Purpose: In germline BRCA1 or BRCA2 (BRCA1/2) mutation carriers, restoration of tumor BRCA1/2 function by a secondary mutation is recognized as a mechanism of resistance to platinum and PARP inhibitors, primarily in ovarian cancer. We evaluated this mechanism of resistance in newly diagnosed patients with BRCA1/2-mutant breast cancer with poor response to neoadjuvant platinum-based therapy.

Experimental Design: PrECOG 0105 was a phase II neoadjuvant study of gemcitabine, carboplatin, and iniparib in patients with stage I–IIIA triple-negative or BRCA1/2 mutation–associated breast cancer (n = 80). All patients underwent comprehensive BRCA1/2 genotyping. For mutation carriers with moderate or extensive residual disease after neoadjuvant therapy, BRCA1/2 status was resequenced in the residual surgical breast tumor tissue.

Results: Nineteen patients had a deleterious germline BRCA1/2 mutation, and four had moderate residual disease at surgery. BRCA1/2 sequencing of residual tissue was performed on three patients. These patients had BRCA1 1479delAG, 3374insGA, and W1712X mutations, respectively, with LOH at these loci in the pretreatment tumors. In the first case, a new BRCA1 mutation was detected in the residual disease. This resulted in a 14–amino acid deletion and restoration of the BRCA1 reading frame. A local relapse biopsy 4 months later revealed the identical reversion mutation, and the patient subsequently died from metastatic breast cancer.

Conclusions: We report a BRCA1 reversion mutation in a patient newly diagnosed with triple-negative breast cancer that developed over 18 weeks of platinum-based neoadjuvant therapy. This was associated with poor therapy response, early relapse, and death. Clin Cancer Res; 23(13); 3365–70. ©2017 AACR.

Restoration of tumor BRCA1 or BRCA2 (BRCA1/2) function by a secondary mutation in these genes is recognized as an acquired mechanism of resistance to platinum and PARP inhibitors and has been reported primarily in ovarian cancer. How frequently this occurs in clinical practice is not well-known but may have significant implications for rational treatment selection in BRCA1/2 mutation–associated cancers. We report a case of a BRCA1 reversion mutation that developed over 18 weeks of neoadjuvant therapy in a newly diagnosed patient with triple-negative breast cancer. This was associated with rapid relapse after platinum-based therapy and death from metastatic breast cancer. Given the growing number of DNA repair–targeted therapeutics currently under development for BRCA1/2 mutation–associated cancers, further characterization of the prevalence, time course, and risk factors for reversion mutation development are warranted, as these events are likely to have greater relevance for clinical practice and drug development.

Individuals with a heterozygous germline mutation in the BRCA1 or BRCA2 (BRCA1/2) genes are predisposed to a range of familial cancers, including breast and ovarian carcinomas (1). The BRCA1/2 genes are tumor suppressors that encode proteins critical for the accurate repair of DNA double-strand breaks by homologous recombination. The interplay between the BRCA1/2 proteins and RAD51, an essential DNA repair protein, preserves genomic stability by avoiding more error-prone DNA repair pathways (2–4). Tumors from patients with deleterious BRCA1/2 mutations usually have loss of the wild-type allele and are thus BRCA1/2-deficient, whereas BRCA1/2 heterozygosity is retained by normal cells (5, 6). Somatic inactivation of the wild-type BRCA1/2 allele and methylation of the BRCA1 promoter domain with subsequent epigenetic silencing are other reported mechanisms (7).

