Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
N Engl J Med. Author manuscript; available in PMC 2015 Dec 4.
Published in final edited form as:
PMCID: PMC4610139
NIHMSID: NIHMS711772
PMID: 26014596

Gene-Panel Sequencing and the Prediction of Breast-Cancer Risk

Associated Data

Supplementary Materials

Advances in sequencing technology have made multigene testing, or “panel testing,” a practical option when looking for genetic variants that may be associated with a risk of breast cancer. In June 2013, the U.S. Supreme Court1 invalidated specific claims made by Myriad Genetics with respect to the patenting of the genomic DNA sequence of BRCA1 and BRCA2. Other companies immediately began to offer panel tests for breast cancer genes that included BRCA1 and BRCA2. The subsequent flourishing of gene-panel testing services (Table 1, and Table S1 in the Supplementary Appendix, available with the full text of this article at NEJM.org) has generated much interest both within the clinical genetics community and in the popular press.2 These panels cover a total of more than 100 genes, and breast cancer is specifically mentioned as an indication for 21 of these genes. However, the fact that the technology is available does not necessarily mean that such tests are appropriate or desirable.

Table 1

Examples of Multigene Testing Panels for Breast Cancer.*

CompanyTestWebsiteGenes Included
Ambry GeneticsBreastNextwww.ambrygen.com/tests/breastnextATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1
CHEK2, MRE11A, MUTYH, NBN, NF1
PALB2, PTEN, RAD50, RAD51C, RAD51D
TP53
BreastHealth UKBreastGenewww.breasthealthuk.com/screening-services/genetic-testing/breastgeneATM, BRCA1, BRCA2, BRIP1, CDH1, CHEK2
PALB2, PTEN, STK11, TP53
CentogeneBreast Ovarian
Cancer Panel
www.centogene.com/centogene/centogene-test-catalogue.phpATM, BARD1, BRIP1, CDH1, CHEK2, MEN1
MLH1, MRE11A, MSH2, MSH6, MUTYH
NBN, PALB2, PMS1, PMS2, RAD50
RAD51C, RAD51D, XRCC2
Emory Genetics
Laboratory
High Risk Breast
Cancer Panel
http://geneticslab.emory.edu/tests/MM201PTEN, STK11, TP53
Fulgent DiagnosticsBreast Ovarian
Cancer NGS
Panel
http://fulgentdiagnostics.com/test/breast-ovarian-cancer-ngs-panel/APC, ATM, ATR, AXIN2, BAP1, BARD1, BLM
BMPR1A, BRCA1, BRCA2, BRIP1, CDH1
CDK4, CDKN2A, CHEK2, CTNNB1
EPCAM, FANCC, HOXB13, MLH1
MRE11A, MSH2, MSH6, MUTYH, NBN
PALB2, PALLD, PMS2, PTEN, RAD50
RAD51, RAD51C, RAD51D, SMAD4
STK11, TP53, VHL, XRCC2, XRCC3
GeneDxOncoGeneDxwww.genedx.com/test-catalog/available-tests/breastovarian-cancer-panelATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1
CHEK2, EPCAM, FANCC, MLH1, MSH2
MSH6, NBN, PALB2, PMS2, PTEN
RAD51C, RAD51D, STK11, TP53, XRCC2
IlluminaTruSight Cancerwww.illumina.com/clinical/translational_genomics/panels/kits.html94 Genes plus 287 SNPs reported to be asso-
ciated with risk of breast cancer
InvitaeHereditary Breast
Cancer, High-
Risk Panel
www.invitae.com/en/physician/panel-detail/PNL0009/BRCA1, BRCA2, CDH1, PALB2, PTEN, STK11
TP53
Myriad GeneticsmyRiskwww.myriad.com/products-services/hereditary-cancers/myrisk-hereditary-cancer/ATM, BARD1, BRCA2, BRIP1, CDH1, CHEK2
NBN, PALB2, PTEN, RAD51C, STK11,
TP53
CD GenomicsGenetic Testing for
the Cancer Suscep-
tibility
www.cd-genomics.com/GeneticvTesting-for-the-Cancer-Susceptibility.htmlNot specified
University of
Washington
BROCA – Cancer
Risk Panel
http://web.labmed.washington.edu/tests/genetics/BROCAAKT1, ATM, BARD1, BRCA1, BRCA2, BRIP1
CDH1, CHEK2, EPCAM, FAM175A
GEN1, MRE11A, MUTYH, NBN, PALB2
PIK3CA, PTEN, RAD50, RAD51C
RAD51D, STK11, TP53, XRCC2
*SNP denotes single-nucleotide polymorphism.
For Myriad Genetics and the University of Washington, only genes for which breast-cancer risk is given as an indication are listed. For a complete list, see Table S1 in the Supplementary Appendix. In several cases, the panels include additional genes, and several companies also offer larger panels. Thus, even if the primary purpose of the test is prediction of the risk of breast cancer, results will often be available (and need to be interpreted) for a larger set of genes than those listed here.

According to the framework proposed by the ACCE (established by the Centers for Disease Control and Prevention), genetic tests should be evaluated on the basis of the four criteria from which the name ACCE is derived: analytical validity, clinical validity, clinical utility, and ethical, legal, and social issues.3 Analytical validity refers to the degree of accuracy with which a test detects the presence or absence of a mutation. Here, however, we focus on the key question of clinical validity: Are the variants the test is intended to identify associated with disease risk, and are these risks well quantified? The validity of the risk estimates is a key determinant of the clinical utility of panel testing, which in turn should inform decisions regarding the adoption of the testing in clinical practice. We do not consider in detail who should undergo testing, what level of risk is associated with any given variant that might be considered clinically useful, or how that risk might be managed. However, broadly similar guidelines for managing the care of women with a family history of breast cancer exist in several countries (Table 2). These guidelines are based on the stratification of patients according to levels of risk and provide guidance on the identification of women to whom screening (by means of mammography or magnetic resonance imaging), risk-reducing medication, and risk-reducing surgery should be offered. These recommendations could be modified to reflect the identification of risk variants through the use of gene-panel testing. Whatever the recommendations for the management of care, the underpinnings of the guidelines should be based on reliable estimates of the risk of cancer.

