An intervention study initiated at age 4 months compared the impact of tamoxifen (25 mg), raloxifene (22.5 mg), and letrozole (2.5 mg) administered by 60-day release subcutaneous pellet on mammary preneoplasia prevalence at age 6 months in conditional genetically engineered mouse models with different Breast cancer 1 (Brca1) gene dosages targeted to mammary epithelial cells and germline Tumor protein P53 (Trp53) haploinsufficiency (10–16/cohort). The proportion of unexposed control mice demonstrating mammary preneoplasia at age 6 months was highest in Brca1fl11/fl11/Cre/p53−/+ (54%) mice followed by Brca1WT/fl11/Cre/p53−/+ mice (30%). By age 12 months, invasive mammary cancers appeared in 80% of Brca1fl11/fl11/Cre/p53−/+and 42% of Brca1WT/fl11/Cre/p53−/+control unexposed mice. The spectrum of cancer histology was similar in both models without somatic mutation of the nongenetically engineered Brca1, Trp53, Brca2, or Death-associated protein kinase 3 (Dapk3) alleles. Two-month exposure to tamoxifen, raloxifene, and letrozole significantly reduced estrogen-mediated tertiary branching by 65%, 71%, and 78%, respectively, in Brca1fl11/fl11/Cre/p53−/+mice at age 6 months. However, only letrozole significantly reduced hyperplastic alveolar nodules (HAN) prevalence (by 52%) and number (by 30%) and invasive cancer appeared despite tamoxifen exposure. In contrast, tamoxifen significantly reduced HAN number by 95% in Brca1WT/fl11/Cre/p53−/+ mice. Control mice with varying combinations of the different genetically modified alleles and MMTV-Cre transgene demonstrated that the combination of Brca1 insufficiency and Trp53 haploinsufficiency was required for appearance of preneoplasia and no individual genetic alteration confounded the response to tamoxifen. In summary, although specific antihormonal approaches showed effectiveness, with Brca1 gene dosage implicated as a possible modifying variable, more effective chemopreventive approaches for Brca1 mutation–induced cancer may require alternative and/or additional agents. Cancer Prev Res; 10(4); 244–54. ©2017 AACR.

Research into breast cancer prevention in the setting of Breast cancer 1 (BRCA1) mutation continues due to considerations of efficacy and side-effects of current approaches (1–3), in conjunction with the possibility of developing more effective chemopreventive methods in the future (4–7). Tamoxifen (1, 2, 8, 9), raloxifene (8, 10), and letrozole (11) have all been proposed as risk-reducing antihormonals for women carrying germline BRCA1 mutations. Conditional genetically engineered mouse models (GEMM) where both Brca1 alleles are disrupted in mammary epithelial cells (MEC) coupled with Tumor protein P53 (Trp53) haploinsufficiency have been recurrently employed to study pathophysiology, prevention, and treatment of mammary cancer related to loss of Brca1 function (5–7, 12, 13). Mammary cancers in these models are predominantly triple negative and demonstrate a range of histologies (5, 12–14). To model the situation in women, where only one BRCA1 allele is mutated (1, 2), here we characterized and compared mammary cancer and preneoplasia pathophysiology and preventive approaches in GEMM with one (Brca1wildtype (WT)/floxed (fl11)/Cre/p53−/+) versus two Brca1 (Brca1floxed (fl)11/fl11/Cre/p53−/+) alleles disrupted in combination with Trp53 haploinsufficiency. Mammary cancer development in mice is typically preceded by the appearance of preneoplastic lesions including hyperplastic alveolar nodules (HAN), a measure that can be reliably and reproducibly quantified on mammary gland whole mounts (5, 13–16). In women, the presence of mammary epithelial cell cytologic atypia has been discussed as an indicator of breast cancer risk (17).

Loss or decreased TRP53 function has been proposed as an early event in Brca1 mutation-related breast cancer progression with BRCA1 loss of heterozygosity (LOH) occurring later and perhaps not required for carcinogenesis (18). Trp53 heterozygosity has been observed to be obligatory for mammary cancer generation in GEMM with both one and two Brca1 alleles disrupted (12, 19). Loss of PTEN (18), pathogenic mutations in Death-associated protein kinase 3 (DAPK3), Transmembrane protein 135 (TMEM135), and GATA binding protein 4 (GATA4; ref. 20), and kindreds showing mutation in both BRCA1 and Breast cancer 2 (BRCA2; ref. 21) have been reported to contribute to human breast cancer generation. Trp53 LOH has been reported in mammary cancer cell cultures derived from Brca1fl11/fl11/Cre/p53−/+ mice (22).

In the mammary gland, hormonal signaling stimulates tertiary branching and MEC proliferation (23, 24). Tamoxifen is reported to reduce levels of MEC proliferation (25). Loss of BRCA1 can enhance estrogen and progesterone signaling in human breast cells (4, 26, 27) and GEMM with disrupted Brca1 alleles also demonstrate abnormal growth responses to estrogen and/or progesterone pathway stimulation (14, 23, 28). Loss of BRCA1 in cancer cells can interrupt the inhibitory action of tamoxifen on estrogen signaling (13, 29). Tamoxifen exposure promoted mammary cancer development in Brca1fl11/fl11/Cre/p53−/+ mice in a previous study (13). Here we specifically examined whether or not loss of Brca1 or any of the other genetic components of the Brca1fl11/fl11/Cre/p53−/+ mice interrupted the inhibitory action of tamoxifen in mammary tissue and compared the impact of tamoxifen to the alternative selective estrogen modulator raloxifene and the nonsteroidal aromatase inhibitor letrozole.

Mouse models and treatment groups

Nulliparous C57Bl/6 female mice carrying different combinations of genetically engineered modifications in Brca1 (30), Trp53 (31), the MMTV-Cre D transgene, which targeted loss of full-length Brca1 to MEC (32), or no genetic modifications (wild-type: WT) were generated in breeding colonies at Georgetown University (Washington, DC). Control and treatment mice for each genotype were derived from the same genetic crosses. Brca1WT/fl11/Cre/p53−/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53 (p53)−/+, and MMTV-Cre (Cre) mice were derived from Brca1fl11/fl11/Cre/p53−/+ mice, maintained since 2002 on a C57Bl/6 background, and bred to C57Bl/6 WT mice from our breeding colony. Strain 129 embryonic stem cells were used for Brca1fl11(NIH) and disrupted Trp53 (AB1) allele generation (NIH; refs. 30, 31). Mice were maintained in barrier zones, weaned (age 3 weeks), genotyped (Transnetyx Inc.), allocated in turn to different observation/treatment groups for inclusion in the study based on genotype, and maintained in single-sex sterilized ventilated cages with corncob bedding (1–5 mice per cage) with ad libitum access to water/chow under 12-hour dark/light cycles at Georgetown University (Washington, DC). Genotypes demonstrated equivalent weights, growth, activity, and fertility. Observation groups (Ob) were aged to 12 months for appearance of tumor. Treatment/control groups were aged to 4 months for tamoxifen (T; 25 mg/60 day/subcutaneous pellet_12.99 mg/kg/day, Innovative Research of America; refs. 13, 15), raloxifene (R; 22.5 mg/60 day/subcutaneous pellet_11.44 mg/kg/day, Innovative Research of America; refs. 33, 34), letrozole (L; 2.5 mg/60 day/subcutaneous pellet_1.29 mg/kg/day, Innovative Research of America; ref. 15), placebo pellet (P; Innovative Research of America), or sham surgery. Initial group size was determined from previous experiments (5, 13, 14) to ensure n ≥ 10 at observation/treatment endpoints. All mice were monitored bi-weekly for health and tumor development until euthanasia and necropsy at either age 6 or 12 months or when largest tumor reached diameter of approximately 2 cm (5, 13, 14). Five mice were excluded from the study for health reasons (open wound/pellet placement site n = 4, ear infection n = 1). There were no significant differences in mouse weights at study endpoints between treatment and control (one-way ANOVA, Sidak multiple comparisons test) or observation (unpaired t test) groups (P < 0.05, GraphPad Prism version 6.01). Genotype numbers (n)/observation/treatment/control group and [mean weights (grams) ± SEM/age in months (m)]: Brca1fl11/fl11/Cre/p53−/+ (n = 71: Ob: n = 10, T: n = 16, R: n = 11, L: n = 11, P: n = 12, S: n = 11; 39.2 ± 2.0/12m, 33.2 ± 0.8/6m), Brca1WT/fl11/Cre/p53−/+(n = 70: Ob: n = 12, T: n = 12, R: n = 11, L: n = 12, P: n = 12, S: n = 11; 34.4 ± 5.0/12m, 33.5 ± 1.3/6m), Brca1fl11/fl11/Cre (n = 25: T: n = 12, S: n = 13; 33.7 ± 1.7/6m), Brca1WT/fl11/Cre (n = 20: T: n = 10, S: n = 10; 30.9 ± 0.8/6m), p53−/+ (n = 20: T: n = 10, S: n = 10; 28.0 ± 0.6/6m), Cre (n = 23: T: n = 11, S: n = 12; 28.0 ± 1.5/6m), WT (n = 22: T: n = 10; S: n = 12; 32 ± 1.0/6m). Georgetown University Animal Care and Use Committee (Protocols 15-020-100220/12-020-100034) and Institutional Biosafety Committee approved all procedures.

Histologic and IHC analyses

HAN numbers per inguinal mammary gland (5, 13–16) and presence of tertiary branching (24) were determined blindly by three independent observers (S.J. Alothman, W. Wang, and P.A. Furth) on mammary gland whole mounts fixed in Carnoy solution and stained in carmine alum. The mean of the three readings was calculated for numerical data and the two concordant readings used for discrepant categorical data. Cancer histology and presence or absence of cytologic atypia and nuclear localized Estrogen Receptor (ER) alpha in MEC were read blindly on hematoxylin and eosin (H&E)-stained sections of formalin-fixed, paraffin-embedded (FFPE) tissue by a board-certified pathologist (B.V. Kallakury). Cytologic atypia was defined as the presence of mild, moderate, or severe abnormal nuclear architecture. IHC was performed on FFPE tissue following antigen retrieval using antibodies directed against proliferating cell nuclear antigen (PCNA; ref.13; 1:300 1-hour room temperature, sc- 7907, Santa Cruz Biotechnology) and ER alpha (refs. 5, 15; 1:225, 1-hour room temperature, sc-542, Santa Cruz Biotechnology). To calculate proliferative indices (PI), MEC percentages demonstrating PCNA nuclear localization within a total population of 500–1,000 MECs were determined blindly by two independent observers (S.J. Alothman and P. A. Furth) and means presented. All available samples were analyzed. N for each treatment/control genotype group included in figure legends. H&E/IHC sections were not available from all mice in some groups due to inadequate fixation technique or expended sample material. Digital photographs were taken using a Nikon Eclipse E800M microscope and DMX1200 camera with the NIS Elements imaging software version 4.30 program (Nikon Corporation).

Genetic analyses for LOH of Brca1 and Trp53 and somatic mutation of Brca2 and Dapk3

DNA samples were prepared for custom DNA capture and sequencing using the SureSelect Target Capture Enrichment System (Agilent Technologies). DNA was isolated from cell culture and tissue sample lysates from Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+ mice using the MasterPureComplete DNA Purification Kit (Epicentre, Illumina Inc.). Custom cRNA baits for the genes of interest (Brca1, Trp53, Brca2, Dapk3, Pten, and Tmem135) were designed using SureDesign software (Agilent Technologies). The genomic coordinates of each gene were derived from the Mus musculus NCBI build 37/UCSC mm9 assembly (MGSCv37, July 2007), and covered the entire transcribed region, including coding exons, introns, 5′ and 3′ untranslated regions, and an additional 10 kb total of contiguous sequence from the 5′ and 3′ ends for each gene. The genomic coordinates for each targeted gene were as follows: Brca1 (chr11:101345078101418269), Trp53 (chr11:69388861-69410375), Brca2 (chr5:151320198-151377324), Dapk3 (chr10:80641008-80660942), Pten (chr19:32827067-32905650), and Tmem135 (chr7:96283231-96492297). The region size of the target sequences totaled 4,59,420 kb over 6,341 cRNA probes (sequences available upon request) resulting in >90% coverage. Libraries were prepared for sequencing using the SureSelectXT Custom 1Kb-499kb Library Kit and SureSelectXT Rreagent Kit (Agilent Technologies). The custom capture DNA libraries were then sequenced on the Illumina MiSeq platform (paired-end 2 × 150 bp) using MiSeq Reagent Kit V2 (Illumina Inc.) with sequences were uploaded to Sequence Read Archive (SRA) BioProject ID PRJNA378147. Sequence reads were processed with a pipeline consisting of the following elements: (i) base calls generated in real-time on the Agilent SureSelect system; (ii) Perl scripts developed in-house to produce demultiplexed FASTQ files by lane and index sequence; (iii) demultiplexed BAM files aligned to the Mus musculus 9 reference genome using BurrowsWheeler Aligner (35). Read-pairs not mapping within +2 SDs of the average library size (*125 + 15 bp for exomes) were removed. All aligned read data were subjected to the following steps: (i) duplicate removal was performed, (i.e., the removal of reads with duplicate start positions; Picard MarkDuplicates, https://broadinstitute.github.io/picard/); (ii) indel realignment was performed (GATK IndelRealigner, https://software.broadinstitute.org/gatk/gatkdocs/org_broadinstitute_gatk_tools_walkers_indels_IndelRealigner.php) resulting in improved base placement and lower false variant calls; (iii) base qualities were recalibrated (GATK Table Recalibration, https://software.broadinstitute.org/gatk/gatkdocs/org_broadinstitute_gatk_tools_walkers_bqsr_BaseRecalibrator.php). Variant detection and genotyping were performed using the UnifiedGenotyper tool (GATK (v3.5) https://software.broadinstitute.org/gatk/gatkdocs/org_broadinstitute_gatk_tools_walkers_genotyper_UnifiedGenotyper.php). Variant data for each sample were formatted (variant call format) as “raw” calls that contain individual genotype data for one or multiple samples, and flagged using the filtration walker (GATK) to mark sites that are of lower quality/false positives, for example, low quality scores (≤ 50), allelic imbalance (≥0.75), long homopolymer runs (> 3), and/or low quality by depth (QD< 20). SNP were visualized using the Integrative Genomics Viewer 2.3.80 (36, 37) to examine for LOH and mutation. Regions demonstrating deviation from the reference genome were identified and compared with regions that were modified in the Brca1 and Trp53 loci in the GEMM (30, 31). For these analyses, DNA was extracted directly from noncancerous mammary gland (MG) or cancer (CA) tissue or primary cultures of noncancerous or cancer MEC under conditional reprogramming culture without feeder cells (CRC) or EpiCult B Mouse Medium (EpiC; Stemcell Technologies; ref. 38). Primary MEC cultures were obtained from mammary glands of Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+ mice aged 8–11months (2011–2012), stored viably at −80°C when not passaged, authenticated (2015) for the presence of Brca1 exon 11 deletion and Cre transgene by RT-PCR and PCR (38), and verified as having predicted genetically engineered deviations from the reference genome in Brca1 and Trp53 genes in this study (2016). Mycoplasma testing has not been performed. Passage numbers (p) reported are from initial isolation (p0). In some cases, samples from a single mouse from noncancerous and cancer tissue and/or under both culture conditions included, indicated by the same superscript number [T, tamoxifen exposure; samples tested: Brca1fl11/fl11/Cre/p53−/+; n = 7 (noncancerous: MG/6m, n = 1; CRCp5/11m, n = 1; cancer: CA/6m, n = 1; CA/6m/T, n = 1; CRCp12/11m, n = 1; CRCp8/8m, n = 11; EpiCp9/8m, n = 11); Brca1WT/fl11/Cre/p53−/+, n = 6 (noncancerous: CRCp4/10m, n = 12; EpiCp3/10m, n = 12; cancer: CRCp7/10m, n = 12; EpiCp17/10m, n = 12; CRC/p12/11m, n = 13; EpiCp11/1m, n = 13; WT, n = 1 (noncancerous: MG/6m)].

