Skip to main content
JNCI Cancer Spectrum logoLink to JNCI Cancer Spectrum
. 2019 Dec 11;4(2):pkz100. doi: 10.1093/jncics/pkz100

BRCA1 Promoter Methylation Status in 1031 Primary Breast Cancers Predicts Favorable Outcomes Following Chemotherapy

Olafur A Stefansson 1,2,3, Holmfridur Hilmarsdottir 2, Kristrun Olafsdottir 1, Laufey Tryggvadottir 1,4, Asgerdur Sverrisdottir 5, Oskar T Johannsson 5, Jon G Jonasson 1,6, Jorunn E Eyfjord 1,2, Stefan Sigurdsson 1,2,
PMCID: PMC7061679  PMID: 32175521

Abstract

Background

Breast Cancer 1 gene (BRCA1) is known to be inactivated in breast tumors by promoter methylation. Tumor cells in patients carrying a germline mutation in BRCA1 are sensitive to cytotoxic drugs that cause DNA double strand breaks. However, very little is known on whether patients with BRCA1 promoter methylated tumors are similarly sensitive to cytotoxic drugs. In this study, we address this by making use of extensive follow-up data on patients treated with cyclophosphamide, methotrexate, and fluorouracil in Iceland between 1976 and 2007.

Methods

We analyzed BRCA1 promoter methylation by pyrosequencing DNA from tumor samples from 1031 patients with primary breast cancer. Of those, 965 were sporadic cases, 61 were BRCA2, and five were BRCA1 germline mutation carriers. All cases were examined with respect to clinicopathological parameters and breast cancer–specific survival in patients treated with cytotoxic drugs. Information on chemotherapy treatment in noncarriers was available for 26 BRCA1 methylated tumors and 857 unmethylated tumors.

Results

BRCA1 was promoter methylated in 29 sporadic tumors or in 3.0% of cases (29 of 965), whereas none of the tumors derived from BRCA germline mutation carriers were promoter methylated. Important to note, patients with BRCA1 promoter methylation receiving chemotherapeutic drug treatment show highly improved breast cancer–specific survival compared with unmethylated controls (hazard ratio = 0.10, 95% confidence interval = 0.01 to 0.75, two-sided P = .02).

Conclusions

BRCA1 promoter methylation is predictive of improved disease outcome in patients receiving cyclophosphamide, methotrexate, and fluorouracil drug treatment. Our results support the use of markers indicative of “BRCAness” in sporadic breast cancers to identify patients that are likely to benefit from the use of DNA-damaging agents.


Germline mutations in the breast cancer–susceptibility gene, Breast Cancer 1 gene (BRCA1), significantly increase the risk of developing breast and ovarian cancer, in addition to other forms of cancer (1). The BRCA1 protein product is involved in DNA double strand–break (DSB) repair by homologous recombination, a highly conserved error-free DNA–repair pathway that uses an intact sister chromatid in late S or G2 phases of the cell cycle for the repair (2).

Tumors lacking BRCA1 or BRCA2 are homologous recombination deficient (HRD) and are characterized by mutational signatures, including indels, rearrangements, and base substitutions (3,4). Based on these mutational signatures, a model, HRDetect, has been developed to predict the BRCA1/BRCA2 deficiency or “BRCAness” of tumors (3).

Loss of the wild-type allele is seen in most tumors arising in BRCA1 mutation carriers (5). These tumors have high HRDetect scores compared with those that do not show loss of heterozygosity at the BRCA1 loci. Tumors without loss of heterozygosity have similar HRDetect scores as noncarriers.

Germline mutations in BRCA genes and somatic mutations in homologous recombination genes are associated with increased sensitivity to platinum chemotherapy and Poly ADP Ribose Polymerase (PARP) inhibitors in breast cancer (6–10) and ovarian cancer (11–15). These agents induce replication fork stalling, creating DNA substrates that are dependent on homologous recombination for replication restart and are essential for the survival of the cell (16).

BRCA1 is sometimes inactivated in breast tumors by promoter methylation (17,18). BRCA1 methylated tumors are associated with the basal-like or triple-negative subtype that is predominant in germline mutation carriers (19,20). Recently BRCA1 methylated tumors were associated with mutational signatures characteristic of tumors arising in BRCA1 germline mutation carriers (4,21).