Table 2

Screening and Treatment Guidelines for Carriers of BRCA1 or BRCA2 Mutations.*

Type of Screening
or Therapy
NCCN
(United States]
NICE
(United Kingdom)]
GC-HBOC–
(Germany)
eviQ Cancer Treatments Online
(Australia)
IKNL–KiMS
(Netherlands)
MammographyRecommended annually
at 30–75 yr of age, or
younger if woman in
family has received
breast-cancer diag-
nosis before 25 yr of
age and MRI is not
available
Recommended for con-
sideration annually at
30–39 yr of age; rec-
ommended once
yearly at 40–69 yr of
age and every 3 yr at
≥70 yr of age
Recommended every 1–2
yr at 49–69 yr of age if
breast density classi-
fied as ACR lor 2,
with ultrasonography
twice yearly
For BRCA1 carriers, recommended
annually at 30–50 yr of age, with
or without ultrasonography
For BRCA2 carriers, recommend-
ed annually at 30–50 yr of age,
with or without ultrasonography;
at >50 yr of age, mammography
recommended annually, with or
without ultrasonography, with
clinical breast examination; if di-
agnosis in family member <35 yr
of age, may recommend individu-
alized schedule
Recommended annually; because risk
of radiation-induced tumors is
greater in young women, first
mammogram recommended
at 30 yr of age
MRIRecommended annually
at 25–75 yr of age,
but earlier if younger
age of onset in any
family member
Recommended annually
at 30–49 yr of age un-
less breast density is
high, in which case
should be continued
until 70 yr of age
Recommended annually
at 25–69 yr of age if
breast density is clas-
sified as ACR >1
For BRCA1 carriers, recommended
annually at 30–50 yr of age, with
or without ultrasonography
For BRCA2 carriers, recommend-
ed annually at 30–50 yr of age,
with or without ultrasonography;
at >50 yr of age, mammography
recommended annually, with or
without ultrasonography, with
clinical breast examination; if di-
agnosis in family member <35 yr
of age, individualized schedule
may be recommended
Recommended annually, starting
25 yr of age
Preventive mastectomyNo definitive guideline,
but “degree of pro-
tection and risks”
should be discussed
No definitive guideline,
but discussions of
potential benefits of
surgery should take
current age into ac-
count
No definitive guideline,
but “degree of protec-
tion and risks” should
be discussed
If performed, recommended at
≤40 yr of age
Recommended at ≥25 yr of age; <5%
of patients are at risk of residual
breast cancer
Preventive oophorectomyIf performed, recom-
mended between 35
and 40 yr of age
No guidelineSalpingo-oophorectomy
recommended at ap-
proximately 40 yr of
age for BRCA1 carriers
and 45 yr of age for
BRCA2 carriers
If performed, recommended at
≤40 yr of age
If performed, recommended at≥35 yr
of age for BRCA1 carriers and ≥40
yr of age for BRCA2 carriers
Oral contraceptiveNo clear directiveNo clear directiveNo clear directiveCombination oral contraceptive not
contraindicated
No clear directive; recommended that
nonsystemic form of contraception
could be discussed
ChemopreventionNo clear directiveProvision of tamoxifen
recommended for
women at high risk
of breast cancer; but
BRCA1 vs. BRCA2
status not discussed
No guidelineRecommendation to consider with
professional on individualized
basis
No guideline
Hormonal therapyNo clear directiveNo guidelineNot indicated for carriers
of BRCA1 or BRCA2
unless preceded by
risk-reducing salpin-
go-oophorectomy
If risk-reducing salpingo-oophorecto-
my performed before meno-
pause, hormone-replacement
therapy should be considered un-
til onset of natural menopause
No guideline
Screening for other
cancers
Prostate-cancer screen-
ing recommended
for BRCA2 mutation
carriers ≥40 yr of age
(consider for BRCA1
mutation carriers)
No guidelineProstate-cancer screen-
ing recommended for
BRCA2 mutation car-
riers 45–50 yr of age
Consideration should be given to an-
nual testing for prostate-specific
antigen and digital rectal exami-
nations for prostate cancer from
approximately 40 yr of age
onward
No guideline
*A summary of guidelines available in the United States is provided by the National Cancer Institute; see Table 12.4 GC-HBOC-AGO denotes German Consortium for Hereditary Breast and Ovarian Cancer-German Gynecological Oncology Group, MRI magnetic resonance imaging, NCCN National Comprehensive Cancer Network, and NICE National Institute for Health and Care Excellence.
The NICE guidelines focus on familial breast cancer only, and they are not confined to the discussion of carriers of BRCA mutations.
ACR denotes American College of Radiologists, with ACR 1 indicating breast-tissue involution, ACR 2 scattered fibroglandular tissue, ACR 3 heterogeneously dense parenchyma, and ACR 4 extremely dense tissue composition.

Before these guidelines are developed, the appropriateness of the tests themselves needs to be considered. The determination of analytical validity for laboratory-developed diagnostic tests falls under the remit of the Clinical Laboratory Improvement Amendments (CLIA) of 1988, but neither clinical validity nor clinical utility is part of the assessment process. Therefore, whereas new drugs without clinical utility will not be approved by the Food and Drug Administration (FDA), gene-panel tests can be adopted without any review of data regarding their clinical utility.5,6 Recent commentaries have suggested ways in which the FDA might become involved in the approval of genomic tests.7, 8 Although we acknowledge the enormity of the task, we propose that a genomic test should not be offered until its clinical validity has been established. We consider below some of the key issues that need to be addressed. Others have argued that establishing clinical validity is a postmarketing pursuit,8 but we believe that failing to require the clinical validation of genomic tests before they are submitted for regulatory approval is likely to lead to substantial misuse of the technology.

Key Issues and General Principles

Several key questions must be addressed in order to establish clinical validity. First, are variants in the gene associated with breast-cancer risk-Second, which variants, or classes of variants, are associated with risk? Third, what is the magnitude of those risks? Fourth, what methods have been used to estimate those risks? We will concentrate on the genes in which rare variants have been proposed to confer a moderate or high risk of breast cancer. For the purpose of this review, we define moderate risk as a risk of breast cancer, defined in terms of disease incidence, that is two to four times as high as that in the general population and high risk as an incidence that is more than four times as high.9 We leave aside the separate question of risk prediction in which profiling based on the genotyping of common polymorphisms is used (see box). We will restrict our attention to the prediction of risk in women unaffected by breast cancer, although somewhat analogous issues apply to testing in affected women. We focus on the question of breast-cancer risk, but similar considerations apply to other cancers. Indeed, some of the genes considered here also confer a predisposition to ovarian cancer, pancreatic cancer, and other cancers, and some of the available panels also include genes putatively involved in a wider range of cancers (Table 3, and Table S2 in the Supplementary Appendix). We leave aside the use of panel testing for the identification of cancer syndromes and for the management of disease in women who have cancer.

Table 3

Genes for Which an Association between Protein-Truncating Variants and Breast-Cancer Risk Has Been Established.