Statistical analyses

The primary endpoints were mammary cancer and presence or absence of HANs. Secondary endpoints included mean age when mammary tumor development reached 2-cm diameter, HAN numbers/inguinal mammary gland, tertiary branching and proliferative index, and cytologic atypia. Statistical analyses were performed with GraphPad Prism version 6.01 using Fisher exact test for prevalence of cancer, HANs, tertiary branching, and cytologic atypia (P < 0.05) and Mann–Whitney test for proliferative indices and HAN number (P < 0.05). Percent change was calculated using the formula (BA/A) × 100, where A was the value for the control group and B the value for the treatment group.

Both Brca1 haploinsufficiency and deficiency led to generation of mammary cancer and preneoplasia in combination with Trp53 haploinsufficiency

As women develop breast cancer when only one BRCA1 allele is mutated, we sought to determine whether the same could be true in mice to more closely model the impact of preventive regimens in mice with loss of only one Brca1 allele. Published work documents that disruption of Trp53 frequently occurs in breast cancers that develop in women carrying BRCA1 mutations (18), contributes to cancer generation in mice with disrupted Brca1 alleles (12, 14, 19, 30), and by itself does not lead to mammary cancer by age 12 months (16). Like Brca1fl11/fl11/Cre/p53−/+ mice, Brca1WT/fl11/Cre/p53−/+ mice developed invasive mammary cancer by age 12 months without significant difference in latency (Fig. 1A and B) and demonstrated HAN development on mammary gland whole mounts (Fig. 1C and D). Both GEMM showed a range of invasive mammary cancer histologic subtypes from more differentiated adenocarcinomas to less differentiated sarcomatoid carcinomas (Fig. 2A–H). HANs appeared similarly (Fig. 2I–J) and mammary epithelial cytologic atypia was found in both (Fig. 2K). Some, but not all atypical MEC demonstrated nuclear-localized ERα in both models (Fig. 2L). There was no significant difference in the percentages of mice from the two different genotypes that demonstrated tertiary branching (Fig. 2M).

Figure 1.

Both Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+ mice developed mammary cancers and HAN by age 12 months. A, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice with palpable mammary cancer by age 12 months. B, Bar graphs comparing mean age ± SEM for mammary cancer development in Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice. C, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice with HAN development by age 12 months. D, Scatter plots showing distributions of HAN number/gland in Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+ mice. Mean number ± SEM shown. *, P < 0.05, Mann–Whitney U Test, one-tailed. Brca1fl11/fl11/Cre/p53−/+, n = 10 mammary cancer/HAN number; Brca1WT/fl11/Cre/p53−/+, n = 12 mammary cancer; n = 8 HAN number.

Figure 1.

Both Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+ mice developed mammary cancers and HAN by age 12 months. A, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice with palpable mammary cancer by age 12 months. B, Bar graphs comparing mean age ± SEM for mammary cancer development in Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice. C, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice with HAN development by age 12 months. D, Scatter plots showing distributions of HAN number/gland in Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+ mice. Mean number ± SEM shown. *, P < 0.05, Mann–Whitney U Test, one-tailed. Brca1fl11/fl11/Cre/p53−/+, n = 10 mammary cancer/HAN number; Brca1WT/fl11/Cre/p53−/+, n = 12 mammary cancer; n = 8 HAN number.

Close modal
Figure 2.

Comparable spectrum of cancer histology and HAN appearance were found in Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+ mice. A–D, Representative H&E images of mammary cancer histology from Brca1fl11/fl11/Cre/p53−/+mice. E–H, Representative H&E images of mammary cancer histology from Brca1WT/fl11/Cre/p53−/+mice. I, Representative images of mammary gland branching patterns and HANs from a Brca1fl11/fl11/Cre/p53−/+mouse at low and high power. J, Representative images of mammary gland branching patterns and HANs from a Brca1WT/fl11/Cre/p53−/+mouse at low and high power. K, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice exhibiting cytologic atypia. L, Representative H&E and ERα IHC images of cytologically atypical MECs from Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice. M, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice exhibiting tertiary branching on mammary gland whole mounts. White arrows indicate HANs. White arrowheads indicate MECs demonstrating cytologic atypia. Black arrowheads indicate atypical cells with nuclear-localized ERα. Black arrows indicate ERα-negative atypical cells. LN, lymph node. H&E and IHC images taken at 40×; Scale bar, 10 μm. Low power whole mount images taken at 0.5× and high power images at 1.0×; Scale bar, 1 mm. Brca1fl11/fl11/Cre/p53−/+ n = 10, tertiary branching, n = 7, cytologic atypia. Brca1WT/fl11/Cre/p53−/+ n = 8, tertiary branching, n = 8, cytologic atypia.

Figure 2.

Comparable spectrum of cancer histology and HAN appearance were found in Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+ mice. A–D, Representative H&E images of mammary cancer histology from Brca1fl11/fl11/Cre/p53−/+mice. E–H, Representative H&E images of mammary cancer histology from Brca1WT/fl11/Cre/p53−/+mice. I, Representative images of mammary gland branching patterns and HANs from a Brca1fl11/fl11/Cre/p53−/+mouse at low and high power. J, Representative images of mammary gland branching patterns and HANs from a Brca1WT/fl11/Cre/p53−/+mouse at low and high power. K, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice exhibiting cytologic atypia. L, Representative H&E and ERα IHC images of cytologically atypical MECs from Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice. M, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice exhibiting tertiary branching on mammary gland whole mounts. White arrows indicate HANs. White arrowheads indicate MECs demonstrating cytologic atypia. Black arrowheads indicate atypical cells with nuclear-localized ERα. Black arrows indicate ERα-negative atypical cells. LN, lymph node. H&E and IHC images taken at 40×; Scale bar, 10 μm. Low power whole mount images taken at 0.5× and high power images at 1.0×; Scale bar, 1 mm. Brca1fl11/fl11/Cre/p53−/+ n = 10, tertiary branching, n = 7, cytologic atypia. Brca1WT/fl11/Cre/p53−/+ n = 8, tertiary branching, n = 8, cytologic atypia.

