We analyzed miR expression in 165 adenocarcinoma and 125 squamous cell carcinoma (SQ) tissue samples from the Environment And Genetics in Lung cancer Etiology (EAGLE) study using a custom oligo array with 440 human mature antisense miRs. We compared miR expression profiles using t tests and F tests and accounted for multiple testing using global permutation tests. We assessed the association of miR expression with tobacco smoking using Spearman correlation coefficients and linear regression models, and with clinical outcome using log-rank tests, Cox proportional hazards, and survival risk prediction models, accounting for demographic and tumor characteristics.Results: MiR expression profiles strongly differed between adenocarcinoma and SQ (Pglobal < 0.0001), particularly in the early stages, and included miRs located on chromosome loci most often altered in lung cancer (e.g., 3p21-22). Most miRs, including all members of the let-7 family, were downregulated in SQ. Major findings were confirmed by quantitative real time-polymerase chain reaction (qRT-PCR) in EAGLE samples and in an independent set of lung cancer cases. In SQ, the low expression of miRs that are downregulated in the histology comparison was associated with 1.2- to 3.6-fold increased mortality risk. A five-miR signature significantly predicted survival for SQ.Conclusions: We identified a miR expression profile that strongly differentiated adenocarcinoma from SQ and had prognostic implications. These findings may lead to histology-based therapeutic approaches. Clin Cancer Res; 16(2); 430–41
- Published
- 2023
5. Supplementary Figure 1 from Pooling Prospective Studies to Investigate the Etiology of Second Cancers
- Author
-
Lindsay M. Morton, Amy Berrington de Gonzalez, Joshua N. Sampson, Joanne Elena, Rochelle E. Curtis, Anne Zeleniuch-Jacquotte, James R. Cerhan, Robert N. Hoover, Margaret A. Tucker, Patricia Hartge, Joseph F. Fraumeni, Mark P. Purdue, Gabriella Andreotti, Laura E. Beane Freeman, Stephanie J. Weinstein, Demetrius Albanes, Kim Robien, Yikyung Park, Meredith S. Shiels, Todd M. Gibson, and Amanda Black
- Abstract
PDF file - 217KB, Supplementary Figure 1. Baseline characteristics of participants for each prospective study, by first primary malignancy diagnosis.
- Published
- 2023
6. Supplementary Tables 1 - 3 from Pooling Prospective Studies to Investigate the Etiology of Second Cancers
- Author
-
Lindsay M. Morton, Amy Berrington de Gonzalez, Joshua N. Sampson, Joanne Elena, Rochelle E. Curtis, Anne Zeleniuch-Jacquotte, James R. Cerhan, Robert N. Hoover, Margaret A. Tucker, Patricia Hartge, Joseph F. Fraumeni, Mark P. Purdue, Gabriella Andreotti, Laura E. Beane Freeman, Stephanie J. Weinstein, Demetrius Albanes, Kim Robien, Yikyung Park, Meredith S. Shiels, Todd M. Gibson, and Amanda Black
- Abstract
PDF file - 23KB, Supplemental Table 1. Characteristics of five prospective epidemiologic cohort studies in this pooled analysis investigating feasibility of studying risk factors for second cancers. Supplemental Table 2. Population-specific standardized incidence ratios^ (SIRs) for first primary malignancies by cohort. Supplemental Table 3. Age-, sex-, and race-standardized incidence rates^ (per 10,000 person-years) for first primary malignancies by cohort.
- Published
- 2023
7. Supplementary Data from MicroRNA Expression Differentiates Histology and Predicts Survival of Lung Cancer
- Author
-
Ena Wang, Lisa M. McShane, Neil E. Caporaso, Angela C. Pesatori, Pier Alberto Bertazzi, Margaret A. Tucker, Francesco M. Marincola, Ilona Linnoila, Alisa M. Goldstein, Maurizia Rubagotti, Andrew W. Bergen, Hui Liu, Jill Koshiol, Melissa Rotunno, Yingdong Zhao, and Maria Teresa Landi
- Abstract
Supplementary Data from MicroRNA Expression Differentiates Histology and Predicts Survival of Lung Cancer
- Published
- 2023
8. Targeted long-read sequencing of the Ewing sarcoma 6p25.1 susceptibility locus identifies germline-somatic interactions with EWSR1-FLI1 binding
- Author
-
Olivia W. Lee, Calvin Rodrigues, Shu-Hong Lin, Wen Luo, Kristine Jones, Derek W. Brown, Weiyin Zhou, Eric Karlins, Sairah M. Khan, Sylvain Baulande, Virginie Raynal, Didier Surdez, Stephanie Reynaud, Rebeca Alba Rubio, Sakina Zaidi, Sandrine Grossetête, Stelly Ballet, Eve Lapouble, Valérie Laurence, Gaelle Pierron, Nathalie Gaspar, Nadège Corradini, Perrine Marec-Bérard, Nathaniel Rothman, Casey L. Dagnall, Laurie Burdett, Michelle Manning, Kathleen Wyatt, Meredith Yeager, Raj Chari, Wendy M. Leisenring, Andreas E. Kulozik, Jennifer Kriebel, Thomas Meitinger, Konstantin Strauch, Thomas Kirchner, Uta Dirksen, Lisa Mirabello, Margaret A. Tucker, Franck Tirode, Gregory T. Armstrong, Smita Bhatia, Leslie L. Robison, Yutaka Yasui, Laura Romero-Pérez, Wolfgang Hartmann, Markus Metzler, W. Ryan Diver, Adriana Lori, Neal D. Freedman, Robert N. Hoover, Lindsay M. Morton, Stephen J. Chanock, Thomas G.P. Grünewald, Olivier Delattre, and Mitchell J. Machiela
- Subjects
Genetics ,Medizin ,Genetics (clinical) - Published
- 2023
9. Gene-Level Associations in Patients With and Without Pathogenic Germline Variants in
- Author
-
Esteban, Astiazaran-Symonds, Cole, Graham, Jung, Kim, Margaret A, Tucker, Christian, Ingvar, Hildur, Helgadottir, Lorenza, Pastorino, Remco, van Doorn, Joshua N, Sampson, Bin, Zhu, William, Bruno, Paola, Queirolo, Giuseppe, Fornarini, Stefania, Sciallero, Brian, Carter, Belynda, Hicks, Amy, Hutchinson, Kristine, Jones, Douglas R, Stewart, Stephen J, Chanock, Neal D, Freedman, Maria Teresa, Landi, Veronica, Höiom, Susana, Puig, Nelleke, Gruis, Xiaohong R, Yang, Paola, Ghiorzo, and Alisa M, Goldstein
- Subjects
Pancreatic Neoplasms ,Germ Cells ,Humans ,Genetic Predisposition to Disease ,Cyclin-Dependent Kinase Inhibitor p16 ,Carcinoma, Pancreatic Ductal ,Cyclin-Dependent Kinase Inhibitor Proteins - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a component of familial melanoma due to germline pathogenic variants (GPVs) inWe analyzed 189 cancer predisposition genes using parametric rare-variant association (RVA) tests and nonparametric permutation tests to identify gene-level associations in PDAC for patients with (In RVA tests,These results suggest that variants in other genes likely play a role in PDAC in all patients and that PDAC in
- Published
- 2022
10. Gene-Level Associations in Patients With and Without Pathogenic Germline Variants in CDKN2A and Pancreatic Cancer
- Author
-
Esteban Astiazaran-Symonds, Cole Graham, Jung Kim, Margaret A. Tucker, Christian Ingvar, Hildur Helgadottir, Lorenza Pastorino, Remco van Doorn, Joshua N. Sampson, Bin Zhu, William Bruno, Paola Queirolo, Giuseppe Fornarini, Stefania Sciallero, Brian Carter, Belynda Hicks, Amy Hutchinson, Kristine Jones, Douglas R. Stewart, Stephen J. Chanock, Neal D. Freedman, Maria Teresa Landi, Veronica Höiom, Susana Puig, Nelleke Gruis, Xiaohong R. Yang, Paola Ghiorzo, and Alisa M. Goldstein
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is a component of familial melanoma due to germline pathogenic variants (GPVs) in CDKN2A. However, it is unclear what role this gene or other genes play in its etiology. MATERIALS AND METHODS We analyzed 189 cancer predisposition genes using parametric rare-variant association (RVA) tests and nonparametric permutation tests to identify gene-level associations in PDAC for patients with ( CDKN2A +) and without ( CDKN2A–) GPV. Exome sequencing was performed on 84 patients with PDAC, 47 CDKN2A+ and 37 CDKN2A–. After variant filtering, various RVA tests and permutation tests were run separately by CDKN2A status. Genes with the strongest nominal associations were evaluated in patients with PDAC from The Cancer Genome Atlas and the UK Biobank (UKB). A secondary analysis including only GPV from UKB was also performed. RESULTS In RVA tests, ERCC4 and RET showed the most compelling evidence as plausible PDAC candidate genes for CDKN2A+ patients. In contrast, the findings in CDKN2A– patients provided evidence for HMBS, EPCAM, and MRE11 as potential new candidate genes and confirmed ATM, BRCA2, and PALB2 as PDAC genes, consistent with findings in The Cancer Genome Atlas and the UKB. As expected, CDKN2A– patients were more likely to harbor GPVs from the 189 genes investigated. When including only GPVs from UKB, significant associations with PDAC were seen for ATM, BRCA2, and CDKN2A. CONCLUSION These results suggest that variants in other genes likely play a role in PDAC in all patients and that PDAC in CDKN2A+ patients has a distinct etiology from PDAC in CDKN2A– patients.
- Published
- 2022
11. Use of nonsteroidal anti-inflammatory drugs and incidence of melanoma in the United States Radiologic Technologists study
- Author
-
Jim Z. Mai, Cari M. Kitahara, Michael R. Sargen, Mark P. Little, Bruce H. Alexander, Martha S. Linet, Margaret A. Tucker, and Elizabeth K. Cahoon
- Subjects
Cancer Research ,Likelihood Functions ,Skin Neoplasms ,Ultraviolet Rays ,Incidence ,Anti-Inflammatory Agents, Non-Steroidal ,Article ,United States ,Cohort Studies ,Oncology ,Risk Factors ,Humans ,Prospective Studies ,Melanoma - Abstract
Although NSAIDs have been associated with both reduced and increased cutaneous melanoma risk, few studies have examined these associations by ultraviolet radiation (UVR) or personal sun-sensitivity. We examined the associations between NSAID use and first primary invasive cutaneous melanoma among 58,227 non-Hispanic white participants in the United States Radiologic Technologists cohort study. Poisson regression was used to calculate rate ratios (RR) and 95% likelihood-based confidence intervals (CI), adjusting for attained age, birth cohort, and ambient UVR. No significant association of melanoma was observed for any use of NSAIDs (RR, 0.87; 95% CI, 0.71–1.09). The relative risks of melanoma for the highest categories of aspirin and other NSAID use (≥5 times per month vs. none) were 0.93 (95% CI, 0.74–1.16) and 1.02 (95% CI, 0.83–1.25), respectively. Further analyses did not reveal dose–response for trends in frequency of NSAID use or interactions with sex, UVR, eye and hair color, and skin complexion. In this large nationwide study, NSAID use was not associated with melanoma risk. Prevention Relevance: NSAIDs have been associated with both reduced and increased melanoma risk. However, few studies have examined the role of UVR or personal sun-sensitivity on these associations. Our findings strengthen the evidence that NSAID use is not associated with melanoma risk, even in sun-sensitive subgroups.
- Published
- 2022
12. Impact of Transcript (p16/p14ARF) Alteration on Cancer Risk in CDKN2A Germline Pathogenic Variant Carriers
- Author
-
Michael R Sargen, Hildur Helgadottir, Xiaohong R Yang, Mark Harland, Jessica N Hatton, Kristine Jones, Belynda D Hicks, Amy Hutchinson, Michael Curry, Margaret A Tucker, Alisa M Goldstein, and Ruth M Pfeiffer
- Subjects
Cancer Research ,Squamous Cell Carcinoma of Head and Neck ,Prevention ,Article ,United States ,Rare Diseases ,Oncology ,Head and Neck Neoplasms ,Risk Factors ,Clinical Research ,Tumor Suppressor Protein p14ARF ,Humans ,Digestive Diseases ,Melanoma ,Lung ,Cyclin-Dependent Kinase Inhibitor p16 ,Aged ,Cancer - Abstract
Background Few studies have evaluated the relationship between CDKN2A germline pathogenic variants (GPV), transcript (p16/p14ARF) alteration, and cancer risk. Methods Standardized incidence ratios (SIRs) comparing cancer risk with the general population were calculated for 385 CDKN2A GPV carriers from 2 large cohorts (259 United States and 126 Swedish individuals) using Poisson regression; statistical significance was defined as P less than .002 (Bonferroni correction). Cumulative incidence is reported for melanoma and nonmelanoma cancer. Results Incidence was increased for melanoma (SIR = 159.8, 95% confidence interval [CI] = 132.1 to 193.2), pancreatic cancer (SIR = 24.1, 95% CI = 14.7 to 39.4), head and neck squamous cell carcinoma (SIR = 16.2, 95% CI = 9.5 to 27.6), and lung cancer (SIR = 5.6, 95% CI = 3.4 to 9.1) in GPV carriers. Similar associations were observed with p16 alteration. Combined p16 and p14ARF alteration was associated with increased incidence of esophageal cancer (SIR = 16.7, 95% CI = 5.7 to 48.9) and malignant peripheral nerve sheath tumor (SIR = 113.0, 95% CI = 16.4 to 780.9), although cancer events were limited (n Conclusion These findings highlight the impact of p16 and p14ARF alteration on cancer risk. Smoking was an important risk factor for smoking-related cancers in our study.
- Published
- 2022
13. Novel MAPK/AKT-impairing germline NRAS variant identified in a melanoma-prone family
- Author
-
Jung Kim, Ketty Peris, Kristie Jones, Eduardo Nagore, Alex Stratigos, Paola Ghiorzo, Bin Zhu, Kevin M. Brown, Belynda Hicks, Mai Xu, Hyunbum Jang, Donato Calista, Dario Consonni, Laura Mendoza, Ruth Nussinov, Douglas R. Stewart, Maria Teresa Landi, Xiaohong Rose Yang, Mingzhen Zhang, Nicholas K. Hayward, Thorkell Andresson, Chiara Menin, Maria Concetta Fargnoli, Michael R. Sargen, Alisa M. Goldstein, Margaret A. Tucker, Susana Puig, Angela Cecilia Pesatori, and Tongwu Zhang
- Subjects
MAPK/ERK pathway ,Proto-Oncogene Proteins B-raf ,Neuroblastoma RAS viral oncogene homolog ,Cancer Research ,Skin Neoplasms ,Familial cancer ,In vitro characterization ,Melanoma ,Molecular modeling ,NRAS gene ,Rare variant ,GTPase ,Biology ,Article ,Germline ,GTP Phosphohydrolases ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Gene ,Protein kinase B ,Germ-Line Mutation ,Genetics (clinical) ,Exome sequencing ,Membrane Proteins ,medicine.disease ,Human genetics ,Oncology ,Cancer research ,Guanosine Triphosphate ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,Proto-Oncogene Proteins c-akt - Abstract
While several high-penetrance melanoma risk genes are known, variation in these genes fail to explain melanoma susceptibility in a large proportion of high-risk families. As part of a melanoma family sequencing study, including 435 families from Mediterranean populations, we identified a novel NRAS variant (c.170A>C, p.D57A) in a melanoma-prone family. This variant is absent in exomes in gnomAD, ESP, UKBiobank, and the 1000 Genomes Project, as well as in 11 273 Mediterranean individuals and 109 melanoma-prone families from the US and Australia. This variant occurs in the GTP-binding pocket of NRAS. Differently from other RAS activating alterations, NRAS D57A expression is unable to activate MAPK-pathway both constitutively and after stimulation but enhances EGF-induced PI3K-pathway signaling in serum starved conditions in vitro. Consistent with in vitro data demonstrating that NRAS D57A does not enrich GTP binding, molecular modeling suggests that the D57A substitution would be expected to impair Mg2+ binding and decrease nucleotide-binding and GTPase activity of NRAS. While we cannot firmly establish NRAS c.170A>C (p.D57A) as a melanoma susceptibility variant, further investigation of NRAS as a familial melanoma gene is warranted.
