Network


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

Hotspot


Dive into the research topics where Emily Hallberg is active.

Publication


Featured researches published by Emily Hallberg.


Journal of Clinical Oncology | 2015

Inherited Mutations in 17 Breast Cancer Susceptibility Genes Among a Large Triple-Negative Breast Cancer Cohort Unselected for Family History of Breast Cancer

Fergus J. Couch; Steven N. Hart; Priyanka Sharma; Amanda Ewart Toland; Xianshu Wang; Penelope Miron; Janet E. Olson; Andrew K. Godwin; V. Shane Pankratz; Curtis Olswold; Seth W. Slettedahl; Emily Hallberg; Lucia Guidugli; Jaime Davila; Matthias W. Beckmann; Wolfgang Janni; Brigitte Rack; Arif B. Ekici; Dennis J. Slamon; Irene Konstantopoulou; Florentia Fostira; Athanassios Vratimos; George Fountzilas; Liisa M. Pelttari; William Tapper; Lorraine Durcan; Simon S. Cross; Robert Pilarski; Charles L. Shapiro; Jennifer R. Klemp

PURPOSE Recent advances in DNA sequencing have led to the development of breast cancer susceptibility gene panels for germline genetic testing of patients. We assessed the frequency of mutations in 17 predisposition genes, including BRCA1 and BRCA2, in a large cohort of patients with triple-negative breast cancer (TNBC) unselected for family history of breast or ovarian cancer to determine the utility of germline genetic testing for those with TNBC. PATIENTS AND METHODS Patients with TNBC (N = 1,824) unselected for family history of breast or ovarian cancer were recruited through 12 studies, and germline DNA was sequenced to identify mutations. RESULTS Deleterious mutations were identified in 14.6% of all patients. Of these, 11.2% had mutations in the BRCA1 (8.5%) and BRCA2 (2.7%) genes. Deleterious mutations in 15 other predisposition genes were detected in 3.7% of patients, with the majority observed in genes involved in homologous recombination, including PALB2 (1.2%) and BARD1, RAD51D, RAD51C, and BRIP1 (0.3% to 0.5%). Patients with TNBC with mutations were diagnosed at an earlier age (P < .001) and had higher-grade tumors (P = .01) than those without mutations. CONCLUSION Deleterious mutations in predisposition genes are present at high frequency in patients with TNBC unselected for family history of cancer. Mutation prevalence estimates suggest that patients with TNBC, regardless of age at diagnosis or family history of cancer, should be considered for germline genetic testing of BRCA1 and BRCA2. Although mutations in other predisposition genes are observed among patients with TNBC, better cancer risk estimates are needed before these mutations are used for clinical risk assessment in relatives.


JAMA Oncology | 2017

Associations Between Cancer Predisposition Testing Panel Genes and Breast Cancer

Fergus J. Couch; Hermela Shimelis; Chunling Hu; Steven N. Hart; Eric C. Polley; Jie Na; Emily Hallberg; Raymond Moore; Abigail Thomas; Jenna Lilyquist; Bingjian Feng; Rachel McFarland; Tina Pesaran; Robert Huether; Holly LaDuca; Elizabeth C. Chao; David E. Goldgar; Jill S. Dolinsky