Close modal

Mammary cancer development in Brca1WT/fl11/Cre/p53−/+ mice occurred without LOH in Brca1 or Trp53

We then tested for Brca1 or Trp53 LOH and mutation of Brca2, Dapk3, Pten, and Tmem135 in noncancerous and cancer tissue from the two GEMM and WT mice. Both intronic and exonic sequences were included in the analyses because pathogenic intronic mutations are reported in humans (39). There were no SNPs in either Brca1 or Trp53 uniquely identified in cancers from Brca1WT/fl11/Cre/p53−/+mice as compared with those from Brca1fl11/fl11/Cre/p53−/+ mice. SNPs were identified in the Brca1 gene in Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+ mice but only in regions that were genetically engineered to introduce lox sites in the Brca1 gene (ref. 30; Fig. 3A). All but two of the cancers from both genotypes showed SNPs in the region of the genetically engineered Trp53 gene (ref. 31; Fig. 3B). SNPs outside this region were detected in one of the two Brca1fl11/fl11/Cre/p53−/+ cancers directly analyzed and in the EpiC, but not CRC, culture of noncancerous MEC from a Brca1WT/fl11/Cre/p53−/+ mouse. The few SNPs identified in Brca2 and Dapk3 genes were also present in the WT tissue analyzed (Fig. 3C and D). Analysis of Pten and Tmem135 gene sequences revealed multiple SNPs present in both GEMM and WT samples (data not shown).

Figure 3.

No Brca1 or Trp53 LOH was found in mammary cancers or noncancerous mammary tissue from Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+ mice. Images showing nucleotide deviations from the mm9 reference genome in mammary cancer and noncancer tissue and primary MECs from Brca1fl11/fl11/Cre/p53−/+, Brca1WT/fl11/Cre/p53−/+,and WT mice viewed through the Integrative Genomics Viewer (IGV) for Brca1 (A), Trp53 (B), Brca2 (C), and Dapk3 (D). Regions of Brca1 and Trp53 that were genetically engineered in the Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice indicated below intron/exon structure. Dark blue lines, LOH; red lines, conflict in heterozygosity (homozygous in WT and heterozygous in sample); light blue lines, unknown mutation; light purple lines, synonymous mutation; pink lines, neutral mutation; *, cancer samples. Black bracket lines to left indicate samples obtained from the same mouse. 1, Brca1fl11/fl11/Cre/p53−/+mammary gland (MG), noncancerous; 2, Brca1fl11/fl11/Cre/p53−/+ primary MEC CRC, noncancerous; 3, Brca1fl11/fl11/Cre/p53−/+primary MEC CRC, cancer; 4, Brca1fl11/fl11/Cre/p53−/+primary MEC CRC, cancer; 5, Brca1fl11/fl11/Cre/p53−/+primary MEC EpiC, cancer; 6, Brca1fl11/fl11/Cre/p53−/+cancer tissue (tamoxifen-exposed); 7, Brca1fl11/fl11/Cre/p53−/+ cancer tissue (control); 8, Brca1WT/fl11/Cre/p53−/+ primary MEC CRC, noncancerous; 9, Brca1WT/fl11/Cre/p53−/+primary MEC, noncancerous; 10, Brca1WT/fl11/Cre/p53−/+primary MEC CRC, cancer; 11, Brca1WT/fl11/Cre/p53−/+primary MEC CRC, cancer; 12, Brca1WT/fl11/Cre/p53−/+primary MEC EpiC, cancer; 13, Brca1WT/fl11/Cre/p53−/+primary MEC EpiC, cancer; 14, WT MG, noncancerous.

Figure 3.

No Brca1 or Trp53 LOH was found in mammary cancers or noncancerous mammary tissue from Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+ mice. Images showing nucleotide deviations from the mm9 reference genome in mammary cancer and noncancer tissue and primary MECs from Brca1fl11/fl11/Cre/p53−/+, Brca1WT/fl11/Cre/p53−/+,and WT mice viewed through the Integrative Genomics Viewer (IGV) for Brca1 (A), Trp53 (B), Brca2 (C), and Dapk3 (D). Regions of Brca1 and Trp53 that were genetically engineered in the Brca1fl11/fl11/Cre/p53−/+and Brca1WT/fl11/Cre/p53−/+mice indicated below intron/exon structure. Dark blue lines, LOH; red lines, conflict in heterozygosity (homozygous in WT and heterozygous in sample); light blue lines, unknown mutation; light purple lines, synonymous mutation; pink lines, neutral mutation; *, cancer samples. Black bracket lines to left indicate samples obtained from the same mouse. 1, Brca1fl11/fl11/Cre/p53−/+mammary gland (MG), noncancerous; 2, Brca1fl11/fl11/Cre/p53−/+ primary MEC CRC, noncancerous; 3, Brca1fl11/fl11/Cre/p53−/+primary MEC CRC, cancer; 4, Brca1fl11/fl11/Cre/p53−/+primary MEC CRC, cancer; 5, Brca1fl11/fl11/Cre/p53−/+primary MEC EpiC, cancer; 6, Brca1fl11/fl11/Cre/p53−/+cancer tissue (tamoxifen-exposed); 7, Brca1fl11/fl11/Cre/p53−/+ cancer tissue (control); 8, Brca1WT/fl11/Cre/p53−/+ primary MEC CRC, noncancerous; 9, Brca1WT/fl11/Cre/p53−/+primary MEC, noncancerous; 10, Brca1WT/fl11/Cre/p53−/+primary MEC CRC, cancer; 11, Brca1WT/fl11/Cre/p53−/+primary MEC CRC, cancer; 12, Brca1WT/fl11/Cre/p53−/+primary MEC EpiC, cancer; 13, Brca1WT/fl11/Cre/p53−/+primary MEC EpiC, cancer; 14, WT MG, noncancerous.

Close modal

Tamoxifen reduced MEC growth parameters, but not the development of cancer or preneoplasia in Brca1fl11/fl11/Cre/p53−/+ mice, but did decrease HAN number in Brca1WT/fl11/Cre/p53−/+ mice

In Brca1fl11/fl11/Cre/p53−/+ mice, tamoxifen exposure significantly reduced prevalence of tertiary branching by 65% and proliferative index (PI) by 60% (Fig. 4). In WT mice, tamoxifen exposure significantly reduced tertiary branching prevalence by 88% and in Brca1WT/fl11/Cre mice by 50%, and PI was significantly reduced in Brca1WT/fl11/Cre mice by 42%. While the magnitude of reduction by tamoxifen in tertiary branching and PI varied across different genotypes, no evidence was found to indicate that Brca1fl11/fl11/Cre/p53−/+ mice were less sensitive to the antihormonal action of tamoxifen than mice carrying more limited genetic modifications. Tamoxifen had no impact on HAN or cancer prevalence or HAN number in Brca1fl11/fl11/Cre/p53−/+ mice (Fig. 5A–E). In contrast, tamoxifen exposure did statistically significantly reduce HAN number in Brca1WT/fl11/Cre/p53−/+ mice by 95% (Fig. 5B). Overall cytologic atypia was detected less frequently in tamoxifen-exposed mice (Fig. 5F). Atypical MECs in tamoxifen-exposed Brca1fl11/fl11/Cre/p53−/+ mice retained nuclear-localized ERα expression (Fig. 5G–J).

Figure 4.

