Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lucie McCoy is active.

Publication


Featured researches published by Lucie McCoy.


The New England Journal of Medicine | 2015

Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors

Jeanette E. Eckel-Passow; Daniel H. Lachance; Annette M. Molinaro; Kyle M. Walsh; Paul A. Decker; Hugues Sicotte; Melike Pekmezci; Terri Rice; Matt L. Kosel; Ivan Smirnov; Gobinda Sarkar; Alissa Caron; Thomas M. Kollmeyer; Corinne Praska; Anisha R. Chada; Chandralekha Halder; Helen M. Hansen; Lucie McCoy; Paige M. Bracci; Roxanne Marshall; Shichun Zheng; Gerald F. Reis; Alexander R. Pico; Brian Patrick O’Neill; Jan C. Buckner; Caterina Giannini; Jason T. Huse; Arie Perry; Tarik Tihan; Mitchell S. Berger

BACKGROUND The prediction of clinical behavior, response to therapy, and outcome of infiltrative glioma is challenging. On the basis of previous studies of tumor biology, we defined five glioma molecular groups with the use of three alterations: mutations in the TERT promoter, mutations in IDH, and codeletion of chromosome arms 1p and 19q (1p/19q codeletion). We tested the hypothesis that within groups based on these features, tumors would have similar clinical variables, acquired somatic alterations, and germline variants. METHODS We scored tumors as negative or positive for each of these markers in 1087 gliomas and compared acquired alterations and patient characteristics among the five primary molecular groups. Using 11,590 controls, we assessed associations between these groups and known glioma germline variants. RESULTS Among 615 grade II or III gliomas, 29% had all three alterations (i.e., were triple-positive), 5% had TERT and IDH mutations, 45% had only IDH mutations, 7% were triple-negative, and 10% had only TERT mutations; 5% had other combinations. Among 472 grade IV gliomas, less than 1% were triple-positive, 2% had TERT and IDH mutations, 7% had only IDH mutations, 17% were triple-negative, and 74% had only TERT mutations. The mean age at diagnosis was lowest (37 years) among patients who had gliomas with only IDH mutations and was highest (59 years) among patients who had gliomas with only TERT mutations. The molecular groups were independently associated with overall survival among patients with grade II or III gliomas but not among patients with grade IV gliomas. The molecular groups were associated with specific germline variants. CONCLUSIONS Gliomas were classified into five principal groups on the basis of three tumor markers. The groups had different ages at onset, overall survival, and associations with germline variants, which implies that they are characterized by distinct mechanisms of pathogenesis. (Funded by the National Institutes of Health and others.).


Nature Genetics | 2009

Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility.

Margaret Wrensch; Robert B. Jenkins; Jeffrey S. Chang; Ru Fang Yeh; Yuanyuan Xiao; Paul A. Decker; Karla V. Ballman; Mitchel S. Berger; Jan C. Buckner; Susan M. Chang; Caterina Giannini; Chandralekha Halder; Thomas M. Kollmeyer; Matthew L. Kosel; Daniel H. Lachance; Lucie McCoy; Brian Patrick O'Neill; Joe Patoka; Alexander R. Pico; Michael D. Prados; Charles P. Quesenberry; Terri Rice; Amanda L. Rynearson; Ivan Smirnov; Tarik Tihan; Joseph L. Wiemels; Ping Yang; John K. Wiencke

The causes of glioblastoma and other gliomas remain obscure. To discover new candidate genes influencing glioma susceptibility, we conducted a principal component–adjusted genome-wide association study (GWAS) of 275,895 autosomal variants among 692 adult high-grade glioma cases (622 from the San Francisco Adult Glioma Study (AGS) and 70 from the Cancer Genome Atlas (TCGA)) and 3,992 controls (602 from AGS and 3,390 from Illumina iControlDB (iControls)). For replication, we analyzed the 13 SNPs with P < 10−6 using independent data from 176 high-grade glioma cases and 174 controls from the Mayo Clinic. On 9p21, rs1412829 near CDKN2B had discovery P = 3.4 × 10−8, replication P = 0.0038 and combined P = 1.85 × 10−10. On 20q13.3, rs6010620 intronic to RTEL1 had discovery P = 1.5 × 10−7, replication P = 0.00035 and combined P = 3.40 × 10−9. For both SNPs, the direction of association was the same in discovery and replication phases.


