14
Phenotype-Genotype Correlation in Familial Breast Cancer Ana Cristina Vargas & Jorge S. Reis-Filho & Sunil R. Lakhani Received: 25 February 2011 /Accepted: 1 March 2011 /Published online: 12 March 2011 # Springer Science+Business Media, LLC 2011 Abstract Familial breast cancer accounts for a small but significant proportion of breast cancer cases worldwide. Identification of the candidate genes is always challeng- ing specifically in patients with little or no family history. Therefore, a multidisciplinary team is required for the proper detection and further management of these patients. Pathologists have played a pivotal role in the cataloguing of genotypic-phenotypic correlations in fam- ilies with hereditary cancer syndromes. These efforts have led to the identification of histological and phenotypic characteristics that can help predict the presence or absence of germline mutations of specific cancer predisposition genes. However, the panoply of cancer phenotypes associated with mutations of genes other than in BRCA1 is yet to be fully characterised; in fact, many cancer syndromes, germline mutations and gene sequence variants are under investigation for their possible morphological associations. Here we review the current understanding of phenotype-genotype correlation in familial breast cancer. Keywords Familial breast cancer . Germline mutation . Phenotype . Pathology Introduction Breast cancer is the commonest non-skin malignancy in women and it is estimated that over a million women will develop breast cancer each year [1]. Familial breast cancer accounts for approximately 7% of all breast cancers and a proportion of these cases are the result of germline mutations in the BRCA1 and BRCA2 genes [2, 3]. Germline mutations in other genes such as TP53, PTEN, CDH1 and ATM also confer a high risk of breast cancer but their mutation frequency only accounts for a small number of inherited breast cancers. In addition, breast cancer genetic predisposition may not be solely explained by the existence of specific germline mutations of high risk genes. In fact, it has been posited that a combination of low-penetrance variants and/or single nucleotide polymorphisms (SNPs) may ultimately modify the risk in the general population or in patients with other mutations (e.g. BRCA1). Additionally, many of these patients do not have or do not record any family history. Consequently, additional tools such as standard morphological and immunohistochemical criteria may allow us to predict the likelihood of a specific genotype. A. C. Vargas : S. R. Lakhani UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia J. S. Reis-Filho The Breakthrough Breast Cancer Research Centre, Institute of Cancer Research, London, UK S. R. Lakhani School of Medicine, The University of Queensland, Brisbane, Australia S. R. Lakhani Pathology Queensland, The Royal Brisbane & Womens Hospital, Brisbane, Australia S. R. Lakhani (*) The University of Queensland Centre for Clinical Research, Royal Brisbane & Womens Hospital, Building 71 (918), Herston 4029 QLD, Australia e-mail: [email protected] J Mammary Gland Biol Neoplasia (2011) 16:2740 DOI 10.1007/s10911-011-9204-6

Phenotype-Genotype Correlation in Familial Breast Cancer

Embed Size (px)

Citation preview

Page 1: Phenotype-Genotype Correlation in Familial Breast Cancer

Phenotype-Genotype Correlation in Familial Breast Cancer

Ana Cristina Vargas & Jorge S. Reis-Filho &

Sunil R. Lakhani

Received: 25 February 2011 /Accepted: 1 March 2011 /Published online: 12 March 2011# Springer Science+Business Media, LLC 2011

Abstract Familial breast cancer accounts for a small butsignificant proportion of breast cancer cases worldwide.Identification of the candidate genes is always challeng-ing specifically in patients with little or no familyhistory. Therefore, a multidisciplinary team is requiredfor the proper detection and further management of thesepatients. Pathologists have played a pivotal role in thecataloguing of genotypic-phenotypic correlations in fam-ilies with hereditary cancer syndromes. These effortshave led to the identification of histological andphenotypic characteristics that can help predict thepresence or absence of germline mutations of specificcancer predisposition genes. However, the panoply of

cancer phenotypes associated with mutations of genesother than in BRCA1 is yet to be fully characterised; infact, many cancer syndromes, germline mutations andgene sequence variants are under investigation for theirpossible morphological associations. Here we review thecurrent understanding of phenotype-genotype correlationin familial breast cancer.

Keywords Familial breast cancer . Germline mutation .

Phenotype . Pathology

Introduction

Breast cancer is the commonest non-skin malignancy inwomen and it is estimated that over a million women willdevelop breast cancer each year [1]. Familial breast canceraccounts for approximately 7% of all breast cancers and aproportion of these cases are the result of germlinemutations in the BRCA1 and BRCA2 genes [2, 3]. Germlinemutations in other genes such as TP53, PTEN, CDH1 andATM also confer a high risk of breast cancer but theirmutation frequency only accounts for a small number ofinherited breast cancers. In addition, breast cancer geneticpredisposition may not be solely explained by the existenceof specific germline mutations of high risk genes. In fact, ithas been posited that a combination of low-penetrancevariants and/or single nucleotide polymorphisms (SNPs)may ultimately modify the risk in the general population orin patients with other mutations (e.g. BRCA1). Additionally,many of these patients do not have or do not record anyfamily history. Consequently, additional tools such asstandard morphological and immunohistochemical criteriamay allow us to predict the likelihood of a specificgenotype.

A. C. Vargas : S. R. LakhaniUQ Centre for Clinical Research, The University of Queensland,Brisbane, Australia

J. S. Reis-FilhoThe Breakthrough Breast Cancer Research Centre,Institute of Cancer Research,London, UK

S. R. LakhaniSchool of Medicine, The University of Queensland,Brisbane, Australia

S. R. LakhaniPathology Queensland, The Royal Brisbane & Women’s Hospital,Brisbane, Australia

S. R. Lakhani (*)The University of Queensland Centre for Clinical Research,Royal Brisbane & Women’s Hospital,Building 71 (918),Herston 4029 QLD, Australiae-mail: [email protected]

J Mammary Gland Biol Neoplasia (2011) 16:27–40DOI 10.1007/s10911-011-9204-6

Page 2: Phenotype-Genotype Correlation in Familial Breast Cancer

BRCA1, BRCA2 and BRCAX

The BRCA1 gene, located on chromosome 17q, has a keyrole in DNA repair, cell-cycle regulation, transcriptionalregulation, chromatin remodeling, DNA decantenation andcontrol of multiple cell cycle checkpoints [4]. Functionsattributed to BRCA2, at 13q, are mainly restricted to DNArecombination and DNA repair through a regulating role inRAD51 activity [5–7]. Loss of BRCA1 or BRCA2 leads to adeficiency in the repair of DNA double-strand breaks byhomologous recombination (HR), leading to potentiallymutagenic repair of DNA lesions by alternative mecha-nisms such as non-homologous end-joining (NHEJ) andsingle strand annealing (SSA). Ultimately, genomic insta-bility is developed, contributing to the cancer predispositiongenerated by loss-of-function mutations in BRCA1 orBRCA2 [7]. Germline mutations in BRCA1 and BRCA2confer an estimated 65% and 45% cumulative lifetime riskof developing breast and an ovarian cancer risk of 39% and11%, respectively [8].

BRCA1

Patients with BRCA1 germline mutations usually develophigh grade Invasive Ductal Carcinomas of No Special Type(IDC-NST) with frequent medullary-like morphology [9,10] and often but not invariably, negative for oestrogenreceptor/ER, progesterone receptor/PR and HER2 over-expression or amplification (triple negative – TN; approx-imately 70%–75%, Fig 1). In multivariate analysis, Lakhaniet al. showed that morphological features predictive ofBRCA1 phenotype include pushing margins, lymphocyticinfiltrate and high mitotic count [10]. As for sporadic TNbreast cancer [11] BRCA1-associated tumours also clusterwithin the ‘basal-like’ intrinsic subtype of breast canceridentified by expression profiling [12, 13]. These tumoursare enriched by the expression of so-called ‘basal’ markersincluding high molecular weight cytokeratins (CK5/6,CK14, CK17 and/or Epidermal Growth Factor Receptor-EGFR/HER1) (Fig. 1) [14] and constitute in a significantproportion, TN tumours with basal-like phenotype (TNBL)[15]. Other myoeptihelial-related markers such as α-SMA,P-cadherin and Caveolins 1 and 2 are commonly expressedin TNBL tumours [16–19]. Basal-like tumours in bothBRCA1 and sporadic breast cancer tend to show higherexpression of p53, a distinct pattern of cell-cycle prolifer-ation markers (i.e. overexpression of cyclin E rather thancyclin D), neuroendocrine markers (chromogranin A andsynaptophysin), stem-cell-phenotype (CD44+/CD24-) andothers such as hypoxia-associated factor; CA9 and FHITprotein compared to TN-non basal-like cancers [20]. ER-associated genes such as BCL2 [21] and Cyclin D1 are

rarely if at all expressed in TNBL tumours (BRCA1 orsporadic) [22, 23] as are antiapoptotic and proapoptoticproteins, BCL2 and BAX, respectively [21, 24].

Increased expression of cell proliferation and cell cyclerelated markers is characteristic of BRCA1 tumours. Cellproliferation as determined by Ki-67 labelling index hasshown that these tumours are highly proliferative (Ki-67>65%) [25]. Van de Groep et al. [26] demonstrated that thecombination of high proliferation (Ki67 expression >25%)and EGFR positivity in women younger than 54 years ofage was predictive of BRCA1 mutation in 82% of the cases[26, 27]. Amplification of MYC also leads to proliferationand this is observed in up to 60% of BRCA1 tumours [25].Frequent over-expression of other cell cycle-related pro-teins such as Cyclin E, A, B1, p27, p16, p21, CDK4, CDK2and CDK1 [16, 28] is also a common feature of thesecancers.

BRCA1-related tumours exhibit TP53 truncating muta-tions in up to 100% of the cases, when detected bysequencing [29–32]. Although TP53 mutations have beenthought to correlate to BRCA1 mutation, recent evidencesuggests that the presence of TP53 mutations in factcorrelates with the ‘basal-like’ phenotype regardless ofBRCA-germline status. However, complex mutations (inser-tions and deletions) were shown to be more common inBRCA1-associated breast cancer than in sporadic TNBLcancers [33]. Similarly, BRCA1 of luminal subtype (i.e. ER+)also harbours a significant prevalence of TP53 mutations(53%), which is significantly higher than luminal non-BRCA1 tumours [33].

BRCA1 tumours can lose expression of luminal cytoker-atins [34] and a recent study has proposed that CK8/18negativity together with ‘basal-like’ phenotype and familyhistory can be used to predict BRCA1 germline mutationstatus [35]. In this study, absence or decreased CK8/18protein expression was independently associated withBRCA1-associated tumours compared to controls (43% vs94%; P<0.0001) [35].