Tamoxifen induced statistically significant reductions in mammary gland tertiary branching and MEC proliferation in Brca1fl11/fl11/Cre/p53-/+and Brca1fl11/fl11/Cre mice. A, Bar graphs comparing percentages of mammary glands demonstrating tertiary branching in control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice. *, P < 0.05, control and tamoxifen-exposed, Fisher exact, one-tailed. BE, Representative mammary gland whole mount images of branching patterns from control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53/+mice. F, Bar graphs comparing mean MEC, PI in control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice. SEM indicated, *, P < 0.05, control and tamoxifen-exposed, Mann–Whitney U Test, one-tailed. G–J, Representative PCNA IHC images of MECs with nuclear-localized PCNA (white arrows) from control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+mice. Whole mount images taken at 1.0×. Scale bar, 1 mm. IHC images taken at 40×. Scale bar, 10 μm. Brca1fl11/fl11/Cre/p53-/+: Control (C), n = 11 (PI, n = 7); tamoxifen-exposed (T) n = 16 (PI = 11); Brca1WT/fl11/Cre/p53-/+: C, n = 10 (PI, n = 6); T: n = 10 (PI, n = 7). Brca1fl11/fl11/Cre: C, n = 11 (PI, n = 10); T: n = 12 (PI, n = 7); Brca1WT/fl11/Cre: C, n = 10 (PI, n = 6); T: 10 (PI, n = 8); p53−/+: C, n = 10 (PI, n = 9); T, n = 10 (PI, n = 9). Cre: C, n = 12 (PI, n = 5); T, n = 11 (PI, n = 8); WT: C, n = 12 (PI = 11); T, n = 10 (PI = 5).

Figure 4.

Tamoxifen induced statistically significant reductions in mammary gland tertiary branching and MEC proliferation in Brca1fl11/fl11/Cre/p53-/+and Brca1fl11/fl11/Cre mice. A, Bar graphs comparing percentages of mammary glands demonstrating tertiary branching in control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice. *, P < 0.05, control and tamoxifen-exposed, Fisher exact, one-tailed. BE, Representative mammary gland whole mount images of branching patterns from control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53/+mice. F, Bar graphs comparing mean MEC, PI in control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice. SEM indicated, *, P < 0.05, control and tamoxifen-exposed, Mann–Whitney U Test, one-tailed. G–J, Representative PCNA IHC images of MECs with nuclear-localized PCNA (white arrows) from control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+mice. Whole mount images taken at 1.0×. Scale bar, 1 mm. IHC images taken at 40×. Scale bar, 10 μm. Brca1fl11/fl11/Cre/p53-/+: Control (C), n = 11 (PI, n = 7); tamoxifen-exposed (T) n = 16 (PI = 11); Brca1WT/fl11/Cre/p53-/+: C, n = 10 (PI, n = 6); T: n = 10 (PI, n = 7). Brca1fl11/fl11/Cre: C, n = 11 (PI, n = 10); T: n = 12 (PI, n = 7); Brca1WT/fl11/Cre: C, n = 10 (PI, n = 6); T: 10 (PI, n = 8); p53−/+: C, n = 10 (PI, n = 9); T, n = 10 (PI, n = 9). Cre: C, n = 12 (PI, n = 5); T, n = 11 (PI, n = 8); WT: C, n = 12 (PI = 11); T, n = 10 (PI = 5).

Close modal
Figure 5.

Tamoxifen had no impact on HAN and cancer development in Brca1fl11/fl11/Cre/p53-/+mice but reduced HAN number in Brca1WT/fl11/Cre/p53-/+mice. A, Bar graphs comparing percentages of whole mounts demonstrating HANs in controls and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+,Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice at age 6 months. B, Scatter plots showing distributions of HAN numbers/gland in control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+mice. Mean number ± SEM shown. *, P < 0.05 Mann–Whitney U Test, one-tailed. C, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice with palpable cancer at age 6 months in controls and tamoxifen exposed. D and E, Representative H&E-stained images of mammary cancer histology from Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+ mice. F, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice exhibiting cytologic atypia at age 6 months in controls and tamoxifen-exposed. G–J, Representative H&E-stained and ERα IHC images of MECs demonstrating cytologic atypia (Aty) from Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+mice. White arrowheads indicate MECs demonstrating cytologic atypia. Black arrowheads indicate atypical cells with nuclear-localized ERα. Black arrows indicate ERα-negative atypical cells. H&E and IHC images taken at 40×. Scale bar, 0.01 mm. Brca1fl11/fl11/Cre/p53-/+: Control (C): n = 11 (Aty, n = 6); Tamoxifen-exposed (T): n = 16 (Aty, n = 10); Brca1WT/fl11/Cre/p53-/+: C: n = 10 (Aty, n = 9); T: n = 10 (Aty, n = 6); Brca1fl11/fl11/Cre: C: n = 11 (Aty, n = 6). T: n = 12 (Aty, n = 7); Brca1WT/fl11/Cre: C: n = 10 (Aty, n = 6); T:10 (Aty, n = 9); p53−/+ C: n = 10 (Aty, n = 10); T: n = 10 (Aty, n = 10); Cre: C: n = 12 (Aty, n = 12); T: n = 11 (Aty, n = 10). WT: C: n = 12 (Aty, n = 5); T: n = 10 (Aty, n = 5).

Figure 5.

Tamoxifen had no impact on HAN and cancer development in Brca1fl11/fl11/Cre/p53-/+mice but reduced HAN number in Brca1WT/fl11/Cre/p53-/+mice. A, Bar graphs comparing percentages of whole mounts demonstrating HANs in controls and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+,Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice at age 6 months. B, Scatter plots showing distributions of HAN numbers/gland in control and tamoxifen-exposed Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+mice. Mean number ± SEM shown. *, P < 0.05 Mann–Whitney U Test, one-tailed. C, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice with palpable cancer at age 6 months in controls and tamoxifen exposed. D and E, Representative H&E-stained images of mammary cancer histology from Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+ mice. F, Bar graphs comparing percentages of Brca1fl11/fl11/Cre/p53-/+, Brca1WT/fl11/Cre/p53-/+, Brca1fl11/fl11/Cre, Brca1WT/fl11/Cre, Trp53−/+, MMTV-Cre, and WT mice exhibiting cytologic atypia at age 6 months in controls and tamoxifen-exposed. G–J, Representative H&E-stained and ERα IHC images of MECs demonstrating cytologic atypia (Aty) from Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+mice. White arrowheads indicate MECs demonstrating cytologic atypia. Black arrowheads indicate atypical cells with nuclear-localized ERα. Black arrows indicate ERα-negative atypical cells. H&E and IHC images taken at 40×. Scale bar, 0.01 mm. Brca1fl11/fl11/Cre/p53-/+: Control (C): n = 11 (Aty, n = 6); Tamoxifen-exposed (T): n = 16 (Aty, n = 10); Brca1WT/fl11/Cre/p53-/+: C: n = 10 (Aty, n = 9); T: n = 10 (Aty, n = 6); Brca1fl11/fl11/Cre: C: n = 11 (Aty, n = 6). T: n = 12 (Aty, n = 7); Brca1WT/fl11/Cre: C: n = 10 (Aty, n = 6); T:10 (Aty, n = 9); p53−/+ C: n = 10 (Aty, n = 10); T: n = 10 (Aty, n = 10); Cre: C: n = 12 (Aty, n = 12); T: n = 11 (Aty, n = 10). WT: C: n = 12 (Aty, n = 5); T: n = 10 (Aty, n = 5).