Nature Genetics | 2012

A low-frequency variant at 8q24.21 is strongly associated with risk of oligodendroglial tumors and astrocytomas with IDH1 or IDH2 mutation

Robert B. Jenkins; Yuanyuan Xiao; Hugues Sicotte; Paul A. Decker; Thomas M. Kollmeyer; Helen M. Hansen; Matthew L. Kosel; Shichun Zheng; Kyle M. Walsh; Terri Rice; Paige M. Bracci; Lucie McCoy; Ivan Smirnov; Joseph S. Patoka; George Hsuang; Joseph L. Wiemels; Tarik Tihan; Alexander R. Pico; Michael D. Prados; Susan M. Chang; Mitchel S. Berger; Alissa Caron; Stephanie R. Fink; Chandralekha Halder; Amanda L. Rynearson; Brooke L. Fridley; Jan C. Buckner; Brian Patrick O'Neill; Caterina Giannini; Daniel H. Lachance

Variants at 8q24.21 have been shown to be associated with glioma development. By means of tag SNP genotyping and imputation, pooled next-generation sequencing using long-range PCR and subsequent validation SNP genotyping, we identified seven low-frequency SNPs at 8q24.21 that were strongly associated with glioma risk (P = 1 × 10−25 to 1 × 10−14). The most strongly associated SNP, rs55705857, remained highly significant after individual adjustment for the other top six SNPs and two previously published SNPs. After stratifying by histological and tumor genetic subtype, the most significant associations of rs55705857 were with oligodendroglial tumors and gliomas with mutant IDH1 or IDH2 (odds ratio (OR) = 5.1, P = 1.1 × 10−31 and OR = 4.8, P = 6.6 × 10−22, respectively). Strong associations were observed for astrocytomas with mutated IDH1 or IDH2 (grades 2–4) (OR = 5.16–6.66, P = 4.7 × 10−12 to 2.2 × 10−8) but not for astrocytomas with wild-type IDH1 and IDH2 (smallest P = 0.26). The conserved sequence block that includes rs55705857 is consistently modeled as a microRNA.


International Journal of Cancer | 2009

IgE, allergy, and risk of glioma: update from the San Francisco Bay Area Adult Glioma Study in the temozolomide era.

Joseph L. Wiemels; David B. Wilson; Chirag G. Patil; Joseph S. Patoka; Lucie McCoy; Terri Rice; Judith A. Schwartzbaum; Amy B. Heimberger; John H. Sampson; Susan M. Chang; Michael D. Prados; John K. Wiencke; Margaret Wrensch

The consistently observed inverse relationship of allergic conditions with glioma risk and our previous demonstration that immunoglobulin E (IgE) levels also were lower in glioma patients than controls suggest that atopic allergy may be related to a mechanism that inhibits or prevents glioma. We sought to extend these results with a new and larger series of patients (n = 535 with questionnaire data; 393 with IgE measures) and controls (n = 532 with questionnaire data; 470 with IgE measures). As expected, glioma cases were less likely than controls to report history of allergies [among self‐reported cases, Odds ratios (OR) = 0.59, 95% confidence interval (CI): 0.41–0.85]. IgE levels also were lower in glioma cases versus controls (OR per unit log IgE = 0.89, 95% CI (0.82–0.98). However, this inverse relationship was only apparent among cases receiving temozolomide, a treatment which became part of the “standard of care” for glioblastoma patients during the study period. Among patients receiving temozolomide, IgE levels in cases whose blood samples were obtained within 30 days of diagnosis were slightly higher than controls, whereas IgE levels in cases whose blood sample was obtained >60 days after diagnosis were significantly lower than controls (OR = 0.80; 95% CI: 0.71–0.89). Thus, although our results robustly confirm the inverse association between allergy and glioma, the results for IgE are affected by temozolomide treatments which may have influenced IgE levels. These results have implications for the study of immunologic factors in glioma as well as for immunotherapy protocols for treating glioma.