BRCA1 tumours have also been evaluated for theexpression of stromal signature-related proteins. For in-stance, members of the Notch and TGF-β signallingpathways such as Jagged 1, TFG-β, osteopontin andosteonectin are higher in BRCA1-associated tumours [15,36]. The c-kit (CD117) reactivity in these tumours varies,ranging from 14.7% [37] to 48.1% of tumour cells [38]. c-kit has been widely used for the characterisation of stromaltumours. However, recent work suggests that it is a markerof luminal progenitor cells in the breast [38]. These stromalmarkers may provide interesting therapeutic avenues (e.g.imatinib as inhibitor for c-kit) for the management ofBRCA1 carriers [37].

Regarding the predictive value of BRCA1 phenotype,numerous studies have assessed the statistical power of

28 J Mammary Gland Biol Neoplasia (2011) 16:27–40

Page 3: Phenotype-Genotype Correlation in Familial Breast Cancer

hormone receptor negativity, age of onset and positivefamily history. Farshid et al. showed that by using onlyhistopathological criteria in absence of any clinical infor-mation, pathologists were able to predict BRCA1–associat-ed breast cancer with high sensitivity and specificity (92%and 86%, respectively) [41]. In particular, ER negativity inearly onset breast cancer identified 29.6% of BRCA1mutations regardless of PR and HER2 status [42, 43].Eisinger et al. also reported that the combination of ERnegativity and high histological grade in early onset breastcancer was the only predictive feature of BRCA1 carrierstatus [44] but adding family history increases the BRCA1-mutation detection rate [25]. Similarly, triple negative and

basal-like phenotypes are highly predictive of germ-lineBRCA1 mutation [15, 45]. Lakhani et al. [15] showed thatthe addition of basal markers to ER negative tumoursincreases the specificity for detecting BRCA1 carriers.However, it must be emphasized that triple negativity withor without expression of basal markers will not identify allBRCA1 associated tumours. ER positive breast cancer, forinstance, comprises 5%–20% of BRCA1-associated breastcancer, in particular if the current ASCO/CAP definition ofER-positivity is adopted. In addition, HER2 amplification[46] and low-grade morphological variants (tubular carci-noma) [47] have been described in 3% and 3.6% of cases,respectively. Finally, the use of basal markers has not

Figure 1 a-b: High Grade In-vasive Ductal Carcinoma withbasal-like features (a) andMedullary-like breast cancer(b) are over-represented inBRCA1-, FANCN/PALB2- andLynch Syndrome-associatedbreast cancer (H&E). c-f: Im-munohistochemical expressionof basal markers. c: EGFR; d:CK14; e: CK5/6; andf: P63 (50X)

J Mammary Gland Biol Neoplasia (2011) 16:27–40 29

Page 4: Phenotype-Genotype Correlation in Familial Breast Cancer

consistently proven to be predictive of BRCA1 phenotype[48]. Therefore, the presence of a TNBL breast cancer in ayoung patient can be very suggestive of BRCA1 germlinemutation, but an alternative phenotype would not excludethe possibility of a BRCA1 germ-line mutation.

It must be noted that BRCA1 pathway is frequentlydysfunctional in sporadic triple negative breast cancer. Lossof nuclear BRCA1 expression is present in approximately15% of sporadic breast cancer and is mainly due to BRCA1gene promoter methylation, ID4 epigenetic inactivation orposttranscriptional down-regulation (e.g.microRNA-182)[49–52]. Therefore, distinction must be made betweensporadic triple negative (Basal or non basal) breast cancerwith loss of BRCA1 due to methylation and BRCA1germline mutation. Regardless of the mechanism of BRCA1inactivation, patients with TNBL cancers are currentlymanaged with the same repertoire of systemic therapies[39, 40], and have a similar rate of pathological response toneoadjuvant chemotherapy [53–56] However, unlike spo-radic TNBL breast cancer, patients with BRCA1 germlinemutations must be identified to receive subsequent man-agement (i.e. risk prevention in the contralateral breast andovaries, screening of family members, genetic counseling,etc.) [57].

BRCA2

BRCA2-associated breast cancer is a very heterogeneousgroup [36, 58]. Although no specific phenotype is yetpredictive of BRCA2-associated tumours, ER+/HER2-tumours comprise the majority of these cases [36]. Inparticular, two morphological features are significant inBRCA2-associated breast cancer; pushing margins and lackof tubule formation [10]. Low and intermediate histologicalgrade is characteristic [24, 46, 58].

Bane et al. have shown that BRCA2-associated tumoursexpress genes involved in signal transduction, cell prolifer-ation, cell adhesion, and extracellular matrix remodelingwith activation of the MAPK signalling pathway [36].FGF1 (Fibroblastic Growth Factor 1) is involved in theMAPK and PI3K signalling activation [61] and increasedlevels of this protein and a related receptor, FGFR2(Fibroblastic Growth Factor Receptor 2) was observed inBRCA2 tumours. FGFR2 expression was observed in 30%of BRCA2 tumours compared to 6% of BRCA1, beinginversely correlated with basal-like phenotype [36].

A greater incidence of TP53 mutations (29%–63%) hasbeen described in BRCA2 tumours when compared tosporadic breast cancer. This is in agreement with thepotential role of TP53 in promoting tumourigenesis notonly in BRCA1 but also in BRCA2-associated breast cancer[62–64].

Loss of Heterozygosity (LOH) at the BRCA2 locushas been identified in a subset of high-grade lobularcarcinomas (pleomorphic variant) [66]. However, theassociation of BRCA2 germline mutation with the devel-opment of pleomorphic lobular carcinoma has not yetbeen studied.

The incidence of precursor lesions (e.g. DCIS) inpatients with BRCA1 and BRCA2 germline mutations hasproven controversial [67–71] but a recent study usingmammography, has described a higher proportion of DuctalCarcinoma In Situ (DCIS) in BRCA2-associated patients[47]. In that study, BRCA2 associated patients had fewerinterval cancers and favorable tumour size compared toBRCA1 patients [47].

BRCAX

BRCAX (non BRCA1/2)-associated breast cancer is a verydiverse group characterised by a similar proportion of ER+/HER2-, HER2+ and TNBL phenotypes [58, 59]. Nohistopathological features have been reproducible in thisgroup [60]. However, an association between invasivelobular carcinomas and BRCAX has been described [9, 65].

Unclassified Mutation Variants (UMVs)

UMVs are clinically challenging since presymptomatictesting is not possible and surveillance can only be basedon the family history [72]. As many as 30% and 50% of thegenetic variants in BRCA1 and BRCA2, respectively, fallunder the category of UMVs [73]. UMVs are mostlymissense sequence variations with not definite role incarcinogenesis [72], but deleterious mutations, may bepathogenic and clinically significant. Therefore, distinctionof patients with deleterious mutations who will benefit fromgenetic counselling is important [74].

Currently, pathological data have a restricted value inclassifying UMVs. Tumours arising in patients withgermline BRCA1/BRCA2-UMVs do not seem to havedistinctive features, with tumours being of high or lowgrade, ductal or mixed morphology with positive ornegative ER, PR and HER2 status [75–77]. However,some studies have highlighted the role of pathology inclassifying UMVs as pathogenic or non-pathogenic [74,78, 79]. By using LOH, family history of carriers and twopathological criteria (Grade 3 and ER negativity) Osorio etal. were able to classify an UMV as neutral or deleterious[74]. Furthermore, pathology has been applied to comparethe degree of inter and intra-familial concordance oftumour phenotype from multiple-case cancer BRCA1 andBRCA2 families [58]. Balleine et al. performed gene

30 J Mammary Gland Biol Neoplasia (2011) 16:27–40

Page 5: Phenotype-Genotype Correlation in Familial Breast Cancer

expression profiling of BRCA1 and BRCA2 pathogenicmutations and UMVs. BRCA1 pathogenic mutationsclustered together but not family members, indicating thatno particular similarities exist between individuals. Aspecific clustering was not observed for BRCA2 tumours.This study demonstrated that breast cancer pathology isvariable even between individuals who carry the sameBRCA mutation [58].

Single Nucleotide Polymorphisms

Much of the genetic variability in breast cancer is notexplained by specific germline mutations, but may be dueto the combination of low-penetrance variants that tend tobe more frequently found in selected populations [80].

Three intronic polymorphisms in the FGFR2 gene,located at 10q26 (rs2981582, rs1219648, and rs2420946),clearly confer breast cancer risk not only in patients withBRCA-UMV or germline mutation carriers but also in thegeneral population [81]. FGFR2 encodes a receptor tyrosinekinase, which plays a role in mammary gland development[82] and is amplified and/or over-expressed in a smallproportion of breast cancer [83]. The SNP rs2981582 isassociated with breast cancer risk in BRCA2 mutationcarriers [84] and in agreement with the role of FGFR2 inER-related carcinogenesis, rs2981582 showed a strongerassociation with low grade ER-positive, node positivebreast cancer. This SNP was also associated with PRpositivity, but not independently from ER status [85]. TheFGFR2 SNP rs1219648 was also associated with ER-positive, node positive breast cancer in the generalpopulation, particularly in women with family history ofbreast cancer [80, 81].

Some other SNPs have been found to correlate with ERpositive breast cancer independently from PR status. SNPrs3803662 (TNRC9/TOX3, at 16q12) was significantlyassociated with ER positive breast cancer in both BRCA1and BRCA2 mutation carriers [84, 85]. The rs13281615located on 8q24 [85] and rs4415084 and rs10941679 on5p12 [86] also conferred risk for ER positive disease in thegeneral population. Interestingly, the MRPS30 gene locatedat 5p12 has been implicated in good outcome tamoxifen-treated patients [87]. None of these SNPs have beenassociated with other histopathological features or overallsurvival [80, 85].

Many other polymorphisms, such as rs13387042(2q35) [80], rs889312 (MAP3K1), rs3817198 (LSP1)[84, 85] and other BRCA1/BRCA2 modifiers such asCASP8 D302H (Caspase 8 coding variant) [88], RAD51135 G.C [89, 90] and the KRAS-variant, rs61764370 T > G[91] have not yet been linked to specific phenotypicalcharacteristics.

Lynch Syndrome and Breast Cancer

Lynch syndrome or Hereditary Non-Polyposis Colorectalcancer syndrome (HNPCC) is associated with germlinemutations in any of the mismatch repair (MMR) genes(MLH1, MSH2, MSH6, PMS2) [92, 93], resulting inmicrosatellite instability (MSI). The MMR pathway repairsbase substitution and insertion-deletion mismatches whicharise from replication errors. As a result, MMR-geneinactivation confers a mutator phenotype [94]. MSIinvolves length abnormalities in DNA microsatellites (shorttandem highly repeated DNA sequences) [95] and is usedas diagnostic marker to detect loss or dysfunction of theMMR genes [94, 96]. MSI in colorectal, as well as in breastcancer, is diagnosed when mutation in at least 1 of the 5NCI (National Cancer Institute)-recommended microsatel-lite sequences (BAT25, BAT26, D2S123, D5S346 andD17S250) [96–98] is present. Gene mutation in 1 or morethan 2 microsatellite sequences is called MSI-low (MSI-L)or MSI-high (MSI-H), respectively [94, 97, 99]. Inborderline cases, additional microsatellite markers must betested [100].

