Purpose: Preclinical data suggest that exposure to PARP inhibitors (PARPi) may compromise benefit to subsequent chemotherapy, particularly platinum-based regimens, in patients with BRCA1/2 mutation carrier ovarian cancer (PBMCOC), possibly through the acquisition of secondary BRCA1/2 mutations. The efficacy of chemotherapy in the PARPi-resistant setting was therefore investigated.

Experimental Design: We conducted a retrospective review of PBMCOC who received chemotherapy following disease progression on olaparib, administered at ≥200 mg twice daily for one month or more. Tumor samples were obtained in the post-olaparib setting where feasible and analyzed by massively parallel sequencing.

Results: Data were collected from 89 patients who received a median of 3 (range 1–11) lines of pre-olaparib chemotherapy. The overall objective response rate (ORR) to post-olaparib chemotherapy was 36% (24 of 67 patients) by Response Evaluation Criteria in Solid Tumors (RECIST) and 45% (35 of 78) by RECIST and/or Gynecologic Cancer InterGroup (GCIG) CA125 criteria with median progression-free survival (PFS) and overall survival (OS) of 17 weeks [95% confidence interval (CI), 13–21] and 34 weeks (95% CI, 26–42), respectively. For patients receiving platinum-based chemotherapy, ORRs were 40% (19 of 48) and 49% (26/53), respectively, with a median PFS of 22 weeks (95% CI, 15–29) and OS of 45 weeks (95% CI, 15–75). An increased platinum-to-platinum interval was associated with an increased OS and likelihood of response following post-olaparib platinum. No evidence of secondary BRCA1/2 mutation was detected in tumor samples of six PARPi-resistant patients [estimated frequency of such mutations adjusted for sample size: 0.125 (95%-CI: 0–0.375)].

Conclusions: Heavily pretreated PBMCOC who are PARPi-resistant retain the potential to respond to subsequent chemotherapy, including platinum-based agents. These data support the further development of PARPi in PBMCOC. Clin Cancer Res; 19(19); 5485–93. ©2013 AACR.

This article is featured in Highlights of This Issue, p. 5259

Translational Relevance

This study provides support for the lack of significant clinical cross-resistance between PARP inhibitors (PARPi), a promising class of novel targeted agents, and platinum-based chemotherapy, the current standard-of-care, in patients with ovarian cancer with germline BRCA1/2 mutations. First, high-response rates to chemotherapy in the PARPi-resistant setting were elicited in heavily pretreated patients. Second, the platinum-free interval remained prognostic of overall survival and predictive of response to platinum-based regimens in the PARPi-resistant setting despite the interim use of olaparib. Secondary BRCA1/2 mutations have been proposed as a common mechanism of resistance to PARPi; however, these were not detected in a series of tumor samples in this study using massively parallel sequencing. This indicates that other resistance mechanisms are likely to be important and merit further investigations. These novel findings provide support for the continued development of PARPi in patients with BRCA1/2-associated ovarian cancer.

The observation that BRCA1/2-deficient cells were exquisitely sensitive to PARP inhibitors (PARPi; ref. 1, 2) led to the clinical testing of this synthetic lethal approach. The proof-of-concept antitumor strategy of using single-agent PARP inhibition in BRCA1 and BRCA2 (BRCA1/2)-mutated tumors has now been validated by emerging safety and efficacy data from phase I–II clinical trials (3–6). Olaparib (AZD2281, KU-0059436) is a selective and potent inhibitor of PARP1/2 with in vitro potency in the nanomolar range and was the first oral PARPi to be used in this context. The efficacy of olaparib was confirmed in a randomized trial in BRCA1/2 mutation-positive patients with platinum-resistant/partially sensitive relapsed epithelial ovarian cancer (EOC; ref. 7). This study also showed an unexpectedly high level of efficacy for pegylated liposomal doxorubicin (PLD) in these patients (7).

The incidence of BRCA1/2 mutations in high-grade serous EOC is estimated at 15% to 18% irrespective of family history. Furthermore, up to 50% of patients with this commonest EOC histologic subtype may have other genetic changes that also lead to homologous recombination deficiency (8–10). The potential for these other mutations to phenocopy the effects of BRCA1/2 mutations has led to the assessment of PARPi monotherapy in patients with sporadic high-grade serous EOC (negative for germline BRCA1/2 mutations). In this patient group, clinical responses to single-agent olaparib have been confirmed in patients with platinum-sensitive relapsed disease (11). A placebo-controlled trial conducted in patients with platinum-sensitive, relapsed high-grade serous EOC (germline BRCA1/2 mutation status unknown in 64%, positive in 22%, and negative in 14% of patients) subsequently assessed the use of single-agent olaparib as maintenance therapy. Treatment with olaparib significantly improved progression free survival (PFS) compared with placebo (8.4 months versus 4.8 months) with a striking HR of 0.35 (95%-CI, 0.25–0.49). However, this effect on PFS did not translate into an overall survival (OS) difference in an interim analysis (12). Possible contributory factors to this lack of OS difference include the genetic admixture of patients with or without germline BRCA1/2 mutations and the reduction of the likelihood of response to subsequent chemotherapy following olaparib treatment. The latter would be consistent with data from preclinical models, which have predicted the development of cross-resistance between PARPi and platinum-based therapies through the acquisition of secondary mutations that restore the BRCA1/2 open reading frame and functional protein expression (13–15). Secondary BRCA1/2 mutations have been associated with clinical platinum resistance and their incidence is reportedly high at more than 46% in patients with platinum-resistant EOC (16). Our previous data additionally suggest a correlation between prior platinum sensitivity and antitumor response to PARPi in patients with BRCA1/2 deficiencies (5). However, there are as yet no published clinical data about the use of chemotherapy, particularly platinum-based regimens, in the PARPi-resistant setting.