Importance Germline pathogenic variants in BRCA1 and BRCA2 predispose to an increased lifetime risk of breast cancer. However, the relevance of germline variants in other genes from multigene hereditary cancer testing panels is not well defined. Objective To determine the risks of breast cancer associated with germline variants in cancer predisposition genes. Design, Setting, and Participants A study population of 65 057 patients with breast cancer receiving germline genetic testing of cancer predisposition genes with hereditary cancer multigene panels. Associations between pathogenic variants in non-BRCA1 and non-BRCA2 predisposition genes and breast cancer risk were estimated in a case-control analysis of patients with breast cancer and Exome Aggregation Consortium reference controls. The women underwent testing between March 15, 2012, and June 30, 2016. Main Outcomes and Measures Breast cancer risk conferred by pathogenic variants in non-BRCA1 and non-BRCA2 predisposition genes. Results The mean (SD) age at diagnosis for the 65 057 women included in the analysis was 48.5 (11.1) years. The frequency of pathogenic variants in 21 panel genes identified in 41 611 consecutively tested white women with breast cancer was estimated at 10.2%. After exclusion of BRCA1, BRCA2, and syndromic breast cancer genes (CDH1, PTEN, and TP53), observed pathogenic variants in 5 of 16 genes were associated with high or moderately increased risks of breast cancer: ATM (OR, 2.78; 95% CI, 2.22-3.62), BARD1 (OR, 2.16; 95% CI, 1.31-3.63), CHEK2 (OR, 1.48; 95% CI, 1.31-1.67), PALB2 (OR, 7.46; 95% CI, 5.12-11.19), and RAD51D (OR, 3.07; 95% CI, 1.21-7.88). Conversely, variants in the BRIP1 and RAD51C ovarian cancer risk genes; the MRE11A, RAD50, and NBN MRN complex genes; the MLH1 and PMS2 mismatch repair genes; and NF1 were not associated with increased risks of breast cancer. Conclusions and Relevance This study establishes several panel genes as high- and moderate-risk breast cancer genes and provides estimates of breast cancer risk associated with pathogenic variants in these genes among individuals qualifying for clinical genetic testing.


Cancer | 2014

Incidence of chronic lymphocytic leukemia and high-count monoclonal B-cell lymphocytosis using the 2008 guidelines.

Timothy G. Call; Aaron D. Norman; Curtis A. Hanson; Sara J. Achenbach; Neil E. Kay; Clive S. Zent; Wei Ding; James R. Cerhan; Kari G. Rabe; Celine M. Vachon; Emily Hallberg; Tait D. Shanafelt; Susan L. Slager

The 1996 National Cancer Institute Working Group (NCI‐WG 96) guidelines classified disease in individuals who had a B‐cell clone with chronic lymphocytic leukemia (CLL) immunophenotype as CLL if their absolute lymphocyte count was ≥5 × 109/L. The 2008 International Workshop on CLL guidelines (IWCLL 2008) classified disease as CLL if the absolute B‐cell count was ≥5 × 109/L or as monoclonal B‐cell lymphocytosis (MBL) if the absolute B‐cell count was <5 × 109/L. The objective of the current study of Olmsted County, Minnesota, was to assess the effects of these changes on incidence rates and presentation from 2000 to 2010.


International Journal of Cancer | 2015

Investigation of gene‐environment interactions between 47 newly identified breast cancer susceptibility loci and environmental risk factors

Anja Rudolph; Roger L. Milne; Thérèse Truong; Julia A. Knight; Petra Seibold; Dieter Flesch-Janys; Sabine Behrens; Ursula Eilber; Manjeet K. Bolla; Qin Wang; Joe Dennis; Alison M. Dunning; Mitul Shah; Hannah Munday; Hatef Darabi; Mikael Eriksson; Judith S. Brand; Janet E. Olson; Celine M. Vachon; Emily Hallberg; J. Esteban Castelao; Angel Carracedo; M.D. Torres; Jingmei Li; Keith Humphreys; Emilie Cordina-Duverger; Florence Menegaux; Henrik Flyger; Børge G. Nordestgaard; Sune F. Nielsen

A large genotyping project within the Breast Cancer Association Consortium (BCAC) recently identified 41 associations between single nucleotide polymorphisms (SNPs) and overall breast cancer (BC) risk. We investigated whether the effects of these 41 SNPs, as well as six SNPs associated with estrogen receptor (ER) negative BC risk are modified by 13 environmental risk factors for BC. Data from 22 studies participating in BCAC were pooled, comprising up to 26,633 cases and 30,119 controls. Interactions between SNPs and environmental factors were evaluated using an empirical Bayes‐type shrinkage estimator. Six SNPs showed interactions with associated p‐values (pint) <1.1 × 10−3. None of the observed interactions was significant after accounting for multiple testing. The Bayesian False Discovery Probability was used to rank the findings, which indicated three interactions as being noteworthy at 1% prior probability of interaction. SNP rs6828523 was associated with increased ER‐negative BC risk in women ≥170 cm (OR = 1.22, p = 0.017), but inversely associated with ER‐negative BC risk in women <160 cm (OR = 0.83, p = 0.039, pint = 1.9 × 10−4). The inverse association between rs4808801 and overall BC risk was stronger for women who had had four or more pregnancies (OR = 0.85, p = 2.0 × 10−4), and absent in women who had had just one (OR = 0.96, p = 0.19, pint = 6.1 × 10−4). SNP rs11242675 was inversely associated with overall BC risk in never/former smokers (OR = 0.93, p = 2.8 × 10−5), but no association was observed in current smokers (OR = 1.07, p = 0.14, pint = 3.4 × 10−4). In conclusion, recently identified BC susceptibility loci are not strongly modified by established risk factors and the observed potential interactions require confirmation in independent studies.