Although breast cancer was excluded in 2002 as partof the Lynch syndrome [101], some studies appear toconfirm the role of MMR germine mutations in thedevelopment of breast cancer in patients with Lynchsyndrome [102–106]. For instance, Shanley et al. foundthat 4 out of 29 individuals with breast cancer and Lynchsyndrome harboured MMR germline mutations [104].Furthermore, MMR gene-variant mutations have alsobeen shown to predispose to breast cancer. For instance,MSH6 rare variants have been identified in 10.3% amongfamilial breast cancer compared to 2% of the generalpopulation [107].

Breast cancer tumours associated with germline muta-tions of the MMR genes and Lynch syndrome can display adistinctive morphological appearance. These have beendescribed as IDC-NST with dense lymphocytic infiltrate[105, 108]. In addition, Walsh et al. [109] showed that 18out of 35 breast tumours from individuals with Lynchsyndrome were significantly more likely to be ER-/PR-,with high mitotic activity, solid growth pattern, confluentnecrosis and low frequency of adjacent DCIS whencompared to non-MMR gene mutation carriers. Thisphenotype, reminiscent of medullary-like breast cancerhas been previously linked to high MSI [110] (Fig. 1). Onthe other hand, tumour type, size, lymphovascular invasion,lymph node metastasis and P53 status were not significant-ly different in breast cancers arising in patients with Lynchsyndrome when compared to the general population.

The most sensitivity pathologic feature of MSI pheno-type in colorectal cancer is the presence of tumourinfiltrating lymphocytes and mucinous–signet ring cell

J Mammary Gland Biol Neoplasia (2011) 16:27–40 31

Page 6: Phenotype-Genotype Correlation in Familial Breast Cancer

morphology [111, 112]. Tumour lymphocytes quantifica-tion has been suggested as a prognostic marker in colorectalcancer [94], which has been partially evaluated in medul-lary breast cancer [119]. Furthermore, unlike mucinouscolorectal carcinomas, mucinous breast cancer do notharbour MSI (as determined by PCR and IHC) [113].Immunohistochemistry (IHC) for MSI-associated breastcancer identifies loss of protein expression of the MMRaffected gene (MLH1, MSH2, MSH6 or PMS2) [104, 113,115]. However, as opposed to colorectal cancer [114], lackof immunohistochemical expression for these markersrarely occurs in breast cancer [113, 119].

Nevertheless, many other studies exclude MMR germlinemutations as responsible for the development of breast cancerin Lynch syndrome patients. Muller et al. [101] reported thatbreast cancer from Lynch syndrome families is sporadic,unrelated to MMR gene defects and Vansen et al. [116]suggested that MMR deficiency accelerates breast cancertumourigenesis in Lynch syndrome patients but is not theinitiating event [117]. Similarly, MSI is extremely infrequentin sporadic breast cancer (2%–3%) [113, 118, 119] with geneinactivation due to methylation as the main mechanism.

CDH1 Germline Mutations

CDHI germline mutations are associated with the autoso-mal dominant cancer-predisposition syndrome, HereditaryDiffuse Gastric Cancer (HDGC) [120], which is associatedwith increased susceptibility for the development ofinvasive lobular carcinoma of the breast. CDH1 encodesfor the E-cadherin protein, a cell-cell adhesion moleculethat has a fundamental role in adhesion, cell differentiation,and cellular signalling [121]. Inactivating mutations, dis-tributed throughout the gene, with no hot spots in anyparticular region, account for the majority of the cases[122]. CDH1 pathogenic variants are uncommon with afrequency of 1.3% in patients with family history of gastric/breast cancer [123]. Female mutation carriers have a 39%lifetime risk of breast cancer [120, 122, 124–126],particularly with lobular morphology (ILC) [127]. Familyhistory of gastric and/or breast cancer is present in thesecases, but can be absent [123]. In addition, family history ofbreast cancer of non-lobular histology (IDC-NST) can befound [128, 129]. In fact, both ILC as well as IDC can beobserved in CDH1 germ-line mutation carriers [120, 130].In these cases the possibility of sporadic ductal carcinomasarising in CDH1 mutation carriers cannot be excluded.

When lobular carcinomas arise inCDH1 mutation carriers,they show the same phenotype observed in sporadic cases,with loss of E-cadherin and β-catenin membranous expres-sion. As in sporadic ILC, E-cadherin aberrant staining [131]has been described. In a case report of an ILC and concurrent

diffuse gastric carcinoma, Zhu et al. showed that the tumourcells retained E-cadherin and β-catenin expression in asignificant proportion of cells. Interestingly, the gastriccarcinoma displayed the same aberrant E-cadherin/β-cateninreactivity observed in the breast tumour [130].

PTEN Germline Mutations

Cowden syndrome (CS) is an autosomal-dominant disorderchacracterised by multiple hamartomatous and neoplasticmucocutaneous lesions, CNS abnormalities and cancerpredisposition in multiple organs (Breast, colon, endome-trium and brain) [132, 133]. Germline mutations in thetumour suppressor gene, PTEN are responsible for CS.PTEN (phosphatase and tensin homologue) is located onchromosome 10q22.3 and encodes a lipid phosphatase witha regulatory role in the phosphoinositol 3-kinase and thePKB/Akt signalling pathways [134–136]. Diagnosis of CSis challenging because many of these patients do not meetthe criteria for diagnosis in the screening setting, includingthe lack of family history [137, 138].

Breast cancer is the most common malignancy in CS,occurring in 22% of women [139, 140], with a lifetime riskof 50% [137, 138]. CS-associated breast cancer usuallyoccurs in young women (mean age 40), but can occur inmen [139, 141] and is bilateral in 25% of the cases [142,143]. No consistent breast cancer phenotype has yet beendescribed for PTEN mutation carriers, but the majority ofthese tumours are of luminal phenotype, characterised byvariable expression of hormone receptors and negativeexpression for HER2, P53 and basal markers. Morpholog-ical type varies, but IDC-NST and mixed ductolobular[144] of low/intermediate histological grade with adjacentDCIS, account for most of the cases. Therefore, CS-associated breast cancer is regarded as favourable histologywith frequent lack of lympho-vascular invasion (LVI) andlymph node metastasis [137, 142, 143, 145, 146]. However,poor prognostic factors such as high histological grade,lymph node metastasis, squamous differentiation, triplenegative phenotype and interval carcinomas can also beenfound [142, 144, 147]. Benign breast lesions, such asfibrocystic disease [143], tubular adenomas and breasthamartomas [142, 148] are part of the spectrum of breastdisease. It must be noted that LOH of the PTEN locusoccurs in sporadic breast cancer [134, 149, 150].

PARP Inhbitors in BRCA and PTEN Familial BreastCancer

Based on the encouraging results of phase I and II clinicaltrials, Poly (ADP-ribose) polymerase 1 (PARP-1) inhibitors

32 J Mammary Gland Biol Neoplasia (2011) 16:27–40

Page 7: Phenotype-Genotype Correlation in Familial Breast Cancer

(PARPis) are likely to play a major role in the treatment ofcancers in BRCA1 and BRCA2 germline mutation carriers[39, 40]. PARPis target Homologous Recombination (HR)deficiency in BRCA1/2 deficient cells. These are highlyselective due to the mechanism of synthetic lethality, whichrequires both, PARP and BRCA genes to be lost in thetumour cells.

PTEN-mutated cancer characteristically shows HR-deficient cells. Therefore, the role of PARPis is currentlybeing explored in this context [151, 152]. HomozygousPTEN mutations with significant truncations of the openreading frame are particularly sensitive to PARP inhibition.However, PTEN missense mutations have not shown suchsensitivity [152]. This may indicate that a proportion offamilial breast cancer associated with PTEN gemlinemutation, will greatly benefit from PARPis. Immunohisto-chemical expression for nuclear PTEN has been suggestedas predictor for tumour response to PARPis in tumour cellsbut further work is required [152].

Li-Fraumeni Syndrome

Li-Fraumeni syndrome (LFS) is a rare autosomal disordercharacterised by an increased susceptibility for the devel-opment of malignant epithelial, mesenchymal and haema-topoietical neoplasms (i.e. breast cancer, sarcomas, braintumours, adrenocortical carcinoma, leukemia among others)[153, 154]. TP53 germline mutations account for thedevelopment of LFS [155] and Li-Fraumeni-like syndrome(LFL) [156] in 75% and 40% of the affected families,respectively [157]. Diagnostic criteria for LFS and LFL areconstantly being redefined because up to 30% of the TP53-mutated families develop tumours others than thoseincluded in the diagnostic criteria [156]. Although thereare over 450 germline mutations involving the TP53 gene,missense mutations of its coding region (exons 2–11) detect95% of LFS and LFL patients [158].

TP53 germline mutations account for approximately 1%of total breast cancer, which is the commonest epithelialneoplasm developed by women with LFS [159]. Thisfrequency increases up to 7% in women less than 30 yearsof age and up to 13% in selected populations [160]. LFSand LFL also account for a high proportion of bilateralbreast cancer (22%) in young women negative for BRCA1/BRCA2 genetic testing [160]. TP53 mutation carriers have alifetime risk for developing cancer of almost 100%, whichis in a significant proportion due to breast cancer [161].

Breast cancer phenotype in patients with TP53 germ-line mutation has been partially characterised in a recentstudy. Wilson et al. showed that HER2 overexpression oramplification was present in 83% of a small cohort (n=12)of breast cancers developed in TP53 mutation carriers,

compared to 19% HER2 amplification in the referencecohort. Regarding hormone receptor status, it was notedthat 42% of the TP53-mutated tumours were triple positive(ER+/PR+/HER2+) compared to only 8% of the referencecohort. No TN tumours were identified in the TP53-associated breast cancer cohort and this was statisticallysignificant. The authors hypothesised that TP53 mutationand HER2 overexpression/amplification may be coacti-vated in the same oncogenic pathway [162]. Despite thelimited data presented in this study, it can be concludedthat a HER2-amplified breast cancer in a very youngwoman (<30 years), especially when there is familyhistory of neoplasms in other organs may warrant TP53germline mutation testing, but more studies are necessaryto confirm these findings. Other breast malignancies, suchas malignant phyllodes have also been associated withLFS [163].

ATM Germline Mutations

Ataxia-telangiectasia is an autosomal recessive disordercharacterised by mutations in the ataxia telangectasiamutated (ATM) gene [164]. The ATM gene encodes a lipidkinase phosphatidylinositol 3-kinase (PI3K) that is involvedin DNA damage signalling, chromatin remodeling, tran-scriptional regulation and apoptosis [165, 166].