Cancer Genetics and Cytogenetics | 2011

Distinct germ line polymorphisms underlie glioma morphologic heterogeneity

Robert B. Jenkins; Margaret Wrensch; Derek R. Johnson; Brooke L. Fridley; Paul A. Decker; Yuanyuan Xiao; Thomas M. Kollmeyer; Amanda L. Rynearson; Stephanie R. Fink; Terri Rice; Lucie McCoy; Chandralekha Halder; Matthew L. Kosel; Caterina Giannini; Tarik Tihan; Brian Patrick O’Neill; Daniel H. Lachance; Ping Yang; Joseph L. Wiemels; John K. Wiencke

Two recent genome-wide association studies reported that single nucleotide polymorphisms (SNPs) in (or near) TERT (5p15), CCDC26 (8q24), CDKN2A/B (9p21), PHLDB1 (11q23), and RTEL1 (20q13) are associated with infiltrating glioma. From these reports, it was not clear whether the single nucleotide polymorphism associations predispose to glioma in general or whether they are specific to certain glioma grades or morphologic subtypes. To identify hypothesized associations between susceptibility loci and tumor subtype, we genotyped two case-control groups composed of the spectrum of infiltrating glioma subtypes and stratified the analyses by type. We report that specific germ line polymorphisms are associated with different glioma subtypes. CCDC26 (8q24) region polymorphisms are strongly associated with oligodendroglial tumor risk (rs4295627, odds ratio [OR] = 2.05, P = 8.3 × 10(-11)) but not glioblastoma risk. The opposite is true of RTEL (20q13) region polymorphisms, which are significantly associated with glioblastoma (rs2297440, OR = 0.56, P = 4.6 × 10(-10)) but not oligodendroglial tumor. The SNPs in or near CCDC26 (8q24) are associated with oligodendroglial tumors regardless of combined 1p and 19q deletion status; however, the association is greatest for those with combined deletion (rs4295627, OR = 2.77, P = 2.6 × 10(-9)). These observations generate hypotheses concerning the possible mechanisms by which specific SNPs (or alterations in linkage disequilibrium with such SNPs) are associated with glioma development.


Nature Genetics | 2017

Genome-wide association study of glioma subtypes identifies specific differences in genetic susceptibility to glioblastoma and non-glioblastoma tumors.

Beatrice Melin; Jill S. Barnholtz-Sloan; Margaret Wrensch; Christoffer Johansen; Dora Il'yasova; Ben Kinnersley; Quinn T. Ostrom; Karim Labreche; Yanwen Chen; Georgina Armstrong; Yanhong Liu; Jeanette E. Eckel-Passow; Paul A. Decker; Marianne Labussière; Ahmed Idbaih; Khê Hoang-Xuan; Anna-Luisa Di Stefano; Karima Mokhtari; Jean-Yves Delattre; Peter Broderick; Pilar Galan; Konstantinos Gousias; Johannes Schramm; Minouk J. Schoemaker; Sarah Fleming; Stefan Herms; Stefanie Heilmann; Markus M. Nöthen; Heinz-Erich Wichmann; Stefan Schreiber

Genome-wide association studies (GWAS) have transformed our understanding of glioma susceptibility, but individual studies have had limited power to identify risk loci. We performed a meta-analysis of existing GWAS and two new GWAS, which totaled 12,496 cases and 18,190 controls. We identified five new loci for glioblastoma (GBM) at 1p31.3 (rs12752552; P = 2.04 × 10−9, odds ratio (OR) = 1.22), 11q14.1 (rs11233250; P = 9.95 × 10−10, OR = 1.24), 16p13.3 (rs2562152; P = 1.93 × 10−8, OR = 1.21), 16q12.1 (rs10852606; P = 1.29 × 10−11, OR = 1.18) and 22q13.1 (rs2235573; P = 1.76 × 10−10, OR = 1.15), as well as eight loci for non-GBM tumors at 1q32.1 (rs4252707; P = 3.34 × 10−9, OR = 1.19), 1q44 (rs12076373; P = 2.63 × 10−10, OR = 1.23), 2q33.3 (rs7572263; P = 2.18 × 10−10, OR = 1.20), 3p14.1 (rs11706832; P = 7.66 × 10−9, OR = 1.15), 10q24.33 (rs11598018; P = 3.39 × 10−8, OR = 1.14), 11q21 (rs7107785; P = 3.87 × 10−10, OR = 1.16), 14q12 (rs10131032; P = 5.07 × 10−11, OR = 1.33) and 16p13.3 (rs3751667; P = 2.61 × 10−9, OR = 1.18). These data substantiate that genetic susceptibility to GBM and non-GBM tumors are highly distinct, which likely reflects different etiology.