PLOS ONE | 2016

RAD51B in Familial Breast Cancer

Liisa M. Pelttari; Sofia Khan; Mikko Vuorela; Johanna I. Kiiski; Sara Vilske; Viivi Nevanlinna; Salla Ranta; Johanna Schleutker; Robert Winqvist; Anne Kallioniemi; Thilo Dörk; Natalia Bogdanova; Jonine D. Figueroa; Paul Pharoah; Marjanka K. Schmidt; Alison M. Dunning; Montserrat Garcia-Closas; Manjeet K. Bolla; Joe Dennis; Kyriaki Michailidou; Qin Wang; John L. Hopper; Melissa C. Southey; Efraim H. Rosenberg; Peter A. Fasching; Matthias W. Beckmann; Julian Peto; Isabel dos-Santos-Silva; Elinor Sawyer; Ian Tomlinson

Common variation on 14q24.1, close to RAD51B, has been associated with breast cancer: rs999737 and rs2588809 with the risk of female breast cancer and rs1314913 with the risk of male breast cancer. The aim of this study was to investigate the role of RAD51B variants in breast cancer predisposition, particularly in the context of familial breast cancer in Finland. We sequenced the coding region of RAD51B in 168 Finnish breast cancer patients from the Helsinki region for identification of possible recurrent founder mutations. In addition, we studied the known rs999737, rs2588809, and rs1314913 SNPs and RAD51B haplotypes in 44,791 breast cancer cases and 43,583 controls from 40 studies participating in the Breast Cancer Association Consortium (BCAC) that were genotyped on a custom chip (iCOGS). We identified one putatively pathogenic missense mutation c.541C>T among the Finnish cancer patients and subsequently genotyped the mutation in additional breast cancer cases (n = 5259) and population controls (n = 3586) from Finland and Belarus. No significant association with breast cancer risk was seen in the meta-analysis of the Finnish datasets or in the large BCAC dataset. The association with previously identified risk variants rs999737, rs2588809, and rs1314913 was replicated among all breast cancer cases and also among familial cases in the BCAC dataset. The most significant association was observed for the haplotype carrying the risk-alleles of all the three SNPs both among all cases (odds ratio (OR): 1.15, 95% confidence interval (CI): 1.11–1.19, P = 8.88 x 10−16) and among familial cases (OR: 1.24, 95% CI: 1.16–1.32, P = 6.19 x 10−11), compared to the haplotype with the respective protective alleles. Our results suggest that loss-of-function mutations in RAD51B are rare, but common variation at the RAD51B region is significantly associated with familial breast cancer risk.


Oncotarget | 2016

Association of breast cancer risk with genetic variants showing differential allelic expression: Identification of a novel breast cancer susceptibility locus at 4q21

Yosr Hamdi; Penny Soucy; Véronique Adoue; Kyriaki Michailidou; Sander Canisius; Audrey Lemaçon; Arnaud Droit; Irene L. Andrulis; Hoda Anton-Culver; Volker Arndt; Caroline Baynes; Carl Blomqvist; Natalia Bogdanova; Stig E. Bojesen; Manjeet K. Bolla; Bernardo Bonanni; Anne Lise Børresen-Dale; Judith S. Brand; Hiltrud Brauch; Hermann Brenner; Annegien Broeks; Barbara Burwinkel; Jenny Chang-Claude; Fergus J. Couch; Angela Cox; Simon S. Cross; Kamila Czene; Hatef Darabi; Joe Dennis; Peter Devilee