ATM is an intermediate-risk breast cancer susceptibilitygene, conferring 2- to 5-fold increased risk of breast cancerfor female relatives who are heterozygous carriers of ATMmutations [167, 168]. A large number of mutationsinvolving the ATM gene have been identified [169], whichare responsible for 2% of familial breast cancer [168]. ATMpolymorphic missense variants also confer a slight increasein breast cancer risk, specifically in patients with familyhistory and bilateral breast cancer [170, 171].

No distinctive histopathology has been associated withATM-associated breast cancer in mutation carriers [172].ATM protein is expressed in the normal breast epithelial cellswith reduced expression in BRCA1 and BRCA2-associatedbreast cancer as well as in sporadic triple negative tumours.No correlation has been found between decreased ATMexpression and other prognostic features such as P53expression [173]. ATM polymorphisms (rs637064) haveshown association with nodal metastasis [174].

Fanconi Anemia Pathway and Germline Mutations

The Fanconi anemia pathway (FA pathway) is comprisedof 13 genes involved in DNA damage response [175,176]. Mutations in genes pertaining to this pathway conferincreased susceptibility to chromosome breakage in

J Mammary Gland Biol Neoplasia (2011) 16:27–40 33

Page 8: Phenotype-Genotype Correlation in Familial Breast Cancer

response to interstrand DNA cross-linking agents [177].Particularly, four Fanconi’s anemia genes (FANCD1,FANCN/PALB2, FANCJ/BRIP1 and FANCC) have beenshown to be intermediate risk breast cancer susceptibilitygenes [178, 179]. FANCN/PALB2 for instance, confers2.3-fold increased risk of breast cancer [180]. However,due to variable penetrance estimation for multiple-casefamilies, the real frequency of FANCN/PALB2 mutation isnot well defined [181]. The FANCD1 gene is in fact,identical to BRCA2 and considered by some authors as thesame gene [175].

Germline mutations as well as mutation variants andsingle nucleotide polymorphisms in the Fanconi anemia’sgenes have been linked to some phenotypical character-istics. For instance, FANCN/PALB2-associated tumours areusually ER and HER2 negative (58% and 93%, respective-ly) with 40% of all cases, being triple-negative phenotype(Fig. 1). Although this group of tumours shows similaritieswith BRCA1-associated breast cancer, it has reported toharbour gene copy changes (aCGH) similar to thoseobserved in BRCA2 tumours [180, 181]. Two FANCI SNPs(rs7168941 and rs8032440) were associated with presenceor absence of nodal metastasis, respectively [174]. Intronicnoncoding SNPs of the FANCJ/BRIP1 gene (rs7220740)and FANCN/PALB2 (rs447529) are associated with positiveexpression of progesterone, but not estrogen receptor [174].However, this last finding was not reproduced in anindependent study [182]. Association with overall survivalhas also been found for SNPs in FANCC (rs1045276) andFANCD1 (rs1801406) [174].

RAD51C, a tumour suppressor gene involved in homol-ogous recombination, [183] is a predisposing gene for aFanconi anemia-like disorder as well as for hereditary breastand/or ovarian cancer (monoallelic mutations) [176, 184,185]. RAD51 functions are regulated by BRCA1 and BRCA2in response to DNA damage. Mutations have been observedin 1.3% of probands from breast and ovarian cancer in apopulation-based study [186]. Although tumour phenotypein RAD51C mutation carriers has not been described,sporadic breast cancer of luminal B-type (ER+/PR-) wascharacterised by high RAD51 gene and protein expression.This was statistically associated with recurrence, distantmetastasis and overall survival [187].

CHEK2 Germline Mutation

The CHEK2 (Checkpoint kinase 2) protein is a cell cyclekinase that phosphorylates p53 and BRCA1 as a result ofDNA damage, inducing cell cycle arrest [188, 189]. Theframeshift mutation, 1100delC, is the most common andresults in loss of the CHEK2 kinase activity. It is associatedwith twofold breast cancer risk in carriers [190] and

increases up to 12.1% in women with bilateral breastcancer of selected populations [191]. For instance, in non-European populations this mutation does not seem to conferan increased in breast cancer risk [192, 193].

CHEK2 (110delC) mutation carriers usually develophigh-grade ductal carcinomas with positive expression ofhormone receptor (ER+/PR+). CHEK2 protein expressionis absent or significantly reduced in these cases [190]. Adifferent CHEK2 mutation (I157T] was reported to bestrongly associated with lobular carcinoma in a series fromPoland; this observation, however, was not reproduced indifferent populations [194, 195].

Summary

Since the identification of BRCA1 and BRCA2 genes,considerable work has been done to unravel thegenotypic-phenotypic correlations between mutations incancer predisposition genes and breast cancer. The stron-gest data are still for BRCA1-associated cancers but data arebeginning to emerge on the phenotypic characteristics ofcancers arising in families with mutations in other cancerpredisposition genes. The correlations do and will continueto play an important role in understanding biologicalfunction, in identification of patients at risk of carryinggermline mutations and in the development of therapeuticstrategies.

Acknowledgements ACV is a clinical fellow funded by the LudwigInstitute for Cancer Research (LICR). JSR-F is funded in part byBreakthrough Breast Cancer Research Centre. JSR-F is the recipientof the 2010 CRUK Future Leaders Prize.

References

1. Bray F, McCarron P, Parkin DM. The changing global patternsof female breast cancer incidence and mortality. Breast CancerRes. 2004;6(6):229–39.

2. Claus EB, Schildkraut JM, Thompson WD, Risch NJ. Thegenetic attributable risk of breast and ovarian cancer. Cancer.1996;77(11):2318–24.

3. Anglian Breast Cancer Study Group. Prevalence and penetranceof BRCA1 and BRCA2 mutations in a population-based seriesof breast cancer cases. Br J Cancer. 2000;83(10):1301–8.

4. Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K,Tavtigian S, et al. A strong candidate for the breast and ovariancancer susceptibility gene BRCA1. Science. 1994;266(5182):66–71.

5. Wooster R, Neuhausen SL, Mangion J, Quirk Y, Ford D,Collins N, et al. Localization of a breast cancer susceptibilitygene, BRCA2, to chromosome 13q12-13. Science. 1994;265(5181):2088–90.

6. Wooster R, Bignell G, Lancaster J, Swift S, Seal S, Mangion J, etal. Identification of the breast cancer susceptibility gene BRCA2.Nature. 1995;378(6559):789–92.

34 J Mammary Gland Biol Neoplasia (2011) 16:27–40

Page 9: Phenotype-Genotype Correlation in Familial Breast Cancer

7. Venkitaraman AR. Cancer susceptibility and the functions ofBRCA1 and BRCA2. Cell. 2002;108(2):171–82.

8. Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE,Hopper JL, et al. Average risks of breast and ovarian cancerassociated with BRCA1 or BRCA2 mutations detected in caseSeries unselected for family history: a combined analysis of 22studies. Am J Hum Genet. 2003;72(5):1117–30.

9. Lakhani SR, Gusterson BA, Jacquemier J, Sloane JP, AndersonTJ, van de Vijver MJ, et al. The pathology of familial breastcancer: histological features of cancers in families not attribut-able to mutations in BRCA1 or BRCA2. Clin Cancer Res.2000;6(3):782–9.

10. Lakhani SR, Jacquemier J, Sloane JP, Gusterson BA, AndersonTJ, van de Vijver MJ, et al. Multifactorial analysis ofdifferences between sporadic breast cancers and cancersinvolving BRCA1 and BRCA2 mutations. J Natl Cancer Inst.1998;90(15):1138–45.

11. Badve S, Dabbs DJ, Schnitt SJ, Baehner FL, Decker T, Eusebi V,et al. Basal-like and triple-negative breast cancers: a criticalreview with an emphasis on the implications for pathologists andoncologists. Mod Pathol 2010.

12. Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, ReesCA, et al. Molecular portraits of human breast tumours. Nature.2000;406(6797):747–52.

13. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H,et al. Gene expression patterns of breast carcinomas distinguishtumor subclasses with clinical implications. Proc Natl Acad SciUSA. 2001;98(19):10869–74.

14. Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, etal. Immunohistochemical and clinical characterization of thebasal-like subtype of invasive breast carcinoma. Clin CancerRes. 2004;10(16):5367–74.

15. Lakhani SR, Reis-Filho JS, Fulford L, Penault-Llorca F, van derVijver M, Parry S, et al. Prediction of BRCA1 status in patientswith breast cancer using estrogen receptor and basal phenotype.Clin Cancer Res. 2005;11(14):5175–80.

16. Foulkes WD, Brunet JS, Stefansson IM, Straume O, ChappuisPO, Begin LR, et al. The prognostic implication of the basal-like(cyclin E high/p27 low/p53+/glomeruloid-microvascular-prolif-eration+) phenotype of BRCA1-related breast cancer. CancerRes. 2004;64(3):830–5.

17. Armes JE, Venter DJ. The pathology of inherited breast cancer.Pathology. 2002;34(4):309–14.

18. Pinilla SM, Honrado E, Hardisson D, Benitez J, Palacios J.Caveolin-1 expression is associated with a basal-like phenotypein sporadic and hereditary breast cancer. Breast Cancer ResTreat. 2006;99(1):85–90.

19. Elsheikh SE, Green AR, Rakha EA, Samaka RM, Ammar AA,Powe D, et al. Caveolin 1 and Caveolin 2 are associated withbreast cancer basal-like and triple-negative immunophenotype.Br J Cancer. 2008;99(2):327–34.

20. Rakha EA, Elsheikh SE, Aleskandarany MA, Habashi HO,Green AR, Powe DG, et al. Triple-negative breast cancer:distinguishing between basal and nonbasal subtypes. Clin CancerRes. 2009;15(7):2302–10.

21. Freneaux P, Stoppa-Lyonnet D, Mouret E, Kambouchner M,Nicolas A, Zafrani B, et al. Low expression of bcl-2 in Brca1-associated breast cancers. Br J Cancer. 2000;83(10):1318–22.

22. Vaziri SA, Tubbs RR, Darlington G, Casey G. Absence ofCCND1 gene amplification in breast tumours of BRCA1mutation carriers. Mol Pathol. 2001;54(4):259–63.

23. Elsheikh S, Green AR, Aleskandarany MA, Grainge M, PaishCE, Lambros MB, et al. CCND1 amplification and cyclin D1expression in breast cancer and their relation with proteomicsubgroups and patient outcome. Breast Cancer Res Treat.2008;109(2):325–35.

24. Palacios J, Honrado E, Osorio A, Cazorla A, Sarrio D, BarrosoA, et al. Immunohistochemical characteristics defined by tissuemicroarray of hereditary breast cancer not attributable to BRCA1or BRCA2 mutations: differences from breast carcinomas arisingin BRCA1 and BRCA2 mutation carriers. Clin Cancer Res.2003;9(10 Pt 1):3606–14.

25. Cortesi L, Turchetti D, Bertoni C, Bellei R, Mangone L, VincetiM, et al. Comparison between genotype and phenotype identifiesa high-risk population carrying BRCA1 mutations. GenesChromosom Cancer. 2000;27(2):130–5.

26. van der Groep P, Bouter A, van der Zanden R, Siccama I, MenkoFH, Gille JJ, et al. Distinction between hereditary and sporadicbreast cancer on the basis of clinicopathological data. J ClinPathol. 2006;59(6):611–7.

27. Gadzicki D, Schubert A, Fischer C, Milde S, Lehmann U,Steinemann D, et al. Histopathological criteria and selectionalgorithms for BRCA1 genetic testing. Cancer Genet Cytogenet.2009;189(2):105–11.

28. Palacios J, Honrado E, Osorio A, Cazorla A, Sarrio D, BarrosoA, et al. Phenotypic characterization of BRCA1 and BRCA2tumors based in a tissue microarray study with 37 immunohis-tochemical markers. Breast Cancer Res Treat. 2005;90(1):5–14.

29. Visscher DW, Sarkar FH, Shimoyama RK, Crissman JD.Correlation between p53 immunostaining patterns and genesequence mutations in breast carcinoma. Diagn Mol Pathol.1996;5(3):187–93.

30. Phillips KA, Nichol K, Ozcelik H, Knight J, Done SJ, GoodwinPJ, et al. Frequency of p53 mutations in breast carcinomas fromAshkenazi Jewish carriers of BRCA1 mutations. J Natl CancerInst. 1999;91(5):469–73.

31. Crook T, Brooks LA, Crossland S, Osin P, Barker KT, Waller J,et al. p53 mutation with frequent novel condons but not amutator phenotype in BRCA1- and BRCA2-associated breasttumours. Oncogene. 1998;17(13):1681–9.

32. Holstege H, Joosse SA, van Oostrom CT, Nederlof PM, de VriesA, Jonkers J. High incidence of protein-truncating TP53mutations in BRCA1-related breast cancer. Cancer Res.2009;69(8):3625–33.

33. Manie E, Vincent-Salomon A, Lehmann-Che J, Pierron G,Turpin E, Warcoin M, et al. High frequency of TP53 mutationin BRCA1 and sporadic basal-like carcinomas but not in BRCA1luminal breast tumors. Cancer Res. 2009;69(2):663–71.

34. Laakso M, Loman N, Borg A, Isola J. Cytokeratin 5/14-positivebreast cancer: true basal phenotype confined to BRCA1 tumors.Mod Pathol. 2005;18(10):1321–8.

35. Mulligan AM, Pinnaduwage D, Bane AL, Bull SB, O'MalleyFP, Andrulis IL. CK8/18 expression, the basal phenotype, andfamily history in identifying BRCA1-associated breast cancerin the Ontario site of the Breast Cancer Family Registry. Cancer2010.

36. Bane AL, Pinnaduwage D, Colby S, Bull SB, O'Malley FP,Andrulis IL. Expression profiling of familial breast cancersdemonstrates higher expression of FGFR2 in BRCA2-associatedtumors. Breast Cancer Res Treat. 2009;117(1):183–91.

37. Domagala P, Huzarski T, Lubinski J, Gugala K, Domagala W.Immunophenotypic predictive profiling of BRCA1-associatedbreast cancer. Virchows Arch 2010.

38. Lim E, Vaillant F, Wu D, Forrest NC, Pal B, Hart AH, et al.Aberrant luminal progenitors as the candidate target populationfor basal tumor development in BRCA1 mutation carriers. NatMed. 2009;15(8):907–13.

39. Lord CJ, Ashworth A. Targeted therapy for cancer using PARPinhibitors. Curr Opin Pharmacol. 2008;8(4):363–9.

40. Tan DS, Marchio C, Reis-Filho JS. Hereditary breast cancer:from molecular pathology to tailored therapies. J Clin Pathol.2008;61(10):1073–82.

J Mammary Gland Biol Neoplasia (2011) 16:27–40 35

Page 10: Phenotype-Genotype Correlation in Familial Breast Cancer

41. Farshid G, Balleine RL, Cummings M, Waring P. Morphology ofbreast cancer as a means of triage of patients for BRCA1 genetictesting. Am J Surg Pathol. 2006;30(11):1357–66.

42. Lidereau R, Eisinger F, Champeme MH, Nogues C, Bieche I,Birnbaum D, et al. Major improvement in the efficacy ofBRCA1 mutation screening using morphoclinical features ofbreast cancer. Cancer Res. 2000;60(5):1206–10.

43. Chang J, Hilsenbeck SG, Sng JH, Wong J, Ragu GC.Pathological features and BRCA1 mutation screening in pre-menopausal breast cancer patients. Clin Cancer Res. 2001;7(6):1739–42.

44. Eisinger F, Nogues C, Guinebretiere JM, Peyrat JP, Bardou VJ,Noguchi T, et al. Novel indications for BRCA1 screening usingindividual clinical and morphological features. Int J Cancer.1999;84(3):263–7.

45. Arnes JB, Brunet JS, Stefansson I, Begin LR, Wong N, ChappuisPO, et al. Placental cadherin and the basal epithelial phenotypeof BRCA1-related breast cancer. Clin Cancer Res. 2005;11(11):4003–11.

46. Lakhani SR, Van De Vijver MJ, Jacquemier J, Anderson TJ,Osin PP, McGuffog L, et al. The pathology of familial breastcancer: predictive value of immunohistochemical markersestrogen receptor, progesterone receptor, HER-2, and p53 inpatients with mutations in BRCA1 and BRCA2. J Clin Oncol.2002;20(9):2310–8.

47. Rijnsburger AJ, Obdeijn IM, Kaas R, Tilanus-Linthorst MM,Boetes C, Loo CE, et al. BRCA1-Associated Breast CancersPresent Differently From BRCA2-Associated and FamilialCases: Long-Term Follow-Up of the Dutch MRISC ScreeningStudy. J Clin Oncol 2010.

48. Collins LC, Martyniak A, Kandel MJ, Stadler ZK, Masciari S,Miron A, et al. Basal cytokeratin and epidermal growth factorreceptor expression are not predictive of BRCA1 mutation statusin women with triple-negative breast cancers. Am J Surg Pathol.2009;33(7):1093–7.

49. Rakha EA, El-Sheikh SE, Kandil MA, El-Sayed ME, GreenAR, Ellis IO. Expression of BRCA1 protein in breast cancerand its prognostic significance. Hum Pathol. 2008;39(6):857–65.

50. Beger C, Pierce LN, Kruger M, Marcusson EG, Robbins JM,Welcsh P, et al. Identification of Id4 as a regulator of BRCA1expression by using a ribozyme-library-based inverse genomicsapproach. Proc Natl Acad Sci USA. 2001;98(1):130–5.

51. Turner NC, Reis-Filho JS, Russell AM, Springall RJ, Ryder K,Steele D, et al. BRCA1 dysfunction in sporadic basal-like breastcancer. Oncogene. 2007;26(14):2126–32.

52. Moskwa P, Buffa FM, Pan Y, Panchakshari R, Gottipati P,Muschel RJ, et al. miR-182-mediated downregulation of BRCA1impacts DNA repair and sensitivity to PARP inhibitors. MolCell. 2011;41(2):210–20.

53. Byrski T, Huzarski T, Dent R, Gronwald J, Zuziak D, CybulskiC, et al. Response to neoadjuvant therapy with cisplatin inBRCA1-positive breast cancer patients. Breast Cancer Res Treat.2009;115(2):359–63.

54. Byrski T, Gronwald J, Huzarski T, Grzybowska E, Budryk M,Stawicka M, et al. Pathologic complete response rates in youngwomen with BRCA1-positive breast cancers after neoadjuvantchemotherapy. J Clin Oncol. 2010;28(3):375–9.

55. Rouzier R, Perou CM, Symmans WF, Ibrahim N, CristofanilliM, Anderson K, et al. Breast cancer molecular subtypes responddifferently to preoperative chemotherapy. Clin Cancer Res.2005;11(16):5678–85.

56. Chappuis PO, Goffin J, Wong N, Perret C, Ghadirian P, ToninPN, et al. A significant response to neoadjuvant chemotherapy inBRCA1/2 related breast cancer. J Med Genet. 2002;39(8):608–10.

57. Simpson PT, Vargas AC, Al-Ejeh F, Khanna KK, Chenevix-Trench G, Lakhani SR. Application of molecular findings to thediagnosis and management of breast disease: recent advancesand challenges. Hum Pathol 2010.

58. Balleine RL, Provan PJ, Pupo GM, Pathmanathan N, CummingsM, Farshid G, et al. Familial concordance of breast cancerpathology as an indicator of genotype in multiple-case families.Genes Chromosom Cancer. 2010;49(12):1082–94.

59. Honrado E, Osorio A, Milne RL, Paz MF, Melchor L, CasconA, et al. Immunohistochemical classification of non-BRCA1/2tumors identifies different groups that demonstrate theheterogeneity of BRCAX families. Mod Pathol. 2007;20(12):1298–306.

60. Da Silva L, Lakhani SR. Pathology of hereditary breast cancer.Mod Pathol. 2010;23 Suppl 2:S46–51.

61. Moffa AB, Tannheimer SL, Ethier SP. Transforming potential ofalternatively spliced variants of fibroblast growth factor receptor2 in human mammary epithelial cells. Mol Cancer Res. 2004;2(11):643–52.

62. Xu X, Qiao W, Linke SP, Cao L, Li WM, Furth PA, et al.Genetic interactions between tumor suppressors Brca1 and p53in apoptosis, cell cycle and tumorigenesis. Nat Genet. 2001;28(3):266–71.

63. Ongusaha PP, Ouchi T, Kim KT, Nytko E, Kwak JC, Duda RB,et al. BRCA1 shifts p53-mediated cellular outcomes towardsirreversible growth arrest. Oncogene. 2003;22(24):3749–58.

64. Cheung AM, Elia A, Tsao MS, Done S, Wagner KU,Hennighausen L, et al. Brca2 deficiency does not impairmammary epithelium development but promotes mammaryadenocarcinoma formation in p53(+/−) mutant mice. CancerRes. 2004;64(6):1959–65.

65. Bane AL, Beck JC, Bleiweiss I, Buys SS, Catalano E, Daly MB,et al. BRCA2 mutation-associated breast cancers exhibit adistinguishing phenotype based on morphology and molecularprofiles from tissue microarrays. Am J Surg Pathol. 2007;31(1):121–8.

66. Simpson PT, Reis-Filho JS, Lambros MB, Jones C, Steele D,Mackay A, et al. Molecular profiling pleomorphic lobularcarcinomas of the breast: evidence for a common moleculargenetic pathway with classic lobular carcinomas. J Pathol.2008;215(3):231–44.

67. Consortium BCL. Pathology of familial breast cancer: differ-ences between breast cancers in carriers of BRCA1 or BRCA2mutations and sporadic cases. Lancet. 1997;349(9064):1505–10.

68. Jacquemler J, Eisinger F, Guinebretiere JM, Stoppa-Lyonnet D,Sobol H. Intraductal component and BRCA1-associated breastcancer. Lancet. 1996;348(9034):1098.

69. Adem C, Reynolds C, Soderberg CL, Slezak JM, McDonnellSK, Sebo TJ, et al. Pathologic characteristics of breastparenchyma in patients with hereditary breast carcinoma,including BRCA1 and BRCA2 mutation carriers. Cancer.2003;97(1):1–11.

70. Claus EB, Petruzella S, Matloff E, Carter D. Prevalence ofBRCA1 and BRCA2 mutations in women diagnosed with ductalcarcinoma in situ. JAMA. 2005;293(8):964–9.

71. Hwang ES, McLennan JL, Moore DH, Crawford BB, EssermanLJ, Ziegler JL. Ductal carcinoma in situ in BRCA mutationcarriers. J Clin Oncol. 2007;25(6):642–7.

72. Gomez Garcia EB, Oosterwijk JC, Timmermans M, van AsperenCJ, Hogervorst FB, Hoogerbrugge N, et al. A method to assessthe clinical significance of unclassified variants in the BRCA1and BRCA2 genes based on cancer family history. Breast CancerRes. 2009;11(1):R8.

73. Pensabene M, Spagnoletti I, Capuano I, Condello C, Pepe S,Contegiacomo A, et al. Two mutations of BRCA2 gene at exon

36 J Mammary Gland Biol Neoplasia (2011) 16:27–40

Page 11: Phenotype-Genotype Correlation in Familial Breast Cancer

and splicing site in a woman who underwent oncogeneticcounseling. Ann Oncol. 2009;20(5):874–8.

74. Osorio A, Milne RL, Honrado E, Barroso A, Diez O, Salazar R,et al. Classification of missense variants of unknown significancein BRCA1 based on clinical and tumor information. Hum Mutat.2007;28(5):477–85.

75. Tommasi S, Pilato B, Pinto R, Monaco A, Bruno M, CampanaM, et al. Molecular and in silico analysis of BRCA1 and BRCA2variants. Mutat Res. 2008;644(1–2):64–70.

76. Spearman AD, Sweet K, Zhou XP, McLennan J, Couch FJ,Toland AE. Clinically applicable models to characterize BRCA1and BRCA2 variants of uncertain significance. J Clin Oncol.2008;26(33):5393–400.

77. Lovelock PK, Healey S, Au W, Sum EY, Tesoriero A, WongEM, et al. Genetic, functional, and histopathological evaluationof two C-terminal BRCA1 missense variants. J Med Genet.2006;43(1):74–83.

78. Goldgar DE, Easton DF, Deffenbaugh AM, Monteiro AN,Tavtigian SV, Couch FJ. Integrated evaluation of DNA sequencevariants of unknown clinical significance: application to BRCA1and BRCA2. Am J Hum Genet. 2004;75(4):535–44.

79. Chenevix-Trench G, Healey S, Lakhani S, Waring P, CummingsM, Brinkworth R, et al. Genetic and histopathologic evaluationof BRCA1 and BRCA2 DNA sequence variants of unknownclinical significance. Cancer Res. 2006;66(4):2019–27.

80. Stacey SN, Manolescu A, Sulem P, Rafnar T, Gudmundsson J,Gudjonsson SA, et al. Common variants on chromosomes 2q35and 16q12 confer susceptibility to estrogen receptor-positivebreast cancer. Nat Genet. 2007;39(7):865–9.

81. Jia C, Cai Y, Ma Y, Fu D. Quantitative assessment of the effectof FGFR2 gene polymorphism on the risk of breast cancer.Breast Cancer Res Treat. 2010;124(2):521–8.

82. Dillon C, Spencer-Dene B, Dickson C. A crucial role for fibroblastgrowth factor signaling in embryonic mammary gland develop-ment. J Mammary Gland Biol Neoplasia. 2004;9(2):207–15.

83. Turner N, Lambros MB, Horlings HM, Pearson A, Sharpe R,Natrajan R, et al. Integrative molecular profiling of triplenegative breast cancers identifies amplicon drivers and potentialtherapeutic targets. Oncogene. 2010;29(14):2013–23.

84. Antoniou AC, Spurdle AB, Sinilnikova OM, Healey S, PooleyKA, Schmutzler RK, et al. Common breast cancer-predispositionalleles are associated with breast cancer risk in BRCA1 andBRCA2 mutation carriers. Am J Hum Genet. 2008;82(4):937–48.

85. Garcia-Closas M, Hall P, Nevanlinna H, Pooley K, Morrison J,Richesson DA, et al. Heterogeneity of breast cancer associationswith five susceptibility loci by clinical and pathologicalcharacteristics. PLoS Genet. 2008;4(4):e1000054.

86. Stacey SN, Manolescu A, Sulem P, Thorlacius S, GudjonssonSA, Jonsson GF, et al. Common variants on chromosome 5p12confer susceptibility to estrogen receptor-positive breast cancer.Nat Genet. 2008;40(6):703–6.

87. Yu K, Ganesan K, Miller LD, Tan P. A modular analysis ofbreast cancer reveals a novel low-grade molecular signature inestrogen receptor-positive tumors. Clin Cancer Res. 2006;12(11Pt 1):3288–96.

88. Engel C, Versmold B, Wappenschmidt B, Simard J, Easton DF,Peock S, et al. Association of the variants CASP8 D302H andCASP10 V410I with breast and ovarian cancer risk in BRCA1and BRCA2 mutation carriers. Cancer Epidemiol Biomark Prev.2010;19(11):2859–68.

89. Antoniou AC, Sinilnikova OM, Simard J, Leone M, DumontM, Neuhausen SL, et al. RAD51 135 G–>C modifies breastcancer risk among BRCA2 mutation carriers: results from acombined analysis of 19 studies. Am J Hum Genet. 2007;81(6):1186–200.

90. Gaudet MM, Kirchhoff T, Green T, Vijai J, Korn JM, GuiducciC, et al. Common genetic variants and modification ofpenetrance of BRCA2-associated breast cancer. PLoS Genet.2010;6(10):e1001183.

91. Hollestelle A, Pelletier C, Hooning M, Crepin E, Schutte M,Look M, et al. Prevalence of the variant allele rs61764370 T>Gin the 3'UTR of KRAS among Dutch BRCA1, BRCA2 and non-BRCA1/BRCA2 breast cancer families. Breast Cancer Res Treat2010.

92. Lynch HT, Smyrk T, Lynch J. An update of HNPCC (Lynchsyndrome). Cancer Genet Cytogenet. 1997;93(1):84–99.

93. Woods MO, Williams P, Careen A, Edwards L, Bartlett S,McLaughlin JR, et al. A new variant database for mismatchrepair genes associated with Lynch syndrome. Hum Mutat.2007;28(7):669–73.

94. Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A,Ruschoff J, et al. Revised Bethesda Guidelines for hereditarynonpolyposis colorectal cancer (Lynch syndrome) and micro-satellite instability. J Natl Cancer Inst. 2004;96(4):261–8.

95. Chiaravalli AM, Furlan D, Facco C, Tibiletti MG, Dionigi A,Casati B, et al. Immunohistochemical pattern of hMSH2/hMLH1in familial and sporadic colorectal, gastric, endometrial andovarian carcinomas with instability in microsatellite sequences.Virchows Arch. 2001;438(1):39–48.

96. Aaltonen LA, Peltomaki P, Leach FS, Sistonen P, Pylkkanen L,Mecklin JP, et al. Clues to the pathogenesis of familial colorectalcancer. Science. 1993;260(5109):812–6.

97. Boland CR, Thibodeau SN, Hamilton SR, Sidransky D, EshlemanJR, Burt RW, et al. A National Cancer Institute Workshop onMicrosatellite Instability for cancer detection and familial predispo-sition: development of international criteria for the determination ofmicrosatellite instability in colorectal cancer. Cancer Res. 1998;58(22):5248–57.

98. Kim H, Piao Z, Kim JW, Choi JS, Kim NK, Lee JM, et al.Expression of hMSH2 and hMLH1 in colorectal carcinomas withmicrosatellite instability. Pathol Res Pract. 1998;194(1):3–9.

99. Dietmaier W, Wallinger S, Bocker T, Kullmann F, Fishel R,Ruschoff J. Diagnostic microsatellite instability: definition andcorrelation with mismatch repair protein expression. Cancer Res.1997;57(21):4749–56.

100. Suraweera N, Duval A, Reperant M, Vaury C, Furlan D, LeroyK, et al. Evaluation of tumor microsatellite instability using fivequasimonomorphic mononucleotide repeats and pentaplex PCR.Gastroenterology. 2002;123(6):1804–11.

101. Muller A, Edmonston TB, Corao DA, Rose DG, Palazzo JP,Becker H, et al. Exclusion of breast cancer as an integral tumorof hereditary nonpolyposis colorectal cancer. Cancer Res.2002;62(4):1014–9.

102. Scott RJ, McPhillips M, Meldrum CJ, Fitzgerald PE, Adams K,Spigelman AD, et al. Hereditary nonpolyposis colorectal cancerin 95 families: differences and similarities between mutation-positive and mutation-negative kindreds. Am J Hum Genet.2001;68(1):118–27.

103. Oliveira Ferreira F, Napoli Ferreira CC, Rossi BM, ToshihikoNakagawa W, Aguilar Jr S, Monteiro Santos EM, et al.Frequency of extra-colonic tumors in hereditary nonpolyposiscolorectal cancer (HNPCC) and familial colorectal cancer (FCC)Brazilian families: an analysis by a Brazilian HereditaryColorectal Cancer Institutional Registry. Fam Cancer. 2004;3(1):41–7.

104. Shanley S, Fung C, Milliken J, Leary J, Barnetson R, SchnitzlerM, et al. Breast cancer immunohistochemistry can be useful intriage of some HNPCC families. Fam Cancer. 2009;8(3):251–5.

105. Westenend PJ, Schutte R, Hoogmans MM, Wagner A, DinjensWN. Breast cancer in an MSH2 gene mutation carrier. HumPathol. 2005;36(12):1322–6.

J Mammary Gland Biol Neoplasia (2011) 16:27–40 37

Page 12: Phenotype-Genotype Correlation in Familial Breast Cancer

106. Barrow E, Robinson L, Alduaij W, Shenton A, Clancy T, LallooF, et al. Cumulative lifetime incidence of extracolonic cancers inLynch syndrome: a report of 121 families with proven mutations.Clin Genet. 2009;75(2):141–9.

107. Wasielewski M, Riaz M, Vermeulen J, van den Ouweland A,Labrijn-Marks I, Olmer R, et al. Association of rare MSH6variants with familial breast cancer. Breast Cancer Res Treat.2010;123(2):315–20.

108. Jensen UB, Sunde L, Timshel S, Halvarsson B, Nissen A,Bernstein I, et al. Mismatch repair defective breast cancer in thehereditary nonpolyposis colorectal cancer syndrome. BreastCancer Res Treat. 2009;120(3):777–82.

109. Walsh MD, Buchanan DD, Cummings MC, Pearson SA, ArnoldST, Clendenning M, et al. Lynch syndrome-associated breastcancers: clinicopathologic characteristics of a case series fromthe colon cancer family registry. Clin Cancer Res. 2010;16(7):2214–24.

110. Schmitt FC, Soares R, Gobbi H, Milanezzi F, Santos-Silva F,Cirnes L, et al. Microsatellite instability in medullary breastcarcinomas. Int J Cancer. 1999;82(5):644–7.

111. Smyrk TC, Watson P, Kaul K, Lynch HT. Tumor-infiltratinglymphocytes are a marker for microsatellite instability incolorectal carcinoma. Cancer. 2001;91(12):2417–22.

112. Greenson JK, Bonner JD, Ben-Yzhak O, Cohen HI, MiselevichI, Resnick MB, et al. Phenotype of microsatellite unstablecolorectal carcinomas: well-differentiated and focally mucinoustumors and the absence of dirty necrosis correlate with micro-satellite instability. Am J Surg Pathol. 2003;27(5):563–70.

113. Lacroix-Triki M, Lambros MB, Geyer FC, Suarez PH, Reis-FilhoJS, Weigelt B. Absence of microsatellite instability in mucinouscarcinomas of the breast. Int J Clin Exp Pathol. 2010;4(1):22–31.

114. Kim H, Jung JK, Park JH, Park C. Immunohistochemicalcharacteristics of colorectal carcinoma with DNA replicationerrors. J Korean Med Sci. 1996;11(2):137–43.

115. Lindor NM, Burgart LJ, Leontovich O, Goldberg RM, CunninghamJM, Sargent DJ, et al. Immunohistochemistry versus microsatelliteinstability testing in phenotyping colorectal tumors. J Clin Oncol.2002;20(4):1043–8.

116. Vasen HF, Morreau H, Nortier JW. Is breast cancer part of thetumor spectrum of hereditary nonpolyposis colorectal cancer?Am J Hum Genet. 2001;68(6):1533–5.

117. de Leeuw WJ, van Puijenbroek M, Tollenaar RA, Cornelisse CJ,Vasen HF, Morreau H. Correspondence re: A. Muller et al.,Exclusion of breast cancer as an integral tumor of hereditarynonpolyposis colorectal cancer. Cancer Res., 62: 1014–1019,2002. Cancer Res 2003;63(5):1148–9

118. Adem C, Soderberg CL, Cunningham JM, Reynolds C, Sebo TJ,Thibodeau SN, et al. Microsatellite instability in hereditary andsporadic breast cancers. Int J Cancer. 2003;107(4):580–2.

119. Khilko N, Bourne P, Qi Y, Ping T. Mismatch repair geneshMLH1 and hMSH2 may not play an essential role in breastcarcinogenesis. Int J Surg Pathol. 2007;15(3):233–41.

120. Pharoah PD, Guilford P, Caldas C. Incidence of gastric cancerand breast cancer in CDH1 (E-cadherin) mutation carriers fromhereditary diffuse gastric cancer families. Gastroenterology.2001;121(6):1348–53.

121. Wijnhoven BP, Dinjens WN, Pignatelli M. E-cadherin-catenincell-cell adhesion complex and human cancer. Br J Surg.2000;87(8):992–1005.

122. Suriano G, Yew S, Ferreira P, Senz J, Kaurah P, Ford JM, et al.Characterization of a recurrent germ line mutation of the E-cadherin gene: implications for genetic testing and clinicalmanagement. Clin Cancer Res. 2005;11(15):5401–9.

123. Schrader KA, Masciari S, Boyd N, Salamanca C, Senz J,Saunders DN, et al. Germline mutations in CDH1 are infrequent

in women with early-onset or familial lobular breast cancers. JMed Genet 2010.

124. Kaurah P, MacMillan A, Boyd N, Senz J, De Luca A, Chun N, etal. Founder and recurrent CDH1 mutations in families withhereditary diffuse gastric cancer. JAMA. 2007;297(21):2360–72.

125. Keller G, Vogelsang H, Becker I, Hutter J, Ott K, Candidus S, etal. Diffuse type gastric and lobular breast carcinoma in a familialgastric cancer patient with an E-cadherin germline mutation. AmJ Pathol. 1999;155(2):337–42.

126. Brooks-Wilson AR, Kaurah P, Suriano G, Leach S, Senz J,Grehan N, et al. Germline E-cadherin mutations in hereditarydiffuse gastric cancer: assessment of 42 new families andreview of genetic screening criteria. J Med Genet. 2004;41(7):508–17.

127. Masciari S, Larsson N, Senz J, Boyd N, Kaurah P, Kandel MJ, etal. Germline E-cadherin mutations in familial lobular breastcancer. J Med Genet. 2007;44(11):726–31.

128. Lei H, Sjoberg-Margolin S, Salahshor S, Werelius B, JandakovaE, Hemminki K, et al. CDH1 mutations are present in bothductal and lobular breast cancer, but promoter allelic variantsshow no detectable breast cancer risk. Int J Cancer. 2002;98(2):199–204.

129. Hemminki K, Granstrom C. Morphological types of breastcancer in family members and multiple primary tumours: ismorphology genetically determined? Breast Cancer Res. 2002;4(4):R7.

130. Zhu ZG, Yu YY, Zhang Y, Ji J, Zhang J, Liu BY, et al. Germlinemutational analysis of CDH1 and pathologic features in familialcancer syndrome with diffuse gastric cancer/breast cancerproband in a Chinese family. Eur J Surg Oncol. 2004;30(5):531–5.

131. Da Silva L, Parry S, Reid L, Keith P, Waddell N, Kossai M, et al.Aberrant expression of E-cadherin in lobular carcinomas of thebreast. Am J Surg Pathol. 2008;32(5):773–83.

132. Eng C. PTEN: one gene, many syndromes. Hum Mutat. 2003;22(3):183–98.

133. Marsh DJ, Coulon V, Lunetta KL, Rocca-Serra P, Dahia PL,Zheng Z, et al. Mutation spectrum and genotype-phenotypeanalyses in Cowden disease and Bannayan-Zonana syndrome,two hamartoma syndromes with germline PTEN mutation. HumMol Genet. 1998;7(3):507–15.

134. Petrocelli T, Slingerland JM. PTEN deficiency: a role inmammary carcinogenesis. Breast Cancer Res. 2001;3(6):356–60.

135. Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, et al.PTEN, a putative protein tyrosine phosphatase gene mutated inhuman brain, breast, and prostate cancer. Science. 1997;275(5308):1943–7.

136. Liaw D, Marsh DJ, Li J, Dahia PL, Wang SI, Zheng Z, et al.Germline mutations of the PTEN gene in Cowden disease, aninherited breast and thyroid cancer syndrome. Nat Genet.1997;16(1):64–7.

137. Bau MG, Arisio R, Cristini G, Bertone E, Campogrande M.Screening-detected breast carcinoma in a patient with Cowdensyndrome. Breast. 2004;13(3):239–41.

138. Depowski PL, Rosenthal SI, Ross JS. Loss of expression of thePTEN gene protein product is associated with poor outcome inbreast cancer. Mod Pathol. 2001;14(7):672–6.

139. Uppal S, Mistry D, Coatesworth AP. Cowden disease: a review.Int J Clin Pract. 2007;61(4):645–52.

140. Tsao H. Update on familial cancer syndromes and the skin. J AmAcad Dermatol. 2000;42(6):939–69. quiz 970–2.

141. Fackenthal JD, Marsh DJ, Richardson AL, Cummings SA, EngC, Robinson BG, et al. Male breast cancer in Cowden syndromepatients with germline PTEN mutations. J Med Genet. 2001;38(3):159–64.

38 J Mammary Gland Biol Neoplasia (2011) 16:27–40

Page 13: Phenotype-Genotype Correlation in Familial Breast Cancer

142. Kriege M, Brekelmans CT, Boetes C, Besnard PE, ZonderlandHM, Obdeijn IM, et al. Efficacy of MRI and mammography forbreast-cancer screening in women with a familial or geneticpredisposition. N Engl J Med. 2004;351(5):427–37.

143. Schrager CA, Schneider D, Gruener AC, Tsou HC, Peacocke M.Clinical and pathological features of breast disease in Cowden'ssyndrome: an underrecognized syndrome with an increased riskof breast cancer. Hum Pathol. 1998;29(1):47–53.

144. Lachlan KL, Lucassen AM, Bunyan D, Temple IK. Cowdensyndrome and Bannayan Riley Ruvalcaba syndrome representone condition with variable expression and age-related pene-trance: results of a clinical study of PTEN mutation carriers. JMed Genet. 2007;44(9):579–85.

145. Rhei E, Kang L, Bogomolniy F, Federici MG, Borgen PI, BoydJ. Mutation analysis of the putative tumor suppressor genePTEN/MMAC1 in primary breast carcinomas. Cancer Res.1997;57(17):3657–9.

146. Braud AC, de Rocquancourt A, Marty M, Espie M. Cowdendisease and Lhermitte Duclos disease, markers of breastcarcinoma: report of two patients. Ann Oncol. 1999;10(10):1241–3.

147. Reifenberger J, Rauch L, Beckmann MW, Megahed M, RuzickaT, Reifenberger G. Cowden's disease: clinical and moleculargenetic findings in a patient with a novel PTEN germlinemutation. Br J Dermatol. 2003;148(5):1040–6.

148. Sabate JM, Gomez A, Torrubia S, Blancas C, Sanchez G, AlonsoMC, et al. Evaluation of breast involvement in relation toCowden syndrome: a radiological and clinicopathological studyof patients with PTEN germ-line mutations. Eur Radiol. 2006;16(3):702–6.

149. Singh B, Ittmann MM, Krolewski JJ. Sporadic breast cancersexhibit loss of heterozygosity on chromosome segment 10q23close to the Cowden disease locus. Genes Chromosom Cancer.1998;21(2):166–71.

150. Bose S, Wang SI, Terry MB, Hibshoosh H, Parsons R. Allelicloss of chromosome 10q23 is associated with tumor progressionin breast carcinomas. Oncogene. 1998;17(1):123–7.

151. Dedes KJ, Wetterskog D, Mendes-Pereira AM, Natrajan R,Lambros MB, Geyer FC, et al. PTEN deficiency in endometrioidendometrial adenocarcinomas predicts sensitivity to PARPinhibitors. Sci Transl Med. 2010;2(53):53ra75.

152. Mendes-Pereira AM, Martin SA, Brough R, McCarthy A,Taylor JR, Kim JS, et al. Synthetic lethal targeting of PTENmutant cells with PARP inhibitors. EMBO Mol Med. 2009;1(6–7):315–22.

153. Li FP, Fraumeni Jr JF. Soft-tissue sarcomas, breast cancer, andother neoplasms. A familial syndrome? Ann Intern Med.1969;71(4):747–52.

154. Malkin D, Li FP, Strong LC, Fraumeni Jr JF, Nelson CE, KimDH, et al. Germ line p53 mutations in a familial syndrome ofbreast cancer, sarcomas, and other neoplasms. Science. 1990;250(4985):1233–8.

155. Li FP, Fraumeni Jr JF, Mulvihill JJ, Blattner WA, Dreyfus MG,Tucker MA, et al. A cancer family syndrome in twenty-fourkindreds. Cancer Res. 1988;48(18):5358–62.

156. Tinat J, Bougeard G, Baert-Desurmont S, Vasseur S, Martin C,Bouvignies E, et al. 2009 version of the Chompret criteria for LiFraumeni syndrome. J Clin Oncol. 2009;27(26):e108–9. authorreply e110.

157. Varley JM. Germline TP53 mutations and Li-Fraumeni syn-drome. Hum Mutat. 2003;21(3):313–20.

158. Yamada H, Shinmura K, Yamamura Y, Kurachi K, Nakamura T,Tsuneyoshi T, et al. Identification and characterization of a novelgermline p53 mutation in a patient with glioblastoma and coloncancer. Int J Cancer. 2009;125(4):973–6.

159. Ruijs MW, Verhoef S, Rookus MA, Pruntel R, van der Hout AH,Hogervorst FB, et al. TP53 germline mutation testing in 180families suspected of Li-Fraumeni syndrome: mutation detectionrate and relative frequency of cancers in different familialphenotypes. J Med Genet. 2010;47(6):421–8.

160. Blanco A, Grana B, Fachal L, Santamarina M, Cameselle-Teijeiro J, Ruiz-Ponte C, et al. Beyond BRCA1 and BRCA2wild-type breast and/or ovarian cancer families: germlinemutations in TP53 and PTEN. Clin Genet. 2010;77(2):193–6.

161. Gonzalez KD, Noltner KA, Buzin CH, Gu D, Wen-Fong CY,Nguyen VQ, et al. Beyond Li Fraumeni Syndrome: clinicalcharacteristics of families with p53 germline mutations. J ClinOncol. 2009;27(8):1250–6.

162. Wilson JR, Bateman AC, Hanson H, An Q, Evans G, Rahman N, etal. A novel HER2-positive breast cancer phenotype arising fromgermline TP53 mutations. J Med Genet. 2010;47(11):771–4.

163. Birch JM, Alston RD, McNally RJ, Evans DG, Kelsey AM, HarrisM, et al. Relative frequency and morphology of cancers in carriersof germline TP53 mutations. Oncogene. 2001;20(34):4621–8.

164. Savitsky K, Bar-Shira A, Gilad S, Rotman G, Ziv Y, VanagaiteL, et al. A single ataxia telangiectasia gene with a product similarto PI-3 kinase. Science. 1995;268(5218):1749–53.

165. Gatei M, Scott SP, Filippovitch I, Soronika N, Lavin MF, WeberB, et al. Role for ATM in DNA damage-induced phosphorylationof BRCA1. Cancer Res. 2000;60(12):3299–304.

166. Shiloh Y. ATM and related protein kinases: safeguarding genomeintegrity. Nat Rev Cancer. 2003;3(3):155–68.

167. Thompson D, Duedal S, Kirner J, McGuffog L, Last J, ReimanA, et al. Cancer risks and mortality in heterozygous ATMmutation carriers. J Natl Cancer Inst. 2005;97(11):813–22.

168. Renwick A, Thompson D, Seal S, Kelly P, Chagtai T, Ahmed M,et al. ATM mutations that cause ataxia-telangiectasia are breastcancer susceptibility alleles. Nat Genet. 2006;38(8):873–5.

169. Paglia LL, Lauge A, Weber J, Champ J, Cavaciuti E, Russo A, etal. ATM germline mutations in women with familial breastcancer and a relative with haematological malignancy. BreastCancer Res Treat. 2010;119(2):443–52.

170. Fletcher O, Johnson N. dos Santos Silva I, Orr N, Ashworth A,Nevanlinna H, et al. Missense variants in ATM in 26,101 breastcancer cases and 29,842 controls. Cancer Epidemiol BiomarkPrev. 2010;19(9):2143–51.

171. Tavtigian SV, Oefner PJ, Babikyan D, Hartmann A, Healey S, LeCalvez-Kelm F, et al. Rare, evolutionarily unlikely missensesubstitutions in ATM confer increased risk of breast cancer. Am JHum Genet. 2009;85(4):427–46.

172. Balleine RL, Murali R, Bilous AM, Farshid G, Waring P, ProvanP, et al. Histopathological features of breast cancer in carriers ofATM gene variants. Histopathology. 2006;49(5):523–32.

173. Tommiska J, Bartkova J, Heinonen M, Hautala L, Kilpivaara O,Eerola H, et al. The DNA damage signalling kinase ATM isaberrantly reduced or lost in BRCA1/BRCA2-deficient and ER/PR/ERBB2-triple-negative breast cancer. Oncogene. 2008;27(17):2501–6.

174. Barroso E, Pita G, Arias JI, Menendez P, Zamora P, Blanco M, etal. The Fanconi anemia family of genes and its correlation withbreast cancer susceptibility and breast cancer features. BreastCancer Res Treat. 2009;118(3):655–60.

175. D'Andrea AD. Susceptibility pathways in Fanconi's anemia andbreast cancer. N Engl J Med. 2010;362(20):1909–19.

176. Levy-Lahad E. Fanconi anemia and breast cancer susceptibilitymeet again. Nat Genet. 2010;42(5):368–9.

177. Auerbach AD. Fanconi anemia and its diagnosis. Mutat Res.2009;668(1–2):4–10.

178. Stratton MR, Rahman N. The emerging landscape of breastcancer susceptibility. Nat Genet. 2008;40(1):17–22.

J Mammary Gland Biol Neoplasia (2011) 16:27–40 39

Page 14: Phenotype-Genotype Correlation in Familial Breast Cancer

179. Berwick M, Satagopan JM, Ben-Porat L, Carlson A, Mah K, HenryR, et al. Genetic heterogeneity among Fanconi anemia hetero-zygotes and risk of cancer. Cancer Res. 2007;67(19):9591–6.

180. Tischkowitz M, Xia B, Sabbaghian N, Reis-Filho JS, Hamel N,Li G, et al. Analysis of PALB2/FANCN-associated breast cancerfamilies. Proc Natl Acad Sci USA. 2007;104(16):6788–93.

181. Tischkowitz M, Xia B. PALB2/FANCN: recombining cancer andFanconi anemia. Cancer Res. 2010;70(19):7353–9.

182. Mavaddat N, Dunning AM, Ponder BA, Easton DF, Pharoah PD.Common genetic variation in candidate genes and susceptibilityto subtypes of breast cancer. Cancer Epidemiol Biomark Prev.2009;18(1):255–9.

183. San Filippo J, Sung P, Klein H. Mechanism of eukaryotichomologous recombination. Annu Rev Biochem. 2008;77:229–57.

184. Vaz F, Hanenberg H, Schuster B, Barker K, Wiek C, Erven V, etal. Mutation of the RAD51C gene in a Fanconi anemia-likedisorder. Nat Genet. 2010;42(5):406–9.

185. Zheng Y, Zhang J, Hope K, Niu Q, Huo D, Olopade OI.Screening RAD51C nucleotide alterations in patients with afamily history of breast and ovarian cancer. Breast Cancer ResTreat. 2010;124(3):857–61.

186. Scully R, Chen J, Plug A, Xiao Y, Weaver D, Feunteun J, et al.Association of BRCA1 with Rad51 in mitotic and meiotic cells.Cell. 1997;88(2):265–75.

187. Barbano R, Copetti M, Perrone G, Pazienza V, Muscarella LA,Balsamo T, et al. High RAD51 mRNA expression characterizeER-positive/PR-negative breast cancers and is associated withpatient's outcome. Int J Cancer 2010.

188. Chehab NH, Malikzay A, Appel M, Halazonetis TD. Chk2/hCds1 functions as a DNA damage checkpoint in G(1) bystabilizing p53. Genes Dev. 2000;14(3):278–88.

189. Lee JS, Collins KM, Brown AL, Lee CH, Chung JH. hCds1-mediated phosphorylation of BRCA1 regulates the DNA damageresponse. Nature. 2000;404(6774):201–4.

190. Wu X, Webster SR, Chen J. Characterization of tumor-associatedChk2 mutations. J Biol Chem. 2001;276(4):2971–4.

191. Vahteristo P, Bartkova J, Eerola H, Syrjakoski K, Ojala S,Kilpivaara O, et al. A CHEK2 genetic variant contributing to asubstantial fraction of familial breast cancer. Am J Hum Genet.2002;71(2):432–8.

192. Iniesta MD, Gorin MA, Chien LC, Thomas SM, Milliron KJ,Douglas JA, et al. Absence of CHEK2*1100delC mutation infamilies with hereditary breast cancer in North America. CancerGenet Cytogenet. 2010;202(2):136–40.

193. Zhang S, Phelan CM, Zhang P, Rousseau F, Ghadirian P,Robidoux A, et al. Frequency of the CHEK2 1100delC mutationamong women with breast cancer: an international study. CancerRes. 2008;68(7):2154–7.

194. Schmidt MK, Tollenaar RA, de Kemp SR, Broeks A, CornelisseCJ, Smit VT, et al. Breast cancer survival and tumor character-istics in premenopausal women carrying the CHEK2*1100delCgermline mutation. J Clin Oncol. 2007;25(1):64–9.

195. de Bock GH, Schutte M, Krol-Warmerdam EM, Seynaeve C,Blom J, Brekelmans CT, et al. Tumour characteristics andprognosis of breast cancer patients carrying the germlineCHEK2*1100delC variant. J Med Genet. 2004;41(10):731–5.

40 J Mammary Gland Biol Neoplasia (2011) 16:27–40