It is currently unclear, however, whether BRCA1 promoter methylation translates to clinical benefits from the use of DNA-damaging agents in patients. We therefore carried out a large retrospective study aimed at determining whether BRCA1 methylation is associated with improved outcomes in survival among chemotherapy-treated patients.

Methods

Study Group

The study group consisted of 1031 patients (women) diagnosed between 1976 and 2007 previously screened for the local BRCA1 c.5074G>A and BRCA2 c.767-771delCAAAT–germline mutations (22,23). In addition to the two BRCA founder mutations, BRCA2 c.767-771delCAAAT and the much rarer BRCA1 c.5074G>A, the only other BRCA mutation of some frequency, c.9976A>T, is not found to be associated with risk of breast or ovarian cancer but rather risk of small cell lung cancer and squamous cell carcinoma of the skin (24). Out of the 1031 cases, 965 were sporadic, 61 were BRCA2 germline mutation carriers, and five were BRCA1 mutation carriers. DNA samples from these patients were extracted from FF tissue samples (n = 417) and adjacent normal breast tissues (n = 91) using a standard phenolchloroform (+ proteinase K) method. DNA derived from formalin-fixed and paraffin-embedded tumors (n = 615) was extracted by deparaffinization using Octane followed by two rounds of ethanol washes and then overnight incubation in digestion buffer (50 mM Tris pH 8.8, 1 mM EDTA and 0.5% Tween, proteinase K).

This work was carried out according to the permits from the Icelandic Data Protection Commission (2006050307) and Bioethics Committee (VSNb2006050001/03–16).

DNA–Methylation Analysis

The EZ-96 DNA Methylation-Gold kit (Zymo Research; D5008) was used to carry out bisulfite conversion of DNA samples derived from tumor and normal breast tissues. Primer design was based on the PyroMark Assay Desing 2.0 software (Qiagen). Pre-PCR reactions, using the Immolase DNA polymerase (Bioline; Bio-21047), along with pyrosequencing (PyroMark Q24; Qiagen) were carried as previously described (25). Four CpG sites were analyzed at genetic positions: chr17: 43125409(GRCh38.p7), chr17: 43125411(GRCh38.p7), chr17: 43125419(GRCh38.p7), chr17: 43125427(GRCh38.p7).

The primers used for pre-PCR were as follows: Forward primer 5-GTAGGGGTTTAGTTATTTGAGAAATTTT-3; reverse biotinylated primer 5-TATCCCTCCCATCCTCTAATTATAC-3. The sequencing primer for the pyrosequencing reaction was as follows: 5-AGTTTTAATTTATTTGTAATTTT-3. Tumor samples were considered to be BRCA1 methylated at median greater than 10% methylation across the four CpGs (median values).

Clinicopathological Parameters and Treatment

Information on patient age, birth date, and date of diagnosis, tumor grade, staging, and chemotherapy treatment given at time of diagnosis were obtained from the nationwide Icelandic Cancer Registry (26,27). Estrogen-receptor (ER) status was based on tissue-microarray (TMA) analysis (n = 664) (20,26) and, where TMA data were not available, we used data derived from routine clinical diagnoses based on dextran-coated charcoal assay used in Iceland in the period 1981 to 1995 (n = 98).

Statistical Analysis

The χ2 and Fisher exact contingency table tests were carried out using base functions in R. The Kaplan–Meier method was used for generating univariable survival curves and the log-rank test was used for estimating P values. Hazard ratios (HRs) were estimated by applying the Cox proportional hazards model using R (survival package). Here, BRCA1–methylation status represents the main variable, whereas year of birth, year of diagnosis, ER status, nodal status, and tumor size are introduced as adjustment covariates. Patients were followed from the date of diagnosis until death or date of last follow-up (December 31, 2016). The endpoint was breast cancer–specific survival, defined as the time from diagnosis to death from breast cancer, as registered on death certificates. Patients who died of causes other than breast cancer were censored at the date of death. The cox.zph function in the survival package for R was used to test the proportional hazards assumption for a Cox regression model fit. All tests were two-sided and a P value of less than .05 was considered statistically significant.

Results

The Incidence of BRCA1 Methylation in Primary Breast Tumors and Normal Tissue

We analyzed BRCA1 promoter CpG methylation in 1031 primary tumors, along with 91 normal breast tissue samples derived from the same cohort. BRCA1 methylation was identified in 29 tumors, none of which were derived from carries of germline mutations in either BRCA1 (n = 5) or BRCA2 (n = 61) (Figure 1). The frequency of BRCA1 promoter methylation in non-BRCA mutated breast tumors is approximately 3% (29 out of 965; 3.0%).