There are significant inter-individual differences in the levels of gene expression. Through modulation of gene expression, cis-acting variants represent an important source of phenotypic variation. Consequently, cis-regulatory SNPs associated with differential allelic expression are functional candidates for further investigation as disease-causing variants. To investigate whether common variants associated with differential allelic expression were involved in breast cancer susceptibility, a list of genes was established on the basis of their involvement in cancer related pathways and/or mechanisms. Thereafter, using data from a genome-wide map of allelic expression associated SNPs, 313 genetic variants were selected and their association with breast cancer risk was then evaluated in 46,451 breast cancer cases and 42,599 controls of European ancestry ascertained from 41 studies participating in the Breast Cancer Association Consortium. The associations were evaluated with overall breast cancer risk and with estrogen receptor negative and positive disease. One novel breast cancer susceptibility locus on 4q21 (rs11099601) was identified (OR = 1.05, P = 5.6x10-6). rs11099601 lies in a 135 kb linkage disequilibrium block containing several genes, including, HELQ, encoding the protein HEL308 a DNA dependant ATPase and DNA Helicase involved in DNA repair, MRPS18C encoding the Mitochondrial Ribosomal Protein S18C and FAM175A (ABRAXAS), encoding a BRCA1 BRCT domain-interacting protein involved in DNA damage response and double-strand break (DSB) repair. Expression QTL analysis in breast cancer tissue showed rs11099601 to be associated with HELQ (P = 8.28x10-14), MRPS18C (P = 1.94x10-27) and FAM175A (P = 3.83x10-3), explaining about 20%, 14% and 1%, respectively of the variance inexpression of these genes in breast carcinomas.


Breast Cancer Research | 2015

Assessment of variation in immunosuppressive pathway genes reveals TGFBR2 to be associated with prognosis of estrogen receptor-negative breast cancer after chemotherapy

Jieping Lei; Anja Rudolph; Kirsten B. Moysich; Sajjad Rafiq; Sabine Behrens; Ellen L. Goode; Paul Pharoah; Petra Seibold; Peter A. Fasching; Irene L. Andrulis; Vessela N. Kristensen; Fergus J. Couch; Ute Hamann; Maartje J. Hooning; Heli Nevanlinna; Ursula Eilber; Manjeet K. Bolla; Joe Dennis; Qin Wang; Annika Lindblom; Arto Mannermaa; Diether Lambrechts; Montserrat Garcia-Closas; Per Hall; Georgia Chenevix-Trench; Mitul Shah; Robert Luben; Lothar Haeberle; Arif B. Ekici; Matthias W. Beckmann

IntroductionTumor lymphocyte infiltration is associated with clinical response to chemotherapy in estrogen receptor (ER) negative breast cancer. To identify variants in immunosuppressive pathway genes associated with prognosis after adjuvant chemotherapy for ER-negative patients, we studied stage I-III invasive breast cancer patients of European ancestry, including 9,334 ER-positive (3,151 treated with chemotherapy) and 2,334 ER-negative patients (1,499 treated with chemotherapy).MethodsWe pooled data from sixteen studies from the Breast Cancer Association Consortium (BCAC), and employed two independent studies for replications. Overall 3,610 single nucleotide polymorphisms (SNPs) in 133 genes were genotyped as part of the Collaborative Oncological Gene-environment Study, in which phenotype and clinical data were collected and harmonized. Multivariable Cox proportional hazard regression was used to assess genetic associations with overall survival (OS) and breast cancer-specific survival (BCSS). Heterogeneity according to chemotherapy or ER status was evaluated with the log-likelihood ratio test.ResultsThree independent SNPs in TGFBR2 and IL12B were associated with OS (P <10−3) solely in ER-negative patients after chemotherapy (267 events). Poorer OS associated with TGFBR2 rs1367610 (G > C) (per allele hazard ratio (HR) 1.54 (95% confidence interval (CI) 1.22 to 1.95), P = 3.08 × 10−4) was not found in ER-negative patients without chemotherapy or ER-positive patients with chemotherapy (P for interaction <10−3). Two SNPs in IL12B (r2 = 0.20) showed different associations with ER-negative disease after chemotherapy: rs2546892 (G > A) with poorer OS (HR 1.50 (95% CI 1.21 to 1.86), P = 1.81 × 10−4), and rs2853694 (A > C) with improved OS (HR 0.73 (95% CI 0.61 to 0.87), P = 3.67 × 10−4). Similar associations were observed with BCSS. Association with TGFBR2 rs1367610 but not IL12B variants replicated using BCAC Asian samples and the independent Prospective Study of Outcomes in Sporadic versus Hereditary Breast Cancer Study and yielded a combined HR of 1.57 ((95% CI 1.28 to 1.94), P = 2.05 × 10−5) without study heterogeneity.ConclusionsTGFBR2 variants may have prognostic and predictive value in ER-negative breast cancer patients treated with adjuvant chemotherapy. Our findings provide further insights into the development of immunotherapeutic targets for ER-negative breast cancer.