Approximately 10% to 20% of invasive triple-negative breast cancers arise in the setting of BRCA1/2 deficiency (8–10). BRCA1/2-deficient breast cancers have decreased capacity to repair DNA, resulting in enhanced sensitivity to chemotherapy with cross-linking agents, such as cisplatin and carboplatin, and susceptibility to PARP inhibitors due to a synthetic lethal interaction (11–13). In a small proof-of-concept trial, the PARP inhibitor olaparib led to objective tumor response in 41% of patients with advanced breast cancer with a deleterious germline BRCA1/2 mutation (14). Clinical trials are now underway to assess the efficacy of PARP inhibitors as single agents and in combination with chemotherapy in the advanced as well as curative settings for triple-negative and BRCA1/2-associated breast cancers. Likewise, platinum agents have shown efficacy for BRCA1/2-associated breast cancers (15, 16), with a recent phase III study demonstrating substantial improvement in the objective response rate and progression-free survival with carboplatin compared with docetaxel in patients with a deleterious BRCA1/2 mutation and advanced disease (17).

With the increased recognition of DNA-damaging therapies as the forefront agents for BRCA1/2-associated tumors, an unexpected mechanism of drug resistance has garnered clinical attention: the restoration of BRCA1/2 function due to secondary mutations of BRCA1/2 in tumors. Given that defects in homologous recombination secondary to an underlying BRCA1/2 mutation lead to cellular sensitivity to DNA-damaging agents, the restoration of DNA repair proficiency results in acquired resistance to these drugs. To date, this drug resistance mechanism has been reported primarily in patients with advanced ovarian cancer who had disease progression after platinum or PARP inhibitor therapy (7, 18–24). In this study, we evaluated this novel mechanism of drug resistance in newly diagnosed patients with early-stage BRCA1/2-mutant breast cancer who had a poor response to platinum-based neoadjuvant chemotherapy.

Clinical trial study design and treatment regimen

PrECOG 0105 was a single-arm phase II neoadjuvant study in patients with newly diagnosed clinical stage I–IIIA triple-negative [estrogen receptor (ER) ≤ 5%, progesterone receptor (PR) ≤ 5%, and human epidermal growth factor receptor 2 (HER2)-negative (0 or 1+ by immunohistochemistry or FISH unamplified)] or BRCA1/2 mutation–associated breast cancer. Eighty patients were treated with carboplatin, gemcitabine, and iniparib, intravenously, for 6 cycles before definitive surgery. A total of 80 patients were enrolled at multiple cancer centers through PrECOG. Details of the clinical trial methods and results of the study endpoints have been published previously (16).

Study endpoints

The primary endpoint of the clinical study was complete pathologic response rate, defined as no invasive carcinoma in the breast or axillary lymph nodes. The extent of residual disease was assessed using the residual cancer burden (RCB) index (25). The RCB index is a validated prognostic marker of distant relapse-free survival in patients who have been treated with neoadjuvant chemotherapy [RCB 0: complete pathologic response (pCR); RCB I: minimal residual disease; RCB II: moderate residual disease; and, RCB III: extensive residual disease); ref. 25].

Molecular analyses

In addition to comprehensive germline BRCA1/2 testing using serum samples, all patients underwent research core biopsies of the primary breast tumor before therapy initiation; five to ten 5-μm tissue sections of fixed or frozen tumor tissue were collected and processed at Myriad Genetics, Inc. DNA extraction from formalin-fixed, paraffin-embedded or frozen tumors was conducted, and next-generation DNA sequencing using a liquid hybridization assay (Agilent SureSelect) followed by Illumina sequencing was performed. Residual disease tissue was collected in patients who went to surgery and did not achieve pCR and processed using the same assay methodology (26).

To determine BRCA1/2 mutation status, variant and large rearrangement detection was performed on sequence from BRCA1 and BRCA2. Complete descriptions of the mutation detection methods have been previously described (27). Only mutations classified as deleterious or suspected deleterious on the basis of previously described criteria were included in the analysis (28).

Baseline tumor gene expression profiling, copy number alterations and measures of genomic scars (homologous recombination deficiency assay) were assessed for any correlation with treatment response to neoadjuvant therapy. For BRCA1/2 mutation carriers with unfavorable response to neoadjuvant therapy, e.g., RCB II, tumor BRCA1/2 status was re-sequenced in the residual surgical tissue.

DNA extractions from two independent samples of the pretreatment tumor specimen were also performed for patient 15 to assess for a reversion mutation. In both samples, there were between 800 and 1,800 unique clonal reads through the region where the reversion mutation was located. On the basis of simulations, we would have 95% confidence to detect the reversion mutation in this number of clonal reads, if the mutation occurred in at least 1 in 250 cells.

Furthermore, SNP data from the homologous recombination deficiency assay was used to calculate the percent tumor content in each sample and to reconstruct the genome over the locus. This information allowed for calculation of predicted read frequency of both germline and somatic alleles.

All germline and tumor BRCA1/2 mutation testing was performed by Myriad Genetics, Inc.

Patients and tumor characteristics

Eighty patients received six cycles of gemcitabine, carboplatin, and iniparib in PrECOG 0105. Nineteen of these 80 patients (24%) carried a deleterious germline mutation in BRCA1, BRCA2, or both genes (Table 1). Four of these 19 patients had ER/PR-positive breast cancer (two BRCA1 carriers and two BRCA2 carriers), and the remaining 15 patients (78.9%) had triple-negative breast cancer. Deletion mutations leading to nonfunctional BRCA1/2 proteins were the primary mechanism (13 patients, 65%), including one patient with bilateral breast cancer with deletion of the entire BRCA1 gene, as well as a patient with deletions in both BRCA1 and BRCA2 genes (BRCA1: 1048delA; BRCA2: 1366delA); both patients had excellent responses to neoadjuvant therapy. Insertions and missense mutations were also observed (three and four patients, respectively). The affected gene and specific location of the germline mutations are shown in Table 2.

Table 1.

Breast cancer subtype and BRCA1/2 mutation status in PrECOG 0105

All patients (N = 80)
Characteristicsn (%)
Breast cancer subtype 
 Triple-negative 77 (96) 
 ER- and/or PR-positive/HER2-negative 3 (4) 
BRCA1/2 mutation status 
 BRCA1 mutation 14 (18) 
 BRCA2 mutation 4 (5) 
 BRCA1 and BRCA2 mutation 1 (1) 
All patients (N = 80)
Characteristicsn (%)
Breast cancer subtype 
 Triple-negative 77 (96) 
 ER- and/or PR-positive/HER2-negative 3 (4) 
BRCA1/2 mutation status 
 BRCA1 mutation 14 (18) 
 BRCA2 mutation 4 (5) 
 BRCA1 and BRCA2 mutation 1 (1) 
Table 2.

PrECOG 0105 patients with BRCA1/2 mutation–associated breast cancer

Patient IDGene mutatedSite of deleterious mutationER/PR statusRCB score
BRCA1 2392insT TN II 
BRCA1 3374insGA TN II 
BRCA1 917delTT ER 0%, PR 1% 
BRCA1 5385insC TN 
BRCA1 3607C>T TN N/A 
BRCA1 del exons 23–24 TN 
BRCA2 7297delCT TN 
BRCA2 8803delC ER 90%, PR 5% 
BRCA1 del exons 5–8 TN 
10 BRCA1 187delAG TN 
11-L breast BRCA1 Deletion of entire BRCA1 gene TN 
11-R breast BRCA1 Deletion of entire BRCA1 gene TN 
12 BRCA1 Q284X969CTdel TN 
13 BRCA1 C61G (300T>G) TN 
14 BRCA2 G819delTG ER 30%, PR 20% 
15 BRCA1 1479delAG TN II 
16 BRCA1 and BRCA2 (one breast lesion) BRCA1: 1048delA TN 
  BRCA2: 1366delA TN 
17 BRCA2 5104delAA TN 
18 BRCA1 W1712X (5255G>A) ER 50%, PR 30% II 
19 BRCA1 IVS16+6T>C TN 
Patient IDGene mutatedSite of deleterious mutationER/PR statusRCB score
BRCA1 2392insT TN II 
BRCA1 3374insGA TN II 
BRCA1 917delTT ER 0%, PR 1% 
BRCA1 5385insC TN 
BRCA1 3607C>T TN N/A 
BRCA1 del exons 23–24 TN 
BRCA2 7297delCT TN 
BRCA2 8803delC ER 90%, PR 5% 
BRCA1 del exons 5–8 TN 
10 BRCA1 187delAG TN 
11-L breast BRCA1 Deletion of entire BRCA1 gene TN 
11-R breast BRCA1 Deletion of entire BRCA1 gene TN 
12 BRCA1 Q284X969CTdel TN 
13 BRCA1 C61G (300T>G) TN 
14 BRCA2 G819delTG ER 30%, PR 20% 
15 BRCA1 1479delAG TN II 
16 BRCA1 and BRCA2 (one breast lesion) BRCA1: 1048delA TN 
  BRCA2: 1366delA TN 
17 BRCA2 5104delAA TN 
18 BRCA1 W1712X (5255G>A) ER 50%, PR 30% II 
19 BRCA1 IVS16+6T>C TN 

Abbreviation: TN, triple-negative.

Response data

Among all study patients, 29 [36.3%; 90% confidence interval (CI), 27%–46%] achieved a pCR. The pCR rate was 33.0% among wild-type BRCA1/2 patients (CI, 23%–44%), 47% among BRCA1/2 mutation carriers (CI, 27%–68%), and 56% among BRCA1/2 mutation carriers with triple-negative breast cancer (CI, 33%–77%). Among patients with germline BRCA1/2 mutations, RCB class was as follows: RCB 0 = 10 (53%); RCB I = 6 (32%); RCB II = 4 (21%); and RCB III = 0 (0%). One BRCA1 mutation carrier was lost to follow-up and never had surgery.

BRCA1 sequencing of residual tissue was available in three of four patients with RCB II response (no consent in one case, patient 1). These three patients had BRCA1 3374insGA (patient 2), 1479delAG (patient 15), and W1721X (patient 18) mutations with LOH at these loci in the pretreatment specimens (Table 3). Patients 2 and 15 had triple-negative breast cancer, whereas patient 18 had ER/PR-positive, HER2-negative breast cancer. No reversion mutations were detected in the residual tumor specimen at the time of surgery in patients 2 and 18. However, a new BRCA1 mutation was detected in the residual surgical tumor specimen in patient 15. This reversion mutation comprised a 42-base pair deletion that overlapped with the original 2-base pair deletion. It resulted in a 14–amino acid deletion and restoration of the BRCA1 reading frame. A relapse biopsy of an ipsilateral axillary lymph node 4 months later revealed an identical reversion mutation (Table 3, Figs. 1 and 2).

Table 3.

Study patients with BRCA1 mutation–associated breast cancer and RCB score of II

Patient No.Deleterious mutation in original breast tumorBRCA1 status in residual surgical tissueBRCA1 status in recurrence (metastatic biopsy)
2392insT N/A; patient did not provide consent N/A 
3374insGA No secondary mutation N/A 
15 1479delAG 42-base pair deletion (Ser454_Lys467del) 42-base pair deletion (Ser454_Lys467del) 
18 W1712X (5255G>A) No secondary mutation N/A 
Patient No.Deleterious mutation in original breast tumorBRCA1 status in residual surgical tissueBRCA1 status in recurrence (metastatic biopsy)
2392insT N/A; patient did not provide consent N/A 
3374insGA No secondary mutation N/A 
15 1479delAG 42-base pair deletion (Ser454_Lys467del) 42-base pair deletion (Ser454_Lys467del) 
18 W1712X (5255G>A) No secondary mutation N/A 

Abbreviation: N/A, not applicable.

Figure 1.

Sequence data from tumor samples of patient 15.

Figure 1.

Sequence data from tumor samples of patient 15.

Close modal
Figure 2.

Schematic of BRCA1 reversion mutation in pretreatment tumor compared with posttreatment and recurrence samples.

Figure 2.

Schematic of BRCA1 reversion mutation in pretreatment tumor compared with posttreatment and recurrence samples.

Close modal

Because of the presence of nontumor DNA in the sample, it was not possible to determine whether there were residual tumor cells after neoadjuvant therapy that retained the original 1479delAG germline mutation and lacked the reversion mutation. However, upon analysis of two samples of extracted DNA in the pretreatment tumor specimen, no reversion mutation was detected. On the basis of our statistical constraints, we conclude that whether the reversion mutation was present in the pretreatment tumor specimen, it was in less than 1 in 250 tumor cells. DNA sequencing of the contralateral breast cancer specimen did not reveal evidence of a somatic reversion mutation. Specifically, 324 unique DNA sequence reads were present, spanning both the germline deletion mutation (1479delAG) and the somatic reversion, with 245 of the DNA sequence reads showing the germline mutation and zero reads showing the somatic reversion mutation.

Predicted and observed allele frequencies were consistent with the original mutation being germline and the subsequent reversion mutation being somatic using SNP data. The presence of the original mutation in the post-reversion mutation samples at levels consistent with residual germline DNA was also confirmation that the original mutation was germline (Appendix 1).

Clinical narrative of patient with BRCA1 reversion mutation

At the age of 25 years, this patient was diagnosed with stage IIIC right breast invasive ductal carcinoma that was ER/PR-negative and HER2-unamplified (i.e., triple-negative). A deleterious 1479delAG BRCA1 mutation was detected at breast cancer diagnosis. She received neoadjuvant chemotherapy with adriamycin and cyclophosphamide, followed by a right breast lumpectomy and axillary lymph node dissection. She was found to have achieved a pCR at the time of surgery with no disease in the lymph nodes. This was followed by adjuvant paclitaxel and whole breast radiation (50.4 Gy).

The patient had an in-breast recurrence 1 year later; this was also high-grade, triple-negative carcinoma for which she proceeded with a right mastectomy, followed by additional cycles of anthracycline- and taxane-based chemotherapy.

Approximately 10 years later, she was diagnosed with a contralateral high-grade, triple-negative breast cancer. She enrolled in PrECOG 0105 and received six cycles of gemcitabine, carboplatin, and iniparib. She proceeded with a left mastectomy upon completion of experimental therapy, and this revealed a 2-cm residual invasive carcinoma of mixed lobular and ductal histology (RCB II) with a negative sentinel lymph node dissection.

Within months of completing neoadjuvant therapy, a recurrent left axillary mass was discovered, prompting additional locoregional and systemic treatment. One year later, she was found to have distant disease spread. Her disease progressed rapidly, and she died less than 2 years after her second primary breast cancer diagnosis.

We report the case of a young woman with a history of bilateral triple-negative breast cancer and a germline BRCA1 1479delAG mutation who, over the course of 18 weeks of neoadjuvant platinum-based therapy, developed a 42-base pair somatic deletion in the BRCA1 gene, leading to a reversion of the underlying 1479delAG deleterious germline BRCA1 mutation. The same somatic BRCA1 reversion mutation was identified in a metastatic focus in an axillary lymph node at the time of locoregional relapse, 4 months after the patient's definitive surgery. To our knowledge, there has been no previous report of a BRCA1 somatic reversion mutation developing over the short span of neoadjuvant chemotherapy in a patient with newly diagnosed, early-stage breast cancer.

Two types of somatic reversion mutations have been described. In the case of our patient, a genetic reversion event leading to deletion of a frameshift mutation resulted in correction of the original frameshift, thereby producing an allele that contains a new mutation, but which now encodes a DNA repair–proficient BRCA1 protein. A second class of mutations involves direct reversion to wild-type of the original mutation. Studies have shown that these types of somatic reversions account for the majority of BRCA1/2 mutation–associated ovarian carcinomas with secondary restoration of the BRCA1 or BRCA2 reading frame (7). These ovarian tumors develop platinum resistance, and may also be resistant to PARP inhibitor therapy (15, 17, 21). Reversion mutations in this setting have been associated with a prior history of breast cancer or with exposure to more than one line of chemotherapy for the treatment of ovarian cancer. This has led to the hypothesis that such somatic mutations may have been present at the time of the ovarian cancer diagnosis in a small number of neoplastic cells that were subsequently selected by additional DNA-damaging therapy (17). Furthermore, somatic reversion mutations have also been reported with cisplatin treatment in breast and pancreatic cancer cell lines that had mutations in BRCA2 (19) and with the PARP inhibitor olaparib for a male BRCA2 mutation carrier with metastatic breast cancer (14).

BRCA1 is a large multidomain protein that includes the “really interesting new gene” (RING) finger, nuclear localization signal (NLS), coiled-coil, and BRCA1 C-terminus (BRCT) domains. Prior BRCA1 somatic reversion mutations have been reported in the RING finger domain and in downstream nucleotide sites (Fig. 3). In ovarian cancer, BRCA1 reversion mutations have been reported at several sites, including six patients with a 185delAG mutation and two patients with a 2594delC mutation (21, 24); however, reversion of the germline BRCA1 1479delAG mutation has not previously been reported in either breast or ovarian cancer.

Figure 3.

Known BRCA1 site–specific mutations with reported somatic reversion mutations.

Figure 3.

Known BRCA1 site–specific mutations with reported somatic reversion mutations.

Close modal

Our study has limitations. Here, we report an association between a somatic BRCA1 reversion mutation and therapeutic failure after neoadjuvant platinum-based therapy for breast cancer. However, we cannot establish a causal relationship, in particular, as our patient's breast cancer history was complex, given she had been treated for a contralateral high-grade breast cancer 10 years prior to her new triple-negative breast cancer diagnosis, and, as such, she was exposed to chemotherapy in the past. Her clinical response to gemcitabine, carboplatin, and iniparib may not be generalizable given her past oncologic history, and, as a result of past therapeutic exposures, she may have had other molecular triggers, besides the BRCA1 reversion, leading to chemoresistant tumor cells. Furthermore, this is a relatively small study, and corroboration in a larger cohort of BRCA1/2 carriers treated with neoadjuvant therapy for breast cancer is warranted.

We report a novel, acquired somatic frameshift mutation in BRCA1 leading to reversion of a 1479delAG germline BRCA1 mutation, which developed during 18 weeks of neoadjuvant platinum-based chemotherapy for triple-negative breast cancer. We conclude that this reversion mutation led to reduced treatment sensitivity, manifested as significant disease burden at definitive surgery and rapid metastatic recurrence shortly after completion of therapy. The timing and etiology of such a reversion mutation are unclear and may have stemmed from genomic instability of the primary tumor given an underlying deficiency in homologous recombination DNA repair. How frequently somatic reversion mutations occur in clinical practice is not well known but may have significant implications for rational treatment selection in BRCA1/2 mutation–associated cancers. Given the growing number of DNA repair–targeted therapies, characterization of the prevalence, time course, and correlates of reversion mutations in BRCA1/2 and other DNA repair genes is essential to guide patient care and drug development.

S. Vinayak reports receiving company-sponsored meals from Tesaro during a clinical trial investigator's meeting. M.L. Telli is a consultant/advisory board member for Myriad Genetics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A. Afghahi, S. Vinayak, R.W. Carlson, J.M. Ford, M.L. Telli

Development of methodology: A. Afghahi, K.M. Timms, J.M. Ford, M.L. Telli

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K.C. Jensen, R.W. Carlson, P.-J. Chang, E.A. Schackmann, A.-R. Hartman, J.M. Ford, M.L. Telli

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Afghahi, K.M. Timms, S. Vinayak, K.C. Jensen, A.-R. Hartman, J.M. Ford, M.L. Telli

Writing, review, and/or revision of the manuscript: A. Afghahi, K.M. Timms, S. Vinayak, K.C. Jensen, A.W. Kurian, R.W. Carlson, E.A. Schackmann, A.-R. Hartman, J.M. Ford, M.L. Telli

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.M. Timms, E.A. Schackmann, M.L. Telli

Study supervision: K.M. Timms, M.L. Telli

The authors gratefully acknowledge research support from the ASCO Conquer Cancer Foundation, Breast Cancer Research Foundation, BRCA Foundation, Stanford Cancer Institute, and Susan G. Komen for the Cure.

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.
Domchek
SM
,
Weber
BL
. 
Clinical management of BRCA1 and BRCA2 mutation carriers
.
Oncogene
2006
;
25
:
5825
31
.
2.
Gudmundsdottir
K
,
Ashworth
A
. 
The roles of BRCA1 and BRCA2 and associated proteins in the maintenance of genomic stability
.
Oncogene
2006
;
25
:
5864
74
.
3.
Moynahan
ME
,
Pierce
AJ
,
Jasin
M
. 
BRCA2 is required for homology-directed repair of chromosomal breaks
.
Mol Cell
2001
;
7
:
263
72
.
4.
Ford
JM
,
Kastan
MB
.
DNA damage response pathways and cancer
. Abeloff's Clinical Oncology, 5th ed.
Elsevier
; 
2013
.
5.
Gudmundsson
J
,
Johannesdottir
G
,
Bergthorsson
JT
,
Arason
A
,
Ingvarsson
S
,
Egilsson
V
, et al
Different tumor types from BRCA2 carriers show wild-type chromosome deletions on 13q12-q13
.
Cancer Res
1995
;
55
:
4830
2
.
6.
Neuhausen
SL
,
Marshall
CJ
. 
Loss of heterozygosity in familial tumors from three BRCA1-linked kindreds
.
Cancer Res
1994
;
54
:
6069
72
.
7.
Silver
DP
,
Livingston
DM
. 
Mechanisms of BRCA1 tumor suppression
.
Cancer Discov
2012
;
2
:
679
84
.
8.
Couch
FJ
,
Hart
SN
,
Sharma
P
,
Toland
AE
,
Wang
X
,
Miron
P
, et al
Inherited mutations in 17 breast cancer susceptibility genes among a large triple-negative breast cancer cohort unselected for family history of breast cancer
.
J Clin Oncol
2015
;
33
:
304
11
.
9.
Gonzalez-Angulo
AM
,
Timms
KM
,
Liu
S
,
Chen
H
,
Litton
JK
,
Potter
J
, et al
Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer
.
Clin Cancer Res
2011
;
17
:
1082
9
.
10.
Young
SR
,
Pilarski
RT
,
Donenberg
T
,
Shapiro
C
,
Hammond
LS
,
Miller
J
, et al
The prevalence of BRCA1 mutations among young women with triple-negative breast cancer
.
BMC Cancer
2009
;
9
:
86
.
11.
Ashworth
A
. 
A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair
.
J Clin Oncol
2008
;
26
:
3785
90
.
12.
Bryant
HE
,
Schultz
N
,
Thomas
HD
,
Parker
KM
,
Flower
D
,
Lopez
E
, et al
Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase
.
Nature
2005
;
434
:
913
7
.
13.
Farmer
H
,
McCabe
N
,
Lord
CJ
,
Tutt
AN
,
Johnson
DA
,
Richardson
TB
, et al
Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy
.
Nature
2005
;
434
:
917
21
.
14.
Tutt
A
,
Robson
M
,
Garber
JE
,
Domchek
SM
,
Audeh
MW
,
Weitzel
JN
, et al
Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial
.
Lancet
2010
;
376
:
235
44
.
15.
Isakoff
SJ
,
Mayer
EL
,
He
L
,
Traina
TA
,
Carey
LA
,
Krag
KJ
, et al
TBCRC009: a multicenter phase II clinical trial of platinum monotherapy with biomarker assessment in metastatic triple-negative breast cancer
.
J Clin Oncol
2015
;
33
:
1902
9
.
16.
Telli
ML
,
Jensen
KC
,
Vinayak
S
,
Kurian
AW
,
Lipson
JA
,
Flaherty
PJ
, et al
Phase II study of gemcitabine, carboplatin, and iniparib as neoadjuvant therapy for triple-negative and BRCA1/2 mutation-associated breast cancer with assessment of a tumor-based measure of genomic instability: PrECOG 0105
.
J Clin Oncol
2015
;
33
:
1895
901
.
17.
Tutt
A
,
Ellis
P
,
Kilburn
L
,
Gilett
C
,
Pinder
S
,
Abraham
J
, et al
The TNT trial: a randomized phase III trial of carboplatin compared with docetaxel for patients with metastatic or recurrent locally advanced triple negative or BRCA1/2 breast cancer [abstract]
. In: Proceedings of the 39th Annual
San Antonio Breast Cancer Symposium; 2014 Dec 9–13
;
San Antonio, TX
. Abstract nr S3-01.
18.
Barber
LJ
,
Sandhu
S
,
Chen
L
,
Campbell
J
,
Kozarewa
I
,
Fenwick
K
, et al
Secondary mutations in BRCA2 associated with clinical resistance to a PARP inhibitor
.
J Pathol
2013
;
229
:
422
9
.
19.
Dhillon
KK
,
Swisher
EM
,
Taniguchi
T
. 
Secondary mutations of BRCA1/2 and drug resistance
.
Cancer Sci
2011
;
102
:
663
9
.
20.
Edwards
SL
,
Brough
R
,
Lord
CJ
,
Natrajan
R
,
Vatcheva
R
,
Levine
DA
, et al
Resistance to therapy caused by intragenic deletion in BRCA2
.
Nature
2008
;
451
:
1111
5
.
21.
Norquist
B
,
Wurz
KA
,
Pennil
CC
,
Garcia
R
,
Gross
J
,
Sakai
W
, et al
Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas
.
J Clin Oncol
2011
;
29
:
3008
15
.
22.
Sakai
W
,
Swisher
EM
,
Jacquemont
C
,
Chandramohan
KV
,
Couch
FJ
,
Langdon
SP
, et al
Functional restoration of BRCA2 protein by secondary BRCA2 mutations in BRCA2-mutated ovarian carcinoma
.
Cancer Res
2009
;
69
:
6381
6
.
23.
Sakai
W
,
Swisher
EM
,
Karlan
BY
,
Agarwal
MK
,
Higgins
J
,
Friedman
C
, et al
Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers
.
Nature
2008
;
451
:
1116
20
.
24.
Swisher
EM
,
Sakai
W
,
Karlan
BY
,
Wurz
K
,
Urban
N
,
Taniguchi
T
. 
Secondary BRCA1 mutations in BRCA1-mutated ovarian carcinomas with platinum resistance
.
Cancer Res
2008
;
68
:
2581
6
.
25.
Symmans
WF
,
Peintinger
F
,
Hatzis
C
,
Rajan
R
,
Kuerer
H
,
Valero
V
, et al
Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy
.
J Clin Oncol
2007
;
25
:
4414
22
.
26.
Telli
ML
,
Timms
KM
,
Reid
J
,
Hennessy
B
,
Mills
GB
,
Jensen
KC
, 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
:
3764
73
.
27.
Timms
KM
,
Abkevich
V
,
Hughes
E
,
Neff
C
,
Reid
J
,
Morris
B
, et al
Association of BRCA1/2 defects with genomic scores predictive of DNA damage repair deficiency among breast cancer subtypes
.
Breast Cancer Res
2014
;
16
:
475
.
28.
Eggington
JM
,
Bowles
KR
,
Moyes
K
,
Manley
S
,
Esterling
L
,
Sizemore
S
, et al
A comprehensive laboratory-based program for classification of variants of uncertain significance in hereditary cancer genes
.
Clin Genet
2014
;
86
:
229
37
.

Supplementary data