Figure 1.

Figure 1.

Box plot of BRCA1–promoter CpG methylation in normal breast tissue and tumors derived from carriers of germline mutations in BRCA1, BRCA2, and noncarriers.

BRCA1 Methylation in Relation to Clinicopathological Parameters

Information on ER status was available for 762 out of the 1031 tumors in our cohort; 700 out of the 762 tumors were derived from noncarriers (Table 1, A). We find that 90.5% of tumors with BRCA1 promoter methylation are negative for ER, or 19 out of 21 (see Table 1, A). Similarly, tumors arising in BRCA1 mutation carriers were exclusively ER negative (five out of five) as previously reported (20), and by TMA analysis, we find that the majority of BRCA1 methylated tumors are of the basal-like subtype (eight out of 12; 67%) (see Table 1, B).

Table 1.

CpG–promoter methylation of BRCA1 gene analyzed with respect to clinicopathological parameters and breast cancer subtypes*

A) ER status with respect to BRCA1 methylation
ER status BRCA1 unmethylated BRCA1 methylated
ER positive 526 (77.5%) 2 (9.5%)
ER negative 153 (22.5%) 19 (90.4%)
Total (N = 700) 679 21
Fisher test, P < .0001

B) Breast cancers subtypes with respect to BRCA1 methylation

Subtype BRCA1 unmethylated BRCA1 methylated

LumA 78 (40%) 0
LumB 61 (31.3%) 1 (8.3%)
HER2 18 (9.2%) 0
Basal-like 34 (17.4%) 8 (66.7%)
5-negative (5NP) 4 (2.1%) 3 (25%)
Total (N = 207) 195 12
 χ2 = 39.0, P < .0001

C) Histological grade with respect to BRCA1 methylation

Histological grade BRCA1 unmethylated BRCA1 methylated

1+ 104 (23.6%) 0
2+ 187 (42.4%) 6 (31.6%)
3+ 150 (34%) 13 (68.4%)
Total (N = 460) 441 19
χ2 = 11.1, P = .0039

D) Tumor size with respect to BRCA1 methylation

Tumor size BRCA1 unmethylated BRCA1 methylated

T1 462 (54.4%) 8 (30.8%)
T2 319 (37.6%) 16 (61.5%)
T3 54 (6.4%) 2 (7.7%)
T4 14 (1.6%) 0
Total (N = 875) 849 26
 χ2 = 6.9, P = .075

E) Nodal status with respect to BRCA1 methylation

Nodal status BRCA1 unmethylated BRCA1 methylated

Negative 422 (51.9%) 17 (65.4%)
Positive 391 (48.1%) 8 (30.6%)
Total (N = 839) 813 26
 Fisher test, P = .15

F) Age at diagnosis

Age BRCA1 unmethylated BRCA1 methylated

≤55 439 (46.9%) 22 (75.9%)
>55 497 (53.1%) 7 (24.1%)
Total (N = 965) 936 29
Fisher test, P = .002
*

ER = estrogen receptor.

BRCA1 promoter-methylated tumors are statistically significantly less differentiated based on histopathological grading (Χ2=11.1, P = .0039) (Table 1, C). However, no association was found with respect to clinical staging in terms of tumor size or nodal status (Table 1, D and E).

The Effects of BRCA1 Methylation with Respect to Responsiveness to Cytotoxic Treatment

To assess the association between BRCA1 methylation and response to cytotoxic chemotherapy, we obtained information on chemotherapy and used time to patient death (breast cancer–specific survival) as a proxy for treatment response. In our cohort, cyclophosphamide, methotrexate, and fluorouracil (CMF) was the most commonly used cytotoxic treatment given at time of diagnosis, and nine of 29 patients with BRCA1 methylated tumors received CMF treatment (Table 2). Information on chemotherapy treatment in noncarriers was available for 26 BRCA1 methylated tumors and 857 unmethylated tumors. In noncarriers, patients with BRCA1 methylated tumors show long-term survival following cytotoxic treatment (HR = 0.10, 95% CI = 0.014 to 0.751, P = .025) (Figure 2, A;Table 3). In contrast, nontreated patients with BRCA1 methylation, compared with nontreated patients without BRCA1 methylated tumors, showed similar time to breast cancer–specific death (Figure 2, B;Table 3). In comparing treated vs nontreated survival curves for non-BRCA mutated cases (Figure 2, A and B), there are notable differences, but these are likely explained by clinical presentation reflecting poor prognosis, and as a result, the treated group is biased toward reduced survival. This, however, does not affect our results, as we find improved survival for patients with BRCA1–methylated tumors after receiving cytotoxic treatment.

Table 2.

Type of chemotherapeutic drugs used during treatment in the cohort of non-BRCA mutation carriers listed out with respect to BRCA1 methylation status*

BRCA1 unmethylated BRCA1 methylated
CMF
242 9
Vincristine
97 2
Taxanes
9 0
Anthracycline
53 0
Any type
294 9
*

CMF = cyclophosphamide, methotrexate, and fluorouracil.

Figure 2.

Figure 2.

Breast cancer–specific survival time in patients treated A) with and B) without cytotoxic chemotherapy analyzed with respect to BRCA1 methylation status. The P values shown reflect log-rank hypothesis testing for differences in survival times with respect to BRCA1 methylation status.

Table 3.

The effect of BRCA1 methylation on time to breast cancer–specific death with and without chemotherapeutic treatment analyzed by multivariate Cox proportional hazards regression*

Treatment HR (95% CI) P
BRCA1 methylation in nontreated patients (n = 391) 1.58 (0.52 to 4.80) .416
BRCA1 methylation in treated patients (n = 183) 0.10 (0.01 to 0.75) .025
*

Adjusted for year of diagnosis and birth, estrogen-receptor status, tumor size, and nodal status. CI = confidence interval; HR = hazard ratio.

In this cohort, nine patients with BRCA1 methylated tumors received chemotherapeutic treatment with CMF (Table 2). However, patients with non-BRCA1 methylated tumors received CMF or other treatments. By restricting our analysis to CMF–treated patients only, we find that the relation between BRCA1 methylation and breast cancer–specific survival holds statistical significance (HR = 0.11, 95% CI = 0.014 to 0.81, P = .03).

Discussion

In this article, we demonstrate that BRCA1 promoter methylation, analyzed in a large cohort of 1031 primary breast tumors, predicts improved breast cancer–specific survival outcomes in patients treated with cytotoxic chemotherapy. No difference is seen in patients that did not receive a cytotoxic treatment, suggesting that BRCA1 promoter methylation is a predictive factor for chemosensitivity but not a prognostic factor. Previous studies have shown that BRCA1 mutation carriers are sensitive to treatment with platinum and PARP inhibitors. However, little is known about the effect of BRCA1 methylation with respect to treatment based on DNA–damaging agents.

The classical CMF combination was the standard treatment for breast cancer patients until it was replaced by anthracyclines and taxanes as adjuvant treatment (28). Although it is less effective than anthracyclines and taxanes, large retrospective studies have shown a clear benefit of CMF, especially in triple-negative breast cancer (29–31). Although both methotrexate and fluorouracil (5-FU) are antimetabolites that block the synthesis of thymidine required for DNA synthesis, cyclophosphamide is an alkylating agent, and its cytotoxic metabolite phosphoramide mustard leads to inter- and intrastrand crosslinks in DNA causing replication fork stalling (32–34). Because of the importance of the DNA DSB repair machinery in DNA–replication restart, phosphoramide mustard treatment is likely to lead to synergistic lethality in BRCA–deficient cells. This mechanism of action is similar to that currently thought to underlie the effectiveness of PARP1 inhibitors and platinum drugs in killing BRCA deficient cells (16). The mechanism of PARP1 inhibition is however more complex than previously expected. In addition to generating persistent single-strand breaks leading to collapse of the DNA–replication fork, PARP1 becomes trapped on DNA when inhibited, forming a cytotoxic lesion leading to replication fork stalling (35). Recently PARP inhibitors were also shown to affect the recruitment of POLQ to DNA DSBs, inhibiting microhomology-mediated end joining or Alt-EJ pathway (36–38).

Recent data have shown that 40–70% of triple-negative breast cancer is HR deficient (3,4,39) and therefore likely to respond to agents causing DNA–replication fork stalling (40–43). Replication fork stalling possibly explains the efficacy of cyclophosphamide in triple-negative tumors (30,31), as these are likely to be HRD.

HRDetect is a recently introduced method that makes use of next-generation sequencing for identifying mutational signatures characteristic of tumors arising in BRCA1 or BRCA2 germline mutation carriers (3). The main idea behind HRDetect is to enable identification of tumors with defective homologous recombination repair. Indeed, Davies et al. found that tumors with BRCA1 methylation were detected as HRD by HRDetect (3). However, it is unclear whether this finding translates to enhanced sensitivity of BRCA1 methylated tumors to treatment with DNA–damaging agents. Our results suggest that, indeed, this does seem to be the case. It is important to note that HRDetect genomic signatures might still persist after restored homologous recombination function, especially in the metastatic setting. To discriminate between tumors that are truly HRD, functional methods such as RAD51 foci staining have been developed for clinical use to improve selection of patients sensitive to DNA–damaging agents (9,10).

Information on HRDetect scores was available for 24 tumors in our cohort, of which four were BRCA1 methylated as previously described ([3]; Supplementary Figure 1, available online). The four BRCA1 methylated tumors all show high HRDetect scores, comparable to values seen in BRCA1 mutation carriers. Two of these four received CMF treatment, and both are still alive today, more than 18 years after their diagnosis.

Recently, in Tutt et al., advanced TNBC tumors where investigated for response to docetaxel and carboplatin, which revealed better response to docetaxel. These tumors, however, were pretreated with drugs that cause DNA lesions that require DNA DSB repair. As suggested by the authors, BRCA1 methylation detected in the primary tumor may have been lost following this first treatment, thereby explaining the lack of response to carboplatin in the advanced tumors. Our study differs from that of Tutt et al. in that we investigate the response to first treatment in our patient cohort (8).

Of interest, we observe substantially lower frequency of BRCA1 methylation than previously reported (17,18,44,45). The discrepancy likely reflects either smaller cohorts in previous studies, leading to a situation where the detection of a single methylated tumor greatly influences the frequency, or use of methods known to have a higher false-positive detection rate, compared with pyrosequencing, such as methylation-specific PCR. In our cohort, 3% of primary breast cancer samples are BRCA1 methylated, which is in agreement with a recent study using data from The Cancer Genome Atlas (21).

Our cohort is based on DNA samples isolated from freshly frozen (FF) tumors (n = 417) and formalin-fixed paraffin-embedded (FFPE) tumors (n = 615). Previous studies have shown higher fraction of BRCA1 methylation in FFPE samples (46). Our analysis, based on pyrosequencing, does not show statistically significant differences in frequency of BRCA1 methylation in noncarriers (n = 965) between FFPE (14 out of 587; 2.4%) and FF (15 out of 378; 4.0%).

A limitation of this study lies in the low number of BRCA1 methylated tumors detected in our cohort. However, we would like to point out that this is the largest cohort studied to date on BRCA1 methylation. Another limitation lies in the use of time to breast cancer–specific death as a proxy for response to treatment.

In summary, we demonstrate that BRCA1 methylation is an important predictive factor of chemosensitivity in breast cancer rather than being a prognostic factor. This provides support for the use of methods aimed at defining BRCAness to identify patients that will derive benefits from DNA–damaging chemotherapy treatment and, possibly, targeted therapy using PARP inhibitors.

Funding

The authors would like to thank The Icelandic Research Fund (www.rannis.is) (14193–051 and 152077–051) and Gongum saman (www.gongumsaman.is) for funding.

Notes

The funders had no role in the design of the study; the collection, analysis, and interpretation of the data; the writing of the manuscript; and the decision to submit the manuscript for publication.

The authors have no disclosures.

Supplementary Material

pkz100_Supplementary_Data

References

  • 1. Lalloo F, Evans DG.. Familial breast cancer. Clin Genet. 2012;82(2):105–114. [DOI] [PubMed] [Google Scholar]
  • 2. Jasin M, Rothstein R.. Repair of strand breaks by homologous recombination. Cold Spring Harb Perspect Biol. 2013;5(11):a012740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Davies H, Glodzik D, Morganella S, et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat Med. 2017;23(4):517–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Nik-Zainal S, Davies H, Staaf J, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature. 2016;534(7605):47–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Maxwell KN, Wubbenhorst B, Wenz BM, et al. BRCA locus-specific loss of heterozygosity in germline BRCA1 and BRCA2 carriers. Nat Commun. 2017;8(1):319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361(2):123–134. [DOI] [PubMed] [Google Scholar]
  • 7. Hahnen E, Lederer B, Hauke J, et al. Germline mutation status, pathological complete response, and disease-free survival in triple-negative breast cancer: secondary analysis of the geparsixto randomized clinical trial. JAMA Oncol. 2017;3(10):1378–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Tutt A, Tovey H, Cheang MCU, et al. Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT trial. Nat Med. 2018;24(5):628–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Cruz C, Castroviejo-Bermejo M, Gutierrez-Enriquez S, et al. RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer. Ann Oncol. 2018;29(5):1203–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Castroviejo-Bermejo M, Cruz C, Llop-Guevara A, et al. A RAD51 assay feasible in routine tumor samples calls PARP inhibitor response beyond BRCA mutation. EMBO Mol Med. 2018;10(12):e9172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Bolton KL, Chenevix-Trench G, Goh C, et al. Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA. 2012;307(4):382–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 2016;375(22):2154–2164. [DOI] [PubMed] [Google Scholar]
  • 13. Swisher EM, Lin KK, Oza AM, et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 2017;18(1):75–87. [DOI] [PubMed] [Google Scholar]
  • 14. Vencken PM, Kriege M, Hoogwerf D, et al. Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann Oncol. 2011;22(6):1346–1352. [DOI] [PubMed] [Google Scholar]
  • 15. Yang D, Khan S, Sun Y, et al. Association of BRCA1 and BRCA2 mutations with survival, chemotherapy sensitivity, and gene mutator phenotype in patients with ovarian cancer. JAMA. 2011;306(14):1557–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Lord CJ, Ashworth A.. PARP inhibitors: Synthetic lethality in the clinic. Science. 2017;355(6330):1152–1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Birgisdottir V, Stefansson OA, Bodvarsdottir SK, et al. Epigenetic silencing and deletion of the BRCA1 gene in sporadic breast cancer. Breast Cancer Res. 2006;8(4):R38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Esteller M, Silva JM, Dominguez G, et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000;92(7):564–569. [DOI] [PubMed] [Google Scholar]
  • 19. Foulkes WD, Stefansson IM, Chappuis PO, et al. Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst. 2003;95(19):1482–1485. [DOI] [PubMed] [Google Scholar]
  • 20. Stefansson OA, Jonasson JG, Olafsdottir K, et al. CpG island hypermethylation of BRCA1 and loss of pRb as co-occurring events in basal/triple-negative breast cancer. Epigenetics. 2011;6(5):638–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Polak P, Kim J, Braunstein LZ, et al. A mutational signature reveals alterations underlying deficient homologous recombination repair in breast cancer. Nat Genet. 2017;49(10):1476–1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Bergthorsson JT, Jonasdottir A, Johannesdottir G, et al. Identification of a novel splice-site mutation of the BRCA1 gene in two breast cancer families: screening reveals low frequency in Icelandic breast cancer patients. Hum Mutat. 1998;(suppl 1):S195–S197. [DOI] [PubMed] [Google Scholar]
  • 23. Thorlacius S, Olafsdottir G, Tryggvadottir L, et al. A single BRCA2 mutation in male and female breast cancer families from Iceland with varied cancer phenotypes. Nat Genet. 1996;13(1):117–119. [DOI] [PubMed] [Google Scholar]
  • 24. Rafnar T, Sigurjonsdottir GR, Stacey SN, et al. Association of BRCA2 K3326* with small cell lung cancer and squamous cell cancer of the skin. J Natl Cancer Inst. 2018;110(9):967–974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Stefansson OA, Hermanowicz S, van der Horst J, et al. CpG promoter methylation of the ALKBH3 alkylation repair gene in breast cancer. BMC Cancer. 2017;17(1):469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Jonasson JG, Stefansson OA, Johannsson OT, et al. Oestrogen receptor status, treatment and breast cancer prognosis in Icelandic BRCA2 mutation carriers. Br J Cancer. 2016;115(7):776–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Sigurdardottir LG, Jonasson JG, Stefansdottir S, et al. Data quality at the Icelandic Cancer Registry: comparability, validity, timeliness and completeness. Acta Oncol. 2012;51(7):880–889. [DOI] [PubMed] [Google Scholar]
  • 28. Munzone E, Curigliano G, Burstein HJ, et al. CMF revisited in the 21st century. Ann Oncol. 2012;23(2):305–311. [DOI] [PubMed] [Google Scholar]
  • 29. Bonadonna G, Moliterni A, Zambetti M, et al. 30 years’ follow up of randomised studies of adjuvant CMF in operable breast cancer: Cohort study. BMJ. 2005;330(7485):217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Colleoni M, Cole BF, Viale G, et al. Classical cyclophosphamide, methotrexate, and fluorouracil chemotherapy is more effective in triple-negative, node-negative breast cancer: results from two randomized trials of adjuvant chemoendocrine therapy for node-negative breast cancer. J Clin Oncol. 2010;28(18):2966–2973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Falo C, Moreno A, Benito E, et al. Primary chemotherapy with cyclophosphamide, methotrexate, and 5-fluorouracil in operable breast carcinoma. Cancer. 2005;103(4):657–663. [DOI] [PubMed] [Google Scholar]
  • 32. Anderson D, Bishop JB, Garner RC, et al. Cyclophosphamide: review of its mutagenicity for an assessment of potential germ cell risks. Mutat Res. 1995;330(1–2):115–181. [DOI] [PubMed] [Google Scholar]
  • 33. Ganesan S, Keating AF.. Phosphoramide mustard exposure induces DNA adduct formation and the DNA damage repair response in rat ovarian granulosa cells. Toxicol Appl Pharmacol. 2015;282(3):252–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Wilson PM, Danenberg PV, Johnston PG, et al. Standing the test of time: targeting thymidylate biosynthesis in cancer therapy. Nat Rev Clin Oncol. 2014;11(5):282–298. [DOI] [PubMed] [Google Scholar]
  • 35. Murai J, Huang SY, Das BB, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72(21):5588–5599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Ceccaldi R, Liu JC, Amunugama R, et al. Homologous-recombination-deficient tumours are dependent on Poltheta-mediated repair. Nature. 2015;518(7538):258–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. D’Andrea AD. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair (Amst). 2018;71:172–176. [DOI] [PubMed] [Google Scholar]
  • 38. Mateos-Gomez PA, Gong F, Nair N, et al. Mammalian polymerase theta promotes alternative NHEJ and suppresses recombination. Nature. 2015;518(7538):254–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Loibl S, Weber KE, Timms KM, et al. Survival analysis of carboplatin added to an anthracycline/taxane-based neoadjuvant chemotherapy and HRD score as predictor of response-final results from GeparSixto. Ann Oncol. 2018;29(12):2341–2347. [DOI] [PubMed] [Google Scholar]
  • 40. Akashi-Tanaka S, Watanabe C, Takamaru T, et al. BRCAness predicts resistance to taxane-containing regimens in triple negative breast cancer during neoadjuvant chemotherapy. Clin Breast Cancer. 2015;15(1):80–85. [DOI] [PubMed] [Google Scholar]
  • 41. Sharma P. Update on the treatment of early-stage triple-negative breast cancer. Curr Treat Options Oncol. 2018;19(5):22. [DOI] [PubMed] [Google Scholar]
  • 42. Telli ML, Hellyer J, Audeh W, et al. Homologous recombination deficiency (HRD) status predicts response to standard neoadjuvant chemotherapy in patients with triple-negative or BRCA1/2 mutation-associated breast cancer. Breast Cancer Res Treat. 2018;168(3):625–630. [DOI] [PubMed] [Google Scholar]
  • 43. Telli ML, Timms KM, Reid J, et al. Homologous Recombination Deficiency (HRD) score predicts response to platinum-containing neoadjuvant chemotherapy in patients with triple-negative breast cancer. Clin Cancer Res. 2016;22(15):3764–3773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Shakeri H, Fakhrjou A, Nikanfar A, et al. Methylation analysis of BRCA1 and APC in breast cancer and its relationship to clinicopathological features. Clin Lab. 2016;62(12):2333–2337. [DOI] [PubMed] [Google Scholar]
  • 45. Xu X, Gammon MD, Zhang Y, et al. BRCA1 promoter methylation is associated with increased mortality among women with breast cancer. Breast Cancer Res Treat. 2009;115(2):397–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Turner NC, Reis-Filho JS, Russell AM, et al. BRCA1 dysfunction in sporadic basal-like breast cancer. Oncogene. 2007;26(14):2126–2132. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

pkz100_Supplementary_Data

Articles from JNCI Cancer Spectrum are provided here courtesy of Oxford University Press

RESOURCES