Journal of Medical Genetics | 2018

The BRCA1 c. 5096G>A p.Arg1699Gln (R1699Q) intermediate risk variant: breast and ovarian cancer risk estimation and recommendations for clinical management from the ENIGMA consortium

Setareh Moghadasi; Huong Meeks; Maaike P.G. Vreeswijk; Linda A.M. Janssen; Åke Borg; Hans Ehrencrona; Ylva Paulsson-Karlsson; Barbara Wappenschmidt; Christoph Engel; Andrea Gehrig; Norbert Arnold; Thomas V O Hansen; Mads Thomassen; Uffe Birk Jensen; Torben A. Kruse; Bent Ejlertsen; Anne-Marie Gerdes; Inge Søkilde Pedersen; Sandrine M. Caputo; Fergus J. Couch; Emily Hallberg; Ans van den Ouweland; J. Margriet Collée; Erik Teugels; Muriel A. Adank; Rob B. van der Luijt; Arjen R. Mensenkamp; Jan C. Oosterwijk; Marinus J. Blok; Nicolas Janin

Background We previously showed that the BRCA1 variant c.5096G>A p.Arg1699Gln (R1699Q) was associated with an intermediate risk of breast cancer (BC) and ovarian cancer (OC). This study aimed to assess these cancer risks for R1699Q carriers in a larger cohort, including follow-up of previously studied families, to further define cancer risks and to propose adjusted clinical management of female BRCA1*R1699Q carriers. Methods Data were collected from 129 BRCA1*R1699Q families ascertained internationally by ENIGMA (Evidence-based Network for the Interpretation of Germline Mutant Alleles) consortium members. A modified segregation analysis was used to calculate BC and OC risks. Relative risks were calculated under both monogenic model and major gene plus polygenic model assumptions. Results In this cohort the cumulative risk of BC and OC by age 70 years was 20% and 6%, respectively. The relative risk for developing cancer was higher when using a model that included the effects of both the R1699Q variant and a residual polygenic component compared with monogenic model (for BC 3.67 vs 2.83, and for OC 6.41 vs 5.83). Conclusion Our results confirm that BRCA1*R1699Q confers an intermediate risk for BC and OC. Breast surveillance for female carriers based on mammogram annually from age 40 is advised. Bilateral salpingo-oophorectomy should be considered based on family history.


Oncotarget | 2017

TP53-based interaction analysis identifies cis-eQTL variants for TP53BP2, FBXO28, and FAM53A that associate with survival and treatment outcome in breast cancer

Rainer Fagerholm; Sofia Khan; Marjanka K. Schmidt; Montserrat Garcia-Closas; Päivi Heikkilä; Jani Saarela; Jonathan Beesley; Maral Jamshidi; Kristiina Aittomäki; Jianjun Liu; H. Raza Ali; Irene L. Andrulis; Matthias W. Beckmann; Sabine Behrens; Fiona Blows; Hermann Brenner; Jenny Chang-Claude; Fergus J. Couch; Kamila Czene; Peter A. Fasching; Jonine D. Figueroa; Giuseppe Floris; Gord Glendon; Qi Guo; Per Hall; Emily Hallberg; Ute Hamann; Bernd Holleczek; Maartje J. Hooning; John L. Hopper