The primary objective of this study was therefore to provide the first clinical evaluation of chemotherapy in patients with BRCA1/2 mutation-associated EOC following the development of disease progression on olaparib treatment. In addition to assessing objective response rates (ORR), associations between baseline clinical factors with OS and response to treatment with platinum-based chemotherapy in the post-olaparib setting were analyzed. To investigate the hypothesis that PARPi-resistant patients may harbor secondary BRCA1/2 mutations, tumor samples were obtained wherever feasible in the post-olaparib (PARPi-resistant) setting before chemotherapy and were examined using massively parallel sequencing.

This is a retrospective study of patients with EOC with germline BRCA1/2 mutations who received chemotherapy following objective evidence of disease progression on olaparib administered at doses 200 mg or more twice daily for more than a month in eight hospitals worldwide between April 2006 and May 2012. The minimum dose of 200 mg twice a day was selected because this was a pharmacologically active dose showed to inhibit both PARP target and pathway where antitumor responses were observed. This was also the dose used in the phase I clinical trial expansion cohort of patients with BRCA1/2-mutated EOC (5). Demonstration of objective evidence of disease progression on olaparib by response evaluation criteria in solid tumors (RECIST) or Gynecologic Cancer InterGroup (GCIG) CA125 criteria was mandatory in these patients and was confirmed by all investigators by site-specific review. Patients without evidence of radiologically measurable disease (using RECIST) were excluded, unless the disease was evaluable using GCIG CA125 criteria (17, 18). Patients who were initially only evaluable using GCIG-CA125 criteria at the start of treatment but assessed using a different CA125 assay during the course of treatment or received intervention to their pleura and/or peritoneum within 28 days before response assessments were excluded from response rate calculations. However, all patients with evaluable disease (by either criterion) were included in the survival analyses. Demographic and clinicopathologic factors, treatment details, and antitumor responses were recorded. All patients had previously undergone BRCA1/2 mutation testing following appropriate genetic counseling and informed consent. The study protocol was approved by the Institutional Review Board or independent ethics committee at each site.

Patients were defined as platinum-sensitive, partially sensitive, and resistant if the treatment-free interval was ≥12, ≥6 and <12, and <6 months, respectively, after their last pre-olaparib platinum-based chemotherapy (19). An additional time interval was calculated, that is, platinum-to-platinum (PTP) interval, which describes the interval from the end of the last pre-olaparib platinum treatment to the start of the first post-olaparib platinum-based chemotherapy. During chemotherapy in the post-olaparib setting, patients had safety evaluations following each treatment cycle and tumor response assessments after every two cycles, as assessed by RECIST criteria, and/or after every cycle with serum CA125 levels using GCIG criteria (17, 18).

Tumor tissue and DNA extraction

Following patient consent, formalin-fixed paraffin-embedded or fresh-frozen tumor tissue was obtained at surgery for relapsed disease where clinically indicated. Sample anonymization, tissue access, and recording of clinical data were undertaken in accordance with the Human Tissue Act and Multicentre Research Ethics Committee Guidelines. Microdissection was conducted as previously described (20). In two additional cases, tumor cells were isolated from ascites by Dynal bead separation. DNA was extracted using the DNeasy Kit (Qiagen).

Massively parallel sequencing of BRCA1, BRCA2, PTEN, and TP53

Two micrograms of DNA was fragmented to 200 bp fragments using a Covaris E-Series instrument and paired-end libraries were prepared using the SureSelect Target Enrichment System for Illumina Paired-End Sequencing Library Kit (Agilent). The libraries were then hybridized to a custom RNA bait library targeting BRCA1, BRCA2, TP53, and PTEN (eArray; Agilent) and quantified using a Bioanalyzer DNA chip (Agilent) and quantitative PCR (qPCR). Final libraries were run on an Illumina HiSeq using a 2 × 76 basepair paired-end strategy. After quality controls, reads were aligned to the human reference genome (GRCh37) using BWA (21). PCR duplicates and off-target reads were subsequently filtered and variants were called using GATK (22, 23). Bam files were visualized using the Integrated Genome Viewer (IGV, Broad).

Statistical analysis

Descriptive and inferential statistics were applied to baseline clinical and laboratory data. To account for the limited sample size, the Laplace and adjusted Wald methods were used to derive the conservative point estimate and 95% confidence interval (95% CI; ref. 24), respectively, of the frequency of secondary BRCA1/2 mutations. The Kaplan–Meier method was used to estimate intervals between milestones along a timeline. PFS and OS following post-PARPi chemotherapy were assessed from first administration of drug. Differences between defined groups of patients were assessed using log-rank (Mantel–Cox) test, multinomial logistic and Cox regression, where appropriate. In the univariate analysis, a P value of 0.10 was adopted as the limit for inclusion in the multivariate analysis; in the latter, P values less than 0.05 were considered significant. All P values presented were two-sided. The SPSS program (Version 16.0) was used for all statistical analysis.

Patient, tumor, and treatment characteristics

A total of 89 BRCA1/2 mutation carriers with EOC were included (Table 1). Sixty-six (74%) patients had germline BRCA1 mutations, whereas 23 (26%) carried BRCA2 mutations. These patients had received a median of three lines of chemotherapy before olaparib treatment. The percentages of patients with platinum-resistant, partially sensitive, and sensitive relapses before olaparib exposure were 40%, 43%, and 17%, respectively. The median interval from initial diagnosis to the start of post-olaparib chemotherapy was 47.2 months (95% CI, 38.1–56.3) and that between the commencement of olaparib and subsequent line of systemic chemotherapy was 7.4 months (95% CI, 6.5–8.3).

Table 1.

Patient, tumor, and treatment characteristics

CharacteristicsValues
Total no. of patients 89 
Age (y), median (range)   51 (31–77) 
Stage at diagnosis—no. (%) 
 1 5 (6) 
 2 7 (8) 
 3 63 (71) 
 4 8 (9) 
 Not known 6 (7) 
Histology grade—no. (%) 
 2 13 (15) 
 3 66 (74) 
 Not known 10 (11) 
Histology—no. (%) 
 Serous papillary 67 (75) 
 Endometrioid 7 (8) 
 Adenocarcinoma not otherwise specified 15 (17) 
Prior no. of lines of systemic chemotherapy (excluding olaparib)—no. (%)  
 1 13 (15) 
 2 30 (34) 
 3 23 (26) 
 ≥4 21 (24) 
 Not known 2 (2) 
BRCA mutation status—no. (%) 
BRCA1 66 (74) 
BRCA2 23 (26) 
Optimally debulked—no. (%) 
 Yes 48 (54) 
 No 23 (26) 
 Not known 18 (20) 
Previous breast cancer—no. (%) 
 Yes 29 (33) 
 Chemotherapy for breast cancer 11 (38) 
 No 60 (67) 
Sensitivity to last pre-PARPi platinum chemotherapy—no. (%) 
 Sensitive (TFI >12 m) 15 (17) 
 Partially sensitive (TFI >6 and ≤12 m) 38 (43) 
 Resistant (TFI <6 m) 36 (40) 
Overall response rate to olaparib—no. of responses/no. evaluable (%) 
 RECIST 34/86 (40) 
 RECIST and/or CA125 41/87 (47) 
CharacteristicsValues
Total no. of patients 89 
Age (y), median (range)   51 (31–77) 
Stage at diagnosis—no. (%) 
 1 5 (6) 
 2 7 (8) 
 3 63 (71) 
 4 8 (9) 
 Not known 6 (7) 
Histology grade—no. (%) 
 2 13 (15) 
 3 66 (74) 
 Not known 10 (11) 
Histology—no. (%) 
 Serous papillary 67 (75) 
 Endometrioid 7 (8) 
 Adenocarcinoma not otherwise specified 15 (17) 
Prior no. of lines of systemic chemotherapy (excluding olaparib)—no. (%)  
 1 13 (15) 
 2 30 (34) 
 3 23 (26) 
 ≥4 21 (24) 
 Not known 2 (2) 
BRCA mutation status—no. (%) 
BRCA1 66 (74) 
BRCA2 23 (26) 
Optimally debulked—no. (%) 
 Yes 48 (54) 
 No 23 (26) 
 Not known 18 (20) 
Previous breast cancer—no. (%) 
 Yes 29 (33) 
 Chemotherapy for breast cancer 11 (38) 
 No 60 (67) 
Sensitivity to last pre-PARPi platinum chemotherapy—no. (%) 
 Sensitive (TFI >12 m) 15 (17) 
 Partially sensitive (TFI >6 and ≤12 m) 38 (43) 
 Resistant (TFI <6 m) 36 (40) 
Overall response rate to olaparib—no. of responses/no. evaluable (%) 
 RECIST 34/86 (40) 
 RECIST and/or CA125 41/87 (47) 

Efficacy of chemotherapy in the setting of PARPi resistance

Of 89 patients, 78 had disease measurable by RECIST and/or GCIG CA125 criteria. Chemotherapy regimens used are detailed in Tables 2 and 3; the majority comprised either a platinum and/or taxane chemotherapy and these were administered through weekly, three-weekly, or monthly schedules. Of note, single-agent paclitaxel chemotherapy was administered weekly. The overall ORR with the use of any first-line chemotherapy following olaparib treatment was 36% (24 of 67 patients) by RECIST and 45% (35 of 78 patients) by RECIST and/or GCIG criteria. After a median follow-up of 113 weeks (26.3 months), the overall median PFS was 17 weeks (95% CI, 13–21) and median OS was 34 weeks (95% CI, 26–42). In comparison, the use of platinum-based regimens resulted in ORRs of 40% (19 of 48 patients) by RECIST criteria and 49% (26 of 53 patients) by RECIST and/or GCIG criteria with a median PFS of 22 weeks (95% CI, 15–29) and a median OS of 45 weeks (95%-CI: 15–75). Objective responses to chemotherapy in the post-PARPi setting were observed regardless of pre-PARPi platinum sensitivity (Table 3). Responses were also observed with the use of other chemotherapeutic regimens; the use of taxane and PLD monotherapies were associated with ORRs by RECIST and/or GCIG criteria of 50% (5 of 10 patients) and 30% (3 of 10 patients), respectively (Table 2). The median overall interval between first diagnosis and death was 66.3 months (95% CI, 53.4–79.2) with a median interval between first diagnosis and the start of olaparib of 35.8 months (95% CI, 32–40).

Table 2.

Types and efficacies of chemotherapies used in the post-olaparib first-line setting

RECISTRECIST and/or GCIG
RegimensEvaluableResponses (%)EvaluableResponses (%)
Platinum-based chemotherapies 48 19 (40) 53 26 (49) 
 Platinum single-agent 3 (50) 3 (50) 
 Platinum-taxane 19 8 (42) 22 14 (64) 
 Platinum-PLD 10 4 (40) 11 5 (45) 
 Platinum-others 13 4 (31) 14 4 (29) 
Taxane single-agent 10 4 (40) 10 5 (50) 
PLD single-agent 0 (0) 10 3 (30) 
Others 1 (25) 1 (20) 
Total 67 24 (36) 78 35 (45) 
RECISTRECIST and/or GCIG
RegimensEvaluableResponses (%)EvaluableResponses (%)
Platinum-based chemotherapies 48 19 (40) 53 26 (49) 
 Platinum single-agent 3 (50) 3 (50) 
 Platinum-taxane 19 8 (42) 22 14 (64) 
 Platinum-PLD 10 4 (40) 11 5 (45) 
 Platinum-others 13 4 (31) 14 4 (29) 
Taxane single-agent 10 4 (40) 10 5 (50) 
PLD single-agent 0 (0) 10 3 (30) 
Others 1 (25) 1 (20) 
Total 67 24 (36) 78 35 (45) 
Table 3.

Breakdown of objective responses to different chemotherapeutic regimens used in the post-PARPi setting according to pre-PARPi platinum sensitivity

Response by RECIST and/or GCIG criteria (events/evaluable; %)
Chemotherapeutic regimen usedPlatinum-resistantPlatinum-partially sensitivePlatinum-sensitive
Platinum-based chemotherapies 5/14 (36) 16/26 (62) 5/13 (38) 
 Platinum single-agent 1/3 (33) 1/2 (50) 1/1 (100) 
 Platinum-taxane 3/5 (60) 9/10 (90) 2/7 (29) 
 Platinum-PLD 1/4 (25) 3/5 (60) 1/2 (50) 
 Platinum-others 0/2 (0) 3/9 (33) 1/3 (33) 
Taxane single-agent 3/8 (38) 2/2 (100) 0/0 (0) 
PLD single-agent 2/6 (33) 1/3 (33) 0/1 (0) 
Others 1/2 (50) 0/3 (0) 0/0 (0) 
Total 11/30 (37) 19/34 (56) 5/14 (36) 
Response by RECIST and/or GCIG criteria (events/evaluable; %)
Chemotherapeutic regimen usedPlatinum-resistantPlatinum-partially sensitivePlatinum-sensitive
Platinum-based chemotherapies 5/14 (36) 16/26 (62) 5/13 (38) 
 Platinum single-agent 1/3 (33) 1/2 (50) 1/1 (100) 
 Platinum-taxane 3/5 (60) 9/10 (90) 2/7 (29) 
 Platinum-PLD 1/4 (25) 3/5 (60) 1/2 (50) 
 Platinum-others 0/2 (0) 3/9 (33) 1/3 (33) 
Taxane single-agent 3/8 (38) 2/2 (100) 0/0 (0) 
PLD single-agent 2/6 (33) 1/3 (33) 0/1 (0) 
Others 1/2 (50) 0/3 (0) 0/0 (0) 
Total 11/30 (37) 19/34 (56) 5/14 (36) 

Factors associated with OS following post-olaparib platinum-based chemotherapy

In the univariate analysis (Table 4), factors significantly associated with OS following post-olaparib platinum-based chemotherapy included best response to olaparib by RECIST and/or GCIG criteria [overall P = 0.031; stable disease (SD) versus progressive disease (PD); P = 0.026; HR = 0.299 (95% CI, 0.103–0.868); partial response (PR)/complete response (CR) versus PD; P = 0.009; HR = 0.257 (95% CI, 0.093–0.709) and the PTP interval [P = 0.018; HR = 0.960 (95%-CI: 0.928–0.993)]. Interestingly, pre-PARPi platinum-sensitivity [measured by the treatment-free interval (TFI) following last pre-olaparib platinum] was not significantly associated with OS (overall P = 0.332).

Table 4.

Table summarizing the association of baseline factors with OS in patients treated with platinum-based chemotherapy in the post-olaparib setting

UnivariateMultivariate
VariablesHRPHRP
Age (y) 0.981 (0.948–1.015) 0.267 
Grade (3 vs. 2) 1.066 (0.325–3.493) 0.916 
Stage at diagnosis (III or IV vs. I or II) 1.072 (0.442–2.599) 0.877 
Serous histology 0.834 (0.293–2.380) 0.735 
Optimally debulked (<1 cm) 0.598 (0.291–1.229) 0.162 
BRCA2 vs. BRCA1 0.970 (0.461–2.042) 0.936 
Previous breast cancer 0.661 (0.341–1.281) 0.220 
Residual disease after first-line treatment 1.665 (0.773–3.588) 0.193 
Olaparib RECIST and/or GCIG response  0.031  0.056 
 SD vs. PD 0.299 (0.103–0.868) 0.026 0.301 (0.104–0.875) 0.027 
 PR/CR vs. PD 0.257 (0.093–0.709) 0.009 0.298 (0.106–0.838) 0.022 
Last pre-PARPi platinum sensitivity  0.332   
 Partial sensitive vs. resistant 0.902 (0.448–1.815) 0.773 
 Sensitive vs. resistant 0.522 (0.214–1.276) 0.154   
PTP interval (mo) 0.960 (0.928–0.993) 0.018 0.962 (0.928–0.997) 0.036 
UnivariateMultivariate
VariablesHRPHRP
Age (y) 0.981 (0.948–1.015) 0.267 
Grade (3 vs. 2) 1.066 (0.325–3.493) 0.916 
Stage at diagnosis (III or IV vs. I or II) 1.072 (0.442–2.599) 0.877 
Serous histology 0.834 (0.293–2.380) 0.735 
Optimally debulked (<1 cm) 0.598 (0.291–1.229) 0.162 
BRCA2 vs. BRCA1 0.970 (0.461–2.042) 0.936 
Previous breast cancer 0.661 (0.341–1.281) 0.220 
Residual disease after first-line treatment 1.665 (0.773–3.588) 0.193 
Olaparib RECIST and/or GCIG response  0.031  0.056 
 SD vs. PD 0.299 (0.103–0.868) 0.026 0.301 (0.104–0.875) 0.027 
 PR/CR vs. PD 0.257 (0.093–0.709) 0.009 0.298 (0.106–0.838) 0.022 
Last pre-PARPi platinum sensitivity  0.332   
 Partial sensitive vs. resistant 0.902 (0.448–1.815) 0.773 
 Sensitive vs. resistant 0.522 (0.214–1.276) 0.154   
PTP interval (mo) 0.960 (0.928–0.993) 0.018 0.962 (0.928–0.997) 0.036 

In the multivariate analysis, the PTP interval remained significant [P = 0.036; HR = 0.962 (95%-CI, 0.928–0.997)], whereas the best response (RECIST and/or GCIG) to olaparib showed a trend to significance [overall P = 0.056; SD versus PD, P = 0.027; HR = 0.301 (95%-CI: 0.104–0.875); PR/CR versus PD, P = 0.022; HR = 0.298 (95%-CI: 0.106–0.838)]. These results are summarized in Table 4.

Predictors of response to post-olaparib platinum-based chemotherapy

The association between post-PARPi platinum-based chemotherapy response (RECIST and/or GCIG) and baseline factors was assessed; these baseline factors included pre-PARPi platinum sensitivity (TFI following last pre-PARPi platinum), best response to olaparib (RECIST and/or GCIG), and the PTP interval (Table 5). The PTP interval was significantly associated with objective response to post-PARPi platinum-based chemotherapy [PR/CR versus PD, P = 0.045, OR = 1.12 (95% CI, 1.01–1.26)]. A trend to significance was observed between the PTP interval and the comparison of SD with PD on post-PARPi platinum-based chemotherapy [P = 0.061, OR = 1.12 (95% CI, 1.11–1.25)]. By contrast, the interval between last pre-PARPi platinum and start of post-PARPi chemotherapy was not significantly associated with the response to non-platinum regimens (CR/PR versus PD, P = 0.57).

Table 5.

Associations between objective response to first-line post-olaparib platinum-based chemotherapy and (i) olaparib response, (ii) pre-olaparib platinum sensitivity (measured by TFI), and (iii) interval between last pre-olaparib and first post-olaparib platinum-based chemotherapies (PTP interval)

Post-olaparib platinum chemotherapy response by RECIST and/or GCIG criteria
SD vs. PDPR/CR vs. PD
POR (95% CI)POR (95% CI)
Olaparib response by RECIST and/or GCIG SD vs. PD 0.105 14 (0.58–338.78) 0.184 7.0 (0.40–123.35) 
 PR/CR vs. PD 0.341 3.6 (0.26–50.33) 0.275 3.4 (0.38—30.66) 
TFI after last pre-PARPi platinum (months) 0.874 1.008 (0.915–1.110) 0.921 1.005 (0.916–1.102) 
PTP interval (months) 0.061 1.117 (0.995–1.254) 0.045 1.124 (1.003–1.264) 
Post-olaparib platinum chemotherapy response by RECIST and/or GCIG criteria
SD vs. PDPR/CR vs. PD
POR (95% CI)POR (95% CI)
Olaparib response by RECIST and/or GCIG SD vs. PD 0.105 14 (0.58–338.78) 0.184 7.0 (0.40–123.35) 
 PR/CR vs. PD 0.341 3.6 (0.26–50.33) 0.275 3.4 (0.38—30.66) 
TFI after last pre-PARPi platinum (months) 0.874 1.008 (0.915–1.110) 0.921 1.005 (0.916–1.102) 
PTP interval (months) 0.061 1.117 (0.995–1.254) 0.045 1.124 (1.003–1.264) 

DNA sequencing of clinical samples

A massively parallel sequencing strategy that focused on four genes, BRCA1, BRCA2, TP53, and PTEN, was used as described previously (25). For each sample, the 0.1 Mb targeted region was sequenced to a median depth of more than 200 times, and more than 99.9% of target regions were covered sufficiently for confident variant calling (depth >10 reads). In 6 patients with objective evidence of disease progression on treatment with olaparib (Supplementary Table S1), secondary BRCA1/2 mutations were not observed [conservative estimated frequency adjusted for sample size: 0.125 (95% CI, 0–0.375)]. Coding mutations in PTEN were not detected; the coding mutations found in BRCA1, BRCA2, and TP53 are summarized in Table 6.

Table 6.

Results of massively parallel sequencing of tumors obtained from the six patients in this study.

BRCA1 mutation reads (%)BRCA2 mutation reads (%)TP53 mutation reads (%)
PatientTime of tumor biopsyBiopsy siteBRCA1 mutation (rs number)Wild-type allelesMutant allelesBRCA2 mutation (rs number)Wild-type allelesMutant allelesTP53 mutationWild-type allelesMutant alleles
Diagnosis Right ovary c.3005delA/p.N1002fs (80357846) 257 (58) 183 (42) c.5946delT p.2003 (80359550) 17 (89) 2 (11) c.949C>T/p.Q317* 169 (76) 52 (24) 
 Diagnosis Left ovary c.3005delA/p.N1002fs (80357846) 79 (24) 244 (76) – – – c.949C>T/p.Q317* 179 (79) 48 (21) 
 Diagnosis LN c.3005delA/p.N1002fs (80357846) 112 (21) 426 (79) – – – c.949C>T/p.Q317* 178 (77) 52 (23) 
 Post-PARPi LN 1 c.3005delA/p.N1002fs (80357846) 102 (18) 453 (82) – – – c.949C>T/p.Q317* 164 (71) 66 (29) 
 Post-PARPi LN 2 c.3005delA/p.N1002fs (80357846) 88 (17) 435 (83) – – – c.949C>T/p.Q317* 160 (73) 59 (27) 
Diagnosis Right ovary c.3760_3761insT/p.K1254fs (80357986) 87 (42) 120 (58) – – – c.659A>G/p.Y220C 15 (42) 21 (58) 
 Post-PARPi Pelvic sidewall tumor c.3760_3761insT/p.K1254fs (80357986) 130 (51) 126 (49)    c.659A>G/p.Y220C 1 (6) 16 (94) 
Diagnosis Left ovary c.66_67delAG/p.E23fs (80357713) 28 (17) 132 (83) — — — – – – 
 Post-PARPi Peritoneal tumor c.66_67delAG/p.E23fs (80357713) 24 (27) 66 (73)       
Post first-line Vaginal vault lesion c.3450_3453delCAAG/p.Q1111fs (80357903) 50 (12) 382 (88) — — — – – – 
 Post-PARPi Para-aortic LN c.3450_3453delCAAG/p.Q1111fs (80357903) 74 (20) 298 (80)       
Post-PARPi Ascites – — — c.8975_9100del/p.Pro2992_Gln3034 (80359736) 167 (61) 104 (38) c.794A>G/p.L265P 60 (79) 16 (21) 
Post-PARPi Ascites c.1961_1962insA/p.K654fs (80357853) 311 (57) 232 (43) — — — c.489insTGCTTG/pS166* 243 (98) 5 (2) 
BRCA1 mutation reads (%)BRCA2 mutation reads (%)TP53 mutation reads (%)
PatientTime of tumor biopsyBiopsy siteBRCA1 mutation (rs number)Wild-type allelesMutant allelesBRCA2 mutation (rs number)Wild-type allelesMutant allelesTP53 mutationWild-type allelesMutant alleles
Diagnosis Right ovary c.3005delA/p.N1002fs (80357846) 257 (58) 183 (42) c.5946delT p.2003 (80359550) 17 (89) 2 (11) c.949C>T/p.Q317* 169 (76) 52 (24) 
 Diagnosis Left ovary c.3005delA/p.N1002fs (80357846) 79 (24) 244 (76) – – – c.949C>T/p.Q317* 179 (79) 48 (21) 
 Diagnosis LN c.3005delA/p.N1002fs (80357846) 112 (21) 426 (79) – – – c.949C>T/p.Q317* 178 (77) 52 (23) 
 Post-PARPi LN 1 c.3005delA/p.N1002fs (80357846) 102 (18) 453 (82) – – – c.949C>T/p.Q317* 164 (71) 66 (29) 
 Post-PARPi LN 2 c.3005delA/p.N1002fs (80357846) 88 (17) 435 (83) – – – c.949C>T/p.Q317* 160 (73) 59 (27) 
Diagnosis Right ovary c.3760_3761insT/p.K1254fs (80357986) 87 (42) 120 (58) – – – c.659A>G/p.Y220C 15 (42) 21 (58) 
 Post-PARPi Pelvic sidewall tumor c.3760_3761insT/p.K1254fs (80357986) 130 (51) 126 (49)    c.659A>G/p.Y220C 1 (6) 16 (94) 
Diagnosis Left ovary c.66_67delAG/p.E23fs (80357713) 28 (17) 132 (83) — — — – – – 
 Post-PARPi Peritoneal tumor c.66_67delAG/p.E23fs (80357713) 24 (27) 66 (73)       
Post first-line Vaginal vault lesion c.3450_3453delCAAG/p.Q1111fs (80357903) 50 (12) 382 (88) — — — – – – 
 Post-PARPi Para-aortic LN c.3450_3453delCAAG/p.Q1111fs (80357903) 74 (20) 298 (80)       
Post-PARPi Ascites – — — c.8975_9100del/p.Pro2992_Gln3034 (80359736) 167 (61) 104 (38) c.794A>G/p.L265P 60 (79) 16 (21) 
Post-PARPi Ascites c.1961_1962insA/p.K654fs (80357853) 311 (57) 232 (43) — — — c.489insTGCTTG/pS166* 243 (98) 5 (2) 

Abbreviation: LN, lymph node.

This is the first study to systematically describe the use of chemotherapy in patients with BRCA1/2-mutated EOC in the PARPi-resistant setting. Our data indicate that patients continue to have the potential to respond to further chemotherapy; the overall ORRs of first-line chemotherapy following PARPi were 36% by RECIST criteria and 45% by RECIST and/or GCIG criteria. For platinum-based regimens, the ORRs were 40% and 49% according to RECIST criteria and RECIST and/or GCIG criteria, respectively. It is noteworthy that similar levels of ORRs were also observed with the use of taxanes and PLD as monotherapies (Tables 2 and 3). Although published data on ORRs are scarce in patients with EOC carrying germline BRCA1/2 mutations in the third-line (and later) settings, the ORRs observed in this study seem to be in keeping with other phase II data for the same regimens in the general EOC population (26). Overall, these data point to significant differences in the mechanisms of resistance between PARPi and chemotherapy including platinum-based agents.

The exploratory analyses involving predictors of response and OS with the use of post-PARPi platinum-based chemotherapy indicate that both endpoints were significantly associated with the PTP interval despite the use of the putatively cross-resistant olaparib during this time interval. If resistance mechanisms to olaparib and platinum-based agents had been significantly overlapping, we would not have expected to observe either a high ORR to platinum-based regimens (approximately 50% in heavily pretreated patients in the fifth-line setting) or a PTP interval which retained prognostic and predictive values despite the interim use of olaparib. In this context, we note that the use of non-cross–resistant treatments to prolong the platinum-free interval seems to be associated with an increased likelihood of response to later rechallenge with platinum in previous studies (27). These data may also prove useful in planning future clinical trials and in the selection of therapy, that is, platinum or non-platinum–based treatments, for patients previously treated with PARPi. For instance, previously platinum-resistant patients who had a prolonged remission with olaparib might be expected to benefit when rechallenged with a platinum-based regimen in the post-PARPi setting. Although our data might suggest that the use of PARP inhibitors could possibly restore platinum sensitivity, there are currently no other preclinical or clinical data to support this hypothesis; further work is certainly warranted in this regard.

Secondary BRCA1/2 mutations leading to restoration of BRCA1/2 function have been associated with platinum resistance and proposed as a common mechanism linking platinum and PARPi resistance. However, in this study, secondary BRCA1/2 mutations were not detected by massively parallel sequencing of tumor samples collected from PARPi-resistant patients despite having achieved sufficient coverage and depth for confident variant calling. The conservative estimated frequency of such mutations in this PARPi-resistant cohort of patients is 0.125 (95% CI, 0–0.375). Although this estimate is adjusted for the limited size of our sample population, we recognize the limitations of this analysis and also that a significant proportion of patients treated with platinum-based regimens did not achieve objective responses. On the basis of the published data, we would have expected to observe a higher frequency of secondary BRCA1/2 mutations and it would clearly be desirable to study a larger patient population to address definitively their clinical relevance (16). There is currently one published case in the literature (reported by our group) of an patient with EOC with BRCA1/2 germline mutation who developed a PARPi-resistant lesion with a secondary BRCA1/2 mutation (which restored the BRCA2 open reading frame; ref. 25); she was not included in the current study as she did not receive post-olaparib chemotherapy. In this study, the failure to observe secondary BRCA1/2 mutations in the PARPi-resistant patients suggests other mechanisms may play a role in mediating PARPi resistance. These might include changes in the activity of the ABC transporter family (these would not be relevant to carboplatin resistance) and/or the loss of 53BP1 function leading to restoration of homologous recombination (28). Such events would have been undetected using our strategy and further evaluation of these mechanisms is certainly warranted in light of our findings.

A potential benefit for the use of PARPi therapy in patients with EOC with germline BRCA1/2 mutations lies in its favorable toxicity profile and route of administration compared with chemotherapy (7), leading to a longer chemotherapy-free interval and associated improvements in quality-of-life (12). Another relevant question is whether PARPi therapy for BRCA1/2 mutation-positive EOC adds a meaningful OS benefit to the current standard of care, i.e., platinum-based chemotherapy. In this series, the relatively short median OS of 34 weeks following post-olaparib chemotherapy is likely to reflect the extent of prior therapy for most patients (3 median lines of pre-olaparib chemotherapy). Moreover, patients in this series could only access olaparib through clinical trials mainly (83%) involving resistant/partially sensitive disease, that is, patients with anticipated shorter OS. The median OS from initial diagnosis of 66.3 months for our patients lies within the range previously reported in observational studies of BRCA1/2-mutated patients with EOC before PARPi became available (29, 30) and randomized trials will be required to properly assess the impact of olaparib on OS in BRCA1/2 mutation-associated EOC patients. In this context, a recently reported subgroup analysis of OS in the olaparib maintenance trial does suggest an OS benefit (HR of 0.74) for patients positive for germline and/or somatic BRCA1/2 mutations which was not evident in the overall population (HR of 0.88; ref 12, 31).

A strength of this study is that it represents the combined experience from several comprehensive cancer centers and data are relatively mature. We were therefore able to collect data from 89 patients despite the strict inclusion of only germline BRCA1/2-carrier EOC patients who were exposed to an adequate dose and duration of olaparib treatment. However, it has the limitations inherent in a retrospective study and central review of responses was not conducted. Nevertheless, all data were reviewed and confirmed by experienced investigators at each site.

In summary, our data indicate that chemotherapy, including platinum, taxane, and anthracycline-based regimens, retains efficacy in heavily pretreated EOC patients with germline BRCA1/2 mutations who have progressed on olaparib. The analyses of prognostic and predictive clinical variables also point to PARPi and platinum-based regimens lacking significant clinical cross-resistance. In addition, the failure to observe secondary BRCA1/2 mutations in this study indicates that other mechanisms of PARPi resistance are likely to be important. Together with evidence from several published studies showing a high level of patient acceptability to single-agent PARPi treatment, these data provide strong support for the further evaluation of single-agent PARPi in patients with EOC with BRCA1/2 mutations.

C. Gourley has commercial research grant from AstraZeneca. J.S. de Bono is employed as a Professor at the Institute of Cancer Research and is a consultant/advisory board member of and has commercial research grant from AstraZeneca. A. Ashworth may benefit financially from the development of PARP inhibitors through patents held jointly with AstraZeneca through the Institute of Cancer Research and rewards to inventors scheme. S.B. Kaye is a consultant/advisory board member of AstraZeneca Advisory Board.

Conception and design: J.E. Ang, T.A. Yap, J.S. de Bono, M.E. Gore

Development of methodology: J.E. Ang, T.A. Yap

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.E. Ang, C. Gourley, C.B. Powell, H. High, R. Shapira-Frommer, V. Castonguay, J. De Greve, T.A. Yap, S. Sandhu, L.-M. Chen, M.L. Friedlander, B. Kaufman, A.M. Oza, U. Matulonis, L.J. Barber, I. Kozarewa, K. Fenwick, I. Assiotis, J.S. de Bono, M.E. Gore, C.J. Lord, A. Ashworth

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.E. Ang, C. Gourley, T.A. Yap, A.M. Oza, L.J. Barber, J. Campbell, L. Chen, J.S. de Bono, C.J. Lord, A. Ashworth

Writing, review, and/or revision of the manuscript: J.E. Ang, C. Gourley, C.B. Powell, H. High, R. Shapira-Frommer, V. Castonguay, J. De Greve, T.A. Yap, S. Sandhu, S. Banerjee, L.-M. Chen, M.L. Friedlander, B. Kaufman, A.M. Oza, U. Matulonis, J.S. de Bono, M.E. Gore, C.J. Lord

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): J.E. Ang, C. Gourley, C.B. Powell, T. Atkinson, C.J. Lord

Study supervision: J.E. Ang, T.A. Yap, J.S. de Bono, M.E. Gore, S.B. Kaye

Presented herein are follow-up data from patients who previously completed AstraZeneca-sponsored studies (ClinicalTrials.gov: NCT00516373, NCT00777582, NCT00628251, NCT00494442, and NCT00679783). The Drug Development Unit of The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, and Drs. C.J. Lord and A. Ashworth are supported in part by program grants from Cancer Research UK. Support was also provided by Experimental Cancer Medicine Centre grants (to The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, and the Edinburgh Cancer Research UK Centre), the National Institute for Health Research Biomedical Research Centre (jointly to The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research), the Wellcome Trust (grant 090952/Z/09/Z; to J.E. Ang). C. Gourley is supported by a Scottish Senior Clinical Fellowship Award.

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.
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
.
2.
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
.
3.
Audeh
MW
,
Carmichael
J
,
Penson
RT
,
Friedlander
M
,
Powell
B
,
Bell-McGuinn
KM
, et al
Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial
.
Lancet
2010
;
376
:
245
51
.
4.
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
.
5.
Fong
PC
,
Yap
TA
,
Boss
DS
,
Carden
CP
,
Mergui-Roelvink
M
,
Gourley
C
, et al
Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval
.
J Clin Oncol
2010
;
28
:
2512
9
.
6.
Fong
PC
,
Boss
DS
,
Yap
TA
,
Tutt
A
,
Wu
P
,
Mergui-Roelvink
M
, et al
Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers
.
N Engl J Med
2009
;
361
:
123
34
.
7.
Kaye
SB
,
Lubinski
J
,
Matulonis
U
,
Ang
JE
,
Gourley
C
,
Karlan
BY
, et al
Phase II, open-label, randomized, multicenter study comparing the efficacy and safety of olaparib, a poly (ADP-ribose) polymerase inhibitor, and pegylated liposomal doxorubicin in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer
.
J Clin Oncol
2012
;
30
:
372
9
.
8.
Cancer Genome Atlas Research Network
. 
Integrated genomic analyses of ovarian carcinoma
.
Nature
2011
;
474
:
609
15
.
9.
Hennessy
BT
,
Timms
KM
,
Carey
MS
,
Gutin
A
,
Meyer
LA
,
Flake
DD
 II
, et al
Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly (ADP ribose) polymerase inhibitors in ovarian cancer
.
J Clin Oncol
2010
;
28
:
3570
6
.
10.
Press
JZ
,
De Luca
A
,
Boyd
N
,
Young
S
,
Troussard
A
,
Ridge
Y
, et al
Ovarian carcinomas with genetic and epigenetic BRCA1 loss have distinct molecular abnormalities
.
BMC Cancer
2008
;
8
:
17
.
11.
Gelmon
KA
,
Tischkowitz
M
,
Mackay
H
,
Swenerton
K
,
Robidoux
A
,
Tonkin
K
, et al
Olaparib in patients with recurrent high-grade serous or poorly differentiated ovarian carcinoma or triple-negative breast cancer: a phase 2, multicentre, open-label, non-randomised study
.
Lancet Oncol
2011
;
12
:
852
61
.
12.
Ledermann
J
,
Harter
P
,
Gourley
C
,
Friedlander
M
,
Vergote
I
,
Rustin
G
, et al
Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer
.
N Engl J Med
2012
;
366
:
1382
92
.
13.
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
.
14.
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
.
15.
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
.
16.
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
.
17.
Rustin
GJ
,
Quinn
M
,
Thigpen
T
,
du Bois
A
,
Pujade-Lauraine
E
,
Jakobsen
A
, et al
Re: New guidelines to evaluate the response to treatment in solid tumors (ovarian cancer)
.
J Natl Cancer Inst
2004
;
96
:
487
8
.
18.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
19.
Agarwal
R
,
Kaye
SB
. 
Ovarian cancer: strategies for overcoming resistance to chemotherapy
.
Nat Rev Cancer
2003
;
3
:
502
16
.
20.
Geyer
FC
,
Weigelt
B
,
Natrajan
R
,
Lambros
MB
,
de Biase
D
,
Vatcheva
R
, et al
Molecular analysis reveals a genetic basis for the phenotypic diversity of metaplastic breast carcinomas
.
J Pathol
2010
;
220
:
562
73
.
21.
Li
H
,
Durbin
R
. 
Fast and accurate short read alignment with Burrows–Wheeler transform
.
Bioinformatics
2009
;
25
:
1754
60
.
22.
McKenna
A
,
Hanna
M
,
Banks
E
,
Sivachenko
A
,
Cibulskis
K
,
Kernytsky
A
, et al
The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data
.
Genome Res
2010
;
20
:
1297
303
.
23.
DePristo
MA
,
Banks
E
,
Poplin
R
,
Garimella
KV
,
Maguire
JR
,
Hartl
C
, et al
A framework for variation discovery and genotyping using next-generation DNA sequencing data
.
Nat Genet
2011
;
43
:
491
8
.
24.
Lewis
JR
,
Sauro
J
. 
When 100% really isn't 100%: improving the accuracy of small-sample estimates of completion rates
.
J Usability Studies
2006
;
1
:
136
50
.
25.
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
.
26.
Hanker
LC
,
Loibl
S
,
Burchardi
N
,
Pfisterer
J
,
Meier
W
,
Pujade-Lauraine
E
, et al
The impact of second to sixth line therapy on survival of relapsed ovarian cancer after primary taxane/platinum-based therapy
.
Ann Oncol
2012
;
23
:
2605
12
.
27.
Spriggs
D
. 
Optimal sequencing in the treatment of recurrent ovarian cancer
.
Gynecol Oncol
2003
;
90
:
S39
44
.
28.
Fojo
T
,
Bates
S
. 
Mechanisms of resistance to PARP inhibitors–three and counting
.
Cancer Discov
2013
;
3
:
20
3
.
29.
Chetrit
A
,
Hirsh-Yechezkel
G
,
Ben-David
Y
,
Lubin
F
,
Friedman
E
,
Sadetzki
S
. 
Effect of BRCA1/2 mutations on long-term survival of patients with invasive ovarian cancer: the national Israeli study of ovarian cancer
.
J Clin Oncol
2008
;
26
:
20
5
.
30.
Gourley
C
,
Michie
CO
,
Roxburgh
P
,
Yap
TA
,
Harden
S
,
Paul
J
, et al
Increased incidence of visceral metastases in scottish patients with BRCA1/2-defective ovarian cancer: an extension of the ovarian BRCAness phenotype
.
J Clin Oncol
2010
;
28
:
2505
11
.
31.
Ledermann
JA
,
Harter
P
,
Gourley
C
,
Friedlander
M
,
Vergote
I
,
Rustin
Gordon JS
, et al
Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer (SOC) and a BRCA mutation (BRCAm)
.
J Clin Oncol
31
, 
2013
(
suppl; abstr 5505
).