GeneMagnitude of Relative
Risk Associated with
Truncating Variants*
Risk
Associated
with
Missense
Variants
Estimated
Relative Risk
(90% Cl)
P ValueAbsolute
Risk by 80
Yr of Age§
CommentsOther Associated CancersReferences
ModerateHigh
%
BRCA1YesYesYes11.475Estimates are based on the
BOADICEA model for a
woman born in 1960
OvaryAntoniou et al.,10 Lee et
al.,11 Chen and Parmi-
giani,12 Mavaddat et
al.13

BRCA2YesYesYes11.776Estimates are based on the
BOADICEA model for a
woman born in 1960;
p.Lys3326Ter in the car-
boxyl terminus is associ-
ated with a lower increase
in risk
Ovary, prostate, pancreasAntoniou et al.,10 Lee
et al.,11 Chen and
Parmigiani,12
Mavaddat et al.13

TP53YesYesYes105 (62–165)Most published risk esti-
mates are subject to as-
certainment bias
Childhood sarcoma, adreno-
cortical carcinoma, brain
tumors
Hisada et al.,14 Hwang
et al.15

PTENUnknownUnknownYesNo reliable
estimate
Published risk estimates are
subject to ascertainment
bias
Thyroid, endometrial
cancer
Bubien et al.,16 Tan
etal.17

CDH1LikelyUnknownUnknown6.6 (2.2–19.9)0.00453Specific to lobular breast
cancer
Diffuse gastric cancerPharoah et al.18

STK11UnknownUnknownUnknownNo reliable
estimate**
Published risk estimates are
subject to ascertainment
bias
Colon, pancreas, ovarian sex
cord-stromal tumors
Hearle et al.19

NF1LikelyUnlikelyUnknown2.6 (2.1–3.2)2.3×l0−1326Estimates are based on co-
hort studies of patients
with neurofibromatosis
type 1††
Malignant tumors of periph-
eral nerve sheath, brain,
central nervous
system
Madanikia et al.,20
Seminog and
Goldacre21

PALB2LikelyUnknownUnknown5.3 (3.0–9.4)4×l0−1045Estimates are based on a me-
ta-analysis of published
case-control and family
studies
PancreasAntoniou et al.,22
Heikkinen et al.,23
Rahman et al.,24
Erkko et al.25

ATMLikelyUnlikelyYes2.8 (2.2–3.7)5×10−1127The p.Val2424Gly variant is
associated with higher risk
than truncating variants
PancreasRenwick et al.,26
Thompson et al.,27
Janin et al.,28 Olsen
et al.29

CHEK2LikelyUnlikelyYes3.0 (2.6–3.5)8×l0−3729Most data for truncating vari-
ants are limited to the vari-
ant c.1100delC; p.Ile157Thr
is associated with an in-
crease in risk that is 1.3
times as high as in the
general population
Lung, although p.Ile157Thr
is associated with reduced
risk
Meijers-Heijboer et
al.,30 CHEK2 Breast
Cancer Case-
Control
Consortium,31
Weischer et al.,32
Kilpivaara et al.33

NBNLikelyUnlikelyUnknown2.7 (1.9–3.7)5×l0−723Almost all data pertain to the
c.657del5 variant in Slavic
populations
UnknownZhang et al.34
*Moderate risk is defined as an average increase that is two to four times as high as that in the general population (on the basis of disease incidence) and high risk as an increase that is more than four times as high. When a quantitative analysis has been performed, “likely” indicates that the lower 90% confidence limit on the relative-risk estimate exceeds the threshold, and “unlikely” indicates that the upper 90% confidence limit on the relative-risk estimate is lower than the threshold.
The majority of missense variants have not been shown to be associated with risk.
Estimates were obtained from the Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm (BOADICEA) risk model for BRCA1 and BRCA2, from a single study for TP53 and CDH1, and from a meta-analysis of multiple studies for the other genes (see the Methods section and Table S3 in the Supplementary Appendix for further details). Note that there is evidence that relative risk declines with age for carriers of mutations in BRCA1,10 CHEK2,11 and ATM27 the evidence for PALB222 is weaker. These represent average relative risks for all ages and may therefore underestimate the relative risk of breast cancer for younger persons and overestimate the relative risk for older persons. The estimates relate for protein-truncating variants, except as noted (see the Methods section in the Supplementary Appendix).
§Absolute risks are risks in the absence of other causes of death. Adjusted estimates that allow for competing mortality will be lower, especially when the risk of other cancers is high (e.g., in BRCA1 and BRCA2). Unless otherwise indicated, the values shown were estimated by applying the estimated relative risks to breast-cancer incidence rates in England for 2003 through 2007.36
Most pathogenic mutations in TP53 are missense variants.
In PTEN mutation carriers, relative risks of breast cancer of 39.1 (90% CI, 26.7 to 54.9) and 25.4 (90% CI, 20.6 to 30.8) have been reported in two studies.16,17 However, estimates were based on selected families with the Cowden syndrome or related syndromes, resulting in an overestimate of risk.
**The cumulative breast-cancer risk for women with the Peutz-Jeghers syndrome has been reported to be 45% by 70 years of age (90% CI, 29–64).19 However, this estimate did not allow for ascertainment, which would result in an overestimate of risk even in families at high risk for disease. Furthermore, the data included patients with the Peutz-Jeghers syndrome in whom no STK11 variant had been identified.
††Risk estimates are based on follow-up data from patients with neurofibromatosis type 1, which is caused by both truncating and missense mutations in NF1 (although the majority are protein-truncating mutations). There are no published risk estimates for mutations in NF1 according to mutation type.

Common versus Rare Variants

Approximately 100 independent common variants (consisting primarily of single-nucleotide polymorphisms [SNPs]) associated with breast-cancer risk have been identified through large-scale genotyping studies. These variants typically have minor allele frequencies higher than 1%, and all confer risks that are less than 1.5 times as high as those in the general population; almost all these polymorphisms occur in noncoding sequences. Some commercial genetic panels include a subset of these SNPs. Thus, at present, there is a reasonably clear distinction between SNPs that confer a small increased susceptibility to breast cancer and variants that confer a moderate-to-high susceptibility as identified through sequencing. However, some sequence variants located in genes classified as conferring a high or moderate risk confer risks that fall below the threshold for moderate risk (i.e., two times as high as that in the general popula-tion). Examples include BRCA2 p.Lys3326Ter and CHEK2 p.Ile157Thr. Use of the term “low risk” for variants conferring a risk that is less than moderate is widespread, but it is not a particularly helpful term for counseling purposes, since carriers of such variants are still at an elevated level of risk.

Types of Genetic Variants

Most panel testing involves identifying the coding sequences and splice junctions of the genes of interest, often in combination with alternative methods used for the detection of large genomic rearrangements.35 Most of the variants identified are single-base substitutions and small insertions or deletions (indels). We refer to all nonsense substitutions, frameshift indels, and variants affecting splicing as protein-truncating variants. For the large majority of genes, most of the evidence on breast-cancer risk relates to protein-truncating variants assumed to result in loss of function.

Statistical Significance and Burden Tests

It is important to establish stringent levels of statistical significance. Although it would be ideal to have specific evidence for every variant detected, most variants for which there is a suspicion of association with a high risk of disease are rare, and the sample sizes required to establish allele-specific associations with risk are so large as to make the task infeasible. Consequently, some form of burden testing is frequently used in which the association between carrying any variant in a specific class and the risk of disease is evaluated. A potential problem with this method is that it does not indicate whether any specific variant identified is associated with disease. It is often assumed that all protein-truncating variants are equally pathogenic; however, all such variants do not confer the same risks. For missense variants, the situation is even more problematic.

Strength of Statistical Evidence for Association

The issue of what constitutes appropriate levels of significance for targeted sequencing has not been extensively discussed. An exomewide significance level of P<2.5×l0−6 is often used for whole-exome studies (calculated on the basis of a Bonferroni correction for approximately 20,000 genes). Since most genes associated with susceptibility to breast cancer are involved in DNA repair (a class involving fewer than 500 genes), more liberal significance levels (on the order of P<0.0001) might be appropriate for genes in this pathway. The use of Bayesian arguments leads to similar thresholds (see the Methods section in the Supplementary Appendix). Although these significance thresholds may be appropriate for a single burden test, more stringent thresholds would be required for calculations involving individual variants. A related question is the precision of the risk estimate. It is clearly undesirable to give a patient an estimate of risk that may be subject to substantial change when additional data are acquired. For the purposes of this review, we consider it to be likely that a given risk will be above (or below) a certain threshold if the 90% confidence limit on the risk estimate exceeds (or is less than) the threshold.

Definition of Risk

Our estimates are presented primarily in terms of average relative risks. We recognize that for purposes of counseling, absolute estimates of risk (projected over a few years or a lifetime) are more useful. However, most studies report estimates of relative risk rather than absolute risk, and absolute risks are more strongly influenced by risk factors for breast cancer, such as a family history of breast cancer, age at menopause, and breast density on mammography. In the case of a rare variant conferring a relative risk of 2 or 4, the corresponding absolute risks of breast cancer would be approximately 18% and 32%, respectively, by the time a patient reached 80 years of age (according to recent U.K. incidence rates),36 in the absence of other causes of death. These risks approximately correspond to the definitions of moderate and high risk familiar to the clinical genetics community.4

It follows that the identification of a variant conferring a relative risk higher than 4, in the absence of any other data, can place a woman in the high-risk category. In contrast, a variant conferring a relative risk of 2 to 4 will place a woman in the high-risk category only if her risk is increased by other factors. For some genes (notably, BRCA1,10 CHEK2,31 and ATM27), there is evidence that the rate ratio declines with age. The published overall relative risk estimates can thus provide a misleading estimate of lifetime risk. Ideally, age-specific estimates are required, but the data available on risks for older women are often limited.

Study Design

Appropriate study design is critical for both the identification of disease-associated alleles and the derivation of reliable risk estimates. Several study designs are available (Table 4). The use of case–control studies for risk estimation involving rare variants can be problematic; family-based methods, including kin–cohort designs and cosegregation analysis, provide alternatives, but these methods also have pitfalls. Furthermore, many studies are based on a few variants that are restricted to specific populations; although it is generally assumed that the risk estimates associated with different truncating variants observed in other populations are similar, it is usually impossible to test this assumption.

Table 4

Study Designs for Estimating the Risks Associated with Rare Variants.

MethodDescriptionAdvantagesDisadvantagesExample
Population-based
case-control
Screening is conducted for vari-
ants in unselected cases of
disease and population-
matched controls
Provides direct estimates of the
relative risk (odds ratio) and is
not biased by other familial
factors
Must be very large since variants are typically rare; bias-
es arise if controls are not appropriately population-
matched (there are large differences in allele fre-
quency among populations) Requires that the same
assay techniques are used for cases and controls to
provide valid tests and estimates, typically with
screening of the full coding sequences in all cases
and controls; large biases may arise if only the vari-
ants identified in the cases are tested in the controls

Requires that the same assay techniques are used for
cases and controls to provide valid tests and esti-
mates, typically with screening of the full coding se-
quences in all cases and controls; large biases may
arise if only the variants identified in the cases are
tested in the controls
CHEK231
Family-based
case-control
In theses case-control studies,
cases are enriched by family
histories
Improves power because of the
higher frequency of variants in
familial cases
Is subject to biased risk estimates
Efforts to correct bias depend on use of additional
assumptions about the modifying effects of other
familial factors
CHEK230
Kin cohortData on cancer occurrence in rel-
atives of carriers in popula-
tion-based series are used to
estimate risks with maxi-
mum-likelihood methods37
Provides estimates without the need
to screen controls; genotype
data in relatives can be incorpo-
rated but are not required
Is limited by the accuracy of the family history; risks
may be overestimated if familial factors are not ac-
counted for
BRCA1/2, PALB210,22
Segregation in
families
Can be applied in families that have
been selected for a strong family
history; controls are not required
Requires samples on multiple persons from the same
family; power is typically very limited
CHEK230
Prospective cohortProvides direct estimates of abso-
lute risk
Requires long-term investment; is prohibitively large
except in the case of high-risk variants; risk esti-
mates may altered by interventions (e.g., prophylac-
tic surgery); risk estimates are affected by other fa-
milial factors
BRCA1/213

Overestimation of Risk

The problems of publication bias, in which negative studies are not published, and winner’s curse, whereby an initial study identifying an association tends to overestimate the risk, should be noted.38 Furthermore, many gene-discovery studies oversample for early-onset cases of disease or cases with a family history. This approach improves power but leads to seriously biased risk estimates unless the ascertainment is allowed for in the analysis. Moreover, risk estimates based on data from highly selected families may not reflect the true “average” risk for all carriers of pathogenic variants, because such biased sampling results in a selection of individuals that are not random with respect to other modifiers of risk.

Evidence of Association for Specific Genes

Here we review several genes for which some evidence of an association with breast cancer has been reported. A summary of the genes for which an association with breast cancer has, in our view, been established is given in Table 3. See Table S2 in the Supplementary Appendix for a list of genes for which an association with breast cancer has been suggested but not established and Table S3 for a summary of the studies used to derive estimates of breast-cancer risk. The Methods section in the Supplementary Appendix summarizes the methods used to derive summary estimates of risk.

BRCA1 and BRCA2

The clinical validity and utility of testing for variants in BRCA1 and BRCA2 are well established. There is overwhelming evidence that most protein-truncating variants in these genes are associated with a high risk of breast cancer and other cancers.10,12,13 Even among protein-truncating variants, however, variant-specific differences in risk have been observed.39 Furthermore, a polymorphic nonsense variant at the carboxyl terminus of BRCA2, p.Lys3326Ter, has been reported to be associated with a relative risk of breast cancer of 1.4 (90% confidence interval [CI], 1.2 to 1.7),40 which is substantially lower than the risks conferred by more proximal truncating variants (Table 3).

TP53, CDH1, PTEN, STK11, and NF1

Mutations in TP53 , CDH1, PTEN, STK11, and NF1 cause pleiotropic tumor syndromes in which breast cancer is only one feature. Germline mutations in TP53 (both protein-truncating and missense mutations) are responsible for the Li– Fraumeni syndrome, in which carriers are predisposed to childhood sarcomas, brain tumors, adrenocortical carcinoma, and other rare cancers, in addition to breast cancer.41 Although the association with breast cancer is not controversial, reliable estimates of risk are lacking; most studies are based on pedigrees in which family members have features of the Li–Fraumeni syndrome and thus are subject to ascertainment bias. However, a study based on carriers of a TP53 mutation identified through probands with childhood sarcoma has also reported a high risk of breast cancer.15 Similar ascertainment biases apply to mutations in PTEN and STK11. Mutations in PTEN are associated with the Cowden syndrome, in which breast cancer is a characteristic of the clinical phenotype,16,17,42 and mutations in STK11 are associated with the Peutz– Jeghers syndrome and an increased risk of breast cancer.19 Protein-truncating variants in CDH1, which are known to be associated with diffuse-type gastric cancer, are also thought to be associated with an increased risk of breast cancer (specifically, the lobular subtype), with a reported relative risk of 6.6 (90% CI, 2.2 to 19.9; P = 0.004).18 Recent cohort studies20,21 have reported an elevated risk of breast cancer in women with neurofibromatosis type 1 (odds ratio, 2.6; 90% CI, 2.1 to 3.2).