- Published
- 2021
14. The Impact of Longitudinal Surveillance on Tumor Thickness for Melanoma-Prone Families with and without Pathogenic Germline Variants of CDKN2A and CDK4
- Author
-
Michael R. Sargen, Xiaohong R. Yang, Alisa M. Goldstein, Margaret A. Tucker, Ruth M. Pfeiffer, and David E. Elder
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,education.field_of_study ,Epidemiology ,business.industry ,Melanoma ,Population ,Familial Melanoma ,medicine.disease ,Germline ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,CDKN2A ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Stage (cooking) ,education ,business ,Generalized estimating equation - Abstract
Background: Skin cancer screening is routinely performed for members of melanoma-prone families, but longitudinal studies evaluating the efficacy of surveillance in this high-risk population are lacking. Methods: We evaluated thickness for first primary melanomas diagnosed in melanoma-prone families (≥2 individuals with melanoma) enrolled in NCT00040352 (NCI familial melanoma study) from 1976 through 2014; enrolled patients received routine skin cancer screening and education about skin self-exams. We used linear and ordinal logistic regression models adjusted for gender and age with a generalized estimating equations approach to report changes in thickness and tumor (T) stage over time, comparing outcomes for NCI cases diagnosed before (pre-study) versus after study participation (prospective) and for NCI cases versus nonfamilial cases [Surveillance, Epidemiology, and End Results (SEER) 9 registries]. Results: Tumor thickness was evaluated for 293 NCI (pre-study = 246; prospective = 47) patients. Compared with NCI pre-study cases, NCI prospective melanomas were thinner (0.6 vs. 1.1 mm; P < 0.001) and more likely to be T1 stage [39/47 (83%) vs. 98/246 (40%); P < 0.001]. Similar findings (P < 0.05) were observed for familial cases with and without germline CDKN2A and CDK4 mutations. Peters–Belson modeling suggested that calendar period effects of decreasing thickness in the general population (SEER 9) did not fully explain thickness trends in NCI families. Conclusions: Participation in a longitudinal surveillance program providing skin cancer screening and education about skin self-exams was associated with thinner melanomas for members of melanoma-prone families. Impact: The study findings support the clinical benefit of screening (physician and self) for this high-risk population.
- Published
- 2021
15. Long-term risk of subsequent cancer incidence among hereditary and nonhereditary retinoblastoma survivors
- Author
-
Lindsay M. Morton, Margaret A. Tucker, David H. Abramson, Johanna M. Seddon, Sara J. Schonfeld, and Ruth A. Kleinerman
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Retinal Neoplasms ,Population ,Article ,Eye cancer ,Paediatric cancer ,03 medical and health sciences ,Cancer epidemiology ,0302 clinical medicine ,Cancer Survivors ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Cumulative incidence ,education ,education.field_of_study ,business.industry ,Retinoblastoma ,Incidence ,Incidence (epidemiology) ,Melanoma ,Neoplasms, Second Primary ,Middle Aged ,Prognosis ,medicine.disease ,United States ,Confidence interval ,Survival Rate ,030220 oncology & carcinogenesis ,Hereditary Retinoblastoma ,Female ,Sarcoma ,business ,Follow-Up Studies - Abstract
Background Increased sarcoma and melanoma risks after hereditary retinoblastoma are well established, whereas less is known about epithelial subsequent malignant neoplasms (SMNs) and risks for multiple (≥2) SMNs. Methods Leveraging long-term follow-up and detailed histologic information, we quantified incident SMN risk among 1128 hereditary and 924 nonhereditary retinoblastoma survivors (diagnosed 1914–2006; follow-up through 2016). Standardised incidence ratios (SIRs) compared cancer risk after retinoblastoma relative to the general population. We estimated cumulative incidence accounting for competing risk of death. Results Hereditary survivors had statistically significantly increased SMN risk (N = 239; SIR = 11.9; 95% confidence interval [CI] 10.4–13.5), with SIRs >80-fold for sarcomas, nasal cavity tumours and pineoblastoma. Significantly increased risks were also observed for melanoma and central nervous system, oral cavity and breast SMNs (SIRs = 3.1–17), but not the uterus, kidney, lung, bladder, pancreas or other types. Cumulative incidence 50 years following hereditary retinoblastoma was 33.1% (95% CI 29.0–37.2) for a first SMN and 6.0% (95% CI 3.8–8.2) for a second SMN. SMN risk was not increased after nonhereditary retinoblastoma (N = 25; SIR = 0.8; 95% CI 0.5–1.2). Conclusion Beyond the established sarcoma and melanoma risks after hereditary retinoblastoma, we demonstrate increased risk for a more limited number of epithelial malignancies than previously suggested. Cumulative incidence estimates emphasise long-term SMN burden after hereditary retinoblastoma.
- Published
- 2021
16. Subsequent Neoplasm Risk Associated With Rare Variants in DNA Damage Response and Clinical Radiation Sensitivity Syndrome Genes in the Childhood Cancer Survivor Study
- Author
-
Bin Zhu, Kristine Jones, Eric Karlins, Gregory T. Armstrong, Stephen W. Hartley, Belynda Hicks, Byron S. Sigel, Jeremy A. Miller, Stephen J. Chanock, Rita E. Weathers, Joshua N. Sampson, Yutaka Yasui, Smita Bhatia, Lucie M. Turcotte, Leslie L. Robison, Diana M. Merino, Margaret A. Tucker, Lindsay M. Morton, Megan N. Frone, Michael Arnold, Wendy M. Leisenring, Casey L. Dagnall, Joseph P. Neglia, Meredith Yeager, Danielle M. Karyadi, Susan A. Smith, Amy Berrington de Gonzalez, and Rebecca M. Howell
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,DNA damage ,business.industry ,Childhood cancer ,Childhood Cancer Survivor Study ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Radiation sensitivity ,030220 oncology & carcinogenesis ,Internal medicine ,Original Reports ,Medicine ,Neoplasm ,business ,Gene - Abstract
PURPOSE Radiotherapy for childhood cancer is associated with elevated subsequent neoplasm (SN) risk, but the contribution of rare variants in DNA damage response and radiation sensitivity genes to SN risk is unknown. PATIENTS AND METHODS We conducted whole-exome sequencing in a cohort of childhood cancer survivors originally diagnosed during 1970 to 1986 (mean follow-up, 32.7 years), with reconstruction of doses to body regions from radiotherapy records. We identified patients who developed SN types previously reported to be related to radiotherapy (RT-SNs; eg, basal cell carcinoma [BCC], breast cancer, meningioma, thyroid cancer, sarcoma) and matched controls (sex, childhood cancer type/diagnosis, age, SN location, radiation dose, survival). Conditional logistic regression assessed SN risk associated with potentially protein-damaging rare variants (SnpEff, ClinVar) in 476 DNA damage response or radiation sensitivity genes with exact permutation-based P values using a Bonferroni-corrected significance threshold of P < 8.06 × 10−5. RESULTS Among 5,105 childhood cancer survivors of European descent, 1,108 (21.7%) developed at least 1 RT-SN. Out-of-field RT-SN risk, excluding BCC, was associated with homologous recombination repair (HRR) gene variants (patient cases, 23.2%; controls, 10.8%; odds ratio [OR], 2.6; 95% CI, 1.7 to 3.9; P = 4.79 × 10−5), most notably but nonsignificantly for FANCM (patient cases, 4.0%; matched controls, 0.6%; P = 9.64 × 10−5). HRR variants were not associated with likely in/near-field RT-SNs, excluding BCC (patient cases, 12.7%; matched controls, 12.9%; P = .92). Irrespective of radiation dose, risk for RT-SNs was also associated with EXO1 variants (patient cases, 1.8%; controls, 0.4%; P = 3.31 × 10−5), another gene implicated in DNA double-strand break repair. CONCLUSION In this large-scale discovery study, we identified novel associations between RT-SN risk after childhood cancer and potentially protein-damaging rare variants in genes involved in DNA double-strand break repair, particularly HRR. With replication, these results could affect screening recommendations for childhood cancer survivors and risk-benefit assessments of treatment approaches.
- Published
- 2020
17. Histologic features of melanoma associated with germline mutations of CDKN2A, CDK4, and POT1 in melanoma-prone families from the United States, Italy, and Spain
- Author
-
Michael R. Sargen, Ruth M. Pfeiffer, Daniela Massi, Cristina Carrera, Donato Calista, Maria Teresa Landi, Emily Y. Chu, Paula Aguilera, Rosalie Elenitsas, Margaret A. Tucker, Alisa M. Goldstein, Xiaohong R. Yang, Miriam Potrony, David E. Elder, Llucia Alos, and Susana Puig
- Subjects
Adult ,Male ,Oncology ,Heterozygote ,medicine.medical_specialty ,Skin Neoplasms ,Telomere-Binding Proteins ,Telomere dysfunction ,Dermatology ,medicine.disease_cause ,Article ,Shelterin Complex ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,CDKN2A ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Neoplasm Invasiveness ,Melanoma ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,Germ-Line Mutation ,Skin ,Mutation ,business.industry ,Tumor-infiltrating lymphocytes ,Cyclin-Dependent Kinase 4 ,Odds ratio ,Middle Aged ,medicine.disease ,United States ,Italy ,Spain ,030220 oncology & carcinogenesis ,Female ,Histopathology ,business - Abstract
BACKGROUND: CDKN2A, CDK4, and POT1 are well-established melanoma-susceptibility genes. OBJECTIVE: We evaluated melanoma histopathology for individuals with germline mutations of CDKN2A, CDK4, and POT1. METHODS: We assessed histopathology for melanomas diagnosed in melanoma-prone families (≥2 individuals with melanoma) from the United States, Italy, and Spain. Comparisons between mutation carriers and non-carriers (no mutation) were adjusted for age, sex, Breslow depth, and correlations among individuals within the same family. RESULTS: Histologic slides were evaluated for 290 melanomas (139 from 132 non-carriers, 122 from 68 CDKN2A carriers, 10 from 6 CDK4 carriers, 19 from 16 POT1 carriers). Superficial spreading was the predominant subtype for all groups. Spitzoid morphology (>25% of tumor) was observed in the majority of invasive melanomas from POT1 carriers (10 of 15 [67%], P
- Published
- 2020
18. Reproductive factors, hormone use, and incidence of melanoma in a cohort of US Radiologic Technologists
- Author
-
Jim Z Mai, Rui Zhang, Michael R Sargen, Mark P Little, Bruce H Alexander, Margaret A Tucker, Cari M Kitahara, and Elizabeth K Cahoon
- Subjects
Male ,Likelihood Functions ,Skin Neoplasms ,Adolescent ,Ultraviolet Rays ,Incidence ,Rehabilitation ,Obstetrics and Gynecology ,Estrogens ,Original Articles ,Reproductive Medicine ,Risk Factors ,Humans ,Female ,Child ,Melanoma ,Reproductive History - Abstract
STUDY QUESTION Are reproductive factors and exogenous hormone use associated with incidence of cutaneous melanoma while accounting for ultraviolet radiation (UVR) exposure across different life periods and sun sensitivity factors? SUMMARY ANSWER Earlier age at menarche and late age at first birth, but not other estrogen-related factors were associated with an increased incidence rate of melanoma, with higher risks observed for earlier age at menarche and light hair color at age 15 years. WHAT IS KNOWN ALREADY Although estrogens have been recognized as photosensitizing, previous studies have reported inconsistent findings for the association of melanoma with estrogen-related factors. Most have not collected detailed skin cancer risk factors and have not thoroughly investigated effect modification by ambient UVR and sun sensitivity. STUDY DESIGN, SIZE, DURATION Participants in the US Radiologic Technologists study, an occupational cohort of 146 022 radiologic technologists (73% women), were included and followed during the four time periods (1983–1989, 1994–1998, 2003–2005 and 2012–2014). PARTICIPANTS/MATERIALS, SETTING, METHODS Non-Hispanic white female participants who completed both the second (baseline) and third questionnaires, and did not report having cancer (except keratinocyte carcinoma) at baseline, were included and followed from their age at completion of the second (baseline) questionnaire until the earlier of first primary cancer diagnosis, including invasive melanoma of the skin, or completion of either the third or fourth questionnaire. Reproductive and exogenous hormonal factors were ascertained from the second (baseline) questionnaire, which also collected information on demographic, lifestyle factors and sun sensitivity factors. Ambient UVR was assigned by linking geocoded residential locations, based on self-reported residential history information collected from the third questionnaire to satellite-based ambient UVR data from the National Aeronautics and Space Administration’s Total Ozone Mapping Spectrometer database. To examine the association of reproductive factors, exogenous hormone use, and first primary invasive melanoma of the skin, we used Poisson regression to calculate rate ratios (RRs) and 95% likelihood-based CIs, adjusting for attained age, birth cohort, lifetime average annual ambient UVR, contraceptives and menopausal hormone therapy use. To address the effect modification of ambient UVR exposure and sun sensitivities on melanoma risk, we conducted likelihood-ratio tests for multiplicative interaction. MAIN RESULTS AND THE ROLE OF CHANCE Over a median follow-up time of 17.1 years, 0.95% of eligible participants had an incident first primary melanoma (n = 444). Higher melanoma incidence rates were observed in participants with older attained age, blue/green/gray eye color, blonde/red/auburn natural hair color at age 15, fair skin complexion, and higher UVR. We found an increased incidence rate of melanoma in women who experienced menarche at an earlier age (13, 12 and LIMITATIONS, REASONS FOR CAUTION Information on reproductive history and exogenous hormone use was self-reported. We did not have information on specific doses or formulations of exogenous hormone medications or breastfeeding. WIDER IMPLICATIONS OF THE FINDINGS Women residing in areas of high ambient UVR and those with blonde/red/auburn natural hair color may constitute an additional high-risk group in need of more frequent skin cancer screening. Identifying susceptible periods of exposure or factors that modify UVR susceptibility may aid in guiding more targeted guidelines for melanoma prevention. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by the Intramural Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, US Department of Health and Human Services. Authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
- Published
- 2022
19. Pooled Analysis of Meningioma Risk Following Treatment for Childhood Cancer
- Author
-
Diana R. Withrow, Harald Anderson, Gregory T. Armstrong, Michael Hawkins, Neige Journy, Joseph P. Neglia, Florent de Vathaire, Margaret A. Tucker, Peter D. Inskip, Alina V. Brenner, Marilyn A. Stovall, Ibrahima Diallo, Amy Berrington de Gonzalez, and Lene H. S. Veiga
- Subjects
Cancer Research ,Oncology - Abstract
ImportanceMeningioma is the most common subsequent neoplasm following cranial irradiation among survivors of childhood cancer, but there are still uncertainties regarding the magnitude of the radiation dose-response association, potential modifiers of radiation risks, and the role of chemotherapy.ObjectiveTo evaluate meningioma risk in survivors of childhood cancer following radiotherapy and chemotherapy and identify possible modifying factors of radiation-associated risk.Design, Setting, and ParticipantsThis international case-control study pooled data from 4 nested case-control studies of survivors of childhood cancer diagnosed between 1942 and 2000, followed through 2016. Cases were defined as participants diagnosed with a subsequent meningioma. Controls were matched to cases based on sex, age at first cancer diagnosis, and duration of follow-up. Data were analyzed from July 2019 to June 2022.ExposuresRadiation dose (Gy) to the meningioma site and cumulative chemotherapy doses, including intrathecal and systemic methotrexate doses.Main Outcomes and MeasuresThe main outcome was subsequent meningioma, assessed using odds ratios (ORs) and excess odds ratios per gray (EOR/Gy).ResultsThe analysis included 273 survivors of childhood cancer who developed meningioma (cases) and 738 survivors who did not (controls), with a total of 1011 individuals (median [IQR] age at first cancer diagnosis 5.0 [3.0-9.2] years; 599 [59.2%] female). Median (IQR) time since first cancer was 21.5 (15.0-27.0) years. Increasing radiation dose was associated with increased risk of meningioma (EOR/Gy, 1.44; 95% CI, 0.62-3.61), and there was no evidence of departure from linearity (P = .90). Compared with survivors who were not exposed to radiation therapy, those who received doses of 24 Gy or more had more than 30-fold higher odds of meningioma (OR, 33.66; 95% CI, 14.10-80.31). The radiation dose-response association was significantly lower among patients treated at age 10 years or older compared with those treated before age 10 years (EOR/Gy, 0.57; 95% CI, 0.18-1.91 vs 2.20; 95% CI, 0.87-6.31; P for heterogeneity = .03). Risk associated with radiation remained significantly elevated 30 years after exposure (EOR/Gy, 3.76; 95% CI, 0.77-29.15). We found an increased risk of meningioma among children who had received methotrexate (OR, 3.43; 95% CI, 1.56-7.57), but no evidence of a dose-response association or interaction with radiation dose.Conclusions and RelevanceThese findings suggest that the meninges are highly radiosensitive, especially for children treated before age 10 years. These results support the reduction in whole-brain irradiation over recent decades and the prioritization of approaches that limit radiation exposure in healthy tissue for children. The persistence of elevated risks of meningiomas for 30 years after cranial radiotherapy could help inform surveillance guidelines.
- Published
- 2022
20. Integrated Analysis of Coexpression and Exome Sequencing to Prioritize Susceptibility Genes for Familial Cutaneous Melanoma
- Author
-
Sally Yepes, Margaret A. Tucker, Hela Koka, Yanzi Xiao, Tongwu Zhang, Kristine Jones, Aurelie Vogt, Laurie Burdette, Wen Luo, Bin Zhu, Amy Hutchinson, Meredith Yeager, Belynda Hicks, Kevin M. Brown, Neal D. Freedman, Stephen J. Chanock, Alisa M. Goldstein, and Xiaohong R. Yang
- Subjects
Skin Neoplasms ,Exome Sequencing ,Humans ,Exome ,Genetic Predisposition to Disease ,Cell Biology ,Dermatology ,Molecular Biology ,Biochemistry ,Melanoma - Abstract
The application of whole-exome sequencing has led to the identification of high- and moderate-risk variants that contribute to cutaneous melanoma susceptibility. However, confirming disease-causing variants remains challenging. We applied a gene coexpression network analysis to prioritize the candidate genes identified from whole-exome sequencing of 34 melanoma-prone families, with at least three affected members sequenced per family (N = 119 cases). A coexpression network was constructed from genotype-tissue expression project, skin melanoma from the cancer genome atlas, and primary melanocyte cultures. We performed module-specific enrichment and focused on modules associated with pigmentation processes because they are the best-studied and most well-known risk factors for melanoma susceptibility. We found that pigmentation-associated modules across the four expression datasets examined were enriched for well-known melanoma susceptibility genes plus genes associated with pigmentation. We also used network properties to prioritize genes within pigmentation modules as candidate susceptibility genes. Integrating information from coexpression network analysis and variant prioritization, we identified 36 genes (such as DCT, TPCN2, TRPM1, ATP10A, and EPHA5) as potential melanoma risk genes in the families. Our approach also allowed us to link families with private gene mutations on the basis of gene coexpression patterns and thereby may provide an innovative perspective in gene identification in high-risk families.
- Published
- 2021
21. Bone and Soft‐Tissue Sarcoma Risk in Long‐Term Survivors of Hereditary Retinoblastoma Treated With Radiation
- Author
-
David H. Abramson, Sara J. Schonfeld, Lindsay M. Morton, Ruth A. Kleinerman, Ethel S. Gilbert, Jeannette R. Wong-Siegel, Byron S. Sigel, Margaret A. Tucker, and Johanna M. Seddon
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Neoplasms, Radiation-Induced ,Adolescent ,Epidemiology ,medicine.medical_treatment ,Retinal Neoplasms ,Bone Neoplasms ,Risk Assessment ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Risk Factors ,Internal medicine ,Original Reports ,medicine ,Humans ,Genetic Predisposition to Disease ,Survivors ,Child ,Survival rate ,Radiotherapy ,business.industry ,Soft tissue sarcoma ,Incidence ,Follow up studies ,Infant, Newborn ,Retinoblastoma ,Infant ,Neoplasms, Second Primary ,Sarcoma ,Middle Aged ,medicine.disease ,Prognosis ,Radiation therapy ,Survival Rate ,030104 developmental biology ,030220 oncology & carcinogenesis ,Child, Preschool ,Hereditary Retinoblastoma ,Female ,business ,Follow-Up Studies - Abstract
PURPOSE Survivors of hereditary retinoblastoma have excellent survival but substantially increased risks of subsequent bone and soft-tissue sarcomas, particularly after radiotherapy. Comprehensive investigation of sarcoma risk patterns would inform clinical surveillance for survivors. PATIENTS AND METHODS In a cohort of 952 irradiated survivors of hereditary retinoblastoma who were originally diagnosed during 1914 to 2006, we quantified sarcoma risk with standardized incidence ratios (SIRs) and cumulative incidence analyses. We conducted analyses separately for bone and soft-tissue sarcomas occurring in the head and neck (in/near the radiotherapy field) versus body and extremities (out of field). RESULTS Of 105 bone and 124 soft-tissue sarcomas, more than one half occurred in the head and neck (bone, 53.3%; soft tissue, 51.6%), one quarter in the body and extremities (bone, 29.5%; soft tissue, 25.0%), and approximately one fifth in unknown/unspecified locations (bone, 17.1%; soft tissue, 23.4%). We noted substantially higher risks compared with the general population for head and neck versus body and extremity tumors for both bone (SIR, 2,213; 95% CI, 1,671 to 2,873 v SIR, 169; 95% CI, 115 to 239) and soft-tissue sarcomas (SIR, 542; 95% CI, 418 to 692 v SIR, 45.7; 95% CI, 31.1 to 64.9). Head and neck bone and soft-tissue sarcomas were diagnosed beginning in early childhood and continued well into adulthood, reaching a 60-year cumulative incidence of 6.8% (95% CI, 5.0% to 8.7%) and 9.3% (95% CI, 7.0% to 11.7%), respectively. In contrast, body and extremity bone sarcoma incidence flattened after adolescence (3.5%; 95% CI, 2.3% to 4.8%), whereas body and extremity soft-tissue sarcoma incidence was rare until age 30, when incidence rose steeply (60-year cumulative incidence, 6.6%; 95% CI, 4.1% to 9.2%), particularly for females (9.4%; 95% CI, 5.1% to 13.8%). CONCLUSION Strikingly elevated bone and soft-tissue sarcoma risks differ by age, location, and sex, highlighting important contributions of both radiotherapy and genetic susceptibility. These data provide guidance for the development of a risk-based screening protocol that focuses on the highest sarcoma risks by age, location, and sex.
- Published
- 2019
22. Enhancing Career Paths for Tomorrow's Radiation Oncologists
- Author
-
Kavita V. Dharmarajan, Mary Gospodarowicz, Andrew D. Trister, Clifton D. Fuller, J.M. Longo, Neha Vapiwala, Joshua Jones, Danielle Rodin, John D. Boice, Reid F. Thompson, Joel W. Goldwein, Joanne B. Weidhaas, C. Norman Coleman, Paul Okunieff, Ronald D. Ennis, James A. Hayman, Alan H. Epstein, Daniel G. Petereit, Mei Ling Yap, Charles R. Thomas, Bhadrasain Vikram, Anthony L. Zietman, May Abdel-Wahab, Jeffrey C. Buchsbaum, Silvia C. Formenti, Lawrence N. Shulman, Mary Helen Barcellos-Hoff, Patrick A. Kupelian, Timur Mitin, Surbhi Grover, and Margaret A. Tucker
- Subjects
Cancer Research ,Palliative care ,MEDLINE ,Global Health ,Health informatics ,Nursing ,Global health ,Humans ,Medicine ,Industrial Development ,Radiology, Nuclear Medicine and imaging ,Medical Informatics Applications ,Biology ,Health policy ,Radiation ,Extramural ,business.industry ,Health Policy ,Palliative Care ,Radiation Oncologists ,United States ,Career Mobility ,Oncology ,Rural Health Services ,Diffusion of Innovation ,Radioactive Hazard Release ,business ,Medical Informatics ,Forecasting - Published
- 2019
23. Risk of therapy-related myelodysplastic syndrome/acute myeloid leukemia after childhood cancer: a population-based study
- Author
-
Lindsay M. Morton, Martha S. Linet, Stephen J. Chanock, Pragati Advani, Clara J K Lam, Byron S. Sigel, Margaret A. Tucker, Sara J. Schonfeld, Graça M. Dores, and Rochelle E. Curtis
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Myeloid ,Childhood cancer ,MEDLINE ,Article ,Therapy-related myelodysplastic syndrome ,Public health surveillance ,Neoplasms ,Internal medicine ,Humans ,Medicine ,Public Health Surveillance ,Child ,business.industry ,Age Factors ,Myeloid leukemia ,Neoplasms, Second Primary ,Hematology ,medicine.disease ,Population based study ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Female ,business ,SEER Program - Published
- 2019
24. A Pragmatic Testing-Eligibility Framework for Population Mutation Screening: The Example of BRCA1/2
- Author
-
Hormuzd A. Katki, June A. Peters, Megan N. Frone, Margaret A. Tucker, Ana F. Best, and Mark H. Greene
- Subjects
0301 basic medicine ,Cascade testing ,education.field_of_study ,medicine.diagnostic_test ,Epidemiology ,business.industry ,Population ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Mutation (genetic algorithm) ,Mutation testing ,Mutation screening ,Medicine ,Family history ,business ,education ,Founder mutation ,Demography ,Genetic testing - Abstract
Background: Eligibility guidelines for genetic testing may be revisited, given technological advances, plummeting costs, and proposals for population mutation screening. A key property of eligibility criteria is the tradeoff between the number of mutation carriers identified versus population members tested. We assess the fractions of mutation carriers identified, versus women undergoing mutation testing, for BRCA1/2 founder mutation screening in U.S. Ashkenazi-Jewish women. Methods: BRCA1/2 carrier probabilities, based on personal/family history, were calculated using the risk-prediction tool BRCAPRO for 4,589 volunteers (102 mutation carriers) in the population-based Washington Ashkenazi Study. For each carrier-probability threshold between 0% and 10%, we compared the percentage of founder mutations detected versus the percentage of women requiring mutation testing. PCR mutation testing was conducted at the NIH for the 3 Ashkenazi-Jewish founder mutations (5382insC and 185delAG in BRCA1, and 6174delT in BRCA2). Results: Identifying 90% of BRCA1/2 founder mutations required testing the 60% of Ashkenazi-Jewish women with carrier probabilities exceeding 0.56%, potentially avoiding mutation testing for approximately 0.7 to 1.1 million U.S. Ashkenazi-Jewish women. Alternatively, testing the 44% whose carrier probability exceeded 0.78% identified 80% of mutation carriers, increasing to 89% of mutation carriers when accounting for cascade testing triggered after mutation-positive daughters were identified by screening. We present data on all carrier-probability thresholds, e.g., a 5% threshold identified 46% of mutation carriers while testing 10% of women. Conclusions: Different carrier-probability thresholds offered diverse tradeoffs between numbers of identified mutation carriers versus women tested. Low carrier-probability thresholds identified 90% of BRCA1/2 founder mutation carriers, without testing approximately 1 million U.S. Ashkenazi-Jewish women with lowest carrier probabilities. Impact: In general, this risk-based framework could provide useful new options to consider during eligibility-criteria development for population mutation screening.
- Published
- 2019
25. Novel loss-of-function variant in DENND5A impedes melanosomal cargo transport and predisposes to familial cutaneous melanoma
- Author
-
Per Johnsson, Margaret A. Tucker, Jiwei Gao, Noémi Nagy, Hanna Eriksson, Joakim Lundeberg, Nicholas P. Tobin, Pär G. Engström, Yitian Zhou, Rong Yu, Jian Zhao, Yihai Cao, Kim Thrane, Veronica Höiom, Muyi Yang, Alisa M. Goldstein, Rainer Tuominen, suzanne Egyhazi-Brage, Lars Bräutigam, and Xiaohong R. Yang
- Subjects
Candidate gene ,Melanosomes ,Skin Neoplasms ,Monophenol Monooxygenase ,Melanoma ,Context (language use) ,Biology ,medicine.disease ,Melanin ,Exome Sequencing ,Genetic predisposition ,Cancer research ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Genetic Predisposition to Disease ,Exome ,Sorting Nexins ,Genetics (clinical) ,Loss function ,Zebrafish ,Melanosome - Abstract
Purpose More than half of the familial cutaneous melanomas have unknown genetic predisposition. This study aims at characterizing a novel melanoma susceptibility gene. Methods We performed exome and targeted sequencing in melanoma-prone families without any known melanoma susceptibility genes. We analyzed the expression of candidate gene DENND5A in melanoma samples in relation to pigmentation and UV signature. Functional studies were carried out using microscopic approaches and zebrafish model. Results We identified a novel DENND5A truncating variant that segregated with melanoma in a Swedish family and 2 additional rare DENND5A variants, 1 of which segregated with the disease in an American family. We found that DENND5A is significantly enriched in pigmented melanoma tissue. Our functional studies show that loss of DENND5A function leads to decrease in melanin content in vitro and pigmentation defects in vivo. Mechanistically, harboring the truncating variant or being suppressed leads to DENND5A losing its interaction with SNX1 and its ability to transport the SNX1-associated vesicles from melanosomes. Consequently, untethered SNX1-premelanosome protein and redundant tyrosinase are redirected to lysosomal degradation by default, causing decrease in melanin content. Conclusion Our findings provide evidence of a physiological role of DENND5A in the skin context and link its variants to melanoma susceptibility.
- Published
- 2021
26. Genotypic vs Phenotypic Risk Assessment for Melanoma
- Author
-
Michael R. Sargen and Margaret A. Tucker
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Genotype ,business.industry ,Melanoma ,MEDLINE ,Articles ,medicine.disease ,Phenotype ,Risk Assessment ,Internal medicine ,Medicine ,Humans ,business ,Risk assessment - Abstract
BACKGROUND: Recent genome-wide association meta-analysis for melanoma doubled the number of previously identified variants. We assessed the performance of an updated polygenic risk score (PRS) in a population of older individuals, where melanoma incidence and cumulative ultraviolet radiation exposure is greatest. METHODS: We assessed a PRS for cutaneous melanoma comprising 55 variants in a prospective study of 12 712 individuals in the ASPirin in Reducing Events in the Elderly Trial. We evaluated incident melanomas diagnosed during the trial and prevalent melanomas diagnosed preenrolment (self-reported). Multivariable models examined associations between PRS as a continuous variable (per SD) and categorical (low-risk [0%-20%], medium-risk [21%-80%], high-risk [81%-100%] groups) with incident melanoma. Logistic regression examined the association between PRS and prevalent melanoma. RESULTS: At baseline, mean participant age was 75 years; 55.0% were female, and 528 (4.2%) had prevalent melanomas. During follow-up (median = 4.7 years), 120 (1.0%) incident cutaneous melanomas occurred, 98 of which were in participants with no history. PRS was associated with incident melanoma (hazard ratio = 1.46 per SD, 95% confidence interval [CI] = 1.20 to 1.77) and prevalent melanoma (odds ratio [OR] = 1.55 per SD, 95% CI = 1.42 to 1.69). Participants in the highest-risk PRS group had increased risk compared with the low-risk group for incident melanoma (OR = 2.51, 95% CI = 1.28 to 4.92) and prevalent melanoma (OR = 3.66, 95% CI = 2.69 to 5.05). When stratifying by sex, only males had an association between the PRS and incident melanoma, whereas both sexes had an association between the PRS and prevalent melanoma. CONCLUSIONS: A genomic risk score is associated with melanoma risk in older individuals and may contribute to targeted surveillance.
- Published
- 2021
27. Benign Tumors in Long-Term Survivors of Retinoblastoma
- Author
-
Jasmine H. Francis, Lindsay M. Morton, Annette C. Moll, Milo van Hoefen Wijsard, Johanna M. Seddon, Sara J. Schonfeld, Margaret A. Tucker, Ruth A. Kleinerman, David H. Abramson, Armida W. M. Fabius, Flora E. van Leeuwen, Ophthalmology, Epidemiology and Data Science, ACS - Diabetes & metabolism, CCA - Cancer Treatment and quality of life, APH - Quality of Care, and APH - Health Behaviors & Chronic Diseases
- Subjects
0301 basic medicine ,Oncology ,subsequent benign tumor ,Cancer Research ,medicine.medical_specialty ,retinoblastoma survivor ,lipoma ,lcsh:RC254-282 ,Article ,retinoblastoma ,Benign tumor ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,leiomyoma ,Epidemiology ,medicine ,RB1 ,Cumulative incidence ,Family history ,cumulative incidence ,Retinoblastoma ,business.industry ,subsequent malignant neoplasms ,Lipoma ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Leiomyoma ,030220 oncology & carcinogenesis ,Hereditary Retinoblastoma ,epidemiology ,hereditary retinoblastoma ,business - Abstract
Hereditary retinoblastoma survivors have substantially increased risk of subsequent malignant neoplasms (SMNs). The risk of benign neoplasms, a substantial cause of morbidity, is unclear. We calculated the cumulative incidence of developing benign tumors at 60 years following retinoblastoma diagnosis among 1128 hereditary (i.e., bilateral retinoblastoma or unilateral with family history, mutation testing was not available) and 924 nonhereditary retinoblastoma survivors diagnosed during 1914–2006 at two US medical centers with follow-up through 2016. Using Cox proportional hazards regression, we compared benign tumor risk by hereditary status and evaluated the association between benign tumors and SMNs. There were 100 benign tumors among 73 hereditary survivors (cumulative incidence = 17.6%, 95% confidence interval [CI] = 12.9–22.8%) and 22 benign tumors among 16 nonhereditary survivors (cumulative incidence = 3.9%, 95%CI = 2.2–6.4%), corresponding to 4.9-fold (95%CI = 2.8–8.4) increased risk for hereditary survivors. The cumulative incidence after hereditary retinoblastoma was highest for lipoma among males (14.0%, 95%CI = 7.7–22.1%) and leiomyoma among females (8.9%, 95%CI = 5.2–13.8%). Among hereditary survivors, having a prior SMN was associated with 3.5-fold (95%CI = 2.0–6.1) increased risk of developing a benign tumor, the reciprocal risk for developing an SMN after a benign tumor was 1.8 (95%CI = 1.1–2.9). These large-scale, long-term data demonstrate an increased risk for benign tumors after hereditary versus nonhereditary retinoblastoma. If confirmed, the association between benign tumors and SMNs among hereditary patients may have implications for long-term surveillance.
- Published
- 2021
28. Frequency of Pathogenic Germline Variants in Cancer-Susceptibility Genes in the Childhood Cancer Survivor Study
- Author
-
Jung Kim, Matthew Gianferante, Danielle M Karyadi, Stephen W Hartley, Megan N Frone, Wen Luo, Leslie L Robison, Gregory T Armstrong, Smita Bhatia, Michael Dean, Meredith Yeager, Bin Zhu, Lei Song, Joshua N Sampson, Yutaka Yasui, Wendy M Leisenring, Seth A Brodie, Kelvin C de Andrade, Fernanda P Fortes, Alisa M Goldstein, Payal P Khincha, Mitchell J Machiela, Mary L McMaster, Michael L Nickerson, Leatrisse Oba, Alexander Pemov, Maisa Pinheiro, Melissa Rotunno, Karina Santiago, Talia Wegman-Ostrosky, W Ryan Diver, Lauren Teras, Neal D Freedman, Belynda D Hicks, Mingyi Wang, Kristine Jones, Amy A Hutchinson, Casey Dagnall, Sharon A Savage, Margaret A Tucker, Stephen J Chanock, Lindsay M Morton, Douglas R Stewart, and Lisa Mirabello
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Genes, Recessive ,Penetrance ,Childhood Cancer Survivor Study ,Wilms Tumor ,Article ,Central Nervous System Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Cancer Survivors ,CDKN2A ,Neoplasms ,Internal medicine ,Exome Sequencing ,Genetic predisposition ,medicine ,Humans ,Genetic Predisposition to Disease ,Age of Onset ,Child ,Germ-Line Mutation ,Exome sequencing ,Aged ,business.industry ,Lymphoma, Non-Hodgkin ,Cancer ,Sarcoma ,Wilms' tumor ,medicine.disease ,Pediatric cancer ,Kidney Neoplasms ,030104 developmental biology ,Case-Control Studies ,030220 oncology & carcinogenesis ,Female ,business - Abstract
Background Pediatric cancers are the leading cause of death by disease in children despite improved survival rates overall. The contribution of germline genetic susceptibility to pediatric cancer survivors has not been extensively characterized. We assessed the frequency of pathogenic or likely pathogenic (P/LP) variants in 5451 long-term pediatric cancer survivors from the Childhood Cancer Survivor Study. Methods Exome sequencing was conducted on germline DNA from 5451 pediatric cancer survivors (cases who survived ≥5 years from diagnosis; n = 5105 European) and 597 European cancer-free adults (controls). Analyses focused on comparing the frequency of rare P/LP variants in 237 cancer-susceptibility genes and a subset of 60 autosomal dominant high-to-moderate penetrance genes, for both case-case and case-control comparisons. Results Of European cases, 4.1% harbored a P/LP variant in high-to-moderate penetrance autosomal dominant genes compared with 1.3% in controls (2-sided P = 3 × 10-4). The highest frequency of P/LP variants was in genes typically associated with adult onset rather than pediatric cancers, including BRCA1/2, FH, PALB2, PMS2, and CDKN2A. A statistically significant excess of P/LP variants, after correction for multiple tests, was detected in patients with central nervous system cancers (NF1, SUFU, TSC1, PTCH2), Wilms tumor (WT1, REST), non-Hodgkin lymphoma (PMS2), and soft tissue sarcomas (SDHB, DICER1, TP53, ERCC4, FGFR3) compared with other pediatric cancers. Conclusion In long-term pediatric cancer survivors, we identified P/LP variants in cancer-susceptibility genes not previously associated with pediatric cancer as well as confirmed known associations. Further characterization of variants in these genes in pediatric cancer will be important to provide optimal genetic counseling for patients and their families.
- Published
- 2021
29. Immune-Related Adverse Events After Immune Checkpoint Inhibitors for Melanoma Among Older Adults
- Author
-
Sara J, Schonfeld, Margaret A, Tucker, Eric A, Engels, Graça M, Dores, Joshua N, Sampson, Meredith S, Shiels, Stephen J, Chanock, and Lindsay M, Morton
- Subjects
Cohort Studies ,Male ,Skin Neoplasms ,Humans ,General Medicine ,Medicare ,Immune Checkpoint Inhibitors ,Melanoma ,United States ,Aged - Abstract
Immune checkpoint inhibitors (ICIs) have improved survival in patients with advanced melanoma but can be associated with a spectrum of immune-related adverse events (AEs), including both autoimmune-related AEs and other immune-related inflammatory AEs. These associations have primarily been evaluated in clinical trials that include highly selected patients, with older adults often underrepresented.To evaluate the association between use of ICIs and immune-related AEs (autoimmune and other immune related) among older patients with cutaneous melanoma.A population-based cohort study was conducted from January 1, 2011, to December 31, 2015. Data were analyzed from January 31 to May 31, 2021. With use of a linked database of Medicare claims and Surveillance, Epidemiology, and End Results (SEER) Program population-based cancer registries, patients of White race diagnosed with stages II-IV or unknown (American Joint Committee on Cancer, AJCC Cancer Staging Manual 6th edition) first primary cutaneous melanoma during 2011-2015, as reported to SEER, and followed up through December 31, 2015, were identified.Immune checkpoint inhibitors for treatment of melanoma.The association between ICIs and immune-related AEs ascertained from Medicare claims data was estimated using multivariable Cox regression with hazard ratios (HRs) and 95% CIs and with cumulative incidence accounting for competing risk of death.The study included 4489 patients of White race with first primary melanoma (3002 men [66.9%]; median age, 74.9 [range, 66.0-84.9] years). During follow-up (median, 1.4 [range, 0-5.0] years), 1576 patients (35.1%) had an immune-related AE on a Medicare claim. Use of ICIs (reported for 418 patients) was associated with autoimmune-related AEs (HR, 2.5; 95% CI, 1.6-4.0), including primary adrenal insufficiency (HR, 9.9; 95% CI, 4.5-21.5) and ulcerative colitis (HR, 8.6; 95% CI, 2.8-26.3). Immune checkpoint inhibitors also were associated with other immune-related AEs (HR, 2.2; 95% CI, 1.7-2.8), including Cushing syndrome (HR, 11.8; 95% CI, 1.4-97.2), hyperthyroidism (HR, 6.3; 95% CI, 2.0-19.5), hypothyroidism (HR, 3.8; 95% CI, 2.4-6.1), hypopituitarism (HR, 19.8; 95% CI, 5.4-72.9), other pituitary gland disorders (HR, 6.0; 95% CI, 1.2-30.2), diarrhea (HR, 3.5; 95% CI, 2.5-4.9), and sepsis or septicemia (HR, 2.2; 95% CI, 1.4-3.3). Most associations were pronounced within 6 months following the first ICI claim and comparable with or without a baseline history of autoimmune disease. The cumulative incidence at 6 months following the first ICI claim was 13.7% (95% CI, 9.7%-18.3%) for autoimmune-related AEs and 46.8% (95% CI, 40.7%-52.7%) for other immune-related AEs.In this cohort study of older adults with melanoma, ICIs were associated with autoimmune-related AEs and other immune-related AEs. Although some findings were consistent with clinical trials of ICIs, others warrant further investigation. As ICI use continues to expand rapidly, ongoing investigation of the spectrum of immune-related AEs may optimize management of disease in patients.
- Published
- 2022
30. Sebaceous Carcinoma Incidence and Survival Among Solid Organ Transplant Recipients in the United States, 1987-2017
- Author
-
Elizabeth K. Cahoon, Margaret A. Tucker, Eric A. Engels, Michael R. Sargen, Alisa M. Goldstein, and Charles F. Lynch
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Carcinogenesis ,Ultraviolet Rays ,Population ,Dermatology ,Organ transplantation ,030207 dermatology & venereal diseases ,03 medical and health sciences ,Immunocompromised Host ,Sebaceous Glands ,Young Adult ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,Registries ,Sebaceous Gland Neoplasms ,education ,Child ,Original Investigation ,Aged ,education.field_of_study ,Proportional hazards model ,business.industry ,Incidence (epidemiology) ,Incidence ,Hazard ratio ,Infant, Newborn ,Infant ,Organ Transplantation ,Middle Aged ,Survival Analysis ,Transplant Recipients ,United States ,Cancer registry ,Transplantation ,Muir-Torre Syndrome ,030220 oncology & carcinogenesis ,Child, Preschool ,Carcinoma, Squamous Cell ,Female ,business ,Cohort study - Abstract
Importance Risk of sebaceous carcinoma (SC), a rare skin cancer associated with Muir-Torre syndrome, is elevated among solid organ transplant recipients (SOTRs). However, population studies evaluating this association and assessing survival for posttransplant cases are lacking, and further understanding of SC epidemiology in this immunosuppressed population could provide etiologic and clinical insights. Objective To assess SC incidence and patient survival after solid organ transplantation. Design, Setting, and Participants This cohort study, conducted from January 1, 1987, to December 31, 2017, used data from the Transplant Cancer Match Study, which links transplant and cancer registry data for 17 states and 1 metropolitan area in the United States. Altogether, these registries account for approximately 46% of all US transplants. Data on demographic and transplant characteristics as well as induction and initial maintenance immunosuppressive therapies were obtained from the transplant registry. Standardized incidence ratios (SIRs) comparing SC incidence among SOTRs to the general population were calculated. Incidence rate ratios (IRRs) comparing SC risk between SOTR subgroups were calculated using multivariate Poisson regression. Cox regression was used to compare overall survival between SC cases in SOTRs and other individuals. Main Outcomes and Measures Sebaceous carcinoma incidence and overall patient survival after transplantation compared with the general population. Results A total of 326 282 transplant procedures were performed for 301 075 patients (No. [%] age at transplant, 126 550 [38.8%] aged 0-44 years; 82 394 [25.3%] aged 45-54 years; 82 082 [25.5%] aged 55-64 years; 35 256 [10.8%] aged ≥65 years; 201 354 male patients [61.7%]; 202 557 White patients [62.1%]). A total of 102 SCs were diagnosed in 301 075 SOTRs, corresponding to a 25-fold increased incidence (SIR, 24.8; 95% CI, 20.2-30.1). Incidence was especially elevated among lung recipients (SIR, 47.7; 95% CI, 20.6-94.0) and after a posttransplant diagnosis of cutaneous squamous cell carcinoma (SIR, 104.0; 95% CI, 62.8-163.0). Among SOTRs, factors independently associated with SC risk included male sex (IRR, 2.46; 95% CI, 1.48-4.07; P
- Published
- 2020
31. Ambient ultraviolet radiation and major salivary gland cancer in the United States
- Author
-
Ruth M. Pfeiffer, Elizabeth K. Cahoon, Margaret A. Tucker, Michael R. Sargen, and Z. Mai
- Subjects
business.industry ,Major Salivary Gland ,medicine ,Cancer research ,General Earth and Planetary Sciences ,Cancer ,medicine.disease ,business ,Ultraviolet radiation ,General Environmental Science - Published
- 2020
32. The Impact of Longitudinal Surveillance on Tumor Thickness for Melanoma-Prone Families with and without Pathogenic Germline Variants of
- Author
-
Michael R, Sargen, Ruth M, Pfeiffer, David E, Elder, Xiaohong R, Yang, Alisa M, Goldstein, and Margaret A, Tucker
- Subjects
Adult ,Male ,Skin Neoplasms ,Cyclin-Dependent Kinase 4 ,Middle Aged ,United States ,Article ,Population Surveillance ,Humans ,Mass Screening ,Self-Examination ,Female ,Genetic Predisposition to Disease ,Longitudinal Studies ,Melanoma ,Cyclin-Dependent Kinase Inhibitor p16 ,Germ-Line Mutation ,Aged ,Neoplasm Staging ,SEER Program - Abstract
BACKGROUND: Skin cancer screening is routinely performed for members of melanoma-prone families, but longitudinal studies evaluating the efficacy of surveillance in this high-risk population are lacking. METHODS: We evaluated thickness for first primary melanomas diagnosed in melanoma-prone families (≥2 individuals with melanoma) enrolled in NCT00040352 (National Cancer Institute [NCI] familial melanoma study) from 1976 through 2014; enrolled patients received routine skin cancer screening and education about skin self-exams. We used linear and ordinal logistic regression models adjusted for gender and age with a generalized estimating equations approach to report changes in thickness and tumor (T) stage over time, comparing outcomes for NCI cases diagnosed before (“pre-study”) vs. after study participation (“prospective”) and for NCI cases vs. non-familial cases (Surveillance, Epidemiology, and End Results [SEER] 9 registries). RESULTS: Tumor thickness was evaluated for 293 NCI (pre-study=246; prospective=47) patients. Compared to NCI pre-study cases, NCI prospective melanomas were thinner (0.6mm vs. 1.1mm; P
- Published
- 2020
33. Role of radiotherapy and chemotherapy in the risk of leukemia after childhood cancer: An international pooled analysis
- Author
-
Cristina Veres, Rita E. Weathers, D.L. Winter, Ibrahima Diallo, Florent de Vathaire, Rebecca M. Howell, Margaret A. Tucker, Carole Rubino, Rodrigue S. Allodji, Mark P. Little, Michael M. Hawkins, Nadia Haddy, Marie-Cécile Le Deley, and Lindsay M. Morton
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Childhood cancer ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cancer Survivors ,Internal medicine ,medicine ,Humans ,Child ,Chemotherapy ,business.industry ,Infant, Newborn ,Myeloid leukemia ,Infant ,Odds ratio ,medicine.disease ,Confidence interval ,Radiation therapy ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Child, Preschool ,Bone marrow ,business - Abstract
Childhood cancer survivors are at increased risk for second primary leukemia (SPL), but there is little consensus on the magnitude of some risk factors because of the small size of previous studies. We performed a pooled analysis of all published studies with detailed treatment data, including estimated active bone marrow (ABM) dose received during radiation therapy and doses of specific chemotherapeutic agents for childhood cancer diagnosed from 1930 through 2000, in order to more thoroughly investigate treatment-related risks of SPL. A total of 147 SPL cases (of which 69% were acute myeloid leukemia [AML]) were individually matched to 522 controls, all from four case-control studies including patients from six countries (France, United Kingdom, United States, Canada, Italy and Netherlands). Odds ratios (OR) and corresponding 95% confidence intervals (CIs) were calculated using conditional logistic regression, and the excess OR per Gray (EOR/Gy) was also calculated. After accounting for the other therapies received, topoisomerase II inhibitor was associated with an increased SPL risk (highest tertile vs none: OR = 10.0, 95% CI: 3.7-27.3). Radiation dose to the ABM was also associated with increased SPL risk among those not receiving chemotherapy (EOR/Gy = 1.6, 95% CI: 0.1-14.3), but not among those who received chemotherapy (CT). SPL were most likely to occur in the first decade following cancer treatment. Results were similar when analyses were restricted to AML. The evidence of interaction between radiation and CT has implications for leukemogenic mechanism. The results for topoisomerase II inhibitors are particularly important given their increasing use to treat childhood cancer.
- Published
- 2020
34. Sebaceous Carcinoma Epidemiology and Genetics: Emerging Concepts and Clinical Implications for Screening, Prevention, and Treatment
- Author
-
Michael R. Sargen, Margaret A. Tucker, Alisa M. Goldstein, Eric A. Engels, Elizabeth K. Cahoon, and Gabriel J. Starrett
- Subjects
Cancer Research ,medicine.medical_specialty ,Skin Neoplasms ,MEDLINE ,Regional Disease ,Disease ,Article ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Epidemiology ,medicine ,Humans ,Sebaceous Gland Neoplasms ,Early Detection of Cancer ,Genetics ,business.industry ,Incidence (epidemiology) ,Incidence ,Cancer ,Genomics ,medicine.disease ,United States ,Oncology ,030220 oncology & carcinogenesis ,Microsatellite Instability ,Skin cancer ,business ,Sebaceous carcinoma ,SEER Program - Abstract
Sebaceous carcinoma is an aggressive skin cancer with a 5-year overall survival rate of 78% for localized/regional disease and 50% for metastatic disease. The incidence of this cancer has been increasing in the United States for several decades, but the underlying reasons for this increase are unclear. In this article, we review the epidemiology and genetics of sebaceous carcinoma, including recent population data and tumor genomic analyses that provide new insights into underlying tumor biology. We further discuss emerging evidence of a possible viral etiology for this cancer. Finally, we review the clinical implications of recent advances in sebaceous carcinoma research for screening, prevention, and treatment.
- Published
- 2020
35. Discovery and fine-mapping of height loci via high-density imputation of GWASs in individuals of African ancestry
- Author
-
Mariaelisa Graff, Zhaoming Wang, David R. Weir, Ann W. Hsing, Joel N. Hirschhorn, Rebecca S. Fine, Wei Zheng, William Maixner, Adebowale Adeyemo, Rick A. Kittles, Rebecca Rohde, Krista A. Zanetti, Elizabeth K. Speliotes, Keri L. Monda, Ingrid B. Borecki, Yaming Shao, Suhn K. Rhie, Sanjay R. Patel, Richard S. Cooper, Jerome I. Rotter, Michael J. Thun, William J. Blot, Brendan J. Keating, Michèle M. Sale, Scott M. Williams, Yingchang Lu, Margaret A. Tucker, Eric A. Klein, Uma Nayak, Curtis A. Pettaway, Yii-Der Ida Chen, Sailaja Vedantam, Pamela J. Schreiner, Phyllis J. Goodman, Babatunde L. Salako, Christine Neslund-Dudas, Andrew B. Singleton, Michael F. Press, Neil E. Caporaso, Maureen Sanderson, Diane M. Becker, Michele K. Evans, Margaret R. Spitz, Christopher I. Amos, Susan Redline, Kurt Lohman, Ching-Ti Liu, Hampton L. Leonard, Kris Monroe, Sun J. Kang, David Van Den Berg, Janet L. Stanford, Lewis H. Kuller, Traci M. Bartz, J. M. Zmuda, Christopher S. Carlson, Ruth J. F. Loos, Joanne M. Jordan, Guillaume Lettre, Youfang Liu, Lara Sucheston, Mike A. Nalls, Shad B. Smith, Suzanne Kolb, Lisa B. Signorello, Daniel Shriner, Tamara B. Harris, Larry D. Atwood, Wei-Min Chen, Oladosu Ojengbede, Kristin A. Rand, Claudia Schurmann, Guanjie Chen, Sharon L.R. Kardia, Graham Casey, Lawrence F. Bielak, Dena G. Hernandez, Talin Haritunians, Rajiv Nadukuru, Elisa V. Bandera, Matthew A. Allison, Amidou N'Diaye, Donna K. Arnett, Olufunmilayo I. Olopade, Ellen W. Demerath, Babette S. Zemel, Margaret Wrensch, Myriam Fornage, John K. Wiencke, George J. Papanicolaou, Sara S. Strom, Temidayo O. Ogundiran, Gregory L. Burke, Erin B. Ware, Marian L. Neuhouser, James G. Wilson, Dhananjay Vaidya, Nicholette D. Palmer, Marguerite R. Irvin, JoAnn E. Manson, Benjamin A. Rybicki, Laurence N. Kolonel, Chad D. Huff, Jennifer J. Hu, Jennifer A. Brody, L. Keoki Williams, Leslie A. Lange, Qing Duan, David S. Siscovick, Yongmei Liu, Leslie Bernstein, Jennifer A. Smith, Latchezar Dimitrov, Sarah J. Nyante, Sue A. Ingles, Yu Han H. Hsu, Xifeng Wu, Heather M. Ochs-Balcom, Jingzhong Ding, David V. Conti, L. Adrienne Cupples, Gerry A. Coetzee, Thomas H. Mosley, Kristin L. Young, Donald W. Bowden, Edmond K. Kabagambe, Christine B. Ambrosone, Karen C. Johnson, Patricia A. Peyser, Stefan Ambs, Victoria L. Stevens, Minhui Chen, Carl D. Langefeld, Sonja I. Berndt, Andrew F. Olshan, Qiuyin Cai, Jessica D. Faul, Mary F. Feitosa, Brian E. Cade, Esther M. John, Ann G. Schwartz, Yan V. Sun, Xinruo Zhang, Stephen J. Chanock, Jason H. Moore, Alexander P. Reiner, Caroline S. Fox, Charles Kooperberg, Anne E. Justice, Jin Li, Lorna H. McNeill, Alex Stram, Jie Yao, Xiuqing Guo, Mara Z. Vitolins, Edward A. Ruiz-Narváez, Abdullah Kutlar, Charleston W. K. Chiang, Jing Hua Zhao, Lisa R. Yanek, Poorva Mudgal, John D. Carpten, Barbara V. Howard, Anselm Hennis, Charles N. Rotimi, Ulrich Broeckel, Albert M. Levin, Jonathan P. Bradfield, Curtis C. Harris, Regina G. Ziegler, Brian E. Henderson, Bruce M. Psaty, Adeyinka G. Falusi, Katherine L. Nathanson, Barbara McKnight, Melinda C. Aldrich, Herman A. Taylor, Melissa Garcia, Degui Zhi, Dezheng Huo, Barry I. Freedman, D. C. Rao, Sylvia Wassertheil-Smoller, Jie Zhou, Adam B. Murphy, Eric E. Schadt, Sandra L. Deming, John S. Witte, Julie R. Palmer, Neil A. Zakai, Adesola Ogunniyi, Yun Li, Mary Cushman, Victoria L. Buchanan, Jorge L. Rodriguez-Gil, Struan F.A. Grant, Timothy D. Howard, Omri Gottesman, Wei Zhao, Kira C. Taylor, Yan Meng, Hayrettin Okut, Hakon Hakonarson, Xiaofeng Zhu, Elizabeth M. Gillanders, Alan B. Zonderman, Lisa Chu, Badri Padhukasahasram, Vaneet Lotay, W. Ryan Diver, Mary K. Wojczynski, Simin Liu, Erwin P. Bottinger, Cameron D. Palmer, Alessandra Chesi, Elise Lim, Laura J. Rasmussen-Torvik, Eirini Marouli, Kari E. North, Salman M. Tajuddin, Christopher A. Haiman, Barbara Nemesure, Loic Le Marchand, Bamidele O. Tayo, and Maggie C.Y. Ng
- Subjects
Male ,0303 health sciences ,030305 genetics & heredity ,High density ,Black People ,Single-nucleotide polymorphism ,Genome-wide association study ,Biology ,Polymorphism, Single Nucleotide ,Body Height ,Article ,Minor allele frequency ,Black or African American ,Europe ,03 medical and health sciences ,Evolutionary biology ,Africa ,Genetics ,Humans ,Female ,Genotyping ,Genetics (clinical) ,Imputation (genetics) ,030304 developmental biology ,Genome-Wide Association Study - Abstract
Although many loci have been associated with height in European ancestry populations, very few have been identified in African ancestry individuals. Furthermore, many of the known loci have yet to be generalized to and fine-mapped within a large-scale African ancestry sample. We performed sex-combined and sex-stratified meta-analyses in up to 52,764 individuals with height and genome-wide genotyping data from the African Ancestry Anthropometry Genetics Consortium (AAAGC). We additionally combined our African ancestry meta-analysis results with published European genome-wide association study (GWAS) data. In the African ancestry analyses, we identified three novel loci (SLC4A3, NCOA2, ECD/FAM149B1) in sex-combined results and two loci (CRB1, KLF6) in women only. In the African plus European sex-combined GWAS, we identified an additional three novel loci (RCCD1, G6PC3, CEP95) which were equally driven by AAAGC and European results. Among 39 genome-wide significant signals at known loci, conditioning index SNPs from European studies identified 20 secondary signals. Two of the 20 new secondary signals and none of the 8 novel loci had minor allele frequencies (MAF) < 5%. Of 802 known European height signals, 643 displayed directionally consistent associations with height, of which 205 were nominally significant (p < 0.05) in the African ancestry sex-combined sample. Furthermore, 148 of 241 loci contained ≤20 variants in the credible sets that jointly account for 99% of the posterior probability of driving the associations. In summary, trans-ethnic meta-analyses revealed novel signals and further improved fine-mapping of putative causal variants in loci shared between African and European ancestry populations.
- Published
- 2020
36. Frequency of Pathogenic Germline Variants in Cancer-Susceptibility Genes in Patients With Osteosarcoma
- Author
-
Danielle M. Karyadi, Michael Dean, Piero Picci, Roberto Tirabosco, Adrienne M. Flanagan, Meredith Yeager, Claudia Maria Hattinger, Ana Patiño-García, Antonio Sergio Petrilli, Leslie L. Robison, Miriam Gutiérrez-Jimeno, Donald A. Barkauskas, Lisa Mirabello, Amy Hutchinson, Smita Bhatia, Sharon A. Savage, Silvia Regina Caminada de Toledo, Nathan Pankratz, Brian D. Carter, Neal D. Freedman, Julie M. Gastier-Foster, Massimo Serra, Stephen J. Chanock, Patricia Valverde, Inci Ergurhan Ilhan, Gregory T. Armstrong, Mandy L. Ballinger, Richard Gorlick, Maria Fernanda Amary, Maisa Pinheiro, Katia Scotlandi, Joshua N. Sampson, Lindsay M. Morton, Robert N. Hoover, Gerardo Mejia-Baltodano, Margaret A. Tucker, David Thomas, Mingyi Wang, Lei Song, Nilgun Kurucu, Logan G. Spector, Fernando Lecanda, Maria Teresa Landi, Susan M. Gapstur, Bin Zhu, Eric Karlins, Roelof Koster, Matthew Gianferante, and Belynda Hicks
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Candidate gene ,Adolescent ,Genetic counseling ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Germline mutation ,CDKN2A ,Internal medicine ,Genetic predisposition ,medicine ,Humans ,Genetic Predisposition to Disease ,030212 general & internal medicine ,Child ,Exome ,Exome sequencing ,Germ-Line Mutation ,Original Investigation ,Aged ,Aged, 80 and over ,Osteosarcoma ,business.industry ,Cancer ,High-Throughput Nucleotide Sequencing ,Middle Aged ,medicine.disease ,030220 oncology & carcinogenesis ,Child, Preschool ,Female ,business - Abstract
IMPORTANCE: Osteosarcoma, the most common malignant bone tumor in children and adolescents, occurs in a high number of cancer predisposition syndromes that are defined by highly penetrant germline mutations. The germline genetic susceptibility to osteosarcoma outside of familial cancer syndromes remains unclear. OBJECTIVE: To investigate the germline genetic architecture of 1244 patients with osteosarcoma. DESIGN, SETTING, AND PARTICIPANTS: Whole-exome sequencing (n = 1104) or targeted sequencing (n = 140) of the DNA of 1244 patients with osteosarcoma from 10 participating international centers or studies was conducted from April 21, 2014, to September 1, 2017. The results were compared with the DNA of 1062 individuals without cancer assembled internally from 4 participating studies who underwent comparable whole-exome sequencing and 27 173 individuals of non-Finnish European ancestry who were identified through the Exome Aggregation Consortium (ExAC) database. In the analysis, 238 high-interest cancer-susceptibility genes were assessed followed by testing of the mutational burden across 736 additional candidate genes. Principal component analyses were used to identify 732 European patients with osteosarcoma and 994 European individuals without cancer, with outliers removed for patient-control group comparisons. Patients were subsequently compared with individuals in the ExAC group. All data were analyzed from June 1, 2017, to July 1, 2019. MAIN OUTCOMES AND MEASURES: The frequency of rare pathogenic or likely pathogenic genetic variants. RESULTS: Among 1244 patients with osteosarcoma (mean [SD] age at diagnosis, 16 [8.9] years [range, 2-80 years]; 684 patients [55.0%] were male), an analysis restricted to individuals with European ancestry indicated a significantly higher pathogenic or likely pathogenic variant burden in 238 high-interest cancer-susceptibility genes among patients with osteosarcoma compared with the control group (732 vs 994, respectively; P = 1.3 × 10(−18)). A pathogenic or likely pathogenic cancer-susceptibility gene variant was identified in 281 of 1004 patients with osteosarcoma (28.0%), of which nearly three-quarters had a variant that mapped to an autosomal-dominant gene or a known osteosarcoma-associated cancer predisposition syndrome gene. The frequency of a pathogenic or likely pathogenic cancer-susceptibility gene variant was 128 of 1062 individuals (12.1%) in the control group and 2527 of 27 173 individuals (9.3%) in the ExAC group. A higher than expected frequency of pathogenic or likely pathogenic variants was observed in genes not previously linked to osteosarcoma (eg, CDKN2A, MEN1, VHL, POT1, APC, MSH2, and ATRX) and in the Li-Fraumeni syndrome-associated gene, TP53. CONCLUSIONS AND RELEVANCE: In this study, approximately one-fourth of patients with osteosarcoma unselected for family history had a highly penetrant germline mutation requiring additional follow-up analysis and possible genetic counseling with cascade testing.
- Published
- 2020
37. Using whole-exome sequencing and protein interaction networks to prioritize candidate genes for germline cutaneous melanoma susceptibility
- Author
-
Sally Yepes, Bin Zhu, Alisa M. Goldstein, Aurelie Vogt, Yanzi Xiao, Amy Hutchinson, Wen Luo, Neal D. Freedman, Meredith Yeager, Laurie Burdette, Hela Koka, Belynda Hicks, Margaret A. Tucker, Xiaohong R. Yang, Kristine Jones, and Stephen J. Chanock
- Subjects
Male ,Candidate gene ,Skin Neoplasms ,lcsh:Medicine ,Context (language use) ,Computational biology ,Biology ,Germline ,Article ,Genetic Heterogeneity ,Exome Sequencing ,Genetics ,Humans ,Exome ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Protein Interaction Maps ,NGLY1 ,lcsh:Science ,Gene ,Melanoma ,Exome sequencing ,Cancer ,Multidisciplinary ,Genetic heterogeneity ,lcsh:R ,High-Throughput Nucleotide Sequencing ,Germ Cells ,Cutaneous melanoma ,lcsh:Q ,Female ,Systems biology - Abstract
Although next-generation sequencing has demonstrated great potential for novel gene discovery, confirming disease-causing genes after initial discovery remains challenging. Here, we applied a network analysis approach to prioritize candidate genes identified from whole-exome sequencing analysis of 98 cutaneous melanoma patients from 27 families. Using a network propagation method, we ranked candidate genes by their similarity to known disease genes in protein–protein interaction networks and identified gene clusters with functional connectivity. Using this approach, we identified several new candidate susceptibility genes that warrant future investigations such as NGLY1, IL1RN, FABP2, PRKDC, and PROSER2. The propagated network analysis also allowed us to link families that did not have common underlying genes but that carried variants in genes that interact on protein–protein interaction networks. In conclusion, our study provided an analysis perspective for gene prioritization in the context of genetic heterogeneity across families and prioritized top potential candidate susceptibility genes in our dataset.