TP53 overexpression is indicative of somatic TP53 mutations and associates with aggressive tumors and poor prognosis in breast cancer. We utilized a two-stage SNP association study to detect variants associated with breast cancer survival in a TP53-dependent manner. Initially, a genome-wide study (n = 575 cases) was conducted to discover candidate SNPs for genotyping and validation in the Breast Cancer Association Consortium (BCAC). The SNPs were then tested for interaction with tumor TP53 status (n = 4,610) and anthracycline treatment (n = 17,828). For SNPs interacting with anthracycline treatment, siRNA knockdown experiments were carried out to validate candidate genes. In the test for interaction between SNP genotype and TP53 status, we identified one locus, represented by rs10916264 (p(interaction) = 3.44 05E010-5; FDR-adjusted p = 0.0011) in estrogen receptor (ER) positive cases. The rs10916264 AA genotype associated with worse survival among cases with ER-positive, TP53-positive tumors (hazard ratio [HR] 2.36, 95% confidence interval [C.I] 1.45 - 3.82). This is a cis-eQTL locus for FBXO28 and TP53BP2; expression levels of these genes were associated with patient survival specifically in ER-positive, TP53-mutated tumors. Additionally, the SNP rs798755 was associated with survival in interaction with anthracycline treatment (p(interaction) = 9.57 05E010-5, FDR-adjusted p = 0.0130). RNAi-based depletion of a predicted regulatory target gene, FAM53A, indicated that this gene can modulate doxorubicin sensitivity in breast cancer cell lines. If confirmed in independent data sets, these results may be of clinical relevance in the development of prognostic and predictive marker panels for breast cancer.


British Journal of Cancer | 2016

Heterogeneity of luminal breast cancer characterised by immunohistochemical expression of basal markers

Hyuna Sung; Montserrat Garcia-Closas; Jenny Chang-Claude; Fiona Blows; H. Raza Ali; Jonine D. Figueroa; Heli Nevanlinna; Rainer Fagerholm; Päivi Heikkilä; Carl Blomqvist; Graham G. Giles; Roger L. Milne; Melissa C. Southey; Catriona McLean; Arto Mannermaa; Veli-Matti Kosma; Vesa Kataja; Reijo Sironen; Fergus J. Couch; Janet E. Olson; Emily Hallberg; Curtis Olswold; Angela Cox; Simon S. Cross; Peter Kraft; Rulla M. Tamimi; A. Heather Eliassen; Marjanka K. Schmidt; Manjeet K. Bolla; Qin Wang

Background:Luminal A breast cancer defined as hormone receptor positive and human epidermal growth factor receptor 2 (HER2) negative is known to be heterogeneous. Previous study showed that luminal A tumours with the expression of basal markers ((cytokeratin (CK) 5 or CK5/6) or epidermal growth factor receptor (EGFR)) were associated with poorer prognosis compared with those that stained negative for basal markers. Prompted by this study, we assessed whether tumour characteristics and risk factors differed by basal marker status within luminal A tumours.Methods:We pooled 5040 luminal A cases defined by immunohistochemistry (4490 basal-negative ((CK5 (or CK5/6))− and EGFR−) and 550 basal-positive ((CK5 (or CK5/6+)) or EGFR+)) from eight studies participating in the Breast Cancer Association Consortium. Case–case comparison was performed using unconditional logistic regression.Results:Tumour characteristics and risk factors did not vary significantly by the expression of basal markers, although results suggested that basal-positive luminal tumours tended to be smaller and node negative, and were more common in women with a positive family history and lower body mass index.Conclusions:Most established breast cancer risk factors were similar in basal-positive and basal-negative luminal A tumours. The non-significant but suggestive differences in tumour features and family history warrant further investigations.

Collaboration


Dive into the Emily Hallberg's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qin Wang

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar

Sabine Behrens

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Matthias W. Beckmann

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Peter A. Fasching

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge