14
Review Breast Cancer Theme Issue REVIEW Inherited Genetic Susceptibility to Breast Cancer The Beginning of the End or the End of the Beginning? Q16 Maya Ghoussaini,* Paul D.P. Pharoah,* y and Douglas F. Easton* From the Departments of Oncology* and Public Health and Primary Care, y Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, United Kingdom Accepted for publication July 22, 2013. Address correspondence to Maya Q15 Ghoussaini, Department of Oncology, Centre for Cancer Genetic Epidemiology, Univer- sity of Cambridge, Worts Causeway, Cambridge CB1 8RN, United Kingdom. E-mail: [email protected]. ac.uk. Genome-wide association studies have identied 72 loci associated with breast cancer susceptibility. Seventeen of these are known to predispose to other cancers. High-penetrance susceptibility loci for breast cancer usually result from coding alterations, principally in genes involved in DNA repair, whereas almost all of the associations identied through genome-wide association studies are found in noncoding regions of the genome and are likely to involve regulation of genes in multiple pathways. However, the genes underlying most associations are not yet known. In this review, we summarize the ndings from genome-wide association studies in breast cancer and describe the genes and mechanisms that are likely to be involved in the tumorigenesis process. We also discuss approaches to ne-scale mapping of susceptibility regions used to identify the likely causal variant(s) underlying the associa- tions, a major challenge in genetic epidemiology. Finally, we discuss the potential impact of such ndings on personalized medicine and future avenues for screening, prediction, and prevention programs. (Am J Pathol 2013, -:1e14; http://dx.doi.org/10.1016/j.ajpath.2013.07.003) Breast Q1 cancer is the most common cancer among women in both developing and developed regions in the world, accounting for 23% of all malignancies in women world- wide. 1 In the United States, the lifetime risk of breast cancer is approximately one in eight. 2 It is also the most common cause of cancer-related deaths worldwide (nearly 400,000 a year). 1 The incidence of breast cancer is approximately twofold higher in rst-degree relatives of breast cancer patients. This risk increases with the number of affected relatives and is much greater for women with relatives affected at a young age. 3e6 The higher prevalence of breast cancer among monozygotic twins of patients compared with dizygotic twins or siblings suggests strongly that genetic, rather than envi- ronmental, factors account for most of the familial aggrega- tion. Molecular genetic studies during the past 20 years have demonstrated that the genetic basis of susceptibility to breast cancer is complex, and is the result of germ-line variation at many different loci. The susceptibility alleles at these loci have widely different frequencies, and confer different disease risks (Figure 1 ½F1 ½F1 ). Genetic variants for breast cancer can be broadly categorized into high-, moderate-, and low-penetrance alleles. The high-penetrance alleles typically confer lifetime risks of breast cancer of >50%, sufcient to warrant intervention to reduce risk (eg, prophylactic surgery or magnetic resonance imaging screening), even in the absence of any other factors. Moderate-penetrance alleles confer risks of greater than twofold, corresponding to lifetime risks of 20% in Western populations, whereas low-penetrance alleles confer smaller increases in risk (ie, 10% to 20% lifetime risk). Rare High-Penetrance Alleles Family-based linkage analysis and positional cloning were used to identify high-penetrance alleles in BRCA1 and BRCA2, two tumor-suppressor genes involved in DNA repair. 7,8 Deleterious alleles of these genes are rare (cumu- lative frequency, approximately 1 in 800 for BRCA1 muta- tions and 1 in 500 for BRCA2 mutations 9 ) and confer a 10- to Supported by the European Communitys Seventh Framework Programme grant 223175 (HEALTH-F2-2009-223175) and Cancer Research UK Q14 . Copyright ª 2013 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2013.07.003 ajp.amjpathol.org The American Journal of Pathology, Vol. -, No. -, - 2013 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 REV 5.2.0 DTD ĸ AJPA1459_proof ĸ 22 August 2013 ĸ 11:15 pm ĸ EO: AJP13_0146

Inherited Genetic Susceptibility to Breast Cancer

Embed Size (px)

Citation preview

Page 1: Inherited Genetic Susceptibility to Breast Cancer

Q16

Q15

Q1

½F1�½F1�

The American Journal of Pathology, Vol. -, No. -, - 2013

1234567891011121314151617181920212223242526272829303132333435363738394041424344454647484950515253545556575859606162

636465666768

ajp.amjpathol.org

Review Breast Cancer Theme Issue 69707172737475767778798081

REVIEWInherited Genetic Susceptibility to Breast Cancer

The Beginning of the End or the End of the Beginning?Maya Ghoussaini,* Paul D.P. Pharoah,*y and Douglas F. Easton*

82838485

From the Departments of Oncology* and Public Health and Primary Care,y Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge,United Kingdom

8687

Accepted for publication

C

P

h

888990919293949596

July 22, 2013.

Address correspondence toMaya Ghoussaini, Departmentof Oncology, Centre for CancerGenetic Epidemiology, Univer-sity of Cambridge, WortsCauseway, Cambridge CB18RN, United Kingdom.E-mail: [email protected].

opyright ª 2013 American Society for Inve

ublished by Elsevier Inc. All rights reserved

ttp://dx.doi.org/10.1016/j.ajpath.2013.07.003

979899100

R

Genome-wide association studies have identified 72 loci associated with breast cancer susceptibility.Seventeen of these are known to predispose to other cancers. High-penetrance susceptibility loci forbreast cancer usually result from coding alterations, principally in genes involved in DNA repair,whereas almost all of the associations identified through genome-wide association studies are found innoncoding regions of the genome and are likely to involve regulation of genes in multiple pathways.However, the genes underlying most associations are not yet known. In this review, we summarize thefindings from genome-wide association studies in breast cancer and describe the genes and mechanismsthat are likely to be involved in the tumorigenesis process. We also discuss approaches to fine-scalemapping of susceptibility regions used to identify the likely causal variant(s) underlying the associa-tions, a major challenge in genetic epidemiology. Finally, we discuss the potential impact of suchfindings on personalized medicine and future avenues for screening, prediction, and preventionprograms. (Am J Pathol 2013, -: 1e14; http://dx.doi.org/10.1016/j.ajpath.2013.07.003)

101

Supported by the European Community’s Seventh Framework Programmegrant 223175 (HEALTH-F2-2009-223175) and Cancer Research UK Q14.

102103104105106107108109110111112113114115116117118119120121122123

Breast cancer is the most common cancer among women inboth developing and developed regions in the world,accounting for 23% of all malignancies in women world-wide.1 In the United States, the lifetime risk of breast canceris approximately one in eight.2 It is also the most commoncause of cancer-related deaths worldwide (nearly 400,000a year).1

The incidence of breast cancer is approximately twofoldhigher in first-degree relatives of breast cancer patients. Thisrisk increases with the number of affected relatives and ismuch greater for women with relatives affected at a youngage.3e6 The higher prevalence of breast cancer amongmonozygotic twins of patients compared with dizygotic twinsor siblings suggests strongly that genetic, rather than envi-ronmental, factors account for most of the familial aggrega-tion. Molecular genetic studies during the past 20 years havedemonstrated that the genetic basis of susceptibility to breastcancer is complex, and is the result of germ-line variation atmany different loci. The susceptibility alleles at these loci havewidely different frequencies, and confer different disease risks(Figure 1). Genetic variants for breast cancer can be broadly

stigative Pathology.

.

EV 5.2.0 DTD � AJPA1459_proof �

categorized into high-, moderate-, and low-penetrance alleles.The high-penetrance alleles typically confer lifetime risks ofbreast cancer of >50%, sufficient to warrant intervention toreduce risk (eg, prophylactic surgery or magnetic resonanceimaging screening), even in the absence of any other factors.Moderate-penetrance alleles confer risks of greater thantwofold, corresponding to lifetime risks of �20% in Westernpopulations, whereas low-penetrance alleles confer smallerincreases in risk (ie, 10% to 20% lifetime risk).

Rare High-Penetrance Alleles

Family-based linkage analysis and positional cloning wereused to identify high-penetrance alleles in BRCA1 andBRCA2, two tumor-suppressor genes involved in DNArepair.7,8 Deleterious alleles of these genes are rare (cumu-lative frequency, approximately 1 in 800 for BRCA1 muta-tions and 1 in 500 for BRCA2mutations9) and confer a 10- to

124

22 August 2013 � 11:15 pm � EO: AJP13_0146

Page 2: Inherited Genetic Susceptibility to Breast Cancer

Q2

½T1�½T1�

3

4

Figure 1 Genetic loci identified for breast cancer by risk allelefrequency and risk conferred.

Ghoussaini et al

125126127128129130131132133134135136137138139140141142143144145146147148149150151152153154155156157158159160161162163164165166167168169170171172173174175176177178179180181182183184185186

187188189190191192193194195196197198199200201202203204205206207208209210211212213214215216217218219220221222223224225226227228229230231232233234235236237238239240241242243244245246247

30-fold increase in the risk of breast cancer in carrierscompared with noncarriers.10 These alleles also predispose toother cancer types, including ovarian and prostate cancers.11

Deleterious alleles in other genes conferring a similar riskto BRCA1 and BRCA2 may exist, but they are likely to berare, because approximately 84% of families with four ormore cases of breast cancer diagnosed at younger than 60years can be accounted for by mutations in BRCA1 orBRCA2.10 A few high-penetrance alleles have been identi-fied as part of inherited cancer symptoms, such as TP53mutations found in Li-Fraumeni cancer syndrome12 andSTK11/LKB1 mutations found in Peutz-Jegher syndrome.13

Rare Moderate-Penetrance Alleles

Moderate-penetrance variants have been found by rese-quencing candidate genes, such as those interacting withBRCA1 and BRCA2, or acting in the same DNA repairpathway. Protein truncating variants in ATM,14 CHEK2,15,16

PALB2,17 and BRIP118 genes have been shown to confera twofold to fourfold risk of breast cancer. Other moderate-penetrance genes have been identified through linkagestudies in families with rare syndromes involving breast andother malignancies. Deleterious alleles of PTEN (Cowdendisease)19 and CDH1 (hereditary diffuse gastric cancer)20

confer relative risks of two to eight. It is likely that othergenetic variations in this category exist, but their identifica-tion will require resequencing in large case-control studies.

The susceptibility alleles previously mentioned are rareand do not reach a frequency of >1% even when combined,suggesting a selective disadvantage, possibly as a result ofhomozygotes being nonviable or seriously disadvantaged.Altogether, they account for <25% of the familial risk ofbreast cancer.21

Common Low-Penetrance Variation

Susceptibility alleles that are common in the general pop-ulation (minor allele frequency of >5%) are associated witha modest increase in breast cancer risk (relative risk of <1.5;no alleles with a relative risk between 1.5 and 2 have beenidentified). Such alleles are generally identified by case-control, association studies. Early association studies usinga candidate gene approach focused on polymorphisms ingenes thought to be relevant to cancer biology throughmechanisms such as cell cycle regulation, apoptosis, andDNA repair. Many positive associations were reported, butfew have been convincingly replicated.22 The mostconvincing reported evidence is for a coding polymorphismin CASP8, D302H, the minor allele of which has beenassociated with a reduced risk of breast cancer.23

During the past 5 years, improvements in genotypingtechnology have made genome-wide association studies(GWASs) possible. In GWASs, a few 100,000 variants thatreport on the most common variation in the genome are

2REV 5.2.0 DTD � AJPA1459_proof

genotyped, allowing associations to be detected withoutprior knowledge of function or location. GWASs have beenextremely successful in identifying susceptibility loci formany common diseases and phenotypes. To date, 72 breastcancer susceptibility loci have been identified throughGWASs and large-scale replication studies.24e36 The asso-ciated single-nucleotide polymorphisms (SNPs) and thenearest genes to each of the loci are summarized in Table 1.These loci, however, explain approximately 14% of theinherited genetic component of breast cancer. It is likely thatthere are many more common alleles with weak effects.

Association of GWAS Loci by Tumor Subtypes

Breast cancer is heterogeneous, and different types of tumorscan vary etiologically, histologically, and clinically.37 Morethan 90% of breast tumors that develop in carriers of BRCA1mutations are estrogen receptor negative (ER�),37 whereasthe common breast cancer susceptibility alleles identified aremore strongly associated with ERþ disease (FGFR2,MAP3K1, 8q24, 5p12, 11q13, 12q24, and 21q21) and lessstrongly, if at all, with ER� disease. Some loci, such as 16q12(TOX3), 12p11 (PTHLH ), and 10q21 (ZNF365), are asso-ciated with both disease types.26,30,31,38e41 Seven loci withcommon susceptibility alleles for ER� breast cancer havebeen identified: 6q25 (near ESR1) was first identified ina GWAS conducted in Chinese women33; 19p13 was initiallyidentified as a modifier of the BRCA1 breast cancer,25 and5p12 was identified through a GWAS in ER� and triple-negative (ER�/progesterone receptor�/HER� Q) breast can-cer.28 Four loci (1q32, MDM4; 1q32, LGR6; 2p24, gene Q

desert; 16q12, FTO) were recently identified after combiningthree GWASs comprising 4193 ER-negative breast cancercases and 35,194 controls.34 Most GWAS loci also modifythe penetrance of breast cancer in BRCA2 carriers, to a similarrelative extent, but only the subset of SNPs associated withER� disease appear to modify the risk in BRCA1 carriers,consistent with the distinct pathological characteristics of thedisease in BRCA1 carriers.

ajp.amjpathol.org - The American Journal of Pathology

248

� 22 August 2013 � 11:15 pm � EO: AJP13_0146

Page 3: Inherited Genetic Susceptibility to Breast Cancer

½F2�½F2�

½F3�½F3�

Genetic Susceptibility to Breast Cancer

249250251252253254255256257258259260261262263264265266267268269270271272273274275276277278279280281282283284285286287288289290291292293294295296297298299300301302303304305306307308309310

311312313314315316317318319320321322323324325326327328329330331332333334335336337338339340341342343344345346347348349350351352353354355356357358359360361362363364365366367368369370371

With the exception of the FGFR2 locus, which alsoshows the strongest association with breast cancer, themechanisms and the genes behind these associations arelargely unknown. However, knowledge about gene functionwithin the regions suggests that a complex and diverse rangeof biological pathways are involved in the commonsusceptibility to breast carcinogenesis. Almost all of the lociare in regions that contain genes that could be involved inbreast cancer development (Figure 2), and it is notable thatseveral of these genes have been shown to be somaticallymutated in breast tumors (MAP3K1, TBX3, NOTCH2, MYC,CASP8, CCND1, and TET2).42 However, in almost allcases, there is no direct evidence that these genes arecausally related to the observed susceptibility.

From Association to Causation and Function

A Major Challenge for Molecular Genetics

There are two key steps to understanding the functional mech-anisms bywhich the risk-associated variant causes breast cancerat a susceptibility locus. The first is to identify the true causalvariant at the locus, and the second is to identify the molecularmechanism by which the risk allele could cause cancer.

Fine-Scale Mapping

Fine-scale mapping aims to identify the causal variant(s) ata susceptibility locus. The set of SNPs genotyped in a GWASare designed to tag common variation across the genome.Thus, most variants found to be associated with disease will becorrelated with multiple other variants in the surroundingregion [called linkage disequilibrium (LD) block or haplotypeblock], for which the boundaries are delimited by recombi-nation hot spots. Haplotype blocks vary substantially in size,the number of SNPs, and the LD structure of those SNPs.

Several steps are required to narrow down a locus ofinterest and pinpoint the causal variant(s). The first step is toobtain a complete list of all variants in the LD block ofinterest. The availability of the 1000 Genomes project(http://www.1000genomes.org, last accessed July 29, 2013)has accelerated this process, and resequencing of the regionis usually no longer needed to search for common variants.Ideally, all of the SNPs correlated with the associatedmarker are then genotyped in a large case-control set, andthe strength of association is compared. The variant with thestrongest statistical evidence for association is the strongestcandidate for being the causal variant. However, because ofthe high correlation among SNPs within a block, multipleSNPs may have equally strong evidence and the true causalvariant may not be definitively identified. Because thepattern of LD often differs among populations, case-controlstudies from multiple populations (notably populations ofEast Asian or African ancestries, in addition to Europeanancestry) can assist with the fine mapping, provided that thesame causal variant segregates in each population

The American Journal of Pathology - ajp.amjpathol.orgREV 5.2.0 DTD � AJPA1459_proof �

(Figure 3). However, complementary approaches are stilllikely to be required to identify a single causal variant.

Because the causative variant is expected to be morestrongly associated with breast cancer risk than other SNPs,finding the causal variant in each of the GWAS loci will alsoimprove the accuracy of disease risk prediction. Bio-informatic analysis SNPs may have a variety of functionaleffects, by either directly altering protein structure (missensevariants or splice site variants) or altering the transcriptionalregulation of nearby genes. In silico bioinformatics analysiscan help identify SNPs more or less likely to have a func-tional effect.43 For example, SNPs in genomic regions thatare highly conserved between species are more likely to befunctionally important, particularly those in noncodingregions. Genome data on many species, including 28vertebrate species, are publicly available to enable suchanalyses to be routine. The ENCyclopedia Of DNA Elements(National Human Genome Research Institute, NIH, http://www.genome.gov/10005107, last accessed July 29, 2013)project has recently provided functional annotation across thehuman genome [histone modification and regulatoryelements, DNA methylation, DNase-hypersensitive sites andfootprints, chromosome-interacting regions, transcriptionfactor (TF) binding sites, regulatory elements, and codingand noncoding RNAs], showing that 80% of the noncodinggenome could be functional.44 Putative causal SNPs can bemapped to see if they lie in specific functional elements, suchas enhancers, silencers, or TF binding sites.

When the LD block of interest contains several genes, itmay be difficult to predict what gene is being targeted by theassociated variant. Because most associations are thought tobe regulatory, the target gene may be identified by evalu-ating whether the SNP genotype is associated with geneexpression. For example, a gene expression database, GENeExpression VARiation, in which the expression of geneswas quantified in immortalized B lymphocytes extractedfrom the 270 HapMap individuals of European, Asian, andAfrican ancestries, has been made publicly available. Thisapproach may be limited by the size of the association withexpression levels, which may be modest. More important,expression may be tissue specific, in which case availabledata in, for example, blood will be of limited value.Emerging data on expression in tumors, through projectssuch as The Cancer Genome Atlas (National Cancer Insti-tute, http://www.cancergenome.nih.gov, last accessed July29, 2013) and the International Cancer Genome ConsortiumCancer Genome Projects (http://www.icgc.org, last accessedJuly 29, 2013), may be more valuable for analysis of genesexpressed in the breast, but ultimately studies in normalbreast tissue may be required.

Functional Experiments

Experiments to determine the functional effect of candidatecausal variants are beyond the scope of this review. Briefly,if an SNP falls in the noncoding region of the genome

3

372

22 August 2013 � 11:15 pm � EO: AJP13_0146

Page 4: Inherited Genetic Susceptibility to Breast Cancer

Table 1 List of the 72 Breast Cancer Susceptibility Loci Identified to Date.

Region SNP Nearest plausible genes Year Per-allele OR P value MAF

1p11 rs11249433 NOTCH2/FCGR1B 2009 1.16 (1.09e1.24) 7 � 10�10 0.391p13 rs11552449 TPN22/BCL2L15 2013 1.07 (1.04e1.09) 1.8 � 10�8 0.171p36 rs616488 PEX14 2013 0.94 (0.92e0.96) 2.0 � 10�10 0.331q32 rs4245739 MDM4 2013 1.14 (1.10e1.18) 2.1 � 10�12 0.261q32 rs6678914 LGR6 2013 1.10 (1.06e1.13) 1.4 � 10�8 0.592p24 rs12710696 desert 2013 1.10 (1.06e1.13) 0.362q14 rs4849887 None 2013 0.91 (0.88e0.94) 3.7 � 10�11 0.102q31 rs2016394 METAP1D 2013 0.95 (0.93e0.97) 1.2 � 10�8 0.482q31 rs1550623 CDCA7 2013 0.94 (0.92e0.97) 3.0 � 10�8 0.162q33 rs1045485 CASP8 2007 0.88 (0.84e0.92) 1.1 � 10�7 0.13

rs10931936 0.88 (0.82e0.94) 0.262q35 rs13387042 IGFBP2, IGFBP5, TPN2 2007 1.20 (1.14e1.26) 1 � 10�13 0.492q35 rs16857609 DIRC3 2013 1.08 (1.06e1.10) 1.1 � 10�15 0.263p24 rs4973768 SLC4A7/NEK10 2009 1.11 (1.08e1.13) 4.1 � 10�23 0.463p24 rs12493607 TGFBR2 2013 1.06 (1.03e1.08) 2.3 � 10�8 0.353p26 rs6762644 ITPR1/EGOT 2013 1.07 (1.04e1.09) 2.2 � 10�12 0.404q24 rs9790517 TET2 2013 1.05 (1.03e1.08) 4.2 � 10�8 0.234q34 rs6828523 ADAM29 2013 0.90 (0.87e0.92) 3.5 � 10�16 0.135p12 rs10941679 MRPS30/HCN1 2008 1.19 (1.13e1.26) 1 � 10�11 0.25

rs9790879 1.10 (1.03e1.17) 0.405p15* rs10069690 TERT/CLPTM1L 2011 1.18 (1.13e1.25) 1.0 � 10�10 0.305q11 rs889312 MAP3K1/MEIR3 2007 1.13 (1.10e1.16) 1 � 10�15 0.285q11 rs10472076 RAB3C 2013 1.05 (1.03e1.07) 2.9 � 10�8 0.385q11 rs1353747 PDE4D 0.92 (0.89e0.95) 2.5 � 10�8 0.105q33 rs1432679 EBF1 2013 1.07 (1.05e1.09) 2.0 � 10�14 0.436p23 rs204247 RANBP9 2013 1.05 (1.03e1.07) 8.3 � 10�9 0.436p25 rs11242675 FOXQ1 2013 0.94 (0.92e0.96) 7.1 � 10�9 0.396q14 rs17530068 None 2012 1.12 (1.08e1.16) 1.1 � 10-9 0.226q25 rs3757318 ESR1 2009 1.21 (1.13e1.31) 2 � 10�15 0.076q25 rs2046210 ESR1 2009 1.11 (1.07e1.16) 3.7 � 10�9 0.347q35 rs720475 ARHGEF5/NOBOX 2013 0.94 (0.92e0.96) 7.0 � 10�11 0.258p12 rs9693444 None 2013 1.07 (1.05e1.09) 9.2 � 10�14 0.328q21 rs6472903 None 2013 0.91 (0.89e0.93) 1.7 � 10�17 0.188q21 rs2943559 HNF4G 2013 1.13 (1.09e1.17) 5.7 � 10�15 0.078q24 rs13281615 MYC 2007 1.08 (1.05e1.11) 5 � 10�12 0.408q24 rs1562430 MYC 2010 1.17 (1.10e1.25) 5 � 10�12 0.408q24 rs11780156 MIR1208 2013 1.07 (1.04e1.10) 3.4 � 10�11 0.169p21 rs1011970 CDKN2A/B 2010 1.09 (1.04e1.14) 2.5 � 10�8 0.179q31 rs865686 KLF4/RAD23B 2011 0.89 (0.85� 0.92) 1.7 � 10�10 0.399q31 rs10759243 None 2013 1.06 (1.03e1.08) 1.2 � 10�8 0.3910p12 rs7072776 MLLT10/DNAJC1 2013 1.07 (1.05e1.09) 4.3 � 10�14 0.2910p12 rs11814448 DNAJC1 2013 1.26 (1.18e1.35) 9.3 � 10�16 0.0210p15 rs2380205 ANKRD16 2010 0.94 (0.91e0.98) 4.6 � 10�7 0.4310q21 rs10995190 ZNF365 2010 0.86 (0.82e0.91) 5.1 � 10�15 0.1510q22 rs704010 ZMIZ1 2010 1.07 (1.03e1.11) 3 � 10�8 0.3910q25 rs7904519 TCF7L2 2013 1.06 (1.04e1.08) 3.1 � 10�8 0.4610q26 rs2981582 FGFR2 2007 1.26 (1.23e1.30) 2 � 10�76 0.3810q26 rs2981579 FGFR2 2010 1.43 (1.35e1.53) 2 � 10�76 0.4210q26 rs11199914 None 2013 0.95 (0.93e0.97) 1.9 � 10�8 0.3211p15 rs3817198 LSP1/H19 2007 1.07 (1.04e1.11) 1 � 10�9 0.3011p15 rs909116 LSP1/H19 2007 1.17 (1.10e1.24) 1 � 10�9 0.3011q13* rs614367 CCND1/FGFs 2010 1.15 (1.10e1.20) 3.2 � 10�15 0.1511q13 rs3903072 OVOL1 2013 0.95 (0.93e0.96) 8.6 � 10�12 0.4711q24 rs11820646 None 2013 0.95 (0.93e0.97) 1.1 � 10�9 0.4112p11 rs10771399 PTHLH 2011 0.79 (0.71e0.87) 4.3 � 10�35 0.1012p13 rs12422552 None 2013 1.05 (1.03e1.07) 3.7 � 10�8 0.2612q22 rs17356907 NTN4 2013 0.91 (0.89e0.93) 1.8 � 10�22 0.30

(table continues)

Ghoussaini et al

4 ajp.amjpathol.org - The American Journal of Pathology

373374375376377378379380381382383384385386387388389390391392393394395396397398399400401402403404405406407408409410411412413414415416417418419420421422423424425426427428429430431432433434

435436437438439440441442443444445446447448449450451452453454455456457458459460461462463464465466467468469470471472473474475476477478479480481482483484485486487488489490491492493494495496

REV 5.2.0 DTD � AJPA1459_proof � 22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 5: Inherited Genetic Susceptibility to Breast Cancer

Table 1 (continued )

Region SNP Nearest plausible genes Year Per-allele OR P value MAF

12q24 rs1292011 TBX3/MAPKAP5 2011 0.92 (0.91e0.94) 5.9 � 10�19 0.4113q13 rs11571833 BRCA2 2013 1.26 (1.14e1.39) 4.9 � 10�8 0.0114q13 rs2236007 PAX9/SLC25A21 2013 0.93 (0.91e0.95) 1.7 � 10�13 0.2114q24 rs999737 RAD51B 2009 0.94 (0.88e0.99) 2 � 10�7 0.2414q24 rs8009944 RAD51B 2009 0.88 (0.82e0.95) 2 � 10�7 0.2414q24 rs2588809 RAD51L1 2013 1.08 (1.05e1.11) 1.4 � 10�10 0.1614q32 rs941764 CCDC88C 2013 1.06 (1.04e1.09) 3.7 � 10�10 0.3416q12 rs12443621 TOX3/LOC643714 2007 1.11 (1.08e1.14) 1 � 10�36 0.4616q12 rs3803662 TOX3/LOC643714 2010 1.20 (1.16e1.24) 1 � 10�36 0.2616q12 rs17817449 MIR1972-2-FTO 2013 0.93 (0.91e0.95) 6.4 � 10�14 0.4016q23 rs13329835 CDYL2 2013 1.08 (1.05e1.10) 2.1 � 10�16 0.2216q22 rs11075995 FTO 2013 1.07 (1.11e1.15) 4.0 � 10�8 0.2417q23 rs6504950 STXBP4/COX11 2008 0.95 (0.92e0.97) 1.4 � 10�8 0.2718q11 rs527616 None 2013 0.95 (0.93e0.97) 1.6 � 10�10 0.3818q11 rs1436904 CHST9 2013 0.96 (0.94e0.98) 3.2 � 10�8 0.4019p13 rs8170 MERIT40 2010 1.26 (1.17e1.35) 2.3 � 10�9 0.1819p13 rs2363956 MERIT40 2010 0.84 (0.80e0.89) 5.5 � 0�9 0.5019p13 rs4808801 SSBP4/ISYNA1/ELL 2013 0.93 (0.91e0.95) 4.6 � 10�15 0.3519q13 rs3760982 KCNN4/ZNF283 2013 1.06 (1.04e1.08) 2.1 � 10�10 0.4620q11 rs2284378 RALY 2012 1.16 (1.01e1.10) 1.1 � 10�8 0.3521q21 rs2823093 NRIP1 2011 0.94 (0.92e0.96) 1.1 � 10�10 0.2722q12 rs132390 EMID1/RHBDD3 2013 1.12 (1.07e1.18) 3.1 � 10�9 0.0422q13 rs6001930 MKL1 2013 1.12 (1.09e1.16) 8.8 � 10�19 0.11

MAF, minor allele frequency.*For each of the 5p15/TERT locus and 11q13/CCND1 locus, three independent SNPs are associated with breast cancer.

Genetic Susceptibility to Breast Cancer

497498499500501502503504505506507508509510511512513514515516517518519520521522523524525526527528529530531532533534535536537538539540541542543544545546547548549550551552553554555556557558

559560561562563564565566567568569570571572573574575576577578579580581582583584585586587588589590591592593594595596597598599600601602603604605606607608609610611612

(promoter, enhancer, or intron) and is predicted to affect thebinding of a TF, experiments such as electrophoreticmobility shift assays can be used to test for a differentialprotein binding pattern. Luciferase transfection assays canbe used to measure the activity of a promoter or theexpression of target genes. Chromatin immunoprecipitationcan be used to establish whether the site of a candidatecausal variant is occupied by a TF in vivo. This approachwas recently used to determine the SNPs that are likely tohave a functional effect in FGFR2.45 Chromosome confor-mation capture can be used to test long-range interactionsthat influence gene expression (eg, between a regulatoryelement containing the causal SNP and the promoter of thesuspected targeted gene). This technique was used for thefine mapping of the 11q13 locus, where the enhancer andsilencer in which the causal variants lie were found tointeract with the promoter and terminator of the CCND1gene.46

In summary, a combination of genetic epidemiologicalcharacteristics, in silico bioinformatics, and functionalexperiments may enable the causal variant (or variants) to beidentified with some certainty.

613614615616617618619

Fine Mapping in Breast Cancer

To date, only four of the 72 known breast cancer suscepti-bility loci have been thoroughly fine mapped in European,Asian, and African American populations.26,45e49 In

The American Journal of Pathology - ajp.amjpathol.orgREV 5.2.0 DTD � AJPA1459_proof �

addition, 19 of the breast cancer susceptibility loci havebeen fine mapped in African Americans.50

FGFR2 Locus

Two variants in intron 2 of FGFR2 on the 10q26 locus werefound to be associated with breast cancer in two differentstudies.26,29 Fine-scale mapping in Europeans, Asians, andAfrican Americans identified eight SNPs as the most likelycandidates for being the causal variant.26,47 The moststrongly associated SNP, rs2981578, lies in an open chro-matin conformation region, where DNA is highly conservedamong mammalian species and was found to mediatedifferential TF binding, leading to an increase in FGFR2expression levels.47 The same SNP was associated withincreased FGFR2 signaling activity in stromal fibroblasts.51

These data altogether suggest that rs2981578 is likely to befunctional. Another correlated SNP, rs7895676, was alsofound to increase FGFR2 expression.47 With the availabilityof a more complete catalog of common variants released bythe 1000 Genome project and fine mapping in larger case-control studies, it is possible that additional causal vari-ant(s) may be identified.

TOX3 Locus

The second strongest GWAS locus lies in a region onchromosome 16q12 containing TOX3 (formerly calledTNRC9) and LOC643714.26,52 The number of candidate

5

620

22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 6: Inherited Genetic Susceptibility to Breast Cancer

print&

web4C=FPO

Figure 2 Mechanisms that are potentially involved in breast cancer susceptibility. Genes in red represent high- and moderate-penetrance mutations. Genesin blue are cancer candidates found in/near common breast cancer susceptibility loci. With the exception of FGFR2, MYC, CASP8, and CCND1, it is unclear ifthose genes are the ones driving the association. Follow-up of the associated loci and functional analysis will confirm or exclude the involvement of thesegenes.

Causative SNP Tag SNP

Haplotype 1

Hap 1 Hap 2 Hap 3Population 2

Population 1

Figure 3 Schematic diagram of the LD pattern in a different pop-ulation. This diagram illustrates the value of using populations from otherethnicities to refine the signal of association. Haplotype 1 in population 1is split into three haplotypes in population 2. In population 1, the causalvariant and the tag SNPs are both in the same haplotype block, in highcorrelation with each other, and are hence both associated with thedisease. In population 2, the causal variant is no longer on the samehaplotype as the tag SNP because they were separated by a recombinationevent. Therefore, if we were to genotype both SNPs in population 2, thecausal variant will remain associated with the disease but not the tag SNP.Assuming that the LD structure is different between populations for thelocus of interest, fine mapping in other ethnicities can help to reduce thenumber of candidate causal variants.

Ghoussaini et al

621622623624625626627628629630631632633634635636637638639640641642643644645646647648649650651652653654655656657658659660661662663664665666667668669670671672673674675676677678679680681682

683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738739740741742743

causal variants was reduced to 13 using data from Europeanand Asian studies, with eight clustering in a highlyconserved region of open chromatin conformation located inthe intergenic region.26,52 Based on these data, it wasimpossible to discriminate which one of the 13 was thecausal variant. In African American studies, these variantshad completely opposite effects, suggesting the existence ofan additional risk variant, not present in Europeans andAsians, although this hypothesis requires confirmation.52

The expression of TOX3 did not vary by the genotypes ofthe original risk variant, rs3803662, in normal and tumorbreast samples.52 Long et al53 found the strongest associa-tion with one of the 13 variants, rs4784227, in Asian andEuropean case-control studies, and demonstrated that thisvariant is functional in luciferase and electrophoreticmobility shift assays. However, unlike the variants previ-ously mentioned, it lies in a region of closed chromatin,emphasizing the difficulty in interpreting functional exper-iments in this context.53 More recently, rs478227 wasassociated with increased FOXA1 binding and a repressiveeffect on TOX3.54

CCND1 Locus (11q13)

The 11q13 locus (rs614367) was among the loci with thestrongest association with breast cancer.32 An extensivefine-scale mapping of this locus using 4405 genotyped andimputed variants was performed in 89,050 Europeansubjects from the Breast Cancer Association Consortium

6REV 5.2.0 DTD � AJPA1459_proof

(BCAC).46 Three independent association signals weredetected and were strictly confined to ERþ tumors;rs554219, in the strongest peak, lies in an enhancer element,increases breast cancer risk, induces differential binding ofa TF, reduces the enhancer activity, and is associated witha decrease in cyclin D1 (CCND1) levels in breast tumors. Asecond, independent SNP in the same peak, rs78540526,lies in the same enhancer element and also appears to befunctional. A third SNP, rs75915166, generates a GATA3

ajp.amjpathol.org - The American Journal of Pathology

744

� 22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 7: Inherited Genetic Susceptibility to Breast Cancer

5

Genetic Susceptibility to Breast Cancer

745746747748749750751752753754755756757758759760761762763764765766767768769770771772773774775776777778779780781782783784785786787788789790791792793794795796797798799800801802803804805806

807808809810811812813814815816817818819820821822823824825826827828829830831832833834835836837838839840841842843844845846847848849850851852853854855856857858859860861862863864865866867

binding site within a silencer element. A long-range inter-action confined to ERþ breast cancer cells was identifiedbetween the enhancer and silencer elements and withCCND1. These associations were replicated in Asian pop-ulations despite their low frequency (<2%), providingfurther support that these SNPs may be causative. The effectsizes of those newly identified SNPs are larger than anypreviously reported breast cancer GWAS loci.46

TERT Locus (5p15)

Fine-scale mapping of the TERT locus was performed inapproximately 103,000 breast cancer cases and controls(European, Asian, and African American ancestries) fromBCAC, approximately 12,000 BRCA1 carriers, and approx-imately 40,000 ovarian cancer cases and their matchingcontrols.49 Three independent association signals wereidentified. SNPs in peak 1 (TERT promoter) were associatedwith telomere length, ERþ and ER� breast cancer, and breastcancer in BRCA1 carriers. SNPs in peak 2 (introns 2 to 4)were associated with telomere length and all breast andovarian cancers. SNPs in peak 3 (introns 2 to 4) were asso-ciated with ER� breast cancer, breast cancer in BRCA1carriers, and ovarian cancer, but not with telomere length.The findings provide evidence for genetic control of telomerelength by common variants across TERT. However, only thedirection of the findings in peak 1 supports the hypothesisthat shorter telomere length increases the risk of breastcancer, suggesting that most of cancer associations identifiedin the TERT locus are likely to be mediated through analternative mechanism involving the TERT gene.49

Biological Characteristics of Known Low-Penetrance Breast Cancer Loci

Insights into Genes and Pathways

For most of the known susceptibility loci, neither the causalvariant nor the gene through which the functional effects ofthat variant are mediated is known. Nevertheless, byconsidering the known functions of all of the genes neara susceptibility locus, which are thus candidates for beingthe functionally relevant gene, some patterns begin toemerge.

Mammary Gland Development

Three of the breast cancer susceptibility loci are in regionswith genes that are candidates because they are involved inmammary gland development: FGFR2 at 10q26, PTHLH at12p11, and TBX3 (T-BoX3) at 12q24. FGFR2, a tyrosinekinase receptor, plays a key role in normal mammary glanddevelopment and stem cell function.55 It is overexpressed andamplified in 5% to 10% of breast tumors. Several missensesomatic mutations, inducing FGFR2 overexpression, havebeen found in multiple cancers.56,57 PTHLH, an oncoprotein

The American Journal of Pathology - ajp.amjpathol.orgREV 5.2.0 DTD � AJPA1459_proof �

expressed in approximately 60% of breast tumors, playsa key role in lactation.58 It has also been suggested thatpatients with PTHLH-positive breast carcinoma are morelikely to develop bone metastasis.58 TBX3 is a downstreamtarget of the WNT/b-catenin pathway (playing a key role instem cell regulation and cell differentiation and prolifera-tion), is amplified and overexpressed in several cancers,including breast cancer, and is found at high levels in plasmafrom breast cancer patients.59 Germ-line mutations thatdisrupt TBX3 are responsible for Ulnar-Mammary syndrome,a condition characterized by defects of several organs,including the mammary glands and the heart.60 Expansion ofbreast cancer stem cells by estrogen has been found to bemediated by FGF/FGFR/TBX3 pathways.59

DNA Repair Pathway

DNA repair processes play a key role in the maintenance ofgenomic integrity. Five susceptibility loci are near genesinvolved in the DNA repair pathway: RAD51B at 14q24,RAD23B at 9q31, MUS81 at 11q13, SSBP4 at 14q13, andBABAM1 at 19p13. RAD51B is involved in the homologousrecombination repair pathway. Overexpression of this genecauses cell cycle G1 delay and cell apoptosis,61 and copynumber variation at 14q24 has been observed consistently infamilies with Li-Fraumeni syndrome.62 RAD23B also hasa key function in recognizing DNA lesions and in the nucle-otide excision repair pathway, as well as in targeting ubiq-uitylated proteins for proteasomal degradation.63 MUS81 isimportant in DNA repair mechanism and in maintaininggenome stability.64 SSBP4 is also involved in DNA repair andrecombination and acts as a tumor-suppressor gene.65

BABAM1 interacts with BRCA1 in the DNA repair processand enforces the BRCA1-dependent DNA damage response.66

Cell Cycle, Differentiation, and Apoptosis

Cell division, differentiation, and apoptosis are similar inboth normal and tumor cells, except that these processes areaberrantly regulated in cancer, leading the cancer cell and itsprogeny into uncontrolled expansion and invasion. Mostbreast cancer susceptibility loci contain genes with a centralfunction in these pathways. These include MDM4 at 1q32,CDKN2A and CDKN2B (CDK Qinhibitors 2A and 2B,respectively) at 9p21, MAP3K1 at 5q11, MRPS30 at 5p12,MYC at 8q24, TCF7L2 at 10q25, MAPKAP/MK5 at 12q24,NOTCH2 at 1p11, CCND1 and FGFs at 11q13, and KRE-MEN1 at 22q12. MDM4 is an oncogene that represses thetranscription of TP53 and TP73 and therefore plays a keyrole in cell cycle regulation and apoptosis.67 CDKN2A andCDKN2B are tumor-suppressor genes that lie adjacent toeach other. The p16 protein encoded by CDKN2A plays animportant role in cell cycle arrest, and its inactivation isa crucial event in the development of several types of humantumors.68 CDKN2B is another cell growth regulator thatcontrols cell cycle G1 progression. Its expression is

7

868

22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 8: Inherited Genetic Susceptibility to Breast Cancer

Q6

Q7

Q8

Ghoussaini et al

869870871872873874875876877878879880881882883884885886887888889890891892893894895896897898899900901902903904905906907908909910911912913914915916917918919920921922923924925926927928929930

931932933934935936937938939940941942943944945946947948949950951952953954955956957958959960961962963964965966967968969970971972973974975976977978979980981982983984985986987988989990991

dramatically induced by transforming growth factor-b,suggesting its involvement in transforming growth factor-beinduced growth inhibition.69,70 MAP3K1 encodes acomponent of the mitogen-activated protein kinase (MAPK)signaling pathway, known for its key role in cell differen-tiation, proliferation, and apoptosis.71 MAP3K1 regulatescell migration and survival, as well as immune systemfunction. MAP3K1-deficient retinas exhibit increased pro-liferation and apoptosis in mice.72 MRPS30 (programed celldeath protein 9/PDCP9) is a key component of the apoptoticsignals in cells.73 MYC is a proto-oncogene that drivesseveral mitogenic signals through activation of the MAPK/ERK pathway.74 MYC is overexpressed in different tumors,including breast and prostate cancers,74 and reducing itsexpression levels using RNA interference inhibits tumorgrowth both in vivo and in vitro.75 The TCF7L2 is a proto-oncogene, an important component of the WNT pathway,and well known for its association with type 2 diabetes.76

The MAPKAPKs have a pivotal role in the cell mitogenicprocesses and survival and are directly regulated byMAPKs.77 MK5/MAPKAP5 was found to regulate thetranslation of MYC and vice versa by a negative feedbackloop.78 NOTCH2 belongs to a family of transmembranereceptors and plays a key role in cell differentiation, pro-liferation, and apoptosis.79 Decreases in NOTCH2 expres-sion are associated with increasing grade of human breastcancer,80 and its signaling is required for anti-tumor im-munity in vivo.81 CCND1 is a key cell cycle regulator, andits amplification and overexpression were documented ina variety of tumors.82 FGFs have diverse roles in regulatingcell proliferation, migration, and differentiation, and theyplay a role in cancer pathogenesis.83 KREMEN1 suppressesthe WNT/b-catenin pathway, and its expression is low orabsent in tumor cells compared with normal tissues. Lack ofKREMEN1 and therefore activation of the WNT/b-cateninpathway is likely to be one of the mechanisms by whichtumorigenesis is enhanced.84

Of these regions, only 8q24 has been the focus ofmultiple functional experiments. Several independent loci,clustered in a gene desert a few hundred kb upstream anddownstream of MYC, have been found to be associated withan increased risk of prostate, breast, colorectal, ovarian,glioblastoma, and bladder cancers, as well as lymphocyticleukemia (National Humane Genome Research Institute,NIH, http://www.genome.gov/gwastudies, last accessed July29, 2013). Functional assessment both in vivo and in vitroidentified enhancer elements in each of the loci, includingmammary and prostate enhancers that interact physicallywith the MYC and PVT-1 promoters through a long-rangechromatin loop and regulate its expression levels.85e87

Growth Factors and Tumor Aggressiveness

Although departure from tissue homeostasis and initiation ofcancer are mainly initiated by oncogenic mutations, growthfactors play a major role in the subsequent steps of tumor

8REV 5.2.0 DTD � AJPA1459_proof

progression, particularly clonal expansion, invasiveness,and colonization of distant organs. Several genes near themost recently identified loci encode a growth factor (NTN4at 12q22) or are involved in tumor aggressiveness andmetastasis (PTH1R at 3p21), FOXQ1 at 6p25, ARHGEF5 at7q35, PAX9 at 14q13, and MKL1 at 22q13. NTN4 encodesa growth factor crucial for the regulation of tumor growth.High expression levels of NTN4 were detected in ERþ

breast cancer but not in ER�, and its use was suggested asa prognosis marker.88 PTH1R is the receptor of PTHLH(also identified in a GWAS). Up-regulation of PTH1R wasfound in patients with invasive breast ductal carcinoma.Silencing PTH1R induced suppression of cell prolifera-tion.89 Forkhead TF (FOXQ1) plays a key role in regulatingepithelial-mesenchymal transition (EMT; crucial formetastasis) in breast cancer cells. Its expression is correlatedwith high-grade breast cancers and is associated with poorprognosis. Silencing FOXQ1 reversed EMT in invasivehuman breast cancer cell lines and decreased their inva-siveness. Conversely, overexpression of FOXQ1 in humanmammary epithelial cells induced an EMT and the acqui-sition of resistance to apoptosis after chemotherapy.90

ARHGEF5 is an oncogene and plays a pivotal role incancer development and breast metastasis through activationof the ARHGEF5/TIM pathway.91 PAX9 encodes a TF thatregulates cell proliferation, invasion, and resistance toapoptosis.92 MLK1 is also important in cell invasion andmetastasis, particularly in breast carcinomas.93

ER Pathway

Estrogen plays a key role in growth and in the maintenanceof reproductive functions. Two breast cancer susceptibilityloci are near genes with pivotal roles in this pathway. One at6q25 is close to ESR1 and the other, at 21q21, is close toNRIP1/RIP140. ESR1 encodes the ERa protein, which isactivated by bound estrogen. NRIP1 is a cofactor of ERaand can inhibit its mitogenic effects. This gene plays animportant role in the regulation of breast cancer cell growth,notably in ER-positive breast tumors.94

Telomere Length

Telomeres are regions of repetitive nucleotides at the end ofchromosomes. They are crucial in maintaining genomicstability by protecting chromosomes from degradation,recombination, and end-to-end fusion.95 Human telomerestend to progressively shorten with each cell division, andthis shortening process is associated with ageing andincreased risk of cancer.96 One of the identified breastcancer susceptibility loci lies at 5p15 and is near TERT,which encodes telomerase reverse transcriptase, the catalyticsubunit of telomerase.28 Abnormal expression of TERT hasbeen reported in several types of cancer, and the region isfrequently amplified in many types of cancer, such as lungand cervical cancers.97,98

ajp.amjpathol.org - The American Journal of Pathology

992

� 22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 9: Inherited Genetic Susceptibility to Breast Cancer

½F4�½F4�

print&

web4C=FPO

Genetic Susceptibility to Breast Cancer

9939949959969979989991000100110021003100410051006100710081009101010111012101310141015101610171018101910201021102210231024102510261027102810291030103110321033103410351036103710381039104010411042104310441045104610471048104910501051105210531054

105510561057105810591060106110621063106410651066

Other Biological Pathways

Many genes near the known susceptibility loci are either ofunknown function or have functions that do not have a clearconnection to breast cancer development (a notable examplebeing the TOX3 locus). These genes could simply not be theones being targeted, but alternatively might be involved insusceptibility through a previously unsuspected mechanism.Future functional analysis should shed some light on thecausal variants within each of these loci and the mechanismsby which they are driving the association.

9

10671068106910701071107210731074107510761077107810791080108110821083108410851086

Common Pathways for Multiple Phenotypes

Although most breast cancer susceptibility loci are diseasespecific, at least 17 loci (at 1q32, 2p24, 2q31, 4q24, 5p12,5p15, 6q25, 8q24, 9p21, 9q31, 10p12, 19p13, 10q26,11p15, 11q13, 12q24, and 14q24) occur in regions associ-ated with other cancers and complex diseases/phenotypes,suggesting a shared mechanism in disease development(Figure 4). Four of these have been intensively discussed.The 5p15 region containing TERT is also associated withglioma, basal cell carcinoma, and lung and pancreaticcancers (http://www.genome.gov/gwastudies). The 8q24region is associated with at least six cancers and one of theloci, rs6983267, has a pleiotropic effect on prostate, colo-rectal, and possibly ovarian cancers (http://www.genome.gov/gwastudies). The 9p21 region containing the twotumor-suppressor genes, CDKN2A and CDKN2B, is

Figure 4 Location of the common breast cancer susceptibility loci together4q24, 5p12, 5p15, 6q25, 8q24, 9p21, 9q31, 10p12, 10q26, 11p15, 11q13, 12qcancers.

The American Journal of Pathology - ajp.amjpathol.orgREV 5.2.0 DTD � AJPA1459_proof �

associated with at least six cancers (breast and pancreaticcancers, lymphoblastic leukemia, melanoma, glioma, basalcell carcinoma, and nasopharyngeal carcinoma), myocardialinfarction, type 2 diabetes, intracranial aneurysm, cutaneousnevi, and sporadic amyotrophic lateral sclerosis (http://www.genome.gov/gwastudies). The 11q13 region containsCCND1 and several FGFs and is associated with breast,prostate, and renal cancers (http://www.genome.gov/gwastudies. The nearest genes to the 12q24 region areTBX3, important for mammary gland development, andMAPKAP, a component of the MAPK pathway that inter-acts with MYC. SNPs in this region are associated withbreast cancer, squamous esophageal carcinoma, liveradenoma, renal cell carcinoma, heart diseases, type 1 dia-betes, blood pressure, and PSA Qlevels (http://www.genome.gov/gwastudies).

Clinical Implications

Although the clinical utility of testing for high-penetrancemutations such as BRCA1 and BRCA2, is well established,the potential clinical use of polygenic prediction for breastcancer is extensively debated.99e102 Taken separately, theSNP associations are too weak to be useful on an individualbasis. However, because their effects seem to combinemultiplicatively, there is potential for the predictive powerto improve substantially as more variants are found.101 Ithas been argued that polygenic risk profiles have limiteddiscrimination for breast cancer, but because the clinical

with the most plausible nearby genes. At least 17 loci (1q32, 2p24, 2q31,24, 14q24, and 19q13) are associated with breast, prostate, and ovarian

9

108710881089109010911092109310941095109610971098109911001101110211031104110511061107110811091110111111121113111411151116

22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 10: Inherited Genetic Susceptibility to Breast Cancer

Q10

Ghoussaini et al

11171118111911201121112211231124112511261127112811291130113111321133113411351136113711381139114011411142114311441145114611471148114911501151115211531154115511561157115811591160116111621163116411651166116711681169117011711172117311741175117611771178

1179118011811182118311841185118611871188118911901191119211931194119511961197119811991200120112021203120412051206120712081209121012111212121312141215121612171218121912201221122212231224122512261227122812291230123112321233123412351236123712381239

utility of a risk prediction model is not restricted todiscrimination, these should be further evaluated in thecontext of risk stratification after combining other riskfactors, such as lifestyle elements and family history.

In a recent study, Pashayan et al103 showed that person-alized screening based on age and polygenic risk factors (18common low-penetrance loci) is more efficient than a stan-dard approach based solely on age. Personalized screeningcould reduce the number of people eligible for screening by24% while detecting most cancers (86%) compared witha program based on age alone.103 Individual risk stratifica-tion would increase the efficiency of the screening program,by intensifying screening in high-risk individuals andsparing unnecessary and even potentially harmful screeningin those with lower risk levels. This increase in efficiencywould be even greater for risk stratification based on the 72loci identified.104

Future Directions

Whole Genome Resequencing

The emerging results from GWASs suggest that a substan-tial fraction, perhaps the majority, of the familial risk ofbreast cancer can be explained by common variants, most ofwhich are yet to be identified. It is highly likely that rarer,higher-risk alleles (such as risk alleles of PALB2) alsoremain to be identified. Unraveling the full architecture ofbreast cancer susceptibility will therefore require a compre-hensive search for rarer variants. Technological advancesover the past few years have rendered whole-exome orwhole-genome resequencing of large cohorts of cases andcontrols increasingly more practical.

Consortia and Collaborations

Studies with large sample sizes are critical to enhancepower, achieve genome-wide significance levels, and yieldadditional insights in terms of replication of the results, newgenetic discoveries, and key biological findings.

The past few years witnessed the formation of multipleinternational consortia and substantial widespread collabo-rations of many multidisciplinary investigators, includingepidemiologists, clinicians, statisticians, bioinformaticians,and biologists. Groups with their individual case-control setsjoined efforts to increase sample size through meta-analysis,and these collaborations resulted in an explosion of newassociations. One of the most successful examples in canceris the Collaborative Oncological Gene-Environment Study,a collaborative project funded by the European Commission,which combined data from four different consortia: BCAC,Prostate Cancer Association Group to Investigate CancerAssociated Alterations in the Genome, the Ovarian CancerAssociation Consortium, and the Consortium of Investigatorsof Modifiers of BRCA1/2. This collaboration has developeda genotyping array (Collaborative Oncological Gene-

10REV 5.2.0 DTD � AJPA1459_proof

Environment Study) of approximately 200,000 SNPs thathas been genotyped in approximately 200,000 cases withbreast, prostate, and ovarian cancers and approximately200,000 controls from around the world. Results from thisproject identified 74 novel breast, prostate, and ovariancancer loci (41 of which are for breast cancer), nearlydoubling the number of the previously established ones. Inaddition, those results provided an accurate assessment of therisk related to each locus and will offer a comprehensive finemapping of the known loci. It will also be possible to evaluategenetic associations with disease survival, clinical outcome,and tumor subsets and to evaluate the combined effect ofgenetic variants and lifestyle risk factors on disease risk.More findings emerging from this project should add greatlyto our understanding of the genetic architecture of breast andother cancers.

GWASs in Other Populations

Breast cancer GWASs have primarily been conducted inwomen of European descent, with the exception of one studyperformed in Chinese women, which indicated importantdifferences in the pattern of genetic loci between ethnicities.These may result from differences in allele frequencies,patterns of LD, or interactions with genetic background orlifestyle risk factors. Further GWASs in non-Europeanpopulations should therefore be extremely fruitful.

Conclusions

In the future, more breast cancer susceptibility alleles arelikely to be identified, and these should, in turn, lead tofurther functional studies and expand our understanding ofthe underlying biological characteristics of breast cancer. Thecandidate genes in susceptibility regions identified thus farappear to cluster into a few biological pathways: mammarygland development, DNA repair, cell cycle control, andestrogen receptor signaling. Genes with unknown function atthese loci will need further work, but their characterizationmight reveal unexpected and important insights into cancerbiology. Loci that are associated with breast cancer and othercancers and traits will be important in revealing biologicalmechanisms that connect pathways and diseases. In additionto the common variants, rare genetic variations identifiedthrough whole-exome and whole-genome sequencing strat-egies will likely expand the catalog of breast cancersusceptibility variations. Further studies to examine struc-tural variation and epigenomics may also uncover moresusceptibility loci. Integration of the germ-line susceptibilitydata with emerging somatic sequence data may be importantto both provide a more comprehensive analysis of suscepti-bility by disease subtype and interpret better the underlyingbiological characteristics. Finally, large-scale populationstudies (preferably prospective) will be required to define thecombined effects of common and rare variants, and lifestyle

ajp.amjpathol.org - The American Journal of Pathology

1240

� 22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 11: Inherited Genetic Susceptibility to Breast Cancer

Q11

12

13

Genetic Susceptibility to Breast Cancer

12411242124312441245124612471248124912501251125212531254125512561257125812591260126112621263126412651266126712681269127012711272127312741275127612771278127912801281128212831284128512861287128812891290129112921293129412951296129712981299130013011302

1303130413051306130713081309131013111312131313141315131613171318131913201321132213231324132513261327132813291330133113321333133413351336133713381339134013411342134313441345134613471348134913501351135213531354135513561357135813591360136113621363

and other risk factors, on disease risk. This should lead tomore rational strategies of disease prevention, while theknowledge of breast cancer etiology gained through thefunctional understanding of disease susceptibility may leadto novel therapeutic approaches for breast cancer patients.

Acknowledgments

We thank Dr. David Meyre and Mel Maranian for com-menting on an early draft of the manuscript and RogerMilne for generating Figure 4.

References

1. Kamangar F, Dores GM, Anderson WF: Patterns of cancer incidence,mortality, and prevalence across five continents: defining priorities toreduce cancer disparities in different geographic regions of the world.J Clin Oncol 2006, 24:2137e2150

2. Wright T, McGechan A: Breast cancer: new technologies for riskassessment and diagnosis. Mol Diagn 2003, 7:49e55

3. Familial breast cancer: collaborative reanalysis of individual datafrom 52 epidemiological studies including 58,209 women with breastcancer and 101,986 women without the disease. Lancet 2001, 358:1389e1399

4. Goldgar DE, Easton DF, Cannon-Albright LA, Skolnick MH:Systematic population-based assessment of cancer risk in first-degreerelatives of cancer probands. J Natl Cancer Inst 1994, 86:1600e1608

5. Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J,Koskenvuo M, Pukkala E, Skytthe A, Hemminki K: Environmentaland heritable factors in the causation of cancer: analyses of cohorts oftwins from Sweden, Denmark, and Finland. N Engl J Med 2000, 343:78e85

6. Peto J, Mack TM: High constant incidence in twins and other rela-tives of women with breast cancer. Nat Genet 2000, 26:411e414

7. Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K,Tavtigian S, Liu Q, Cochran C, Bennett LM, Ding W: A strongcandidate for the breast and ovarian cancer susceptibility geneBRCA1. Science 1994, 266:66e71

8. Wooster R, Bignell G, Lancaster J, Swift S, Seal S, Mangion J,Collins N, Gregory S, Gumbs C, Micklem G: Identification of thebreast cancer susceptibility gene BRCA2. Nature 1995, 378:789e792

9. Antoniou AC, Cunningham AP, Peto J, Evans DG, Lalloo F,Narod SA, Risch HA, Eyfjord JE, Hopper JL, Southey MC,Olsson H, Johannsson O, Borg A, Pasini B, Radice P, Manoukian S,Eccles DM, Tang N, Olah E, Anton-Culver H, Warner E, Lubinski J,Gronwald J, Gorski B, Tryggvadottir L, Syrjakoski K,Kallioniemi OP, Eerola H, Nevanlinna H, Pharoah PD, Easton DF:The BOADICEA model of genetic susceptibility to breast andovarian cancers: updates and extensions. Br J Cancer 2008, 98:1457e1466

10. Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE,Hopper JL, Loman N, Olsson H, Johannsson O, Borg A, Pasini B,Radice P, Manoukian S, Eccles DM, Tang N, Olah E, Anton-Culver H, Warner E, Lubinski J, Gronwald J, Gorski B, Tulinius H,Thorlacius S, Eerola H, Nevanlinna H, Syrjakoski K, Kallioniemi OP,Thompson D, Evans C, Peto J, Lalloo F, Evans DG, Easton DF:Average risks of breast and ovarian cancer associated with BRCA1 orBRCA2 mutations detected in case series unselected for familyhistory: a combined analysis of 22 studies. Am J Hum Genet 2003,72:1117e1130

11. King MC, Marks JH, Mandell JB: Breast and ovarian cancer risks dueto inherited mutations in BRCA1 and BRCA2. Science 2003, 302:643e646

The American Journal of Pathology - ajp.amjpathol.orgREV 5.2.0 DTD � AJPA1459_proof �

12. Malkin D, Li FP, Strong LC, Fraumeni JF Jr., Nelson CE, Kim DH,Kassel J, Gryka MA, Bischoff FZ, Tainsky MA: Germ line p53mutations in a familial syndrome of breast cancer, sarcomas, andother neoplasms. Science 1990, 250:1233e1238 Q

13. Hemminki A, Markie D, Tomlinson I, Avizienyte E, Roth S,Loukola A, Bignell G, Warren W, Aminoff M, Hoglund P,Jarvinen H, Kristo P, Pelin K, Ridanpaa M, Salovaara R, Toro T,Bodmer W, Olschwang S, Olsen AS, Stratton MR, de la Chapelle A,Aaltonen LA: A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature 1998, 391:184e187

14. Renwick A, Thompson D, Seal S, Kelly P, Chagtai T, Ahmed M,North B, Jayatilake H, Barfoot R, Spanova K, McGuffog L,Evans DG, Eccles D, Easton DF, Stratton MR, Rahman N: ATMmutations that cause ataxia-telangiectasia are breast cancer suscepti-bility alleles. Nat Genet 2006, 38:873e875

15. Bell DW, Varley JM, Szydlo TE, Kang DH, Wahrer DC,Shannon KE, Lubratovich M, Verselis SJ, Isselbacher KJ,Fraumeni JF, Birch JM, Li FP, Garber JE, Haber DA: Heterozygousgerm line hCHK2 mutations in Li-Fraumeni syndrome. Science 1999,286:2528e2531

16. Meijers-Heijboer H, van den Ouweland A, Klijn J, Wasielewski M,de Snoo A, Oldenburg R, et al: CHEK2-Breast Cancer Consortium:Low-penetrance susceptibility to breast cancer due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 mutations. Nat Genet2002, 31:55e59 Q

17. Rahman N, Seal S, Thompson D, Kelly P, Renwick A, Elliott A,Reid S, Spanova K, Barfoot R, Chagtai T, Jayatilake H, McGuffog L,Hanks S, Evans DG, Eccles D, Easton DF, Stratton MR: PALB2,which encodes a BRCA2-interacting protein, is a breast cancersusceptibility gene. Nat Genet 2007, 39:165e167

18. Seal S, Thompson D, Renwick A, Elliott A, Kelly P, Barfoot R,Chagtai T, Jayatilake H, Ahmed M, Spanova K, North B,McGuffog L, Evans DG, Eccles D, Easton DF, Stratton MR,Rahman N: Truncating mutations in the Fanconi anemia J geneBRIP1 are low-penetrance breast cancer susceptibility alleles. NatGenet 2006, 38:1239e1241

19. Nelen MR, Padberg GW, Peeters EA, Lin AY, van den Helm B,Frants RR, Coulon V, Goldstein AM, van Reen MM, Easton DF,Eeles RA, Hodgsen S, Mulvihill JJ, Murday VA, Tucker MA,Mariman EC, Starink TM, Ponder BA, Ropers HH, Kremer H,Longy M, Eng C: Localization of the gene for Cowden disease tochromosome 10q22-23. Nat Genet 1996, 13:114e116

20. Guilford P, Hopkins J, Harraway J, McLeodM,McLeod N, Harawira P,Taite H, Scoular R, Miller A, Reeve AE: E-cadherin germline mutationsin familial gastric cancer. Nature 1998, 392:402e405

21. Easton DF: How many more breast cancer predisposition genes arethere? Breast Cancer Res 1999, 1:14e17

22. Pharoah PD, Tyrer J, Dunning AM, Easton DF, Ponder BA: Asso-ciation between common variation in 120 candidate genes and breastcancer risk. PLoS Genet 2007, 3:e42

23. Cox A, Dunning AM, Garcia-Closas M, Balasubramanian S,Reed MW, Pooley KA, et al: A common coding variant in CASP8 isassociated with breast cancer risk. Nat Genet 2007, 39:352e358

24. Ahmed S, Thomas G, Ghoussaini M, Healey CS, Humphreys MK,Platte R, et al: Newly discovered breast cancer susceptibility loci on3p24 and 17q23.2. Nat Genet 2009, 41:585e590

25. Antoniou AC, Wang X, Fredericksen ZS, McGuffog L, Tarrell R,Sinilnikova OM, et al: A locus on 19p13 modifies risk of breastcancer in BRCA1 mutation carriers and is associated with hormonereceptor-negative breast cancer in the general population. Nat Genet2010, 42:885e892

26. Easton DF, Pooley KA, Dunning AM, Pharoah PD, Thompson D,Ballinger DG, et al: Genome-wide association study identifies novelbreast cancer susceptibility loci. Nature 2007, 447:1087e1093

27. Ghoussaini M, Fletcher O, Michailidou K, Turnbull C, Schmidt MK,Dicks E, et al: Genome-wide association analysis identifies three newbreast cancer susceptibility loci. Nat Genet 2012, 44:312e318

11

1364

22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 12: Inherited Genetic Susceptibility to Breast Cancer

Ghoussaini et al

13651366136713681369137013711372137313741375137613771378137913801381138213831384138513861387138813891390139113921393139413951396139713981399140014011402140314041405140614071408140914101411141214131414141514161417141814191420142114221423142414251426

1427142814291430143114321433143414351436143714381439144014411442144314441445144614471448144914501451145214531454145514561457145814591460146114621463146414651466146714681469147014711472147314741475147614771478147914801481148214831484148514861487

28. Haiman CA, Chen GK, Vachon CM, Canzian F, Dunning A,Millikan RC, et al: A common variant at the TERT-CLPTM1L locusis associated with estrogen receptor-negative breast cancer. Nat Genet2011, 43:1210e1214

29. Hunter DJ, Kraft P, Jacobs KB, Cox DG, Yeager M, Hankinson SE,Wacholder S, Wang Z, Welch R, Hutchinson A, Wang J, Yu K,Chatterjee N, Orr N, Willett WC, Colditz GA, Ziegler RG, Berg CD,Buys SS, McCarty CA, Feigelson HS, Calle EE, Thun MJ,Hayes RB, Tucker M, Gerhard DS, Fraumeni JF Jr., Hoover RN,Thomas G, Chanock SJ: A genome-wide association study identifiesalleles in FGFR2 associated with risk of sporadic postmenopausalbreast cancer. Nat Genet 2007, 39:870e874

30. Stacey SN, Manolescu A, Sulem P, Rafnar T, Gudmundsson J,Gudjonsson SA, et al: Common variants on chromosomes 2q35 and16q12 confer susceptibility to estrogen receptor-positive breastcancer. Nat Genet 2007, 39:865e869

31. Stacey SN, Manolescu A, Sulem P, Thorlacius S, Gudjonsson SA,Jonsson GF, et al: Common variants on chromosome 5p12 confersusceptibility to estrogen receptor-positive breast cancer. Nat Genet2008, 40:703e706

32. Turnbull C, Ahmed S, Morrison J, Pernet D, Renwick A,Maranian M, Seal S, Ghoussaini M, Hines S, Healey CS, Hughes D,Warren-Perry M, Tapper W, Eccles D, Evans DG, Hooning M,Schutte M, van den Ouweland A, Houlston R, Ross G, Langford C,Pharoah PD, Stratton MR, Dunning AM, Rahman N, Easton DF:Genome-wide association study identifies five new breast cancersusceptibility loci. Nat Genet 2010, 42:504e507

33. Zheng W, Long J, Gao YT, Li C, Zheng Y, Xiang YB, Wen W,Levy S, Deming SL, Haines JL, Gu K, Fair AM, Cai Q, Lu W,Shu XO: Genome-wide association study identifies a new breastcancer susceptibility locus at 6q25.1. Nat Genet 2009, 41:324e328

34. Garcia-ClosasM, Couch FJ, Lindstrom S, Michailidou K, Schmidt MK,Brook MN, et al: Genome-wide association studies identify four ERnegative-specific breast cancer risk loci. Nat Genet 2013, 45:392

35. Michailidou K, Hall P, Gonzalez-Neira A, Ghoussaini M, Dennis J,Milne RL, et al: Large-scale genotyping identifies 41 new lociassociated with breast cancer risk. Nat Genet 2013, 45:353e361.361e1-2

36. Siddiq A, Couch FJ, Chen GK, Lindstrom S, Eccles D, Millikan RC,et al: A meta-analysis of genome-wide association studies of breastcancer identifies two novel susceptibility loci at 6q14 and 20q11.Hum Mol Genet 2012, 21:5373e5384

37. Lakhani SR, van de Vijver MJ, Jacquemier J, Anderson TJ, Osin PP,McGuffog L, Easton DF: The pathology of familial breast cancer:predictive value of immunohistochemical markers estrogen receptor,progesterone receptor, HER-2, and p53 in patients with mutations inBRCA1 and BRCA2. J Clin Oncol 2002, 20:2310e2318

38. Antoniou AC, Spurdle AB, Sinilnikova OM, Healey S, Pooley KA,Schmutzler RK, et al: Common breast cancer-predisposition allelesare associated with breast cancer risk in BRCA1 and BRCA2 muta-tion carriers. Am J Hum Genet 2008, 82:937e948

39. Antoniou AC, Beesley J, McGuffog L, Sinilnikova OM, Healey S,Neuhausen SL, et al: Common breast cancer susceptibility alleles andthe risk of breast cancer for BRCA1 and BRCA2 mutation carriers:implications for risk prediction. Cancer Res 2010, 70:9742e9754

40. Garcia-Closas M, Hall P, Nevanlinna H, Pooley K, Morrison J,Richesson DA, et al; Australian Ovarian Cancer Management Group,Kathleen Cuningham Foundation Consortium For Research IntoFamilial Breast Cancer: Heterogeneity of breast cancer associationswith five susceptibility loci by clinical and pathological characteris-tics. PLoS Genet 2008, 4:e1000054

41. Garcia-Closas M, Chanock S: Genetic susceptibility loci for breastcancer by estrogen receptor status. Clin Cancer Res 2008, 14:8000e8009

42. Stephens PJ, Tarpey PS, Davies H, Van LP, Greenman C,Wedge DC, et al: The landscape of cancer genes and mutationalprocesses in breast cancer. Nature 2012, 486:400e404

12REV 5.2.0 DTD � AJPA1459_proof

43. Xu Z, Taylor JA: SNPinfo: integrating GWAS and candidate geneinformation into functional SNP selection for genetic associationstudies. Nucleic Acids Res 2009, 37:W600eW605

44. Dunham I, Kundaje A, Aldred SF, Collins PJ, Davis CA, Doyle F,et al: An integrated encyclopedia of DNA elements in the humangenome. Nature 2012, 489:57e74

45. Meyer KB, Maia AT, O’Reilly M, Teschendorff AE, Chin SF,Caldas C, Ponder BA: Allele-specific up-regulation of FGFR2increases susceptibility to breast cancer. PLoS Biol 2008, 6:e108

46. French JD, Ghoussaini M, Edwards SL, Meyer KB, Michailidou K,Ahmed S, et al: Functional variants at the 11q13 risk locus for breastcancer regulate cyclin D1 expression through long-range enhancers.Am J Hum Genet 2013, 92:489e503

47. Udler MS, Meyer KB, Pooley KA, Karlins E, Struewing JP, Zhang J,Doody DR, Macarthur S, Tyrer J, Pharoah PD, Luben R, Bernstein L,Kolonel LN, Henderson BE, Le ML, Ursin G, Press MF, Brennan P,Sangrajrang S, Gaborieau V, Odefrey F, Shen CY, Wu PE,Wang HC, Kang D, Yoo KY, Noh DY, Ahn SH, Ponder BA,Haiman CA, Malone KE, Dunning AM, Ostrander EA, Easton DF:FGFR2 variants and breast cancer risk: fine-scale mapping usingAfrican American studies and analysis of chromatin conformation.Hum Mol Genet 2009, 18:1692e1703

48. Udler MS, Tyrer J, Easton DF: Evaluating the power to discriminatebetween highly correlated SNPs in genetic association studies. GenetEpidemiol 2010, 34:463e468

49. Bojesen SE, Pooley KA, Johnatty SE, Beesley J, Michailidou K,Tyrer JP, et al: Multiple independent variants at the TERT locus areassociated with telomere length and risks of breast and ovariancancer. Nat Genet 2013, 45:371e372

50. Chen F, Chen GK, Millikan RC, John EM, Ambrosone CB,Bernstein L, Zheng W, Hu JJ, Ziegler RG, Deming SL, Bandera EV,Nyante S, Palmer JR, Rebbeck TR, Ingles SA, Press MF, Rodriguez-Gil JL, Chanock SJ, Le ML, Kolonel LN, Henderson BE, Stram DO,Haiman CA: Fine-mapping of breast cancer susceptibility loci char-acterizes genetic risk in African Americans. Hum Mol Genet 2011,20:4491e4503

51. Huijts PE, van Dongen M, de Goeij MC, van Moolenbroek AJ,Blanken F, Vreeswijk MP, de Kruijf EM, Mesker WE, van Zwet EW,Tollenaar RA, Smit VT, van Asperen CJ, Devilee P: Allele-specificregulation of FGFR2 expression is cell type-dependent and mayincrease breast cancer risk through a paracrine stimulus involvingFGF10. Breast Cancer Res 2011, 13:R72

52. Udler MS, Ahmed S, Healey CS, Meyer K, Struewing J, Maranian M,et al: Fine scale mapping of the breast cancer 16q12 locus. Hum MolGenet 2010, 19:2507e2515

53. Long J, Cai Q, Shu XO, Qu S, Li C, Zheng Y, et al: Identification ofa functional genetic variant at 16q12.1 for breast cancer risk: resultsfrom the Asia Breast Cancer Consortium. PLoS Genet 2010, 6:e1001002

54. Cowper-Sal LR, Zhang X, Wright JB, Bailey SD, Cole MD,Eeckhoute J, Moore JH, Lupien M: Breast cancer risk-associatedSNPs modulate the affinity of chromatin for FOXA1 and alter geneexpression. Nat Genet 2012, 44:1191e1198

55. Pond AC, Bin X, Batts T, Roarty K, Hilsenbeck S, Rosen JM:Fibroblast growth factor receptor signaling is essential for normalmammary gland development and stem cell function. Stem Cells2013, 31:178e189

56. Adnane J, Gaudray P, Dionne CA, Crumley G, Jaye M,Schlessinger J, Jeanteur P, Birnbaum D, Theillet C: BEK and FLG,two receptors to members of the FGF family, are amplified in subsetsof human breast cancers. Oncogene 1991, 6:659e663

57. Moffa AB, Tannheimer SL, Ethier SP: Transforming potential ofalternatively spliced variants of fibroblast growth factor receptor 2 inhuman mammary epithelial cells. Mol Cancer Res 2004, 2:643e652

58. Wysolmerski JJ, Broadus AE: Hypercalcemia of malignancy: thecentral role of parathyroid hormone-related protein. Annu Rev Med1994, 45:189e200

ajp.amjpathol.org - The American Journal of Pathology

1488

� 22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 13: Inherited Genetic Susceptibility to Breast Cancer

Genetic Susceptibility to Breast Cancer

14891490149114921493149414951496149714981499150015011502150315041505150615071508150915101511151215131514151515161517151815191520152115221523152415251526152715281529153015311532153315341535153615371538153915401541154215431544154515461547154815491550

1551155215531554155515561557155815591560156115621563156415651566156715681569157015711572157315741575157615771578157915801581158215831584158515861587158815891590159115921593159415951596159715981599160016011602160316041605160616071608160916101611

59. Lomnytska M, Dubrovska A, Hellman U, Volodko N,Souchelnytskyi S: Increased expression of cSHMT, Tbx3 and utro-phin in plasma of ovarian and breast cancer patients. Int J Cancer2006, 118:412e421

60. Bamshad M, Lin RC, Law DJ, Watkins WC, Krakowiak PA,Moore ME, Franceschini P, Lala R, Holmes LB, Gebuhr TC,Bruneau BG, Schinzel A, Seidman JG, Seidman CE, Jorde LB:Mutations in human TBX3 alter limb, apocrine and genital devel-opment in ulnar-mammary syndrome. Nat Genet 1997, 16:311e315

61. Havre PA, Rice M, Ramos R, Kmiec EB: HsRec2/Rad51L1, a proteininfluencing cell cycle progression, has protein kinase activity. ExpCell Res 2000, 254:33e44

62. Shlien A, Tabori U, Marshall CR, Pienkowska M, Feuk L,Novokmet A, Nanda S, Druker H, Scherer SW, Malkin D: Excessivegenomic DNA copy number variation in the Li-Fraumeni cancerpredisposition syndrome. Proc Natl Acad Sci U S A 2008, 105:11264e11269

63. Dantuma NP, Heinen C, Hoogstraten D: The ubiquitin receptorRad23: at the crossroads of nucleotide excision repair and proteaso-mal degradation. DNA Repair (Amst) 2009, 8:449e460

64. Murfuni I, Nicolai S, Baldari S, Crescenzi M, Bignami M,Franchitto A, Pichierri P: The WRN and MUS81 proteins limit celldeath and genome instability following oncogene activation. Onco-gene 2013, 32:610e620

65. Castro P, Liang H, Liang JC, Nagarajan L: A novel, evolutionarilyconserved gene family with putative sequence-specific single-stranded DNA-binding activity. Genomics 2002, 80:78e85

66. Feng L, Huang J, Chen J: MERIT40 facilitates BRCA1 localizationand DNA damage repair. Genes Dev 2009, 23:719e728

67. Toledo F, Wahl GM: MDM2 and MDM4: p53 regulators as targets inanticancer therapy. Int J Biochem Cell Biol 2007, 39:1476e1482

68. Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K,Tavtigian SV, Stockert E, Day RS III, Johnson BE, Skolnick MH: Acell cycle regulator potentially involved in genesis of many tumortypes. Science 1994, 264:436e440

69. Caldon CE, Daly RJ, Sutherland RL, Musgrove EA: Cell cyclecontrol in breast cancer cells. J Cell Biochem 2006, 97:261e274

70. Wu DT, Bitzer M, Ju W, Mundel P, Bottinger EP: TGF-betaconcentration specifies differential signaling profiles of growtharrest/differentiation and apoptosis in podocytes. J Am Soc Nephrol2005, 16:3211e3221

71. Zhang W, Liu HT: MAPK signal pathways in the regulation of cellproliferation in mammalian cells. Cell Res 2002, 12:9e18

72. Mongan M, Wang J, Liu H, Fan Y, Jin C, Kao WY, Xia Y: Loss ofMAP3K1 enhances proliferation and apoptosis during retinal devel-opment. Development 2011, 138:4001e4012

73. Cavdar KE, Ranasinghe A, Burkhart W, Blackburn K, Koc H,Moseley A, Spremulli LL: A new face on apoptosis: death-associatedprotein 3 and PDCD9 are mitochondrial ribosomal proteins. FEBSLett 2001, 492:166e170

74. Sears RC: The life cycle of C-myc: from synthesis to degradation.Cell Cycle 2004, 3:1133e1137

75. Wang YH, Liu S, Zhang G, Zhou CQ, Zhu HX, Zhou XB, Quan LP,Bai JF, Xu NZ: Knockdown of c-Myc expression by RNAi inhibitsMCF-7 breast tumor cells growth in vitro and in vivo. Breast CancerRes 2005, 7:R220eR228

76. Grant SF, ThorleifssonG, Reynisdottir I, Benediktsson R,ManolescuA,Sainz J, Helgason A, Stefansson H, Emilsson V, Helgadottir A,Styrkarsdottir U,Magnusson KP,Walters GB, Palsdottir E, Jonsdottir T,Gudmundsdottir T, Gylfason A, Saemundsdottir J, Wilensky RL,Reilly MP, Rader DJ, Bagger Y, Christiansen C, Gudnason V,Sigurdsson G, Thorsteinsdottir U, Gulcher JR, Kong A, Stefansson K:Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk oftype 2 diabetes. Nat Genet 2006, 38:320e323

77. Cargnello M, Roux PP: Activation and function of the MAPKs andtheir substrates, the MAPK-activated protein kinases. Microbiol MolBiol Rev 2011, 75:50e83

The American Journal of Pathology - ajp.amjpathol.orgREV 5.2.0 DTD � AJPA1459_proof �

78. Kress TR, Cannell IG, Brenkman AB, Samans B, Gaestel M,Roepman P, Burgering BM, Bushell M, Rosenwald A, Eilers M: TheMK5/PRAK kinase and Myc form a negative feedback loop that isdisrupted during colorectal tumorigenesis. Mol Cell 2011, 41:445e457

79. Fiuza UM, Arias AM: Cell and molecular biology of Notch. JEndocrinol 2007, 194:459e474

80. O’Neill CF, Urs S, Cinelli C, Lincoln A, Nadeau RJ, Leon R, Toher J,Mouta-Bellum C, Friesel RE, Liaw L: Notch2 signaling inducesapoptosis and inhibits human MDA-MB-231 xenograft growth. Am JPathol 2007, 171:1023e1036

81. Sugimoto K, Maekawa Y, Kitamura A, Nishida J, Koyanagi A,Yagita H, Kojima H, Chiba S, Shimada M, Yasutomo K: Notch2signaling is required for potent antitumor immunity in vivo. JImmunol 2010, 184:4673e4678

82. Stacey DW: Cyclin D1 serves as a cell cycle regulatory switch inactively proliferating cells. Curr Opin Cell Biol 2003, 15:158e163

83. Ornitz DM, Itoh N: Fibroblast growth factors. Genome Biol 2001, 2.REVIEWS3005

84. Nakamura T, Nakamura T, Matsumoto K: The functions and possiblesignificance of Kremen as the gatekeeper of Wnt signalling indevelopment and pathology. J Cell Mol Med 2008, 12:391e408

85. Ahmadiyeh N, Pomerantz MM, Grisanzio C, Herman P, Jia L,Almendro V, He HH, Brown M, Liu XS, Davis M, Caswell JL,Beckwith CA, Hills A, Macconaill L, Coetzee GA, Regan MM,Freedman ML: 8q24 Prostate, breast, and colon cancer risk loci showtissue-specific long-range interaction with MYC. Proc Natl Acad SciU S A 2010, 107:9742e9746

86. Sotelo J, Esposito D, Duhagon MA, Banfield K, Mehalko J, Liao H,Stephens RM, Harris TJ, Munroe DJ, Wu X: Long-range enhancerson 8q24 regulate c-Myc. Proc Natl Acad Sci U S A 2010, 107:3001e3005

87. Meyer KB, Maia AT, O’Reilly M, Ghoussaini M, Prathalingam R,Porter-Gill P, Ambs S, Prokunina-Olsson L, Carroll J, Ponder BA:A functional variant at a prostate cancer predisposition locus at8q24 is associated with PVT1 expression. PLoS Genet 2011, 7:e1002165

88. Esseghir S, Kennedy A, Seedhar P, Nerurkar A, Poulsom R, Reis-Filho JS, Isacke CM: Identification of NTN4, TRA1, and STC2 asprognostic markers in breast cancer in a screen for signal sequenceencoding proteins. Clin Cancer Res 2007, 13:3164e3173

89. Liang H, Zhong Y, Huang Y, Chen G: Type 1 receptor parathyroidhormone (PTH1R) influences breast cancer cell proliferation andapoptosis induced by high levels of glucose. Med Oncol 2012, 29:439e445

90. Qiao Y, Jiang X, Lee ST, Karuturi RK, Hooi SC, Yu Q: FOXQ1regulates epithelial-mesenchymal transition in human cancers. CancerRes 2011, 71:3076e3086

91. Debily MA, Camarca A, Ciullo M, Mayer C, El Marhomy S, Ba I,Jalil A, Anzisi A, Guardiola J, Piatier-Tonneau D: Expression andmolecular characterization of alternative transcripts of the ARH-GEF5/TIM oncogene specific for human breast cancer. Hum MolGenet 2004, 13:323e334

92. Lee JC, Sharma M, Lee YH, Lee NH, Kim SY, Yun JS, Nam SY,Hwang PH, Jhee EC, Yi HK: Pax9 mediated cell survival in oralsquamous carcinoma cell enhanced by c-myb. Cell Biochem Funct2008, 26:892e899

93. Muehlich S, Wang R, Lee SM, Lewis TC, Dai C, Prywes R:Serum-induced phosphorylation of the serum response factorcoactivator MKL1 by the extracellular signal-regulated kinase 1/2pathway inhibits its nuclear localization. Mol Cell Biol 2008, 28:6302e6313

94. White KA, Yore MM, Deng D, Spinella MJ: Limiting effects ofRIP140 in estrogen signaling: potential mediation of anti-estrogeniceffects of retinoic acid. J Biol Chem 2005, 280:7829e7835

95. Liu Z, Ma H, Wei S, Li G, Sturgis EM, Wei Q: Telomere length andTERT functional polymorphisms are not associated with risk of

13

1612

22 August 2013 � 11:16 pm � EO: AJP13_0146

Page 14: Inherited Genetic Susceptibility to Breast Cancer

Ghoussaini et al

161316141615161616171618161916201621162216231624162516261627

162816291630163116321633163416351636163716381639164016411642

squamous cell carcinoma of the head and neck. Cancer EpidemiolBiomarkers Prev 2011, 20:2642e2645

96. Baird DM: Variation at the TERT locus and predisposition for cancer.Expert Rev Mol Med 2010, 12:e16

97. Kang JU, Koo SH, Kwon KC, Park JW, Kim JM: Gain at chromo-somal region 5p15.33, containing TERT, is the most frequent geneticevent in early stages of non-small cell lung cancer. Cancer GenetCytogenet 2008, 182:1e11

98. Zhang A, Maner S, Betz R, Angstrom T, Stendahl U, Bergman F,Zetterberg A, Wallin KL: Genetic alterations in cervical carcinomas:frequent low-level amplifications of oncogenes are associated withhuman papillomavirus infection. Int J Cancer 2002, 101:427e433

99. Gail MH: Discriminatory accuracy from single-nucleotide poly-morphisms in models to predict breast cancer risk. J Natl Cancer Inst2008, 100:1037e1041

14REV 5.2.0 DTD � AJPA1459_proof

100. Gail MH: Value of adding single-nucleotide polymorphism geno-types to a breast cancer risk model. J Natl Cancer Inst 2009, 101:959e963

101. Ghoussaini M, Pharoah PD: Polygenic susceptibility to breast cancer:current state-of-the-art. Future Oncol 2009, 5:689e701

102. Pharoah PD, Antoniou A, Bobrow M, Zimmern RL, Easton DF,Ponder BA: Polygenic susceptibility to breast cancer and implicationsfor prevention. Nat Genet 2002, 31:33e36

103. Pashayan N, Duffy SW, Chowdhury S, Dent T, Burton H, Neal DE,Easton DF, Eeles R, Pharoah P: Polygenic susceptibility to prostateand breast cancer: implications for personalised screening. Br JCancer 2011, 104:1656e1663

104. Burton H, Chowdhury S, Dent T, Hall A, Pashayan N, Pharoah P:Public health implications from COGS and potential for risk strati-fication and screening. Nat Genet 2013, 45:349e351

ajp.amjpathol.org - The American Journal of Pathology� 22 August 2013 � 11:16 pm � EO: AJP13_0146