Close modal

Raloxifene impact was similar to tamoxifen in Brca1fl11/fl11/Cre/p53−/+ mice, whereas letrozole significantly reduced but did not eradicate HAN development

Raloxifene statistically significantly reduced tertiary branching in Brca1fl11/fl11/Cre/p53−/+ mice by 71%, but was without significant impact on HAN prevalence or number (Fig. 6A and B). In contrast, letrozole statistically significantly reduced tertiary branching by 78%, HAN prevalence by 52%, and HAN number by 75% (Fig. 6A and B). In Brca1WT/fl11/Cre/p53−/+ mice, the impact of raloxifene and letrozole were relatively similar (Fig. 6C). Cancer appeared in control (Fig. 6D) but not raloxifene- or letrozole-exposed mice. Letrozole was more effective than either tamoxifen or raloxifene in reducing preneoplasia in Brca1fl11/fl11/Cre/p53−/+ mice but HANs were not entirely eradicated and cytologic atypia persisted. Similar results were found in Brca1WT/fl11/Cre/p53−/+mice (Fig. 6E–V). Atypical MECs in letrozole- and raloxifene-exposed Brca1fl11/fl11/Cre/p53−/+ and Brca1WT/fl11/Cre/p53−/+mice retained nuclear-localized ERα expression (insets, Fig. 6G, J, M, P, S, V).

Figure 6.

Letrozole reduced HAN development in Brca1fl11/fl11/Cre/p53-/+ mice while raloxifene showed comparable impact to tamoxifen. A, Bar graphs comparing percentage of Brca1fl11/fl11/Cre/p53-/+ mice aged 6 months with tertiary branching, HANs, cytologic atypia and palpable cancer in controls versus raloxifene or letrozole exposure. *, P < 0.05 Fisher Exact, one-tailed. B, Scatter plots showing distributions of HAN numbers/gland in control, raloxifene- and letrozole-exposed Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+mice. Mean number ± SEM shown. *, P < 0.05 Mann–Whitney U Test, one tailed. C, Bar graphs comparing percentage of Brca1WT/fl11/Cre/p53-/+mice aged 6 months with tertiary branching, HANs, cytologic atypia and palpable cancer in controls versus raloxifene or letrozole exposure. D, Cancer histology from control Brca1fl11/fl11/Cre/p53-/+ mice. E–J, Representative images of mammary gland branching patterns, HANs, and cytologic atypia from control Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+ mice. Insets demonstrate presence (black arrowheads) and absence (black arrows) of nuclear-localized ERα in atypical MECs. K–P, Representative images of mammary gland branching patterns, HANs and cytologic atypia from raloxifene-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+ mice. Insets demonstrate presence (black arrowheads) and absence (black arrows) of nuclear-localized ERα in atypical MECs. Q–V, Representative images of mammary gland branching patterns, HANs and cytologic atypia from letrozole-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+ mice. Insets demonstrate presence (black arrowheads) and absence (black arrows) of nuclear-localized ERα in atypical MECs. White arrows indicate HANs. White arrowheads indicate MECs demonstrating cytologic atypia. Whole mount images taken at 1×. Scale bar, 1 mm. H and E, and IHC images taken at 40×. Scale bar, 10 μm. Brca1fl11/fl11/Cre/p53-/+: Control (C): n = 12; raloxifene (R): n = 11; letrozole (L): n = 10; Brca1WT/fl11/Cre/p53-/+: C, n = 12; R, n = 11; L, n = 12.

Figure 6.

Letrozole reduced HAN development in Brca1fl11/fl11/Cre/p53-/+ mice while raloxifene showed comparable impact to tamoxifen. A, Bar graphs comparing percentage of Brca1fl11/fl11/Cre/p53-/+ mice aged 6 months with tertiary branching, HANs, cytologic atypia and palpable cancer in controls versus raloxifene or letrozole exposure. *, P < 0.05 Fisher Exact, one-tailed. B, Scatter plots showing distributions of HAN numbers/gland in control, raloxifene- and letrozole-exposed Brca1fl11/fl11/Cre/p53-/+and Brca1WT/fl11/Cre/p53-/+mice. Mean number ± SEM shown. *, P < 0.05 Mann–Whitney U Test, one tailed. C, Bar graphs comparing percentage of Brca1WT/fl11/Cre/p53-/+mice aged 6 months with tertiary branching, HANs, cytologic atypia and palpable cancer in controls versus raloxifene or letrozole exposure. D, Cancer histology from control Brca1fl11/fl11/Cre/p53-/+ mice. E–J, Representative images of mammary gland branching patterns, HANs, and cytologic atypia from control Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+ mice. Insets demonstrate presence (black arrowheads) and absence (black arrows) of nuclear-localized ERα in atypical MECs. K–P, Representative images of mammary gland branching patterns, HANs and cytologic atypia from raloxifene-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+ mice. Insets demonstrate presence (black arrowheads) and absence (black arrows) of nuclear-localized ERα in atypical MECs. Q–V, Representative images of mammary gland branching patterns, HANs and cytologic atypia from letrozole-exposed Brca1fl11/fl11/Cre/p53-/+ and Brca1WT/fl11/Cre/p53-/+ mice. Insets demonstrate presence (black arrowheads) and absence (black arrows) of nuclear-localized ERα in atypical MECs. White arrows indicate HANs. White arrowheads indicate MECs demonstrating cytologic atypia. Whole mount images taken at 1×. Scale bar, 1 mm. H and E, and IHC images taken at 40×. Scale bar, 10 μm. Brca1fl11/fl11/Cre/p53-/+: Control (C): n = 12; raloxifene (R): n = 11; letrozole (L): n = 10; Brca1WT/fl11/Cre/p53-/+: C, n = 12; R, n = 11; L, n = 12.

Close modal

Pharmacologic approaches to breast cancer prevention can be understood in degrees of risk reduction for a specific individual and agent weighed against possible side effects and potential harms (40). Currently, tamoxifen and raloxifene are recommended by the United States Preventive Services Task Force (41). The American Society of Clinical Oncology Clinical Practice Guidelines adds the possibility of an aromatase inhibitor (exemestane) for postmenopausal women (42). Both groups agree that current evidence fails to show any impact of antihormonal agents on prevention of ER-negative breast cancer or all-cause mortality. Women who carry BRCA1 mutations are of particular concern due to their defined genetic risk factor and their higher proportion of ER-negative breast cancers. Risk-reducing mastectomy and salpingo-oophorectomy offer the highest likelihood of reducing both cancer diagnosis and all-cause mortality (43).

One of the criticisms of the Brca1fl11/fl11/Cre/p53−/+mouse model has been that both Brca1 alleles are disrupted while in women only one Brca1 allele is mutated. It has been suggested that MECs with only one disrupted BRCA1 allele may be more sensitive to antihormonal therapies than cells with two disrupted BRCA1 alleles (26). Here we tested the impact of antihormonals on preneoplasia development with both one and two Brca1 alleles and found evidence of increased responsiveness to tamoxifen in Brca1WT/fl11/Cre/p53−/+ as compared with Brca1fl11/fl11/Cre/p53−/+ mice. There has been significant discussion around whether or not Brca1 LOH is required for cancer development (18, 19). Significantly we determined that Brca1 LOH was not required for mammary cancer development and found no evidence that Trp53 LOH was required for cancer development even though this has been previously reported for this model (22).

Letrozole has been proposed as an alternative antihormonal in women carrying BRCA1 or BRCA2 mutations (11). We found letrozole to be more effective than either tamoxifen or raloxifene in reducing preneoplasia in Brca1fl11/fl11/Cre/p53−/+mice. Because we did not follow cohorts of mice after antihormonal therapy was completed as was done previously (13), we cannot say whether or not the interim antihormonal therapy would have had a significant impact on later cancer generation. However, the persistence of HANs after antihormone exposure suggests that these mice remained at risk for cancer development even when statistically significant reductions in prevalence of preneoplasia was found. Retention of nuclear-localized ERα in cytologically atypical cells following antihormone exposure indicated that the therapy did not interrupt presence of this cell population. Whether or not these cells contribute to cancer generation in either this model or in women who develop triple-negative cancer remains an open question.

Women interested in pharmacologic approaches for breast cancer prevention look for approaches that are highly effective without significant side effects (40). Even if antihormone sensitivity is greater with loss of a single as compared with both Brca1 alleles, BRCA1 LOH is well documented in breast cancers developing in women who carry one mutated BRCA1 allele (18). It is prudent for investigators to seek chemopreventive approaches that would be effective under both genetic settings, as the precise genetic makeup of each breast epithelial cell in a population is not easily defined. Use of both Brca1WT/fl11/Cre/p53−/+ and Brca1fl11/fl11/Cre/p53−/+ mouse models in chemoprevention studies has the advantage of discerning how proposed agents may act when Brca1 is deficient as compared with only haploinsufficient.

In summary, these studies report that antihormonal agents can significantly impact mammary disease prevalence in mouse models of Brca1 mutation but fail to eradicate preneoplasia. While loss of one as compared with two Brca1 alleles may increase responsiveness to antihormonal agents, the call for more effective approaches persists.

No potential conflicts of interest were disclosed.

Conception and design: S.J. Alothman, W. Wang, P.A. Furth

Development of methodology: S.J. Alothman, W. Wang, D.S. Goerlitz, Md. Islam, A. Kishore, B. Kallakury, P.A. Furth

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.J. Alothman, W. Wang, D.S. Goerlitz, Md. Islam, A. Kishore, R.I. Azhar, B. Kallakury, P.A. Furth

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.J. Alothman, W. Wang, X. Zhong, A. Kishore, R.I. Azhar, B. Kallakury, P.A. Furth

Writing, review, and/or revision of the manuscript: S.J. Alothman, W. Wang, Md. Islam, X. Zhong, A. Kishore, B. Kallakury, P.A. Furth

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): W. Wang, Md. Islam, A. Kishore, P.A. Furth

Study supervision: P.A. Furth

This work was supported by grant NIH NCI RO1 CA112176 (to P.A. Furth), King Abdullah Scholarship Program, Ministry of Higher Education, Kingdom of Saudi Arabia (to S.J. Alothman), and NIH NCI 5P30CA051008 (Histology and Tissue, Genomics and Epigenomics, and Animal Shared Resources).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Couch
FJ
,
Nathanson
KL
,
Offit
K
. 
Two decades after BRCA: setting paradigms in personalized cancer care and prevention
.
Science
2014
;
343
:
1466
70
.
2.
Hartmann
LC
,
Lindor
NM
. 
The role of risk-reducing surgery in hereditary breast and ovarian cancer
.
N Engl J Med
2016
;
374
:
454
68
.
3.
Metcalfe
KA
,
Dennis
CL
,
Poll
A
,
Armel
S
,
Demsky
R
,
Carlsson
L
, et al
Effect of decision aid for breast cancer prevention on decisional conflict in women with a BRCA1 or BRCA2 mutation: a multisite, randomized, controlled trial
.
Genet Med
2016
Sep 1.
[Epub ahead of print]
.
4.
Communal
L
,
Vilasco
M
,
Hugon-Rodin
J
,
Courtin
A
,
Mourra
N
,
Lahlou
N
, et al
Proliferation and ovarian hormone signaling are impaired in normal breast tissues from women with BRCA1 mutations: benefit of a progesterone receptor modulator treatment as a breast cancer preventive strategy in women with inherited BRCA1 mutations
.
Oncotarget
2016
;
7
:
45317
30
.
5.
Nakles
RE
,
Kallakury
BVS
,
Furth
PA
. 
The PPARγ agonist efatutazone increases the spectrum of well-differentiated mammary cancer subtypes initiated by loss of full-length BRCA1 in association with TP53 haploinsufficiency
.
Am J Pathol
2013
;
182
:
1976
85
.
6.
Sigl
V
,
Owusu-Boaitey
K
,
Joshi
PA
,
Kavirayani
A
,
Wirnsberger
G
,
Novatchkova
M
, et al
RANKL/RANK control Brca1 mutation-driven mammary tumors
.
Cell Res
2016
;
26
:
761
74
.
7.
To
C
,
Kim
EH
,
Royce
DB
,
Williams
CR
,
Collins
RM
,
Risingsong
R
, et al
The PARP inhibitors, veliparib and olaparib, are effective chemopreventive agents for delaying mammary tumor development in BRCA1-deficient mice
.
Cancer Prev Res
2014
;
7
:
698
707
.
8.
Nelson
HD
,
Fu
R
,
Goddard
K
,
Mitchell
JP
,
Okinaka-Hu
L
,
Pappas
M
, et al
Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: systematic review to update the U.S. Preventive Services Task Force Recommendation
.
Rockville, MD
:
Agency for Healthcare Research and Quality (US)
; 
2013
. Available from: http://www.ncbi.nlm.nih.gov/books/NBK179201/.
9.
Xu
L
,
Zhao
Y
,
Chen
Z
,
Wang
Y
,
Chen
L
,
Wang
S
. 
Tamoxifen and risk of contralateral breast cancer among women with inherited mutations in BRCA1 and BRCA2: a metaanalysis
.
Breast Cancer
2015
;
22
:
327
34
.
10.
Sismondi
P
,
D'Alonzo
M
,
Pecchio
S
,
Bounous
VE
,
Robba
E
,
Biglia
N
. 
Chemoprevention or mastectomy for women at high risk of developing breast cancer
.
Maturitas
2015
;
82
:
271
3
.
11.
Pujol
P
,
Lasset
C
,
Berthet
P
,
Dugast
C
,
Delaloge
S
,
Fricker
JP
, et al
Uptake of a randomized breast cancer prevention trial comparing letrozole to placebo in BRCA1/2 mutations carriers: the LIBER trial
.
Fam Cancer
2012
;
11
:
77
84
.
12.
Diaz-Cruz
ES
,
Cabrera
MC
,
Nakles
R
,
Rutstein
BH
,
Furth
PA
. 
BRCA1 deficient mouse models to study pathogenesis and therapy of triple negative breast cancer
.
Breast Dis
2010
;
32
:
85
97
.
13.
Jones
LP
,
Li
M
,
Halama
ED
,
Ma
Y
,
Lubet
R
,
Grubbs
CJ
, et al
Promotion of mammary cancer development by tamoxifen in a mouse model of Brca1-mutation- related breast cancer
.
Oncogene
2005
;
24
:
3554
62
.
14.
Jones
LP
,
Tilli
MT
,
Assefnia
S
,
Torre
K
,
Halama
ED
,
Parrish
A
, et al
Activation of estrogen signaling pathways collaborates with loss of Brca1 to promote development of ERalpha-negative and ERalpha-positive mammary preneoplasia and cancer
.
Oncogene
2008
;
27
:
794
802
.
15.
Dabydeen
SA
,
Kang
K
,
Díaz-Cruz
ES
,
Alamri
A
,
Axelrod
ML
,
Bouker
KB
, et al
Comparison of tamoxifen and letrozole response in mammary preneoplasia of ER and aromatase overexpressing mice defines an immune-associated gene signature linked to tamoxifen resistance
.
Carcinogenesis
2015
;
36
:
122
32
.
16.
Díaz-Cruz
ES
,
Furth
PA
. 
Deregulated estrogen receptor alpha and p53 heterozygosity collaborate in the development of mammary hyperplasia
.
Cancer Res
2010
;
70
:
3965
74
.
17.
Said
SM
,
Visscher
DW
,
Nassar
A
,
Frank
RD
,
Vierkant
RA
,
Frost
MH
, et al
Flat epithelial atypia and risk of breast cancer: A Mayo cohort study
.
Cancer
2015
;
121
:
1548
55
.
18.
Martins
FC
,
De
S
,
Almendro
V
,
Gönen
M
,
Park
SY
,
Blum
JL
, et al
Evolutionary pathways in BRCA1-associated breast tumors
.
Cancer Discov
2012
;
2
:
503
11
.
19.
Liu
Y
,
Yen
HY
,
Austria
T
,
Pettersson
J
,
Peti-Peterdi
J
,
Maxson
R
, et al
A mouse model that reproduces the developmental pathways and site specificity of the cancers associated with the human BRCA1 mutation carrier state
.
EBioMedicine
2015
;
2
:
1318
30
.
20.
Natrajan
R
,
Mackay
A
,
Lambros
MB
,
Weigelt
B
,
Wilkerson
PM
,
Manie
E
, et al
A whole genome massively parallel sequencing analysis of BRCA1 mutant oestrogen receptor negative and -positive breast cancers
.
J Pathol
2012
;
227
:
29
41
.
21.
Caldes
T
,
de la Hoya
M
,
Tosar
A
,
Sulleiro
S
,
Godino
J
,
Ibañez
D
, et al
A breast cancer family from Spain with germline mutations in both the BRCA1 and BRCA2 genes
.
J Med Genet
2002
;
39
:
e44
.
22.
Brodie
SG
,
Xu
X
,
Qiao
W
,
Li
WM
,
Cao
L
,
Deng
CX
. 
Multiple genetic changes are associated with mammary tumorigenesis in Brca1 conditional knockout mice
.
Oncogene
2001
;
20
:
7514
23
.
23.
Ma
Y
,
Katiyar
P
,
Jones
LP
,
Fan
S
,
Zhang
Y
,
Furth
PA
, et al
The breast cancer susceptibility gene BRCA1 regulates progesterone receptor signaling in mammary epithelial cells
.
Mol Endocrinol
2006
;
20
:
14
34
.
24.
Welsh
J
,
Zinser
LN
,
Mianecki-Morton
L
,
Martin
J
,
Waltz
SE
,
James
H
, et al
Age-related changes in the epithelial and stromal compartments of the mammary gland in normocalcemic mice lacking the vitamin D3 receptor
.
PLoS One
2011
;
6
:
e16479
.
25.
Bramley
M
,
Clarke
RB
,
Howell
A
,
Evans
DGR
,
Armer
T
,
Baildam
AD
, et al
Effects of oestrogens and anti-oestrogens on normal breast tissue from women bearing BRCA1 and BRCA2 mutations
.
Br J Cancer
2006
;
94
:
1021
8
.
26.
Gorski
JJ
,
Kennedy
RD
,
Hosey
AM
,
Harkin
DP
. 
The complex relationship between BRCA1 and ERalpha in hereditary breast cancer
.
Clin Cancer Res
2009
;
15
:
1514
8
.
27.
Savage
KI
,
Matchett
KB
,
Barros
EM
,
Cooper
KM
,
Irwin
GW
,
Gorski
JJ
, et al
BRCA1 deficiency exacerbates estrogen-induced DNA damage and genomic instability
.
Cancer Res
2014
;
74
:
2773
84
.
28.
Li
W
,
Xiao
C
,
Vonderhaar
BK
,
Deng
CX
. 
A role of estrogen/ERalpha signaling in BRCA1 associated tissue-specific tumor formation
.
Oncogene
2007
;
26
:
7204
12
.
29.
Wen
J
,
Li
R
,
Lu
Y
,
Shupnik
MA
. 
Decreased BRCA1 confers tamoxifen resistance in breast cancer cells by altering estrogen receptor-coregulator interactions
.
Oncogene
2009
;
28
:
575
86
.
30.
Xu
X
,
Wagner
KU
,
Larson
D
,
Weaver
Z
,
Li
C
,
Ried
T
, et al
Conditional mutation of Brca1 in mammary epithelial cells results in blunted ductal morphogenesis and tumour formation
.
Nat Genet
1999
;
22
:
37
43
.
31.
Donehower
LA
,
Harvey
M
,
Slagle
BL
,
McArthur
MJ
,
Montgomery
CA
,
Butel
JS
, et al
Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours
.
Nature
1992
;
356
:
215
21
.
32.
Wagner
KU
,
Wall
RJ
,
St-Onge
L
,
Gruss
P
,
Wynshaw-Boris
A
,
Garrett
L
, et al
Cre mediated gene deletion in the mammary gland
.
Nucleic Acids Res
1997
;
25
:
4323
30
.
33.
Westphal
C
,
Schubert
C
,
Prelle
K
,
Penkalla
A
,
Fliegner
D
,
Petrov
G
, et al
Effects of estrogen, an ERα agonist and raloxifene on pressure overload induced cardiac hypertrophy
.
PLoS One
2012
;
7
:
e50802
.
34.
Zeng
Y
,
Yokohira
M
,
Saoo
K
,
Takeuchi
H
,
Chen
Y
,
Yamakawa
K
, et al
Inhibition of prostate carcinogenesis in probasin/SV40 T antigen transgenic rats by raloxifene, an antiestrogen with anti-androgen action, but not nimesulide, a selective cyclooxygenase- 2 inhibitor
.
Carcinogenesis
2005
;
26
:
1109
16
.
35.
Li
H
,
Durbin
R
. 
Fast and accurate short read alignment with Burrows-Wheeler transform
.
Bioinformatics
2010
;
26
:
589
95
.
36.
Robinson
JT
,
Thorvaldsdóttir
H
,
Winckler
W
,
Guttman
M
,
Lander
ES
,
Getz
G
, et al
Integrative genomics viewer
.
Nat Biotechnol
2011
;
29
:
24
6
.
37.
Thorvaldsdóttir
H
,
Robinson
JT
,
Mesirov
JP
. 
Integrative Genomics Viewer (IGV): high performance genomics data visualization and exploration
.
Brief Bioinform
2013
;
14
:
178
92
.
38.
Alamri
AM
,
Kang
K
,
Groeneveld
S
,
Wang
W
,
Zhong
X
,
Kallakury
B
, et al
Primary cancer cell culture: mammary-optimized vs conditional reprogramming
.
Endocr Relat Cancer
2016
;
23
:
535
54
.
39.
Hainaut
P
,
Pfeifer
GP
. 
Somatic TP53 Mutations in the Era of Genome Sequencing
.
Cold Spring Harb Perspect Med
2015
;
6
:
a026179
.
40.
Burns
RB
,
Schonberg
MA
,
Tung
NM
,
Libman
H
. 
Should we offer medication to reduce breast cancer risk?: Grand rounds discussion from Beth Israel Deaconess Medical Center
.
Ann Intern Med
2016
;
165
:
194
204
.
41.
Moyer
VA
. 
Medications for risk reduction of primary breast cancer in women: U.S. Preventive Services Task Force recommendation statement
.
Ann Intern Med
2013
;
159
:
698
708
.
42.
Visvanathan
K
,
Hurley
P
,
Bantug
E
,
Brown
P
,
Col
NF
,
Cuzick
J
, et al
Use of pharmacologic interventions for breast cancer risk reduction: American Society of Clinical Oncology clinical practice guideline
.
J Clin Oncol
2013
;
31
:
2942
62
.
43.
Nelson
HD
,
Pappas
M
,
Zakher
B
,
Mitchell
JP
,
Okinaka-Hu
L
,
Fu
R
. 
Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer in women: a systematic review to update the U.S. Preventive Services Task Force recommendation
.
Ann Intern Med
2014
;
160
:
255
66
.