PALB2, CHEK2, ATM, NBN, and Related Genes

There is strong evidence that protein-truncating variants in four other genes involved in DNA repair confer an increased risk of breast cancer. Among these genes, mutations in PALB2 appear to confer the highest risks. A large family-based study estimated the risk of breast cancer to be approximately six times as high among carriers as compared with noncarriers,22 although two case–control studies based on the Finnish founder variant, c.1592delT, estimated somewhat lower risks.23,25 In a meta-analysis of these estimates the combined relative risk was 5.3 (90% CI, 3.0 to 9.4). Thus, although PALB2 mutations may fall into the high-risk category (in which the risk of cancer is more than four times as high as that in the general population), the confidence limits are too wide to be certain. Most of the data for CHEK2 relate to the c.1100delC variant, which is found fairly frequently in Northern European populations.30 On the basis of two large case– control analyses, we calculated an estimated relative risk of breast cancer of 3.0 (90% CI, 2.6 to 3.5).31,32 Truncating variants in ATM have been evaluated in both case–control studies (with selected cases)26 and cohort studies of relatives of patients with ataxia–telangiectasia.2729 In a meta-analysis of the three largest cohort studies of relatives of patients with ataxia–telangiectasia, the estimated relative risk of breast cancer was 2.8 (90% CI, 2.2 to 3.7; P = 4.7×10−11), a value similar to that for truncating variants in CHEK2.

In NBN, one protein-truncating variant, c.657del5, is sufficiently common in some Eastern European populations to allow its evaluation in a case–control study. A meta-analysis of 10 studies reported strong evidence of an association with breast-cancer risk for this variant (summary relative risk, 2.7; 90% CI, 1.9 to 3.7; P = 5×10−7).34 More limited evidence is available for two other DNA-repair genes, MRE11A and RAD50, which encode proteins that form an evolutionarily conserved complex with NBN.4348

Mutations in three other DNA repair genes, RAD51C, RAD51D, and BRIP1, have shown clear evidence of an association with ovarian cancer.4953 However, in each case, the evidence for association with breast cancer is limited. Recent exome studies and targeted sequencing studies have suggested that breast cancer is associated with deleterious variants in FANCC,54 FANCM,55 and XRCC2.56 In none of these instances, however, does the evidence reach the threshold level (P<0.0001) that we propose for DNA-repair genes. The recent findings of deleterious mutations in RECQL in women with a strong family history of breast cancer, however, suggests that this gene confers susceptibility to breast cancer.57,58

Other Genes

The panels currently marketed for the prediction of risk of cancer contain many other genes, most of which have been included by virtue of their relevance to rare mendelian cancer syndromes. Variants in some of these genes may also be associated with breast cancer. Mutations in DNA mismatch-repair genes (MLH1, MSH2, MSH6, and PMS2) may be associated with breast cancer, but in a recent review, Win et al.59 concluded that the evidence was equivocal. It has also been suggested that MUTYH variants that confer a predisposition to polyposis colorectal cancer may confer a predisposition to breast cancer, but a recent case–control study reported no association.60 Another recent study suggested that carriers of MEN1 mutations may be at increased risk for breast cancer.61 A recent case–control study has reported an association between rare variants in PPM1D and breast cancer.62 However, this association does not reach our proposed significance threshold, and, in addition, the sequence variants are observed as mosaics in lymphocytes and are not inherited. There is currently no clear evidence of an association between breast cancer and any other gene.

Missense Variants

With the exception of TP53, the assessment of the risk of breast cancer from missense variants is much more problematic than it is for protein-truncating variants. Some missense variants in specific domains of BRCA1 and BRCA2 confer high risks of breast and ovarian cancer, but the great majority do not.63,64 For these genes, algorithms based on conservation, pedigree data, and analysis of tumor subtype can be used to predict the pathogenicity of some variants.63,65,66 Similar considerations may apply to ATM and CHEK2 — missense variants falling in key functional domains and at positions that show a high degree of species conservation are more likely to be associated with increased risk.67 However, even for BRCA1 and BRCA2, the breast-cancer risk associated with the large majority of missense variants remains unknown; such variants are referred to as variants of unknown significance. Moreover, clearly pathogenic missense variants need not be associated with the same risk as truncating variants. For example, the CHEK2 missense variant p.Ile157Thr confers a lower risk of breast cancer than the CHEK2 c.1100delC truncating variant,33 whereas ATM p.Val2424Gly appears to be associated with a higher risk of breast cancer than truncating variants (8.0; 90% CI, 2.8 to 22.5; P = 0.0005).68 A more systematic approach to this problem would involve defining risks on the basis of variant classes that are defined through prediction algorithms based on in silico data. However, even though existing data provide good evidence that missense variants falling at highly conserved positions in several genes confer disease risk, and that such variants may make an important contribution to the heritability of breast cancer,69 no system has been established for use in the classification of variants that would allow such estimates of risk to be used clinically.

Risk Modifiers and Absolute Risks

For the purposes of genetic counseling, relative risks need to be converted into absolute risks. For an “average” mutation carrier, absolute risks can be calculated in a straightforward manner by combining the estimated relative risk with population incidence rates. The results are illustrated in Figure 1 for carriers of mutations in PALB2 and CHEK2.

An external file that holds a picture, illustration, etc.
Object name is nihms711772f1.jpg
Predicted Cumulative Risk of Breast Cancer for a Carrier of a Deleterious Mutation in PALB2 and for a Deleterious Mutation in CHEK2

Solid red lines represent summary estimates, and red dashed lines the upper and lower 90% confidence limits. The absolute risks were estimated by applying the estimates of relative risk to the rates for the incidence of breast cancer in England from 2003 through 2007 (obtained from the database Cancer Incidence in Five Continents, volume X).36 The solid blue lines represent the cumulative risks according to these population incidence rates (i.e., corresponding to a relative risk of 1). Estimates ignore competing mortality (i.e., they represent the cumulative risks in the absence of death from another cause). The dashed horizontal black lines represent lifetime risks that are twice and four times as high as the population average. Thus, a “typical” carrier of the CHEK2 mutation is likely to fall into the category of moderate risk. The best estimate for carriers of the PALB2 mutation places them in the high-risk category, but the confidence interval for the estimate is such that their risk may be moderate. These estimates constitute average cumulative risks (for a woman not selected for other risk factors) and are modified by other risk factors, including family history.

However, the calculation of the absolute risk associated with a given variant must also account for the risk associated with other genetic factors, lifestyle, and family history. There is strong evidence that the absolute risk of breast cancer in carriers of BRCA1, BRCA2, PALB2, and CHEK2 mutations is higher among women with a strong family history of breast cancer.10,22,30,70 It has also been shown that the absolute risk of breast cancer in carriers of BRCA1 and BRCA2 mutations depends on the risks associated with their single-nucleotide–polymorphism (SNP) profile.71 A broader question is that of how the risks associated with genetic variants should be combined with risk factors associated with lifestyle. Several studies indicate that the risks associated with common SNPs and other risk factors combine in a multiplicative rather than an additive fashion,7274 and it would be reasonable to assume that rare variants combine with other risk factors in a similar manner. The evidence regarding the combined effects of genetic and lifestyle factors is both limited and conflicting for variants in BRCA1 and BRCA2,75 and no evidence is available for other genes. In addition, absolute risks need to be adjusted for competing risks in analyses of mortality, a factor that may be important in to our understanding of genes associated with cancers other than breast cancer.

Almost all the available data relate to women of European ancestry. At present, it is unclear whether the available estimates of relative risk can be safely extrapolated to women of other ancestries or to populations with different incidences of breast cancer.

Conclusions

We have discussed some of the difficulties of assigning risk to rare variants and reviewed the genes for which the evidence of association with breast cancer is sufficiently robust to be incorporated into personalized risk prediction. Variants that are predicted to truncate BRCA1 and BRCA2 (together with a subset of missense variants) confer a high risk of breast cancer; PALB2 and perhaps PTEN may also fall in this category, but the evidence is insufficient to place them confidently in the category of high risk rather than moderate risk. For TP53, both missense and protein-truncating variants are associated with substantially increased risks of breast cancer. Genes that fall into the category of moderate risk (for which fully deleterious mutations confer a risk of breast cancer that is two to four times as high as that in the general population) include CHEK2, ATM, and NF1. There is clear evidence for an association with risk of cancer for STK11, CDH1, and NBN, but the risk estimates are too imprecise for categorization. Estimates of risk for PTEN, STK11, and CDH1 are derived entirely from studies of selected patients identified through specialized clinics and may be seriously overestimated. We found insufficient evidence to establish any other genes as conferring a predisposition for breast cancer and would caution against their use in the prediction of breast-cancer risk. As the costs of sequencing decline, it is inevitable that the use of gene-panel testing, and indeed whole-exome and whole-genome sequencing, will become widespread. Therefore, there is an urgent need for much larger, well-designed population- and family-based studies in diverse populations that will provide reliable estimates of risk for the purpose of counseling. The systematic collection of data from ongoing use of panel testing linked to the epidemiologic and clinical data may also make an important contribution. Other genes that convey susceptibility to breast cancer (and perhaps rarer variants in noncoding sequences) will probably be identified and may be added to genetic-testing panels. Panel testing can make a useful contribution to prediction of a woman’s risk of breast cancer, but end users need to be aware of the limitations of these panels.

Supplementary Material

Supplement1

Acknowledgments

Supported by grants from Cancer Research UK (A11174, to Dr. Antoniou), the National Institutes of Health (CA116167, CA176785, and CA192393, to Dr. Couch), the Breast Cancer Research Foundation (to Drs. Couch, Nathanson, and Robson), Susan G. Komen (to Drs. Foulkes, Chenevix-Trench, and Domchek), the Cancer Research Society–Quebec Breast Cancer Foundation (to Dr. Foulkes), the National Health and Medical Research Council (to Dr. Chenevix-Trench), and the Basser Research Center (to Dr. Domchek).

We thank Paul James, Ingrid Winship, and Christi van Asperen for assistance with the summary of clinical guidelines.

Footnotes

Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.