Genetic Epidemiology | 2013

Analysis of 60 Reported Glioma Risk SNPs Replicates Published GWAS Findings but Fails to Replicate Associations From Published Candidate‐Gene Studies

Kyle M. Walsh; Erik Anderson; Helen M. Hansen; Paul A. Decker; Matt L. Kosel; Thomas M. Kollmeyer; Terri Rice; Shichun Zheng; Yuanyuan Xiao; Jeffrey S. Chang; Lucie McCoy; Paige M. Bracci; Joseph L. Wiemels; Alexander R. Pico; Ivan Smirnov; Daniel H. Lachance; Hugues Sicotte; Jeanette E. Eckel-Passow; John K. Wiencke; Robert B. Jenkins; Margaret Wrensch

Genomewide association studies (GWAS) and candidate‐gene studies have implicated single‐nucleotide polymorphisms (SNPs) in at least 45 different genes as putative glioma risk factors. Attempts to validate these associations have yielded variable results and few genetic risk factors have been consistently replicated. We conducted a case‐control study of Caucasian glioma cases and controls from the University of California San Francisco (810 cases, 512 controls) and the Mayo Clinic (852 cases, 789 controls) in an attempt to replicate previously reported genetic risk factors for glioma. Sixty SNPs selected from the literature (eight from GWAS and 52 from candidate‐gene studies) were successfully genotyped on an Illumina custom genotyping panel. Eight SNPs in/near seven different genes (TERT, EGFR, CCDC26, CDKN2A, PHLDB1, RTEL1, TP53) were significantly associated with glioma risk in the combined dataset (P < 0.05), with all associations in the same direction as in previous reports. Several SNP associations showed considerable differences across histologic subtype. All eight successfully replicated associations were first identified by GWAS, although none of the putative risk SNPs from candidate‐gene studies was associated in the full case‐control sample (all P values > 0.05). Although several confirmed associations are located near genes long known to be involved in gliomagenesis (e.g., EGFR, CDKN2A, TP53), these associations were first discovered by the GWAS approach and are in noncoding regions. These results highlight that the deficiencies of the candidate‐gene approach lay in selecting both appropriate genes and relevant SNPs within these genes.


Carcinogenesis | 2010

Inherited variation in immune genes and pathways and glioblastoma risk

Judith A. Schwartzbaum; Yuanyuan Xiao; Yanhong Liu; Spyros Tsavachidis; Mitchel S. Berger; Melissa L. Bondy; Jeffrey S. Chang; Susan M. Chang; Paul A. Decker; Bo Ding; Sarah J. Hepworth; Richard S. Houlston; Fay J. Hosking; Robert B. Jenkins; Matthew L. Kosel; Lucie McCoy; Patricia A. McKinney; Kenneth Muir; Joe Patoka; Michael D. Prados; Terri Rice; Lindsay B. Robertson; Minouk J. Schoemaker; Sanjay Shete; Anthony J. Swerdlow; Joseph L. Wiemels; John K. Wiencke; Ping Yang; Margaret Wrensch

To determine whether inherited variations in immune function single-nucleotide polymorphisms (SNPs), genes or pathways affect glioblastoma risk, we analyzed data from recent genome-wide association studies in conjunction with predefined immune function genes and pathways. Gene and pathway analyses were conducted on two independent data sets using 6629 SNPs in 911 genes on 17 immune pathways from 525 glioblastoma cases and 602 controls from the University of California, San Francisco (UCSF) and a subset of 6029 SNPs in 893 genes from 531 cases and 1782 controls from MD Anderson (MDA). To further assess consistency of SNP-level associations, we also compared data from the UK (266 cases and 2482 controls) and the Mayo Clinic (114 cases and 111 controls). Although three correlated epidermal growth factor receptor (EGFR) SNPs were consistently associated with glioblastoma in all four data sets (Mantel–Haenzel P values = 1 × 10−5 to 4 × 10−3), independent replication is required as genome-wide significance was not attained. In gene-level analyses, eight immune function genes were significantly (minP < 0.05) associated with glioblastoma; the IL-2RA (CD25) cytokine gene had the smallest minP values in both UCSF (minP = 0.01) and MDA (minP = 0.001) data sets. The IL-2RA receptor is found on the surface of regulatory T cells potentially contributing to immunosuppression characteristic of the glioblastoma microenvironment. In pathway correlation analyses, cytokine signaling and adhesion–extravasation–migration pathways showed similar associations with glioblastoma risk in both MDA and UCSF data sets. Our findings represent the first systematic description of immune genes and pathways that characterize glioblastoma risk.