- Published
- 2020
38. Ambient Ultraviolet Radiation and Sebaceous Carcinoma Incidence in the United States, 2000–2016
- Author
-
Zhi-Ming Mai, Eric A. Engels, Michael R. Sargen, Ruth M. Pfeiffer, Margaret A. Tucker, Elizabeth K. Cahoon, and Alisa M. Goldstein
- Subjects
Cancer Research ,education.field_of_study ,medicine.medical_specialty ,integumentary system ,business.industry ,Incidence (epidemiology) ,Population ,fungi ,medicine.disease ,Rate ratio ,Brief Communication ,Dermatology ,Cancer registry ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Epidemiology ,medicine ,Skin cancer ,Risk factor ,business ,education ,Sebaceous carcinoma - Abstract
Sebaceous carcinoma (SC) is an aggressive skin tumor. Although ultraviolet radiation (UVR) is an important risk factor for some skin cancer types, no population-level study has evaluated for an association between UVR and SC risk. Herein, we examined satellite-based ambient UVR in relation to SC incidence using Surveillance, Epidemiology, and End Results 18 cancer registry data (2000–2016). There were 3503 microscopically confirmed cases of SC diagnosed during the study period. For non-Hispanic whites, there was an association between increasing ambient UVR and SC risk (incidence rate ratio [per UVR quartile] = 1.15, 95% confidence interval = 1.11 to 1.19; two-sided P
- Published
- 2020
39. Low-frequency variation near common germline susceptibility loci are associated with risk of Ewing sarcoma
- Author
-
Sakina Zaidi, Didier Surdez, Javier Alonso, Gregory T. Armstrong, Gaëlle Pierron, Nadège Corradini, Nathaniel Rothman, Markus Metzler, Valérie Laurence, Kathleen Wyatt, Kristine Jones, Heinrich Kovar, Brian D. Carter, Eve Lapouble, Weiyin Zhou, Leslie L. Robison, Rebeca Alba Rubio, Thomas Kirchner, Wendy M. Leisenring, David G. Cox, Manuela Krumbholz, Lisa Mirabello, Smita Bhatia, Olivier Delattre, Casey L. Dagnall, Lindsay M. Morton, Neal D. Freedman, Stéphanie Reynaud, Stelly Ballet, Udo Kontny, Thomas Meitinger, Sandrine Grossetête-Lalami, Robert N. Hoover, Shu-Hong Lin, Ana Patiño-García, Joshua N. Sampson, Margaret A. Tucker, Laurie Burdett, Olivier Mirabeau, Perrine Marec Bérard, Mitchell J. Machiela, Jeremy A. Miller, Stephen J. Chanock, Thomas G. P. Grunewald, Jennifer Kriebel, Jean Michon, Meredith Yeager, Uta Dirksen, Wolfgang Hartmann, Andreas E. Kulozik, Javed Khan, Konstantin Strauch, Franck Tirode, Eric Karlins, Anna González-Neira, Michelle Manning, NIH - National Cancer Institute (NCI) (Estados Unidos), Agence Nationale de la Recherche (Francia), Unión Europea. Comisión Europea. 7 Programa Marco, Deutsche Forschungsgemeinschaft (Alemania), Instituto de Salud Carlos III, Comunidad Foral de Navarra (España), National Cancer Institute (United States), Agence Nationale de la Recherche (France), 7º Programa Marco - Comisión Europea, Deutsche Forschungsgemeinschaft, Instituto de Salud Carlos III - ISCIII, and Gobierno de Navarra
- Subjects
0301 basic medicine ,Heredity ,Epidemiology ,Medizin ,Genome-wide association study ,Linkage Disequilibrium ,0302 clinical medicine ,Medizinische Fakultät ,Medicine and Health Sciences ,Odds Ratio ,Genetics ,Multidisciplinary ,Cancer Risk Factors ,Prostate Cancer ,Prostate Diseases ,Sarcoma ,Genomics ,Genetic Mapping ,Oncology ,030220 oncology & carcinogenesis ,Medicine ,Research Article ,Urology ,Science ,Ewing Sarcoma ,Locus (genetics) ,Variant Genotypes ,Sarcoma, Ewing ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Genetic variation ,Genome-Wide Association Studies ,Humans ,Genetic Predisposition to Disease ,ddc:610 ,Allele ,Alleles ,Genetic association ,Colorectal Cancer ,Haplotype ,Cancers and Neoplasms ,Biology and Life Sciences ,Computational Biology ,Genetic Variation ,Human Genetics ,Genome Analysis ,Minor allele frequency ,Genitourinary Tract Tumors ,030104 developmental biology ,Germ Cells ,Genetic Loci ,Medical Risk Factors ,Chromosome 20 ,Genome-Wide Association Study - Abstract
PLOS ONE 15(9), e0237792 (2020). doi:10.1371/journal.pone.0237792, Published by PLOS, San Francisco, California, US
- Published
- 2020
- Full Text
- View/download PDF
40. Genome-wide association meta-analyses combining multiple risk phenotypes provide insights into the genetic architecture of cutaneous melanoma susceptibility
- Author
-
Eduardo Nagore, Caroline Hayward, Christopher I. Amos, Douglas F. Easton, Zaida García-Casado, Julie Lang, Anjali K. Henders, Veronica Höiom, Lisa Bowdler, Kathryn P. Burdon, Laura Del Regno, Nick Orr, Per Arne Andresen, Tongwu Zhang, Rose Yang, Myriam Brossard, Eric K. Moses, Dirk Schadendorf, Laura Cattaneo, Casey Rowe, Essen-Heidelberg Investigators, Hans-Joachim Schulze, Jamie E Craig, D. Timothy Bishop, Anne E. Cust, Johan Hansson, David E. Elder, Nelleke A. Gruis, Donato Calista, Wei V. Chen, Abrar A. Qureshi, Amy Hutchinson, Pilar Galan, Leanne Wallace, Jennifer H. Barrett, Nilesh J. Samani, Maria Teresa Landi, Per Helsing, Andreas Hadjisavvas, Fengju Song, Celia Requena, Elizabeth M. Gillanders, Arantxa Rodriguez, Joan Anton Puig-Butille, Blair H. Smith, Mark Smithers, Michael Malasky, Marianna Sanna, Miriam Potrony, Maria A. Loizidou, Evangelos Evangelou, Richard A. Scolyer, Karen A. Pooley, Rachel E. Neale, Mario Falchi, Adèle C. Green, Monica Rodolfo, Ketty Peris, Licia Rivoltini, Mark M. Iles, Catherine M. Olsen, Alexander J. Stratigos, Tadeusz Dębniak, Weiyin Zhou, H. Peter Soyer, Xin Li, Margaret A. Tucker, Rajesh Kumar, Håkan Olsson, Anders Molven, Nicholas G. Martin, Anthony J. Swerdlow, Aurelie Vogt, Lars A. Akslen, Stuart MacGregor, Sarah V. Ward, Hamida Mohamdi, Bruna Dalmasso, Grant W. Montgomery, Rona M. MacKie, Esther Azizi, Gonçalo R. Abecasis, Chiara Menin, Alison M. Dunning, Ibd investigators, Kevin M. Brown, Jiali Han, Veryan Codd, Graham J. Mann, Nicholas K. Hayward, Marko Hočevar, Eitan Friedman, Richard A. Sturm, Paola Queirolo, Qtwin Investigators, Lawrie Wheeler, Lars G. Fritsche, Shenying Fang, Kiarash Khosrotehrani, Nicole A Kukutsch, Pol Gimenez-Xavier, Belynda Hicks, Matthew Zawistowski, Alessia Visconti, Jessica Harris, Chad M. Brummett, Paola Ghiorzo, andMe, David G. Hunter, Veronique Bataille, Julia Newton-Bishop, Srdjan Novaković, Amfs Investigators, Siranoush Manoukian, Jianxin Shi, Mitchell J. Machiela, G. Mark Lathrop, Josep Malvehy, Katerina P. Kypreou, Susana Puig, Dale R. Nyholt, I. Stefanaki, Maria Concetta Fargnoli, Lisa A. Simms, Kerrie McAloney, M. Malt, Adam J. Trower, Matthew Law, Lei Song, Paul D.P. Pharoah, Christian Ingvar, Jiyeon Choi, Alisa M. Goldstein, Jeffrey E. Lee, Mark Harland, Cristina Pellegrini, Daniela Massi, Sarah Simi, Scott D. Gordon, Jacobo Martinez, Florence Demenais, Kristine Jones, Graham L. Radford-Smith, David C. Whiteman, Lorenza Pastorino, Lisa Elefanti, Arcangela De Nicolo, Mario Mandalà, Juliette Randerson-Moor, Q-Mega, Jan Lubinski, Stephen J. Chanock, Marie-Françoise Avril, David L. Duffy, Helen Gogas, Nienke van der Stoep, Peter A. Kanetsky, Georgina V. Long, National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), University of Leeds, QIMR Berghofer Medical Research Institute, National and Kapodistrian University of Athens (NKUA), Ospedale Policlinico San Martino [Genoa], Universita degli studi di Genova, Toxicité environnementale, cibles thérapeutiques, signalisation cellulaire (T3S - UMR_S 1124), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Maurizio Bufalini Hospital, University of L'Aquila [Italy] (UNIVAQ), Fondazione IRCCS Istituto Nazionale Tumori - National Cancer Institute [Milan], Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana [Espagne] (FISABIO), Cyprus Institute of Neurology and Genetics, University of Athens Medical School [Athens], The University of Sydney, Universitat de Barcelona (UB), Azienda Ospedaliera Ospedale Papa Giovanni XXIII [Bergamo, Italy], University of Michigan [Ann Arbor], University of Michigan System, Instituto Valenciano de Oncologia, Veneto Institute of Oncology IOV-IRCCS [Padua, Italy], IRCCS Istituto Nazionale dei Tumori [Milano], Ninewells Hospital and Medical School [Dundee], Cyprus Institute (CyI), Fondazione 'Policlinico Universitario A. Gemelli' [Rome], Università degli Studi di Firenze = University of Florence [Firenze] (UNIFI), University of Queensland [Brisbane], Haukeland University Hospital, University of Bergen (UiB), Geisel School of Medicine at Dartmouth, Oslo University Hospital [Oslo], Service de Dermatologie [CHU Cochin], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Sackler Faculty of Medicine, Tel Aviv University [Tel Aviv], King‘s College London, Menzies School of Health Research [Australia], Charles Darwin University, The University of Texas M.D. Anderson Cancer Center [Houston], University of Leicester, Flinders University [Adelaide, Australia], West Pomeranian University of Technology Szczecin, Equipe 3: EREN- Equipe de Recherche en Epidémiologie Nutritionnelle (CRESS - U1153), Université Sorbonne Paris Nord-Centre de Recherche Épidémiologie et Statistique Sorbonne Paris Cité (CRESS (U1153 / UMR_A_1125 / UMR_S_1153)), Conservatoire National des Arts et Métiers [CNAM] (CNAM)-Université Sorbonne Paris Cité (USPC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Conservatoire National des Arts et Métiers [CNAM] (CNAM)-Université Sorbonne Paris Cité (USPC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), National Human Genome Research Institute (NHGRI), Leiden University Medical Center (LUMC), Karolinska Institutet [Stockholm], Queensland University of Technology [Brisbane] (QUT), Institute of Oncology Ljubljana, Harvard T.H. Chan School of Public Health, Lund University [Lund], German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), University of Glasgow, Statistical Genetics, and Intramural Research Program of the Division of Cancer Epidemiology and Genetics, National Cancer Institute (NCI), National Institutes of Health (NIH) United States Department of Health & Human ServicesNational Institutes of Health (NIH) - USAU19CA148112SEARCH team (Cancer Research UK) C490/A16561AOCS (US Army Medical Research and Material Command) DAMD17-01-1-0729Canadian Institutes of Health Research (CIHR)Cancer Council Victoria Queensland Cancer Fund Cancer Council New South Wales Cancer Council South Australia Cancer Council Western Australia Cancer Council Tasmania National Health and Medical Research Council of AustraliaID400413ID400281National Health and Medical Research Council of Australia National Health and Medical Research Council of Australia National Health and Medical Research Council of Australia National Health and Medical Research Council of Australia Intramural Research Program of the Division of Cancer Epidemiology and Genetics, NCI, NIH, DHHS Ministry of Health, Italy Associazione Italiana per la Ricerca sul Cancro (AIRC)IG 15460Spanish Fondo de Investigaciones Sanitarias grant - FEDER 'Una manera de hacer Europa' PI15/00716PI15/00956CIBER de Enfermedades Raras of the Instituto de Salud Carlos III, Spain - European Development Regional Fund 'A way to achieve Europe' ERDF AGAUR of the Catalan Government, Spain 2014_SGR_603European CommissionEuropean Commission Joint Research CentreLSHC-CT-2006-018702European Union (EU) 'Fundacio La Marato de TV3', Catalonia, Spain 201331-30'Fundacion Cientifica de la Asociacion Espanola Contra el Cancer', Spain GCB15152978SOENCERCA Programme/Generalitat de Catalunya Italian Ministry of the University and Scientific Research (PRIN-2012 grant) 2012JJX494Melanoma Research Alliance United States Department of Health & Human ServicesNational Institutes of Health (NIH) - USANIH National Cancer Institute (NCI)CA88363CA83115CA122838CA87969CA055075CA100264CA133996CA49449National Health and Medical Research Council of Australia200071241944339462380385389927389875389891389892389938443036442915442981496610496675496739552485552498APP1049894Cancer Council Queensland Cancer Institute New South Wales Australian GovernmentDepartment of Industry, Innovation and ScienceCooperative Research Centres (CRC) Programme Australian Cancer Research Foundation Wellcome TrustWT084766/Z/08/ZNational Health and Medical Research Council of Australia Australian Research Council Department of Health and Human Services (DHHS)
- Subjects
Sequence Variants ,Male ,medicine.medical_specialty ,Skin Neoplasms ,Genotype ,Medizin ,Identifies 3 ,Genome-wide association study ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Malignant-melanoma ,0302 clinical medicine ,Genetics ,medicine ,Genetic predisposition ,Nevus ,Humans ,Hla Class-i ,Genetic Predisposition to Disease ,Polymorphism ,Melanoma ,030304 developmental biology ,Telomere Length ,Cancer ,0303 health sciences ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Loci ,Pigmentation ,E-cadherin ,Single Nucleotide ,medicine.disease ,Genetic architecture ,Attributable Fraction ,3. Good health ,Phenotype ,Female ,Genetic Loci ,Genome-Wide Association Study ,Cutaneous melanoma ,Medical genetics ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,030217 neurology & neurosurgery ,Familial Melanoma - Abstract
Meta-analysis of 36,760 cases and 375,188 controls identifies 54 loci associated with susceptibility to cutaneous melanoma. Further analysis combining nevus count and hair color GWAS results provide insights into the genetic architecture of melanoma.Most genetic susceptibility to cutaneous melanoma remains to be discovered. Meta-analysis genome-wide association study (GWAS) of 36,760 cases of melanoma (67% newly genotyped) and 375,188 controls identified 54 significant (P < 5 x 10(-8)) loci with 68 independent single nucleotide polymorphisms. Analysis of risk estimates across geographical regions and host factors suggests the acral melanoma subtype is uniquely unrelated to pigmentation. Combining this meta-analysis with GWAS of nevus count and hair color, and transcriptome association approaches, uncovered 31 potential secondary loci for a total of 85 cutaneous melanoma susceptibility loci. These findings provide insights into cutaneous melanoma genetic architecture, reinforcing the importance of nevogenesis, pigmentation and telomere maintenance, together with identifying potential new pathways for cutaneous melanoma pathogenesis.
- Published
- 2020
41. Pediatric melanoma in melanoma-prone families
- Author
-
Xiaohong R. Yang, Mary C. Fraser, Kelsey C. Stidd, Margaret A. Tucker, and Alisa M. Goldstein
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Sun protection ,Population ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,CDKN2A ,hemic and lymphatic diseases ,Internal medicine ,medicine ,In patient ,education ,neoplasms ,Fisher's exact test ,education.field_of_study ,business.industry ,Melanoma ,medicine.disease ,digestive system diseases ,stomatognathic diseases ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Pediatric melanoma ,Skin surveillance ,symbols ,business - Abstract
Background In the United States, only approximately 0.4% of all melanomas are diagnosed in patients aged Methods For this non-population-based study, families were followed prospectively for up to 40 years. A total of 60 families with ≥ 3 patients with melanoma were included for analysis: 30 CDKN2A mutation-positive (CDKN2A+) and 30 CDKN2A mutation-negative (CDKN2A-) families. Age at the time of first melanoma and number of melanomas were obtained for each patient and summarized by family or sets (CDKN2A + vs CDKN2A-). For set comparisons and categorical variables (occurrence of melanoma in pediatric patients, number of melanomas, number of patients with single or multiple melanomas), the Pearson chi-square or Fisher exact test was used. Results Regardless of CDKN2A status, melanoma-prone families were found to have 6-fold to 28-fold higher percentages of patients with pediatric melanoma compared with the general population of patients with melanoma in the United States. Within CDKN2A + families, pediatric patients with melanoma were significantly more likely to have multiple melanomas compared with their relatives who were diagnosed at age >20 years (71% vs 38%, respectively; P = .004). CDKN2A + families had significantly higher percentages of pediatric patients with melanoma compared with CDKN2A- families (11.1% vs 2.5%; P = .004). Conclusions These observations have implications for the prevention of melanoma as well as clinical care for its early detection. Children in melanoma-prone families should have careful sun protection from an early age and skin surveillance to reduce their risk of melanoma.
- Published
- 2018
42. Phenocopies in melanoma-prone families with germ-line CDKN2A mutations
- Author
-
Margaret A. Tucker, Alisa M. Goldstein, Veronica Höiom, Hildur Helgadottir, Xiaohong R. Yang, and Håkan Olsson
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Skin Neoplasms ,Adolescent ,Population ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,CDKN2A ,hemic and lymphatic diseases ,Internal medicine ,Cyclin-Dependent Kinase Inhibitor p18 ,Humans ,Medicine ,Genetic Predisposition to Disease ,Prospective Studies ,Child ,education ,Melanoma ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,Germ-Line Mutation ,Genetics (clinical) ,Phenocopy ,education.field_of_study ,Mutation ,business.industry ,Squamous cell skin cancer ,Middle Aged ,medicine.disease ,digestive system diseases ,stomatognathic diseases ,Phenotype ,030104 developmental biology ,Child, Preschool ,030220 oncology & carcinogenesis ,Relative risk ,Medical genetics ,Female ,business - Abstract
Purpose: Carriers of CDKN2A mutations have high risks of melanoma and certain other cancers. In this study we examined the occurrence of tumors among CDKN2A wild type (wt) members of melanoma-prone families with CDKN2A mutations. Methods: Swedish and US melanoma-prone families with CDKN2A mutations were included. Data was collected on tumors diagnosed among family members. Among the CDKN2A mutated families, members with CDKN2A wt status who were diagnosed with melanoma were designated phenocopies. Results: Of patients with melanoma in the CDKN2A mutated families (n = 266), 7.1%, were seen among members with CDKN2A wt status (phenocopy rate). Among the CDKN2A wt family members of the CDKN2A mutated families (n = 256), 7.4% were diagnosed with melanoma. The prospective relative risk for melanomas was significantly higher among the CDKN2A wt subjects compared with population-based controls (7.4 (95% confidence interval 1.7–33.2)), while no elevated risks of nonmelanoma cancers were seen and their offspring did not have significantly elevated risks of melanoma or other cancers. Conclusion: Members of CDKN2A mutation carrying families who test negative for their family’s mutation have moderately increased risk for melanoma and should, in addition to being considered for continuing dermatologic surveillance, be encouraged to follow sun safety recommendations and practice skin self-exams. (Less)
- Published
- 2018
43. Recommendations for Long-Term Follow-up of Adults with Heritable Retinoblastoma
- Author
-
Annette C. Moll, Lindsay M. Morton, Charlotte J. Dommering, Michael Walsh, Machteld I. Bosscha, Kevin C. Oeffinger, Ruth A. Kleinerman, Ira J. Dunkel, Danielle Novetsky Friedman, Flora E. van Leeuwen, Mary-Louise C. Greer, Petra Temming, Dana Barnea, Pim de Graaf, Margaret A. Tucker, Guillermo L. Chantada, Armida W. M. Fabius, Suzanne Laughlin, Emily S. Tonorezos, David H. Abramson, Wijnanda A. Kors, Jasmine H. Francis, Ophthalmology, Human genetics, Radiology and nuclear medicine, Pediatric surgery, CCA - Cancer Treatment and quality of life, APH - Health Behaviors & Chronic Diseases, APH - Quality of Care, Epidemiology and Data Science, and ACS - Diabetes & metabolism
- Subjects
Pediatrics ,medicine.medical_specialty ,Retinal Neoplasms ,Population ,Medizin ,MEDLINE ,Guidelines as Topic ,Disease ,Global Health ,Risk Assessment ,Article ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Risk Factors ,medicine ,Humans ,Genetic Predisposition to Disease ,education ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Retinoblastoma ,business.industry ,Incidence (epidemiology) ,Incidence ,medicine.disease ,Ophthalmology ,030221 ophthalmology & optometry ,Sarcoma ,Risk assessment ,business ,Follow-Up Studies - Abstract
OBJECTIVE: Generate recommendations for long-term follow-up for adult survivors of heritable retinoblastoma.DESIGN: We convened a meeting of providers from retinoblastoma centers around the world to review the state of the science and to evaluate the published evidence.SUBJECTS: Retinoblastoma is a rare childhood cancer of the retina. Approximately forty percent of retinoblastoma cases are heritable, due to a germline mutation in RB1. Dramatic improvements in treatment and supportive care have resulted in a growing adult survivor population. Survivors of heritable retinoblastoma, however, have significantly increased risk of subsequent malignant neoplasms, particularly bone and soft tissue sarcomas, uterine leiomyosarcoma, melanomas, and radiotherapy-related central nervous system tumors, which are associated with excess morbidity and mortality. In spite of these risks, no surveillance recommendations for this population are currently in place and surveillance practices vary widely by center.METHODS: Following the Institute of Medicine procedure for clinical practice guideline development, a PubMed, EMBASE, and Web of Science search was performed, resulting in 139 papers; after abstract and full text review, 37 papers underwent detailed data abstraction to quantify risk and evidence regarding surveillance, if available. During an in-person meeting, evidence was presented and discussed, resulting in consensus recommendations.MAIN OUTCOME MEASURES: Diagnosis and mortality from subsequent neoplasm.RESULTS: While evidence for risk of subsequent neoplasm, especially sarcoma and melanoma, was significant, evidence supporting routine testing of asymptomatic survivors was not identified. Skin examination for melanoma and prompt evaluation of signs and symptoms of head and neck disease were determined to be prudent.CONCLUSIONS: This review of the literature confirmed some of the common second cancers in retinoblastoma survivors, but found little evidence for a benefit to currently available surveillance for these malignancies. Future research should incorporate international partners, patients, and family members.
- Published
- 2019
44. Genome-wide association study identifies multiple new loci associated with Ewing sarcoma susceptibility
- Author
-
Ana Patiño-García, Thomas G. P. Grunewald, David G. Cox, Lindsay M. Morton, Michelle Manning, Sandrine Grossetête-Lalami, Gaëlle Pierron, Nadège Corradini, Stephen J. Chanock, Kathleen Wyatt, Udo Kontny, Gregory T. Armstrong, Jean Michon, Sakina Zaidi, Didier Surdez, Wolfgang Hartmann, Heinrich Kovar, Olivier Delattre, Jennifer Kriebel, Nathalie Gaspar, Perrine Marec Bérard, Valérie Laurence, Casey L. Dagnall, Thomas Kirchner, Stéphanie Reynaud, Uta Dirksen, Nathaniel Rothman, Konstantin Strauch, Margaret A. Tucker, Lisa Mirabello, Smita Bhatia, Markus Metzler, Laurie Burdett, Neal D. Freedman, Rebeca Alba Rubio, Franck Tirode, Andreas E. Kulozik, Meredith Yeager, Kristine Jones, Javier Alonso, Stelly Ballet, Olivier Mirabeau, Eve Lapouble, Robert N. Hoover, Weiyin Zhou, Wendy M. Leisenring, Leslie L. Robison, Piero Picci, Javed Khan, Thomas Meitinger, Anna González-Neira, Eric Karlins, Mitchell J. Machiela, Unión Europea, TIRODE, Franck, Clinical Genetics Branch, Division of Cancer Epidemiology & Genetics, National Institutes of Health [Bethesda] (NIH)-National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), Laboratory for Pediatric Sarcoma Biology [Munich, Germany], Ludwig-Maximilian-Universität München Pathologisches Institut [Germany], German Cancer Consortium [Heidelberg] (DKTK), German Cancer Research Center - Deutsches Krebsforschungszentrum [Heidelberg] (DKFZ), Unité de Génétique Somatique, Institut Curie [Paris], Unité de génétique et biologie des cancers (U830), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Département d'Oncologie Médicale [Institut Curie, Paris], Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, Frederick National Laboratory for Cancer Research (FNLCR), Unité de Génétique Somatique [Institut Curie, Paris], Children’s Cancer Research Institute [Vienna, Austria], Département de cancérologie de l'enfant et de l'adolescent [Gustave Roussy], Institut Gustave Roussy (IGR), Division of Pediatric Hematology, Oncology and Stem Cell Transplantation [Aechen, Germany], Genotyping Unit (CeGen), Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Istituto Ortopedico Rizzoli [Bologna, Italy], Unidad de Tumores Sólidos Infantiles – Unidad de Investigación Biomédica [Madrid, Spain], Instituto de Salud Carlos III [Madrid] (ISC)- Instituto de Investigación en Enfermedades Raras (IIER), Center for Applied Medical Research [Plamplona] (CIMA), Universidad de Navarra [Pamplona] (UNAV), Institut d'hématologie et d'oncologie pédiatrique [CHU - HCL] (IHOPe), Hospices Civils de Lyon (HCL), Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Clinical Genetics Branch, National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH)-National Institutes of Health [Bethesda] (NIH), Department of Epidemiology and Cancer Control [Memphis, TN, USA], Cancer Prevention and Clinical Statistics Program [Seattle, WA, USA], Institute for Cancer Outcomes and Survivorship [Birmingham, AL, USA], University Children's Hospital of Heidelberg [Heidelberg, Germany], Research Unit of Molecular Biology [Neuherberg, Germany], Institute of Epidemiology [Neuherberg] (EPI), German Research Center for Environmental Health - Helmholtz Center München (GmbH), German Center for Diabetes Diseases [Neuherberg, Germany] (DZD), Institute of Human Genetics [Neuherberg] (IHG), Helmholtz Zentrum München = German Research Center for Environmental Health, Institute of Human Genetics [Munich, Germany], Technische Universität Munchen - Université Technique de Munich [Munich, Allemagne] (TUM), Pediatric Oncology and Hematology [Erlangen, Germany], University Hospital Erlangen = Uniklinikum Erlangen, Gerhard-Domagk-Institute of Pathology, Westfälische Wilhelms-Universität Münster = University of Münster (WWU), Institute of Genetic Epidemiology [Neuherberg, Germany], Chair of Genetic Epidemiology [Munich, Germany] (IBE), Institute of Pathology [Munich, Germany], University Children's Hospital of Essen [Essen, Germany], This work was supported by the Intramural Research Program of the U.S. NationalCancer Institute and the Intramural Research Program of the American Cancer Society.This work was supported by grants from the Institut Curie, the Inserm, the LigueNationale Contre le Cancer (Equipe labellisée, Carte d’Identité des Tumeurs programand Recherche Epidémiologique 2009 program), the ANR-10-EQPX-03 from the AgenceNationale de la Recherche, the European PROVABES (ERA-649 NET TRANSCAN JTC2011), and ASSET (FP7-HEALTH-2010-259348) projects. This research was supportedby FP7 grant 'EURO EWING Consortium' No. 602856 and the following associations:Courir pour Mathieu, Dans les pas du Géant, Les Bagouzamanon, Enfants et Santé, M lavie avec Lisa, Lulu et les petites bouilles de lune, les Amis de Claire, l’Etoile de Martin andthe Société Française de lutte contre les Cancers et les leucémies de l’Enfant et del’adolescent. The laboratory of T. G. P. Grünewald is supported by grants from the‘Verein zur Förderung von Wissenschaft und Forschung an der Medizinischen Fakultätder LMU München (WiFoMed)’, by LMU Munich’s Institutional Strategy LMU excellentwithin the framework of the German Excellence Initiative, the ‘Mehr LEBEN für krebskranke Kinder—Bettina-Bräu-Stiftung’, the Walter Schulz Foundation, the WilhelmSander-Foundation (2016.167.1), and by the German Cancer Aid (DKH-111886 andDKH-70112257). D. Surdez is supported by SiRIC (Grant « INCa-DGOS-4654). Wethank the following clinicians for providing samples used in this study: C. Alenda, F.Almazán, D. Ansoborlo, L. Aymerich, L. Benboukbher, C. Beléndez, C. Berger, C. Bergeron, P. Biron, J.Y. Blay, E. Bompas, H. Bonnefoi, P. Boutard, B. Bui-Nguyen, D.Chauveaux, C. Calvo, A. Carboné, C. Clement, T. Contra, N. Corradini, A.S. Defachelles,V. Gandemer-Delignieres, A. Deville, A. Echevarria, J. Fayette, M. Fraga, D. Frappaz, J.L.Fuster, P. García-Miguel, J.C. Gentet, P. Kerbrat, V. Laithier, V. Laurence, P. Leblond, O.Lejars, R. López-Almaraz, B. López-Ibor, P. Lutz, J.F. Mallet, L. Mansuy, P. Marec Bérard,G. Margueritte, A. Marie Cardine, C. Melero, L. Mignot, F. Millot, O. Minckes, G.Margueritte, C. Mata, M.E. Mateos, M. Melo, C. Moscardó, M. Munzer, B. Narciso, A.Navajas, D. Orbach, C. Oudot, H. Pacquement, C. Paillard, Y. Perel, T. Philip, C. Piguet,M.I. Pintor, D. Plantaz, E. Plouvier, S. Ramirez-Del-Villar, I. Ray-Coquard, Y. Reguerre,M. Rios, P. Rohrlich, H. Rubie, A. Sastre, G. Schleiermacher, C. Schmitt, P. Schneider, L.Sierrasesumaga, C. Soler, N. Sirvent, S. Taque, E. Thebaud, A. Thyss, R. Tichit, J.J. Uriz, J.P. Vannier, F. Watelle-Pichon. This work was supported by the Instituto de SaludCarlos III (PI16CIII/00026) and the Asociación Pablo Ugarte, Fundación Sonrisa deAlex, ASION-La Hucha de Tomás, Sociedad Española de Hematología y OncologíaPediátricas. The Childhood Cancer Survivor Study is supported by the NationalCancer Institute (CA55727, G.T. Armstrong, Principal Investigator), with funding forgenotyping from the Intramural Research Program of the National Institutes ofHealth, National Cancer Institute. The KORA study was initiated and financed by theHelmholtz Zentrum München—German Research Center for Environmental Health,which is funded by the German Federal Ministry of Education and Research (BMBF) andby the State of Bavaria. Furthermore, KORA research was supported within the MunichCenter of Health Sciences (MC-Health), Ludwig-Maximilians-Universität, as part ofLMUinnovativ, Helmholtz-Zentrum München (HZM), University Hospital Erlangen [Germany], Westfälische Wilhelms-Universität Münster (WWU), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), and Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Candidate gene ,Oncogene Proteins, Fusion ,Medizin ,General Physics and Astronomy ,Genome-wide association study ,Cell Cycle Proteins ,[SDV.BBM.BM] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,lcsh:Science ,Genetics ,Multidisciplinary ,Nuclear Proteins ,3. Good health ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Homeobox Protein Nkx-2.2 ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Quality Control ,Risk ,Genotype ,Science ,European Continental Ancestry Group ,Quantitative Trait Loci ,Single-nucleotide polymorphism ,Locus (genetics) ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Sarcoma, Ewing ,Biology ,Quantitative trait locus ,Polymorphism, Single Nucleotide ,Article ,White People ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Humans ,Genetic Predisposition to Disease ,Allele ,Alleles ,Cell Proliferation ,Homeodomain Proteins ,Proto-Oncogene Protein c-fli-1 ,Gene Expression Profiling ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,General Chemistry ,Zebrafish Proteins ,Pediatric cancer ,030104 developmental biology ,Expression quantitative trait loci ,lcsh:Q ,RNA-Binding Protein EWS ,Transcription Factors ,Genome-Wide Association Study - Abstract
Ewing sarcoma (EWS) is a pediatric cancer characterized by the EWSR1-FLI1 fusion. We performed a genome-wide association study of 733 EWS cases and 1346 unaffected individuals of European ancestry. Our study replicates previously reported susceptibility loci at 1p36.22, 10q21.3 and 15q15.1, and identifies new loci at 6p25.1, 20p11.22 and 20p11.23. Effect estimates exhibit odds ratios in excess of 1.7, which is high for cancer GWAS, and striking in light of the rarity of EWS cases in familial cancer syndromes. Expression quantitative trait locus (eQTL) analyses identify candidate genes at 6p25.1 (RREB1) and 20p11.23 (KIZ). The 20p11.22 locus is near NKX2-2, a highly overexpressed gene in EWS. Interestingly, most loci reside near GGAA repeat sequences and may disrupt binding of the EWSR1-FLI1 fusion protein. The high locus to case discovery ratio from 733 EWS cases suggests a genetic architecture in which moderate risk SNPs constitute a significant fraction of risk., Ewing sarcoma (EWS) is a rare pediatric bone cancer typically involving the EWSR1-FLI1 fusion. Here the authors perform a genome-wide association study and report three new EWS risk loci that reside near GGAA repeat sequences, and identify candidate genes (RREB1 and KIZ) from eQTL analysis.
- Published
- 2018
45. Mutual Risks of Cutaneous Melanoma and Specific Lymphoid Neoplasms: Second Cancer Occurrence and Survival
- Author
-
Rochelle E. Curtis, Margaret A. Tucker, Sara J. Schonfeld, Lindsay M. Morton, Diana R. Withrow, Graça M. Dores, and Megan M. Herr
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Skin Neoplasms ,Lymphoma ,Population ,Follicular lymphoma ,Risk Assessment ,Gastroenterology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,education ,Melanoma ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,education.field_of_study ,business.industry ,Proportional hazards model ,Incidence ,Hazard ratio ,Cancer ,Neoplasms, Second Primary ,Articles ,Middle Aged ,Plasma cell neoplasm ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female ,Marginal zone B-cell lymphoma ,business ,Diffuse large B-cell lymphoma ,SEER Program - Abstract
Background It is unclear whether the established association between cutaneous melanoma (CM) and lymphoid neoplasms (LNs) differs across LN subtypes. This study quantifies risk for developing CM after specific LNs and, conversely, for developing specific LNs after CM, as well as assessing clinical impact. Methods We identified a cohort of Caucasian adults (age 20-83 years) initially diagnosed with CM or LN, as reported to 17 US population-based cancer registries, 2000-2014. Standardized incidence ratios (SIRs) quantified second cancer risk. We assessed impact of second cancer development on risk of all-cause mortality using Cox regression. Results Among 151 949 one-or-more-year survivors of first primary LN, second primary CM risk was statistically significantly elevated after chronic lymphocytic leukemia/small lymphocytic lymphoma (SIR = 1.96, 95% confidence interval [CI] = 1.74 to 2.21), follicular lymphoma (SIR = 1.32, 95% CI = 1.09 to 1.58), and plasma cell neoplasms (SIR = 1.33, 95% CI = 1.07 to 1.63). Risks for these same subtypes were statistically significantly elevated among 148 336 survivors of first primary CM (SIR = 1.44, 95% CI = 1.25 to 1.66; SIR = 1.47, 95% CI = 1.21 to 1.77; SIR = 1.25, 95% CI = 1.06 to 1.47; respectively). Risk for CM was statistically significantly elevated after diffuse large B-cell lymphoma (SIR = 1.22, 95% CI = 1.02 to 1.45) and Hodgkin lymphoma (SIR = 1.75, 95% CI = 1.33 to 2.26), but the reciprocal relationship was not observed. There were no statistically significant associations between marginal zone lymphoma and CM. Among survivors of most LN subtypes, CM statistically significantly increased risk of death (hazard ratio [HR] range = 1.52, 95% CI = 1.25 to 1.85, to 2.46, 95% CI = 1.45 to 4.16). Among survivors of CM, LN statistically significantly increased risk of death (HR range = 1.75, 95% CI = 1.15 to 2.65, to 6.28, 95% CI = 5.00 to 7.88), with the highest risks observed for the most aggressive LN subtypes. Conclusions Heterogeneous associations between CM and specific LN subtypes provide novel insights into the etiology of these malignancies, with the mutual association between CM and certain LN suggesting shared etiology. Development of second primary CM or LN substantially reduces overall survival.
- Published
- 2018
46. Increased Risk of Skin Cancer in 1,851 Long-Term Retinoblastoma Survivors
- Author
-
Jasmine H. Francis, Johanna M. Seddon, Sara J. Schonfeld, Ruth A. Kleinerman, Lindsay M. Morton, David H. Abramson, and Margaret A. Tucker
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Skin Neoplasms ,Adolescent ,Retinal Neoplasms ,Dermatology ,Biochemistry ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Cancer Survivors ,Risk Factors ,Humans ,Medicine ,Cumulative incidence ,Melanoma ,Molecular Biology ,Aged ,business.industry ,Retinoblastoma ,Incidence (epidemiology) ,Cell Biology ,Middle Aged ,medicine.disease ,humanities ,030104 developmental biology ,Standardized mortality ratio ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Hereditary Retinoblastoma ,Cohort ,Female ,Skin cancer ,business - Abstract
Patients with hereditary retinoblastoma are at risk for developing cutaneous melanoma, but little is known about the role of sun exposure or other factors, and the incidence of nonmelanoma skin cancer (NMSC) is poorly understood. We investigated the incidence of melanoma and NMSC in a cohort of 1,851 White, long-term retinoblastoma survivors (1,020 hereditary and 831 nonhereditary) diagnosed during 1914‒2006. During follow-up through 2016, 33 hereditary and 7 nonhereditary survivors developed melanoma, and 26 hereditary and 9 nonhereditary survivors developed NMSC. Most NMSCs were on the head/neck, whereas melanomas were more broadly distributed with patterns similar to melanoma-prone families. For both outcomes, the median age at diagnosis was ~20 years younger among hereditary survivors than among nonhereditary survivors. At 50 years after retinoblastoma diagnosis, the cumulative incidence in hereditary survivors was 4.5% for melanoma and 3.7% for NMSC; for nonhereditary survivors, it was 0.7% and 1.5%, respectively. Sun sensitivity and phenotypic characteristics generally did not vary by skin cancer status. Hereditary retinoblastoma survivors have an increased risk for melanoma and NMSC that occurred earlier than that observed among nonhereditary survivors, likely reflecting genetic factors. These findings among White retinoblastoma survivors support consensus-based recommendations for skin cancer screening and sun protection starting at young ages and continuing long term.
- Published
- 2021
47. Genome-wide association study identifies theGLDC/IL33locus associated with survival of osteosarcoma patients
- Author
-
Mandy L. Ballinger, Massimo Serra, Jay S. Wunder, Silvia Regina Caminada de Toledo, Julie M. Gastier-Foster, Logan G. Spector, Richard Gorlick, Roberto Tirabosco, Irene L. Andrulis, Fernando Lecanda, Stephen J. Chanock, Antonio Sergio Petrilli, Robert N. Hoover, Orestis A. Panagiotou, William Wheeler, Katia Scotlandi, Claudia Maria Hattinger, Ana Patiño-García, Donald A. Barkauskas, Lisa Mirabello, Sholom Wacholder, Meredith Yeager, Adrienne M. Flanagan, Margaret A. Tucker, Nalan Gokgoz, Eric Karlins, Roelof Koster, D. Hicks Belynda, David Thomas, Piero Picci, and Sharon A. Savage
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Proportional hazards model ,business.industry ,Hazard ratio ,Single-nucleotide polymorphism ,Locus (genetics) ,Genome-wide association study ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Osteosarcoma ,Allele ,business ,Survival rate - Abstract
Survival rates for osteosarcoma, the most common primary bone cancer, have changed little over the past three decades and are particularly low for patients with metastatic disease. We conducted a multi-institutional genome-wide association study (GWAS) to identify germline genetic variants associated with overall survival in 632 patients with osteosarcoma, including 523 patients of European ancestry and 109 from Brazil. We conducted a time-to-event analysis and estimated hazard ratios (HR) and 95% confidence intervals (CI) using Cox proportional hazards models, with and without adjustment for metastatic disease. The results were combined across the European and Brazilian case sets using a random-effects meta-analysis. The strongest association after meta-analysis was for rs3765555 at 9p24.1, which was inversely associated with overall survival (HR = 1.76; 95% CI 1.41-2.18, p = 4.84 × 10-7 ). After imputation across this region, the combined analysis identified two SNPs that reached genome-wide significance. The strongest single association was with rs55933544 (HR = 1.9; 95% CI 1.5-2.4; p = 1.3 × 10-8 ), which localizes to the GLDC gene, adjacent to the IL33 gene and was consistent across both the European and Brazilian case sets. Using publicly available data, the risk allele was associated with lower expression of IL33 and low expression of IL33 was associated with poor survival in an independent set of patients with osteosarcoma. In conclusion, we have identified the GLDC/IL33 locus on chromosome 9p24.1 as associated with overall survival in patients with osteosarcoma. Further studies are needed to confirm this association and shed light on the biological underpinnings of this susceptibility locus.
- Published
- 2017
48. GWAS follow-up study of esophageal squamous cell carcinoma identifies potential genetic loci associated with family history of upper gastrointestinal cancer
- Author
-
Jin-Hu Fan, Neal D. Freedman, Wei Li Han, Laurie Burdett, Fu You Zhou, You-Lin Qiao, Ling Fen Ji, Christian C. Abnet, Stephen J. Chanock, Wen-Qing Li, Xin Song, Li-Dong Wang, Xiao-You Han, Li Sun, Peng Yuan Zheng, Alisa M. Goldstein, Nan Hu, Xue Min Li, Min Jie Wu, Hui Meng, Shuang Lv, Lemin Wang, Zhaoming Wang, Hua Su, Tao Jiang, Margaret A. Tucker, Ti Ding, Philip R. Taylor, Tang Juan Zhang, Chaoyu Wang, Guo Qiang Kong, Paula L. Hyland, Carol Giffen, Shou Jia Hu, Sanford M. Dawsey, Zong Min Fan, and Xue Ke Zhao
- Subjects
0301 basic medicine ,Oncology ,Male ,medicine.medical_specialty ,China ,Esophageal Neoplasms ,Science ,Genome-wide association study ,Single-nucleotide polymorphism ,Upper gastrointestinal cancer ,Bioinformatics ,Esophageal squamous cell carcinoma ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,0302 clinical medicine ,Asian People ,Polymorphism (computer science) ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Family history ,Gastrointestinal Neoplasms ,Neoplasm Staging ,Multidisciplinary ,business.industry ,Follow up studies ,Cancer ,medicine.disease ,030104 developmental biology ,Genetic Loci ,030220 oncology & carcinogenesis ,Medicine ,Female ,Esophageal Squamous Cell Carcinoma ,business ,Follow-Up Studies ,Genome-Wide Association Study - Abstract
Based on our initial genome-wide association study (GWAS) on esophageal squamous cell carcinoma (ESCC) in Han Chinese, we conducted a follow-up study to examine the single nucleotide polymorphisms (SNPs) associated with family history (FH) of upper gastrointestinal cancer (UGI) cancer in cases with ESCC. We evaluated the association between SNPs and FH of UGI cancer among ESCC cases in a stage-1 case-only analysis of the National Cancer Institute (NCI, 541 cases with FH and 1399 without FH) and Henan GWAS (493 cases with FH and 869 without FH) data (discovery phase). The top SNPs (or their surrogates) from discovery were advanced to a stage-2 evaluation in additional Henan subjects (2801 cases with FH and 3136 without FH, replication phase). A total of 19 SNPs were associated with FH of UGI cancer in ESCC cases with P −5 in the stage-1 meta-analysis of NCI and Henan GWAS data. In stage-2, the association for rs79747906 (located at 18p11.31, P = 5.79 × 10−6 in discovery) was replicated (P = 0.006), with a pooled-OR of 1.59 (95%CI: 1.11-2.28). We identified potential genetic variants associated with FH of UGI cancer. Our findings may provide important insights into new low-penetrance susceptibility regions involved in the susceptibility of families with multiple UGI cancer cases.
- Published
- 2017
49. Thyroid Cancer Following Childhood Low-Dose Radiation Exposure: A Pooled Analysis of Nine Cohorts
- Author
-
M. Jacob Adams, Erik Holmberg, Leslie L. Robison, Florent de Vathaire, Robert Johansson, Ruth A. Kleinerman, Marie Lundell, Ritsu Sakata, Jay H. Lubin, Margaret A. Tucker, Peter D. Inskip, Lene H.S. Veiga, Roy E. Shore, Siegal Sadetzki, Lena Damber, Arthur B. Schneider, and Michael M. Hawkins
- Subjects
Male ,medicine.medical_specialty ,Pediatrics ,Neoplasms, Radiation-Induced ,Adolescent ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Context (language use) ,Risk Assessment ,Biochemistry ,030218 nuclear medicine & medical imaging ,Cohort Studies ,03 medical and health sciences ,Sex Factors ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Humans ,Thyroid Neoplasms ,Child ,Survival rate ,Thyroid cancer ,business.industry ,Biochemistry (medical) ,Thyroid ,Age Factors ,Dose-Response Relationship, Radiation ,Radiation Exposure ,Prognosis ,medicine.disease ,3. Good health ,Survival Rate ,Dose–response relationship ,medicine.anatomical_structure ,Child, Preschool ,030220 oncology & carcinogenesis ,Relative risk ,Female ,business ,Risk assessment ,Cohort study - Abstract
The increased use of diagnostic and therapeutic procedures that involve radiation raises concerns about radiation effects, particularly in children and the radiosensitive thyroid gland.Evaluation of relative risk (RR) trends for thyroid radiation doses0.2 gray (Gy); evidence of a threshold dose; and possible modifiers of the dose-response, e.g., sex, age at exposure, time since exposure.Pooled data from nine cohort studies of childhood external radiation exposure and thyroid cancer with individualized dose estimates, ≥1000 irradiated subjects or ≥10 thyroid cancer cases, with data limited to individuals receiving doses0.2 Gy.Cohorts included the following: childhood cancer survivors (n = 2); children treated for benign diseases (n = 6); and children who survived the atomic bombings in Japan (n = 1). There were 252 cases and 2,588,559 person-years in irradiated individuals and 142 cases and 1,865,957 person-years in nonirradiated individuals.There were no interventions.Incident thyroid cancers.For both0.2 and0.1 Gy, RRs increased with thyroid dose (P0.01), without significant departure from linearity (P = 0.77 and P = 0.66, respectively). Estimates of threshold dose ranged from 0.0 to 0.03 Gy, with an upper 95% confidence bound of 0.04 Gy. The increasing dose-response trend persisted45 years after exposure, was greater at younger age at exposure and younger attained age, and was similar by sex and number of treatments.Our analyses reaffirmed linearity of the dose response as the most plausible relationship for "as low as reasonably achievable" assessments for pediatric low-dose radiation-associated thyroid cancer risk.
- Published
- 2017
50. Rare Germline Copy Number Variations and Disease Susceptibility in Familial Melanoma
- Author
-
Xijun Zhang, Margaret A. Tucker, Stephen J. Chanock, Lei Song, Laura Burke, Hunter Bennett, Laurie Burdette, Casey L. Dagnall, Daoud Meerzaman, Yanzi Xiao, Belynda Hicks, Bin Zhu, Chih Hao Hsu, Chunhua Yan, Meredith Yeager, Alisa M. Goldstein, Paula L. Hyland, Xiaohong R. Yang, Qing-Rong Chen, Neal D. Freedman, Jianxin Shi, and Weiyin Zhou
- Subjects
Male ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Skin Neoplasms ,DNA Copy Number Variations ,endocrine system diseases ,Single-nucleotide polymorphism ,Genome-wide association study ,Dermatology ,Biology ,Real-Time Polymerase Chain Reaction ,Risk Assessment ,Biochemistry ,Article ,Germline ,03 medical and health sciences ,Germline mutation ,CDKN2A ,mental disorders ,Gene duplication ,Humans ,Genetic Predisposition to Disease ,Copy-number variation ,Melanoma ,neoplasms ,Molecular Biology ,Germ-Line Mutation ,Genetics ,Sequence Analysis, RNA ,Incidence ,Cell Biology ,Pedigree ,030104 developmental biology ,Female ,Genome-Wide Association Study ,Comparative genomic hybridization - Abstract
Mounting evidence suggests that copy number variations (CNVs) can contribute to cancer susceptibility. The main goal of this study was to evaluate the role of germline CNVs in melanoma predisposition in high-risk melanoma families. We used genome-wide tiling comparative genomic hybridization and single nucleotide polymorphism arrays to characterize CNVs in 335 individuals (240 melanoma cases) from American melanoma-prone families (22 with germline CDKN2A or CDK4 mutations). We found that the global burden of overall CNVs (or deletions or duplications separately) was not significantly associated with case-control or CDKN2A/CDK4 mutation status after accounting for the familial dependence. However, we identified several rare CNVs that either involved known melanoma genes (e.g., PARP1, CDKN2A) or cosegregated with melanoma (duplication on 10q23.23, 3p12.2 and deletions on 8q424.3, 2q22.1) in families without mutations in known melanoma high-risk genes. Some of these CNVs were correlated with expression changes in disrupted genes based on RNASeq data from a subset of melanoma cases included in the CNV study. These results suggest that rare cosegregating CNVs may influence melanoma susceptibility in some melanoma-prone families and genes found in our study warrant further evaluation in future genetic analyses of melanoma.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.