References

1. Assoc. for Molecular Pathology v. Myriad Genetics, Inc. 569 U.S. 2013 ( https://supreme.justia.com/cases/federal/us/569/12-398).
3. Haddow JE, Palomaki GE. ACCE: a model process for evaluating data on emerging genetic tests. In: Khoury MJ, Little J, Burke W, editors. Human genome epidemiology: a scientific foundation for using genetic information to improve health and prevent disease. Oxford, United Kingdom: Oxford University Press; 2003. pp. 217–233. [Google Scholar]
4. National Institute for Health and Care Excellence. Familial breast cancer: classification and care of people at risk of familial breast cancer and management of breast cancer and related risks in people with a family history of breast cancer. London: National Collaborating Center for Cancer; 2013. ( http://www.cancer.gov/types/breast/hp/breast-ovarian-genetics-pdq#section/all). [PubMed] [Google Scholar]
5. Hayes DF, Allen J, Compton C, et al. Breaking a vicious cycle. Sci Transl Med. 2013;5 cm6. [PubMed] [Google Scholar]
6. Sharfstein J. FDA regulation of laboratory-developed diagnostic tests: protect the public, advance the science. JAMA. 2015;313:667–668. [PubMed] [Google Scholar]
7. Lander ES. Cutting the Gordian helix — regulating genomic testing in the era of precision medicine. N Engl J Med. 2015;372:1185–1186. [PubMed] [Google Scholar]
8. Evans BJ, Burke W, Jarvik GP. The FDA and genomic tests — getting regulation right. N Engl J Med. 2015;372:2258–2264. [PMC free article] [PubMed] [Google Scholar]
9. Hollestelle A, Wasielewski M, Martens JW, Schutte M. Discovering moderate-risk breast cancer susceptibility genes. Curr Opin Genet Dev. 2010;20:268–276. [PubMed] [Google Scholar]
10. Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case series unselected for family history: a combined analysis of 22 studies. Am J Hum Genet. 2003;72:1117–1130. [PMC free article] [PubMed] [Google Scholar]
11. Lee AJ, Cunningham AP, Kuchenbaecker KB, Mavaddat N, Easton DF, Antoniou AC. BOADICEA breast cancer risk prediction model: updates to cancer incidences, tumour pathology and Web interface. Br J Cancer. 2014;110:535–545. [PMC free article] [PubMed] [Google Scholar]
12. Chen S, Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance. J Clin Oncol. 2007;25:1329–1333. [PMC free article] [PubMed] [Google Scholar]
13. Mavaddat N, Peock S, Frost D, et al. Cancer risks for BRCA1 and BRCA2 mutation carriers: results from prospective analysis of EMBRACE. J Natl Cancer Inst. 2013;105:812–822. [PubMed] [Google Scholar]
14. Hisada M, Garber JE, Fung CY, Fraumeni JF, Jr, Li FP. Multiple primary cancers in families with Li-Fraumeni syndrome. J Natl Cancer Inst. 1998;90:606–611. [PubMed] [Google Scholar]
15. Hwang SJ, Lozano G, Amos CI, Strong LC. Germline p53 mutations in a cohort with childhood sarcoma: sex differences in cancer risk. Am J Hum Genet. 2003;72:975–983. [PMC free article] [PubMed] [Google Scholar]
16. Bubien V, Bonnet F, Brouste V, et al. High cumulative risks of cancer in patients with PTEN hamartoma tumour syndrome. J Med Genet. 2013;50:255–263. [PubMed] [Google Scholar]
17. Tan MH, Mester JL, Ngeow J, Rybicki LA, Orloff MS, Eng C. Lifetime cancer risks in individuals with germline PTEN mutations. Clin Cancer Res. 2012;18:400–407. [PMC free article] [PubMed] [Google Scholar]
18. Pharoah PD, Guilford P, Caldas C. Incidence of gastric cancer and breast cancer in CDH1 (E-cadherin) mutation carriers from hereditary diffuse gastric cancer families. Gastroenterology. 2001;121:1348–1353. [PubMed] [Google Scholar]
19. Hearle N, Schumacher V, Menko FH, et al. Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin Cancer Res. 2006;12:3209–3215. [PubMed] [Google Scholar]
20. Madanikia SA, Bergner A, Ye X, Blakeley JO. Increased risk of breast cancer in women with NF1. Am J Med Genet A. 2012;158A:3056–3060. [PMC free article] [PubMed] [Google Scholar]
21. Seminog OO, Goldacre MJ. Age-specific risk of breast cancer in women with neurofibromatosis type 1. Br J Cancer. 2015;112:1546–1548. [PMC free article] [PubMed] [Google Scholar]
22. Antoniou AC, Casadei S, Heikkinen T, et al. Breast-cancer risk in families with mutations in PALB2. N Engl J Med. 2014;371:497–506. [PMC free article] [PubMed] [Google Scholar]
23. Heikkinen T, Kärkkäinen H, Aaltonen K, et al. The breast cancer susceptibility mutation PALB2 1592delT is associated with an aggressive tumor phenotype. Clin Cancer Res. 2009;15:3214–3222. [PubMed] [Google Scholar]
24. Rahman N, Seal S, Thompson D, et al. PALB2, which encodes a BRCA2-interacting protein, is a breast cancer susceptibility gene. Nat Genet. 2007;39:165–167. [PMC free article] [PubMed] [Google Scholar]
25. Erkko H, Xia B, Nikkilä J, et al. A recurrent mutation in PALB2 in Finnish cancer families. Nature. 2007;446:316–319. [PubMed] [Google Scholar]
26. Renwick A, Thompson D, Seal S, et al. ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet. 2006;38:873–875. [PubMed] [Google Scholar]
27. Thompson D, Duedal S, Kirner J, et al. Cancer risks and mortality in heterozygous ATM mutation carriers. J Natl Cancer Inst. 2005;97:813–822. [PubMed] [Google Scholar]
28. Janin N, Andrieu N, Ossian K, et al. Breast cancer risk in ataxia telangiectasia (AT) heterozygotes: haplotype study in French AT families. Br J Cancer. 1999;80:1042–1045. [PMC free article] [PubMed] [Google Scholar]
29. Olsen JH, Hahnemann JM, Børresen-Dale AL, et al. Breast and other cancers in 1445 blood relatives of 75 Nordic patients with ataxia telangiectasia. Br J Cancer. 2005;93:260–265. [PMC free article] [PubMed] [Google Scholar]
30. Meijers-Heijboer H, van den Ouweland A, Klijn J, et al. Low-penetrance susceptibility to breast cancer due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 mutations. Nat Genet. 2002;31:55–59. [PubMed] [Google Scholar]
31. CHEK2 Breast Cancer Case-Control Consortium. CHEK2*1100delC and susceptibility to breast cancer: a collaborative analysis involving 10,860 breast cancer cases and 9,065 controls from nine studies. Am J Hum Genet. 2004;74:1175–1182. [PMC free article] [PubMed] [Google Scholar]
32. Weischer M, Nordestgaard BG, Pharoah P, et al. CHEK2*1100delC heterozygosity in women with breast cancer associated with early death, breast cancer-specific death, and increased risk of a second breast cancer. J Clin Oncol. 2012;30:4308–4316. [PMC free article] [PubMed] [Google Scholar]
33. Kilpivaara O, Vahteristo P, Falck J, et al. CHEK2 variant I157T may be associated with increased breast cancer risk. Int J Cancer. 2004;111:543–547. [PubMed] [Google Scholar]
34. Zhang G, Zeng Y, Liu Z, Wei W. Significant association between Nijmegen breakage syndrome 1 657del5 polymorphism and breast cancer risk. Tumour Biol. 2013;34:2753–2757. [PubMed] [Google Scholar]
35. Hogervorst FB, Nederlof PM, Gille JJ, et al. Large genomic deletions and duplications in the BRCA1 gene identified by a novel quantitative method. Cancer Res. 2003;63:1449–1453. [PubMed] [Google Scholar]
36. Cancer incidence in five continents. Vol. X. Lyon, France: International Agency for Research on Cancer; 2013. ( http://www.iarc.fr/en/publications/pdfs-online/epi/sp164/index.php). [Google Scholar]
37. Easton DF, Hopper JL, Thomas DC, et al. Breast cancer risks for BRCA1/2 carriers. Science. 2004;306:2187–2191. [PubMed] [Google Scholar]
38. Kraft P. Curses — winner’s and otherwise — in genetic epidemiology. Epidemiology. 2008;19:649–651. [PubMed] [Google Scholar]
39. Thompson D, Easton D. Variation in cancer risks, by mutation position, in BRCA2 mutation carriers. Am J Hum Genet. 2001;68:410–419. [PMC free article] [PubMed] [Google Scholar]
40. Michailidou K, Hall P, Gonzalez-Neira A, et al. Large-scale genotyping identifies 41 new loci associated with breast cancer risk. Nat Genet. 2013;45:353–361. [PMC free article] [PubMed] [Google Scholar]
41. Malkin D, Li FP, Strong LC, et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science. 1990;250:1233–1238. [PubMed] [Google Scholar]
42. Liaw D, Marsh DJ, Li J, et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet. 1997;16(1):64–67. [PubMed] [Google Scholar]
43. Heikkinen K, Rapakko K, Karppinen SM, et al. RAD50 and NBS1 are breast cancer susceptibility genes associated with genomic instability. Carcinogenesis. 2006;27:1593–1599. [PMC free article] [PubMed] [Google Scholar]
44. Tommiska J, Seal S, Renwick A, et al. Evaluation of RAD50 in familial breast cancer predisposition. Int J Cancer. 2006;118:2911–2916. [PubMed] [Google Scholar]
45. Mosor M, Ziółkowska-Suchanek I, Roznowski K, Baranowska M, Januszkiewicz-Lewandowska D, Nowak J. RAD50 gene mutations are not likely a risk factor for breast cancer in Poland. Breast Cancer Res Treat. 2010;123:607–609. [PubMed] [Google Scholar]
46. He M, Di GH, Cao AY, et al. RAD50 and NBS1 are not likely to be susceptibility genes in Chinese non-BRCA1/2 hereditary breast cancer. Breast Cancer Res Treat. 2012;133:111–116. [PubMed] [Google Scholar]
47. Bartkova J, Tommiska J, Oplustilova L, et al. Aberrations of the MRE11-RAD50-NBS1 DNA damage sensor complex in human breast cancer: MRE11 as a candidate familial cancer-predisposing gene. Mol Oncol. 2008;2:296–316. [PMC free article] [PubMed] [Google Scholar]
48. Damiola F, Pertesi M, Oliver J, et al. Rare key functional domain missense substitutions in MRE11A, RAD50, and NBN contribute to breast cancer susceptibility: results from a Breast Cancer Family Registry case-control mutation-screening study. Breast Cancer Res. 2014;16:R58. [PMC free article] [PubMed] [Google Scholar]
49. Pelttari LM, Heikkinen T, Thompson D, et al. RAD51C is a susceptibility gene for ovarian cancer. Hum Mol Genet. 2011;20:3278–3288. [PubMed] [Google Scholar]
50. Loveday C, Turnbull C, Ruark E, et al. Germline RAD51C mutations confer susceptibility to ovarian cancer. Nat Genet. 2012;44:475–476. [PubMed] [Google Scholar]
51. Loveday C, Turnbull C, Ramsay E, et al. Germline mutations in RAD51D confer susceptibility to ovarian cancer. Nat Genet. 2011;43:879–882. [PMC free article] [PubMed] [Google Scholar]
52. Pelttari LM, Kiiski J, Nurminen R, et al. A Finnish founder mutation in RAD51D: analysis in breast, ovarian, prostate, and colorectal cancer. J Med Genet. 2012;49:429–432. [PMC free article] [PubMed] [Google Scholar]
53. Rafnar T, Gudbjartsson DF, Sulem P, et al. Mutations in BRIP1 confer high risk of ovarian cancer. Nat Genet. 2011;43:1104–1107. [PubMed] [Google Scholar]
54. Thompson ER, Doyle MA, Ryland GL, et al. Exome sequencing identifies rare deleterious mutations in DNA repair genes FANCC and BLM as potential breast cancer susceptibility alleles. PLoS Genet. 2012;8(9):e1002894. [PMC free article] [PubMed] [Google Scholar]
55. Kiiski JI, Pelttari LM, Khan S, et al. Exome sequencing identifies FANCM as a susceptibility gene for triple-negative breast cancer. Proc Natl Acad Sci U S A. 2014;111:15172–15177. [PMC free article] [PubMed] [Google Scholar]
56. Park DJ, Lesueur F, Nguyen-Dumont T, et al. Rare mutations in XRCC2 increase the risk of breast cancer. Am J Hum Genet. 2012;90:734–739. [PMC free article] [PubMed] [Google Scholar]
57. Cybulski C, Carrot-Zhang J, Kluźniak W, et al. Germline RECQL mutations are associated with breast cancer susceptibility. Nat Genet. 2015 Apr 27; (Epub ahead of print). [PubMed] [Google Scholar]
58. Sun J, Wang Y, Xia Y, et al. Mutations in RECQL gene are associated with predisposition to breast cancer. PLoS Genet. 2015;11(5):e1005228. [PMC free article] [PubMed] [Google Scholar]
59. Win AK, Lindor NM, Jenkins MA. Risk of breast cancer in Lynch syndrome: a systematic review. Breast Cancer Res. 2013;15:R27. [PMC free article] [PubMed] [Google Scholar]
60. Out AA, Wasielewski M, Huijts PE, et al. MUTYH gene variants and breast cancer in a Dutch case-control study. Breast Cancer Res Treat. 2012;134:219–227. [PMC free article] [PubMed] [Google Scholar]
61. Dreijerink KM, Goudet P, Burgess JR, Valk GD. Breast-cancer predisposition in multiple endocrine neoplasia type 1. N Engl J Med. 2014;371:583–584. [PMC free article] [PubMed] [Google Scholar]
62. Ruark E, Snape K, Humburg P, et al. Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer. Nature. 2013;493:406–410. [PMC free article] [PubMed] [Google Scholar]
63. Easton DF, Deffenbaugh AM, Pruss D, et al. A systematic genetic assessment of 1,433 sequence variants of unknown clinical significance in the BRCA1 and BRCA2 breast cancer-predisposition genes. Am J Hum Genet. 2007;81:873–883. [PMC free article] [PubMed] [Google Scholar]
64. Lindor NM, Guidugli L, Wang X, et al. A review of a multi-factorial probability-based model for classification of BRCA1 and BRCA2 variants of uncertain significance (VUS) Hum Mutat. 2012;33:8–21. [PMC free article] [PubMed] [Google Scholar]
65. Goldgar DE, Easton DF, Deffenbaugh AM, Monteiro AN, Tavtigian SV, Couch FJ. Integrated evaluation of DNA sequence variants of unknown clinical significance: application to BRCA1 and BRCA2. Am J Hum Genet. 2004;75:535–544. [PMC free article] [PubMed] [Google Scholar]
66. Plon SE, Eccles DM, Easton D, et al. Sequence variant classification and reporting: recommendations for improving the interpretation of cancer susceptibility genetic test results. Hum Mutat. 2008;29:1282–1291. [PMC free article] [PubMed] [Google Scholar]
67. Le Calvez-Kelm F, Lesueur F, Damiola F, et al. Rare, evolutionarily unlikely missense substitutions in CHEK2 contribute to breast cancer susceptibility: results from a breast cancer family registry case-control mutation-screening study. Breast Cancer Res. 2011;13:R6. [PMC free article] [PubMed] [Google Scholar]
68. Goldgar DE, Healey S, Dowty JG, et al. Rare variants in the ATM gene and risk of breast cancer. Breast Cancer Res. 2011;13:R73. [PMC free article] [PubMed] [Google Scholar]
69. Tavtigian SV, Chenevix-Trench G. Growing recognition of the role for rare missense substitutions in breast cancer susceptibility. Biomark Med. 2014;8:589–603. [PMC free article] [PubMed] [Google Scholar]
70. Johnson N, Fletcher O, Naceur-Lombardelli C, dos Santos Silva I, Ashworth A, Peto J. Interaction between CHEK2*1100delC and other low-penetrance breast-cancer susceptibility genes: a familial study. Lancet. 2005;366:1554–1557. [PubMed] [Google Scholar]
71. Antoniou AC, Spurdle AB, Sinilnikova OM, et al. Common breast cancer-predisposition alleles are associated with breast cancer risk in BRCA1 and BRCA2 mutation carriers. Am J Hum Genet. 2008;82:937–948. [PMC free article] [PubMed] [Google Scholar]
72. Nickels S, Truong T, Hein R, et al. Evidence of gene-environment interactions between common breast cancer susceptibility loci and established environmental risk factors. PLoS Genet. 2013;9(3):e1003284. [PMC free article] [PubMed] [Google Scholar]
73. Campa D, Kaaks R, Le Marchand L, et al. Interactions between genetic variants and breast cancer risk factors in the Breast and Prostate Cancer Cohort Consortium. J Natl Cancer Inst. 2011;103:1252–1263. [PMC free article] [PubMed] [Google Scholar]
74. Rudolph A, Milne RL, Truong T, et al. Investigation of gene-environment interactions between 47 newly identified breast cancer susceptibility loci and environmental risk factors. Int J Cancer. 2015;136(6):E685–E696. [PMC free article] [PubMed] [Google Scholar]
75. Friebel TM, Domchek SM, Rebbeck TR. Modifiers of cancer risk in BRCA1 and BRCA2 mutation carriers: systematic review and meta-analysis. J Natl Cancer Inst. 2014;106 dju091. [PMC free article] [PubMed] [Google Scholar]
-