Neuro-oncology | 2013

Inherited variant on chromosome 11q23 increases susceptibility to IDH-mutated but not IDH-normal gliomas regardless of grade or histology

Terri Rice; Shichun Zheng; Paul A. Decker; Kyle M. Walsh; Paige M. Bracci; Yuanyuan Xiao; Lucie McCoy; Ivan Smirnov; Joseph S. Patoka; Helen M. Hansen; George Hsuang; Joseph L. Wiemels; Tarik Tihan; Alexander R. Pico; Michael D. Prados; Susan M. Chang; Mitchel S. Berger; Alissa Caron; Stephanie R. Fink; Thomas M. Kollmeyer; Amanda L. Rynearson; Jesse S. Voss; Matthew L. Kosel; Brooke L. Fridley; Daniel H. Lachance; Jeanette E. Eckel-Passow; Hugues Sicotte; Brian Patrick O'Neill; Caterina Giannini; John K. Wiencke

INTRODUCTION Recent discoveries of inherited glioma risk loci and acquired IDH mutations are providing new insights into glioma etiology. IDH mutations are common in lower grade gliomas and secondary glioblastomas and uncommon in primary glioblastomas. Because the inherited variant in 11q23 has been associated with risk of lower grade glioma and not with glioblastomas, we hypothesized that this variant increases susceptibility to IDH-mutated gliomas, but not to IDH-wild-type gliomas. METHODS We tested this hypothesis in patients with glioma and controls from the San Francisco Adult Glioma Study, the Mayo Clinic, and Illumina controls (1102 total patients, 5299 total controls). Case-control additive associations of 11q23 risk alleles (rs498872, T allele) were calculated using logistic regression, stratified by tumor IDH status (mutated or wild-type) and by histology and grade. We also adjusted for the recently discovered 8q24 glioma risk locus rs55705857 G allele. RESULTS The 11q23 glioma risk locus was associated with increased risk of IDH-mutated gliomas of all histologies and grades (odds ratio [OR] = 1.50; 95% confidence interval [CI] = 1.29-1.74; P = 1.3X10(-7)) but not with IDH-wild-type gliomas of any histology or grade (OR = 0.91; 95% CI = 0.81-1.03; P = 0.14). The associations were independent of the rs55705857 G allele. CONCLUSION A variant at the 11q23 locus increases risk for IDH-mutated but not IDH-wild-type gliomas, regardless of grade or histology.


International Journal of Cancer | 2012

HMG CoA Reductase Inhibitors, NSAIDs and Risk of Glioma

Jennifer S. Ferris; Lucie McCoy; Alfred I. Neugut; Margaret Wrensch; Rose Lai

3‐Hydroxy‐3‐methylglutaryl coenzyme A (HMG CoA) reductase inhibitors (statins) have shown inverse associations with cancer risks, but the results have been inconsistent. As there are no previous published data in brain tumors, we conducted a case–control study to investigate statin therapy and risk of glioma. We further evaluated the use of nonsteriodal anti‐inflammatory drugs (NSAIDs) and risk of these tumors. We recruited newly diagnosed glioma cases and frequency matched controls at Columbia University and the University of California San Francisco. Standardized questions on statins and NSAIDs were used at both institutions. Intakes of these drugs were defined as >6 months of at least twice weekly use versus less than this amount or never use. From July 2007 to January 2010, we recruited a total of 517 cases and 400 controls. Simvastatin and lovastatin showed significant inverse associations with glioma (odds ratio [OR] = 0.49, 95% confidence interval [CI] 0.30, 0.81 and OR = 0.47, 95% CI 0.24, 0.93, respectively). For NSAIDs, aspirin use was also inversely related to glioma risk (OR = 0.68, 95% CI 0.49, 0.96). Both statins and NSAIDs showed significant inverse trends between the duration of drug use and glioma risk (trend tests p = 0.03 and p = 0.02, respectively), and drug intake for >120 months demonstrated the most significant associations for both types of medication. The inverse association between statin therapy and risk of glioma supports the roles of Ras/Rho GTPases or inflammatory cytokines in gliomagenesis, and a similar relationship between NSAIDs and glioma highlights the importance of cyclo‐oxygenase 2 in glioma pathogenesis.

Collaboration


Dive into the Lucie McCoy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Terri Rice

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kyle M. Walsh

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge