AZD-9574

PARP Inhibitors in Epithelial Ovarian Cancer

Kristin N. Taylor and Ramez N. Eskander*

Recent Patents on Anti-Cancer Drug Discovery
Rebecca and John Moores Cancer Center, Department of Reproductive Medicine, Division of Gynecologic Oncology, University of California, San Diego, La Jolla, CA, USA

A R T I C L E H I S T O R Y
Abstract: Background: Ovarian cancer remains the most common lethal gynecologic malignancy. The therapeutic gains with the use of traditional cytotoxic chemotherapy in advanced stage disease remain limited, reflecting the need for novel therapies. Poly(ADP-ribose) polymerase (PARP) inhibitors have recently demonstrated a significant therapeutic effect in patients with recurrent, high grade serous ovar- ian cancer, both in the treatment of existing disease and in prolonging the disease-free interval.
Objective: The purpose of this article is to discuss PARP inhibitor use in patients with advanced stage ovarian cancer, and to extensively review the existing clinical literature and related patents.

Methods: A comprehensive PUBMED literature review was conducted to identify all published phase

Received: July 04, 2017
Revised: November 28, 2017
Accepted: December 01, 2017

DOI: 10.2174/1574892813666171204094822
2 and phase 3 clinical trials involving PARP inhibitors in advanced epithelial ovarian cancer. Further, several patents related to PARP inhibitor use, companion diagnostic tests, and the development of bio- markers to predict PARP inhibitor responsiveness are described.
Results: PARP inhibitors have demonstrated significant clinical activity in both BRCA deficient and wild-type patient cohorts, with all three FDA-approved PARP inhibitors demonstrating efficacy irre- spective of BRCA mutation status in patients with advanced epithelial ovarian cancer.
Conclusion: PARP inhibitors have emerged as an exciting new drug class in the treatment of epithelial ovarian cancer. Ongoing studies are aimed at improving our ability to identify ideal candidates for PARP inhibitor therapy, as well as to identify and target mechanisms of drug resistance, and novel combinatorial approaches.

Keywords: BRCA mutation, homologous recombination deficiency, PARP inhibitor, poly (ADP-ribose) polymerase, ovarian cancer, synthetic lethality.

INTRODUCTION
It is anticipated that there will be 22,440 new cases of ovarian cancer in the United States in 2017, with 14,080 deaths [1]. Treatment for patients with advanced stage dis- ease includes maximal effort cytoreductive surgery followed by combination with platinum-based chemotherapy, to which most patients initially respond. However, approximately 70% of patients will ultimately develop disease recurrence and will require additional systemic chemotherapy [2]. Within this cohort of patients, subsequent therapeutic inter- ventions traditionally result in progressively shorter disease- free intervals, with the development of platinum resistance portending a poor prognosis. Given the limited therapeutic gains achieved with traditional cytotoxic agents, the explora- tion of novel therapeutic paradigms is warranted in an effort to improve oncologic outcome and patient-related quality of life.

*Address correspondence to this author at the 3855 Health Sciences Drive #0987, La Jolla, CA 92093 USA; Tel: 001-858-822-6199;
Fax: 001-858-822-6319; E-mail: [email protected]
BRCA
The identification of BRCA1 and BRCA2 germline muta- tions as conferring an increased risk for breast and ovarian cancer revolutionized the approach to both the prevention and treatment of these cancers [3-6]. Patients with a germline BRCA1/2 mutation constitute up to 18% of high grade serous Ovarian Cancer (OC) patients and are more likely to have platinum-sensitive disease, and a longer disease specific sur- vival [7]. As these are tumor suppressor genes, a second so- matic mutation of the wild-type copy of the BRCA1/2 gene, with a Loss of Heterozygosity (LOH), can lead to the devel- opment of cancer in germline mutation carriers [8].
The BRCA1 and BRCA2 proteins play an important role in the repair of DNA Double-Strand Breaks (DSBs) via the high-fidelity Homologous Recombination (HR) pathway. In the absence of HR pathway activity, DNA repair relies on error-prone nonhomologous end-joining (NHEJ) which is an alternative for repairing DSBs [9]. Cancer cells lacking func- tional BRCA1 or 2 proteins are considered deficient in ho- mologous recombination (HRD; Homologous Recombina- tion Deficiency) and display chromosomal instability [10].

2212-3970/18 $100.00+.00 © 2018 Bentham Science Publishers

PARP INHIBITION AND SYNTHETIC LETHALITY
Poly(ADP-ribose) polymerase (PARP) 1 and 2 are en- zymes that play an important role in the repair of Single Strand Breaks (SSBs) in DNA, primarily through the Base Excision Repair (BER) pathway [11]. PARP binds DNA at the location of the excised base, and when activated, recruits other DNA repair proteins [12]. Pharmacologic PARP inhi- bition blocks the catalytic activity of PARP, preventing re- pair of SSBs and leading to the development of DSBs at the replication fork, which are toxic to the cell, particularly those with HRD [10].
Another effect of PARP inhibition is PARP trapping, whereby PARP1 and PARP2 can be trapped on damaged DNA and unable to recruit DNA repair proteins, blocking DNA repair and ultimately leading to cell death [13]. Al- though all PARP inhibitors oppose the catalytic activity of PARP, there are striking differences in their abilities to trap PARP, which are related to the size and structure of the molecule and are correlated with the potency of the specific PARP inhibitor when used as a single agent. This further accounts for the significant differences in dosing among PARP inhibitors, with the most potent PARP trapping agent talazoparib being dosed at 1mg daily, as compared to 300mg or greater daily with all other PARP inhibitors [14]. PARP trapping is also the likely mechanism by which PARP inhibi- tors exert their cytotoxic effects in HR-proficient cells [15].
BRCA mutation or PARP inhibition occurring independ- ently in a cell is not detrimental, but when combined in one cell, becomes lethal. This phenomenon, known as synthetic lethality, is the ability to cause cell death, which is “synthe- sized”, or created, when the two such events are combined. Given this concept of synthetic lethality, PARP inhibition has been evaluated in BRCA1/2-deficient tumor cells, with up to a 1000-fold increased sensitivity relative to BRCA wild-type tumor cells [10, 16]. This effect was also seen in a mouse xenograft model using BRCA2-deficient tumors cells which, when treated with PARP inhibitor for five days, dis- played significant regression relative to BRCA wild-type tumors [17]. These impressive findings catalyzed the inves- tigation of PARP inhibitors in human clinical trials, begin- ning with olaparib.
OLAPARIB
Olaparib is the most extensively studied PARP inhibitor, with an initial patent filing in breast and pancreatic cancers (US20160051517) [18]. On December 19, 2014, olaparib was the first PARP inhibitor to be FDA-approved for use in patients with germline BRCA-mutated high grade serous ovarian, fallopian tube, or primary peritoneal carcinoma fol- lowing three or more prior lines of therapy (Table 1) [19-24]. A second indication for use has more recently been granted by the FDA on August 17, 2017, for maintenance following a complete or partial response to platinum-based chemother- apy, irrespective of BRCA mutation status.
EARLY EFFICACY DATA
Early Phase I and II trials of olaparib monotherapy dem- onstrated promising efficacy signals in cohorts with recurrent OC, with a response rate ranging from 31-41% in those with
a germline BRCA1/2 mutation [25-29], and 24% in patients with no identifiable BRCA mutation [28], at a dose of 400mg twice daily (BID). Subsequently, two larger Phase II trials demonstrated a benefit in Progression-Free Survival (PFS) in the maintenance setting, and a significant tumor response in the treatment setting, the latter of which led to the FDA ap- proval of olaparib as the first PARP inhibitor for the treat- ment of recurrent ovarian cancer in patients harboring a BRCA germline mutation (Table 1).
The first, Study 19, was a randomized, double-blind, pla- cebo-controlled, Phase II study of the use of olaparib as maintenance therapy in the setting of recurrent, platinum- sensitive OC [20]. Patients were eligible regardless of BRCA mutation status, and must have received two or more prior lines of chemotherapy, with a Partial (PR) or Complete Re- sponse (CR) to the last platinum-based chemotherapy. Two hundred sixty-five patients were enrolled and randomized to olaparib 400mg bid or placebo. Those who received olaparib had a significantly prolonged progression-free survival (PFS) (8.4 months vs. 4.8 months, hazard ratio (HR) 0.35, 95% confidence interval (CI) 0.25-0.49, p < 0.001). This PFS ad- vantage persisted in all subgroup analyses. In a planned analysis of BRCA germline mutation carriers, the benefit was magnified, with a nearly 8-month improvement in median PFS (11.2 months vs. 4.3 months, HR 0.18, 95% CI 0.10- 0.31, p < 0.0001). Despite the above data, the FDA Oncol- ogy Drug Advisory Committee (ODAC) rejected the applica- tion for a maintenance olaparib label in this setting. An analysis of Overall Survival (OS) at 77% maturity did not demonstrate a significant difference between the olaparib and placebo groups (median OS 29.8 months vs. 27.8 months, HR 0.73, 95% CI 0.55-0.96, p = 0.025, which did not meet threshold for significance of p = 0.0095) [30]. However, with the exclusion of those in the placebo group who received olaparib after progression of disease, there was a significant OS benefit in the olaparib group among BRCA mutation carriers (HR 0.52, 95% CI 0.28-0.97), suggesting a
confounding effect [30].
A subsequent single-arm trial, Study 42, evaluated ola- parib as treatment for patients with recurrent, platinum- resistant OC and a germline BRCA1/2 mutation [19]. One hundred ninety-three patients were administered olaparib 400mg bid until disease progression or toxicity. The Objec- tive Response Rate (ORR) was 31.1% (95% CI 24.6-38.1%), with stable disease of at least 8 weeks in 40% of patients. Median PFS and OS were 7 months and 16.6 months, re- spectively. In a subgroup analysis of 137 patients with three or more prior lines of chemotherapy, olaparib was found to be similarly effective, with an ORR of 34%. After consider- ing the data from this heavily pre-treated cohort, the FDA granted accelerated approval for olaparib, specifically for treatment as fourth-line chemotherapy in germline BRCA mu- tation carriers. A companion diagnostic blood test, BRACA- nalysis CDx (Myriad) (US6895337) [31], was approved in parallel to identify patients carrying BRCA gene mutations.
PREDICTORS OF RESPONSE TO OLAPARIB
Although olaparib is thought to be most effective in BRCA-mutated tumors, the role of olaparib in those with sporadic OC has been explored. An early Phase II study was

Drug Indications for Use Trial Design Patient Population Regimen Outcome Grade 3/4 AEs Ref.

Olaparib Treatment in recurrent OC, with 3+ prior regimens (germline BRCA1/2
mutation) Study 42 (NCT01078662) Phase II, single-arm -Recurrent, platinum- resistant OC
-Germline
BRCA1/2
-1+ prior regi- mens Olaparib 400mg bid as treatment ORR:
-31.1% (overall)
-34% (3+ prior regimens) Anemia, abdominal pain, fatigue [19]
Maintenance in recurrent, platinum- sensitive OC (any BRCA mutation status) Study 19 (NCT00753545) Phase II, randomized -Recurrent, platinum- sensitive OC
-Any BRCA
mutation status
-2+ prior regi- mens Olaparib 400mg bid vs. placebo as maintenance PFS (months):
-Olaparib = 8.4
-Placebo = 4.8 Nausea, fa- tigue, vomit- ing, anemia [20]
SOLO-2 (NCT01874353) Phase III, RCT -Recurrent, platinum- sensitive OC
-PR or CR to recent platinum chemotherapy
-Germline
BRCA1/2
-2+ prior regi- mens Olaparib 300mg bid vs. placebo as maintenance PFS (months):
-Olaparib 19.1
-Placebo 5.5 Anemia, neutropenia, fatigue, nau- sea, vomiting [21]

Ruca- parib Treatment in recurrent OC, with 2+ prior regimens (germline or somatic BRCA1/2
mutation) Study 10 (Part 2A)
(NCT01482715) Phase I (dose- escalation), Phase II (expansion, single-arm) -Recurrent, platinum- sensitive OC - Germline BRCA1/2
-2-4 prior regi- mens Rucaparib 600mg bid as treatment ORR: 59.5% Fatigue, ane- mia, elevated AST/ALT [22]
ARIEL2 (Part 1) (NCT01891344) Phase II, single-arm -Recurrent, platinum- sensitive OC - Any BRCA mu- tation status
-1+ prior regi-
mens Rucaparib 600mg bid as treatment PFS (months):
-Germline or so- matic BRCA1/2 = 12.8
-LOH high = 5.7
-LOH low = 5.2 Anemia, elevated AST/ALT [23]

Niraparib Maintenance in platinum- sensitive OC, 2+ prior regi- mens (any BRCA muta- tion status) NOVA (NCT01847274) Phase III, randomized- controlled trial -Recurrent, platinum- sensitive OC
-Any BRCA1/2
status
-2+ prior regi- mens Niraparib 300mg daily vs. placebo as maintenance PFS (months):
-Germline BRCA1/2
= 21.0 niraparib vs.
5.5 placebo
-Non-germline
BRCA1/2 = 9.3
niraparib vs. 3.3 placebo
-HRD-positive sub- group = 12.9
niraparib vs. 3.8
placebo Thrombocy- topenia, ane- mia, neutro- penia [24]
Abbreviations:
ALT = Alanine Transaminase; AST = Aspartate Transaminase; AE = Adverse Event; CI = Confidence Interval; LOH = Loss of Heterozygosity; OC = Ovarian Cancer; ORR = Objective Response Rate; PFS = Progression-Free Survival

the first to demonstrate efficacy of olaparib in patients with sporadic OC [28]. However, the ORR was higher in those with a BRCA mutation relative to sporadic OC (ORR 41% vs. 24%, respectively). A planned subgroup analysis of Study 19 by BRCA mutation status demonstrated that, al-
though olaparib was beneficial for both groups, the effect was more pronounced in patients with known BRCA muta- tions (median PFS 11.2 months for olaparib vs. 4.3 months for placebo, HR 0.18, 95% CI 0.10-0.31, p < 0.0001) com- pared to those with sporadic OC (median PFS 7.4 months vs.

5.5 months, respectively; HR 0.54, 95% CI 0.34-0.85, p = 0.0075) [32]. This PFS advantage was similar in a sub- group analysis of the 10% of patients with a somatic rather than germline BRCA mutation (HR 0.23 vs. 0.17 in somatic vs. germline BRCA mutations, respectively) [33].
Sensitivity to platinum also appears to play a role in re- sponsiveness to olaparib in patients with high grade epithe- lial OC. In a Phase I trial of 50 germline BRCA mutation carriers, the ORR differed according to platinum sensitivity, at 61%, 42%, and 15% for platinum-sensitive, -resistant, and
-refractory patients, respectively [26]. A Phase II trial dem- onstrated a more marked variation of 50-60% in platinum- sensitive compared to 4% in platinum-resistant patients [28]. Further studies may better elucidate the role of platinum sen- sitivity as a predictor of response to PARP inhibition.
PHASE 3 MONOTHERAPY TRIALS
Ongoing prospective, Phase III, Randomized-Controlled Trials (RCTs) will seek to confirm the benefit of olaparib in germline BRCA1/2 mutation carriers with OC, with a pri- mary endpoint of PFS (Table 2) [34-36]. All three of the study of olaparib in ovarian cancer (SOLO) trials utilize a dose of 300mg bid in a tablet form (four tablets daily) in- stead of the previous dosage of 400mg bid in a capsule for- mulation (totaling 16 capsules daily), with data supporting equivalent efficacy but improved tolerability [34].
SOLO-1 (NCT01844986) is the first trial to investigate the use of olaparib in newly-diagnosed OC patients. Patients who experienced a partial or complete response to adjuvant platinum-based chemotherapy are randomized 2:1 to ola- parib or placebo maintenance, with a target enrollment of
344 patients. SOLO-2 (NCT01874353) was developed to assess the therapeutic efficacy of olaparib therapy in patients with BRCA deficient, platinum sensitive recurrent ovarian cancer following complete or partial response to most recent platinum based therapy [35]. Two hundred ninety-five pa- tients with recurrent, platinum-sensitive OC, at least two prior lines of platinum-based chemotherapy, and a partial or complete response to the most recent platinum-based chemo- therapy, were randomized 2:1 to olaparib or placebo mainte- nance. Patients in the olaparib group had a significantly pro- longed median PFS of 19.1 months compared to 5.5 months in the placebo group (HR 0.30, 95% CI 0.22-0.41, p < 0.0001). Secondary endpoints of time to first subsequent therapy, PFS2, and time to second subsequent therapy were also significantly longer in the olaparib group. Together with data from Study 19, these results led to the FDA approval for olaparib as maintenance therapy in patients with platinum- sensitive OC, regardless of BRCA mutation status. SOLO-3 (NCT02282020) is the only Phase III trial investigating ola- parib monotherapy as treatment for recurrent disease. Pa- tients with recurrent, platinum-sensitive OC with two or more prior lines of platinum-based chemotherapy are ran- domized 2:1 to receive olaparib or physician’s choice stan- dard-of-care chemotherapy, with a target enrollment of 411 patients.
The potential benefit of re-exposure to PARP inhibition is being explored in a Phase IIIb, RCT of patients with recur- rent, platinum-sensitive OC and any BRCA mutation status who had previously received PARP inhibitor therapy
(OReO, NCT03106987). Patients are randomized 2:1 to ola- parib or placebo maintenance, with a target enrollment of 416.
ADVERSE EVENTS
Consideration of toxicity associated with olaparib ther- apy is critical, particularly in patients who have had multiple prior lines of chemotherapy and are unlikely to be cured. Mild Adverse Events (AEs) have been relatively common across studies, with some rare but serious AEs. In Study 19, nearly all patients experienced at least one AE, most of which were grade 1/2, with nausea (68% vs. 35%), fatigue (49% vs. 38%), vomiting (32% vs. 14%), and anemia (17% vs. 5%) being more frequent in the olaparib group. Grade 3/4 AEs occurred in 35.3% of the olaparib group and 20.3% of the placebo group, with more dose interruptions (27.9% vs. 8.6%, respectively) and reductions (22.8% vs. 4.7%, respec- tively) in the olaparib group [20]. Nausea, fatigue, vomiting, and anemia were the most common AEs in Study 42 as well, occurring in 62%, 60%, 39%, and 32% of patients, respec- tively. Grade 3/4 AEs were more frequent than in Study 19, occurring in 54% of patients. The most common were ane- mia (17%), abdominal pain (7%), and fatigue (6%), with 40% of patients experiencing a dose interruption or reduc- tion. Two patients developed acute myeloid leukemia (AML), and one developed myelodysplastic syndrome (MDS) [19]. In SOLO-2, a similar profile of mostly grade 1/2 AEs was noted and generally improved throughout treatment [37]. Anemia was the most common grade 3/4 toxicity (19% olaparib vs. 2% placebo), with others occur- ring in 10% or fewer patients in either treatment arm. Dose interruptions occurred in 4% of the olaparib group compared to 18% of the placebo group, and dose reductions in 25% and 3%, respectively. MDS and AML were rare complica- tions, occurring in four patients in each group.
HEALTH-RELATED QUALITY OF LIFE (HRQOL)
The impact of olaparib on health-related quality of life (HRQoL) was evaluated in both Study 19 and SOLO-2, with no detrimental effects identified with olaparib monotherapy. The FACT-O, FACT/NCCN Ovarian Symptom Index (FOSI), and the Trial Outcome Index (TOI) were adminis- tered at baseline and every 28 days during therapy in Study
19. Most patients (81%) reported “no change” in their HRQoL from baseline to the completion of therapy, and there were no significant differences between treatment groups at either timepoint [38]. In SOLO-2, the FACT-O TOI was used to assess HRQoL, and time without symptoms or toxicity (TWiST) was also compared between the olaparib and placebo groups. Similarly, HRQoL did not significantly decline from baseline to 12 months in either group, and TWiST was longer in the olaparib group compared to pla- cebo (13.5 months vs. 7.2 months, difference 6.3 months, 95% CI 2.9-8.6, p < 0.001) [39].
EARLY COMBINATION TRIALS
In addition to its use as monotherapy, olaparib has been studied in combination with cytotoxic chemotherapy, anti- angiogenic agents, and immunotherapy. Two Phase I/II trials have evaluated olaparib in combination with platinum-based

Table 2. Ongoing Phase III Trials of PARP Inhibitors in Ovarian Cancer.

Drug Trial Design Patient Population Regimen Primary Outcome Preliminary Data (If Any) Ref. (If Any)
SOLO1 (NCT01844986) Phase III, RCT -Newly-diagnosed OC
-PR or CR to platinum chemotherapy Olaparib 300mg bid vs. placebo as maintenance PFS
-Germline BRCA1/2
SOLO2 (NCT01874353) Phase III, RCT -Recurrent, platinum- sensitive OC
-PR or CR to recent platinum chemotherapy Olaparib 300mg bid vs. placebo as maintenance PFS PFS (months):
-Olaparib 19.1
-Placebo 5.5 [21]
-Germline BRCA1/2
Olaparib (mono- therapy) -2+ prior regimens
SOLO3 (NCT02282020) Phase III, RCT -Recurrent, platinum- sensitive OC
-Germline BRCA1/2 Olaparib 300mg bid vs. physician choice chemo- therapy as treatment PFS
-2+ prior regimens
OReO (NCT03106987) Phase IIIb, RCT -Recurrent, platinum- sensitive OC Olaparib 300mg bid vs. placebo as maintenance PFS
-Previously treated with PARP inhibitor
-Any BRCA mutation status
PAOLA-1 (NCT02477644) Phase III, RCT -Newly-diagnosed OC
-PR or CR to platinum chemotherapy with bevacizumab Olaparib 300mg bid vs. placebo as maintenance (in addition to bevaci- zumab) PFS
-Planned bevacizumab maintenance
-Any BRCA mutation status

Olaparib (com- bination therapy) COCOS (NCT02502266) Phase II/III, RCT -Recurrent, platinum- resistant OC
-Germline BRCA1/2 Four arms (as treatment):
-Olaparib alone
-Cediranib alone OS
-1-3 prior regimens -Olaparib + cediranib
-Physician choice che- motherapy
NCT02446600 Phase III, RCT -Recurrent, platinum- sensitive OC Three arms (as treat- ment): PFS
-Germline BRCA1/2 -Olaparib alone
-Any BRCA mutation status -Olaparib + cediranib
-Physician choice che- motherapy

Rucaparib ARIEL3 (NCT01968213) Phase III, RCT -Recurrent, platinum- sensitive OC
-Any BRCA mutation status
-2+ prior regimens Rucaparib 600mg bid vs. placebo as maintenance PFS PFS (months):
-BRCA mutant (germline or so- matic) with ruca- parib 16.6
-HRD-positive with rucaparib 13.6 [36]
-Intent-to-treat with rucaparib 10.8
-Intent-to-treat with placebo 5.4
Table (2) contd….

Drug Trial Design Patient Population Regimen Primary Outcome Preliminary Data (If Any) Ref. (If Any)
ARIEL4 (NCT02855944) Phase III, RCT -Recurrent OC
-Germline or somatic
BRCA1/2
-2+ prior regimens Rucaparib 600mg bid vs. chemotherapy PFS

Niraparib PRIMA (NCT02655016) Phase III, RCT -Newly-diagnosed OC
-PR or CR to platinum chemotherapy
-HRD-positive Niraparib 300mg daily vs. placebo as main- tenence PFS

Veliparib GOG 3005 (NCT02470585) Phase III, RCT -Newly-diagnosed OC
-Any BRCA mutation status Three arms:
-Chemotherapy (car- boplatin/paclitaxel) and placebo maintenance
-Chemotherapy with veliparib and placebo maintenance
-Chemotherapy with veliparib and veliparib maintenance PFS

Abbreviations:
CR = Complete Response; HRD = Homologous Recombination Deficiency; OC = Ovarian Cancer; OS = Overall Survival; PR = Partial Response; PFS = Progression-Free Survival; RCT = Randomized Controlled Trial.

chemotherapy in recurrent OC. In one small Phase Ib/II trial, patients were administered escalating doses of olaparib bid (three out of the four weeks) together with weekly car- boplatin (AUC 2) and paclitaxel (60mg/m2), with a Maxi- mum Tolerated Dose (MTD) of 150mg bid. Of 12 evaluable patients, there were four with CR and two with PR; the most common grade 3 toxicities were neutropenia, lymphopenia, anemia, fatigue, and one case of myelodysplastic syndrome [40]. In a Phase I/Ib trial of 45 BRCA1/2 mutation carriers, including 37 with OC, olaparib 400mg bid was administered days 1-7 with carboplatin (AUC 5, day 1) every 21 days, followed by the same dose of olaparib continued as mainte- nance. The objective response rate for the ovarian cancer cohort was 41%, with overall grade 3/4 AEs including neu- tropenia (42%), thrombocytopenia (20%), and anemia (16%) [41]. The addition of olaparib to platinum-based chemother- apy was shown to be of benefit in a randomized Phase II trial of 162 patients with recurrent, platinum-sensitive OC (irre- spective of BRCA mutation status) with up to three prior platinum-based regimens [42]. Patients were randomized 1:1 to one of two regimens: Olaparib 200mg bid (days 1-10) with carboplatin AUC 4 (day 1) and paclitaxel 175mg/m2 (day 1) in a 21-day cycle, followed by olaparib 400mg bid continued as maintenance or standard chemotherapy alone (carboplatin AUC 6 and paclitaxel 175mg/m2 day 1, every 21 days). The primary endpoint, PFS, was significantly pro- longed in the olaparib plus chemotherapy group relative to chemotherapy alone (median PFS 12.2 months vs. 9.6 months, respectively; HR 0.51, 95% CI 0.34-0.77, p = 0.0015), with a more pronounced benefit in those with a BRCA mutation (median PFS not reached vs. 9.7 months, HR 0.21, 95% CI 0.05-0.55). There was no OS benefit with combination therapy relative to chemotherapy alone (median OS 33.8 vs. 37.6 months, HR 1.17, 95% CI 0.79-1.73). Ad-
verse events that were at least 10% higher in the olaparib group included alopecia (74%), nausea (69%), neutropenia (49%), diarrhea (42%), headache (33%), peripheral neuropa- thy (31%), and dyspepsia (26%), and were largely grade 1/2 (with the exception of grade 3 neutropenia). Discontinuation rates due to adverse events were similar between the groups. An ongoing Phase III clinical trial (PAOLA-1, NCT02477644) is investigating the use of olaparib as main- tenance therapy in newly-diagnosed OC. Following a com- plete or partial response to adjuvant carboplatin, paclitaxel, and bevacizumab, patients are randomized to olaparib 300mg bid vs. placebo in combination with bevacizumab maintenance, with a primary endpoint of PFS and anticipated enrollment of 612.
Numerous studies have evaluated olaparib in conjunction with anti-angiogenic agents, specifically drugs that inhibit the action of Vascular Endothelial Growth Factor (VEGF). Olaparib was found to be safe in combination with bevaci- zumab, a VEGF antagonist, in a small Phase I trial, with no serious AEs or Dose-Limiting Toxicities (DLTs) noted [43]. Cediranib, a VEGF receptor antagonist, has also been found to be tolerable, with data also suggesting a benefit of combi- nation therapy. After establishing an MTD of olaparib 200mg bid in combination with cediranib 30mg daily in a Phase I study [44], the same regimen was compared to ola- parib 200mg bid alone in a randomized Phase II trial of 90 patients with recurrent, platinum-sensitive OC and any BRCA mutation status. There was a significant improvement in PFS among patients in the combination group relative to olaparib monotherapy (median PFS 17.7 months vs. 9.0 months, HR 0.42, 95% CI 0.23-0.76, p = 0.005). Grade 3/4
AEs including hypertension, fatigue, and diarrhea were more
common in the combination group, though each occurred in fewer than 20% of patients [45].

OTHER ONGOING COMBINATION TRIALS
Four Phase II/III trials are underway to try and identify patients who are likely to benefit from the combinatorial approach of olaparib with cediranib (Table 2). Two Phase II trials are investigating the efficacy of olaparib with cediranib in platinum-resistant patients, specifically by BRCA mutation status. OCTOVA (NCT03117933) randomizes germline BRCA mutation carriers to receive olaparib alone, olaparib with cediranib, or weekly paclitaxel. CONCERTO (NCT02889900) is a single-arm trial of only BRCA negative patients following at least three prior lines of chemotherapy, with administration of olaparib/cediranib combination ther- apy. With anticipated enrollment of 680 patients, COCOS is a Phase II/III randomized trial of platinum-resistant patients with four treatment arms: olaparib monotherapy, cediranib monotherapy, olaparib plus cediranib, or standard chemo- therapy alone (NCT02502266). One Phase III study includes only patients with recurrent, platinum-sensitive OC who will receive olaparib monotherapy, olaparib with cediranib, or standard chemotherapy alone, with a primary endpoint of PFS and target enrollment of 450 subjects (NCT02446600).
Additional trials are evaluating a novel combinatorial ap- proach with immune checkpoint inhibitors. The results of a Phase I dose-escalation study of durvalumab (a PD-L1 in- hibitor) with olaparib or cediranib have recently been pub- lished [46]. Durvalumab 10mg/kg every 2 weeks or 1500mg every 4 weeks was administered to twenty-six women as a doublet with escalating doses of either olaparib or cediranib.
There were no dose-limiting toxicities in the 12 women re- ceiving durvalumab and olaparib, and there was an 83% dis- ease control rate, including two partial responses and eight patients exhibiting stable disease. A Phase I/II trial is under- way evaluating tremelimumab (anti-CTLA-4) alone or in combination with olaparib in patients with persistent OC following one line of chemotherapy (NCT02485990). An- other Phase I/II trial, specific to BRCA mutations carriers with recurrent disease, is also investigating the combination of tremelimumab and olaparib, with the addition of durvalu- mab (NCT02953457).
RUCAPARIB
Rucaparib is a PARP inhibitor that was granted FDA ap- proval on December 19, 2016, for the treatment of patients with recurrent, BRCA-mutated (germline or somatic) ovarian cancer who have received two or more prior lines of chemo- therapy (US20060009517) [47] (Table 3) [48-56]. Results from two multicenter trials led to approval of rucaparib in this setting (Table 1).
PRIOR PHASE II TRIALS
Study 10 was a Phase I/II, single-arm trial that estab- lished tolerability and efficacy of rucaparib in the treatment of recurrent OC [22]. Part 1 (Phase I) included 56 patients with advanced solid tumors and established an optimal dose of 600mg bid, with manageable toxicities. In Part 2A (Phase II), 42 germline BRCA1/2 mutation carriers with recurrent,

Table 3. Recent Patents on Three FDA-Approved PARP Inhibitors.

Target Patent No. Patent title Assignee Pub. Year Ref.

Olaparib US20070179160 Use of RNAI inhibiting PARP activity for the manufacture of a medicament for the treatment of cancer University of Sheffield 2014 [48]
US20120010204 Phthalazinone derivatives Kudos Pharmaceuticals Ltd. 2014 [49]
US20060142231 DNA damage repair inhibitors for treat- ment of cancer Institute of Cancer Research, Kudos Pharmaceuticals Ltd. 2006 [50]
US20050059663 Phthalainone derivatives Kudos Pharmaceuticals Ltd, May- bridge Ltd. 2008 [51]
US20090270617 Phthalainone derivative Kudos Pharmaceuticals Ltd. 2012 [52]
US20020183325 Phthalainone derivative Kudos Pharmaceuticals Ltd. 2006 [53]
US20080200469 Phthalainone derivatives Kudos Pharmaceuticals Ltd, May- bridge Ltd. 2011 [54]

Rucaparib US20060009517 Tricyclic inhibitors of poly(ADP-ribose) polyerases Agouron Pharmaceuticals, Inc., Cancer Research Campaign Tech- nology Ltd. 2006 [47]
WO2016028689 High dose strength tablets of rucaparib Clovis Oncology, Inc. 2016 [55]

Niraparib US8071623 Amide substituted indazoles as poly(ADP-ribose) polymerase(PARP) inhibitors Instituto De Richerche Di Biologia Molecolare P. Angeletti Spa 2011 [56]

platinum-sensitive OC (with two to four prior lines of che- motherapy) were administered rucaparib 600mg bid until disease progression or toxicity. The ORR was 59.5%, with all patients experiencing an AE; the most common were fa- tigue (86%), nausea (83%), anemia (71%), elevations in as- partate and alanine transaminases (AST/ALT) (57%), or vomiting (55%). Fatigue, anemia, and elevated AST/ALT were the most frequent grade 3/4 AEs (26%, 31%, and 7%, respectively), with dose interruptions in 64% and dose reduc- tions in 69% of patients.
ARIEL2 is a Phase II, single-arm, two-part study de- signed to assess the efficacy of rucaparib based on tumor Homologous Recombination Deficiency (HRD) signature. In Part 1, 192 patients with recurrent, platinum-sensitive OC with one or more prior lines of chemotherapy were classified into one of three groups based on tumor next-generation se- quencing: BRCA mutant (germline or somatic), Loss of Het- erozygosity (LOH) high (BRCA wild-type), or LOH low (BRCA wild-type). This HRD assay was developed by Foun- dation Medicine, together with Clovis (WO2015108986) [57]. Patients were administered rucaparib 600mg bid until progression or toxicity. Median PFS was 12.8 months in the BRCA mutant group, 5.7 months in the LOH high group, and
5.2 months in the LOH low group. This difference was sig- nificant in the BRCA mutant group (HR 0.27, 95% CI 0.16- 0.44, p < 0.0001) compared to the LOH low group; a similar, though non-significant trend was seen in the LOH high group (HR 0.62,95% CI 0.42-0.90, p = 0.011) relative to the LOH low group. There was also a significant benefit in the median duration of response in the BRCA mutant group (9.2 months) and LOH high group (10.8 months) relative to the LOH low group (5.6 months). The overall AE profile was similar to that of Study 10, but with slightly less frequent grade 3/4 AEs, with anemia (21%) and elevated AST/ALT (12%) being most common [23]. An analysis of tumor biop- sies demonstrated an association between methylation of BRCA1 or RAD51C and high LOH, with promising ORR (52.4% and 75%, respectively) and PFS (7.4 months and
11.1 months, respectively). These data suggest that methyla- tion of BRCA1 or RAD51C could potentially be used as pre- dictors of sensitivity to rucaparib, pending further study [58]. ARIEL2 Part 2 specifically includes heavily pre-treated pa- tients who have received at least three prior lines of chemo- therapy, with eligibility extended to platinum-resistant and - refractory patients. Although this trial is still ongoing, pre- liminary data incorporating Parts 1 and 2 demonstrate a dif- ference in PFS by platinum sensitivity among BRCA muta- tion carriers (median PFS 12.7 months in platinum-sensitive,
7.3 months in platinum-resistant, and 5.0 months in plati- num-refractory) [59].
ONGOING TRIALS
ARIEL3 is a Phase III, double-blind RCT that was de- signed to prospectively validate the findings of ARIEL2 by evaluating patient response to treatment according to the following molecular signatures: BRCA mutant (germline or somatic), HRD-positive (including BRCA mutant and BRCA wild-type with LOH high), and intent-to-treat (all enrolled
patients) (Table 2). Five hundred sixty-four patients with platinum-sensitive, recurrent OC, and at least two prior lines of chemotherapy, were enrolled and randomized to rucaparib or placebo maintenance immediately following a complete or partial response to platinum-based chemotherapy. This trial demonstrated a significant PFS benefit of rucaparib in the BRCA mutant group (median PFS 16.6 months, HR 0.23, p < 0.0001), HRD-positive group (median PFS 13.6 months, HR 0.32, p < 0.0001), and intent-to-treat group (median PFS
10.8 months, HR 0.36, p < 0.0001) relative to placebo (me- dian PFS 5.4 months) [36]. An exploratory analysis in BRCA wild-type only demonstrated a maintained benefit of ruca- parib in both the HRD-positive (median PFS 9.7 months, HR 0.44, p < 0.0001) and HRD-negative groups (median PFS
6.7 months, HR 0.58, p = 0.0049). These encouraging results suggest that the indication for the use of rucaparib in the treatment of recurrent ovarian cancer may soon be broad- ened, with a supplemental new drug application recently submitted to the FDA for use as a maintenance therapy.
ARIEL4 (NCT02855944) is a Phase III study of rucaparib compared to chemotherapy in BRCA-mutated, recurrent OC following two or more prior lines of therapy, with a primary endpoint of PFS and target enrollment of 345 patients. Ruca- parib is also being explored in combination with atezolizumab (a PD-L1 inhibitor) in a Phase Ib trial of patients with recur- rent, platinum-sensitive OC (NCT03101280).
NIRAPARIB
Niraparib was the most recently approved PARP inhibi- tor, receiving accelerated approval by the FDA on March 27, 2017, for use in patients with recurrent, platinum-sensitive OC (at least two prior chemotherapy regimens) (Table 3). A Phase I dose-escalation trial established the recommended Phase II dose of 300mg daily, and demonstrated antitumor activity in advanced solid tumors [60]. A Phase III, double- blind, randomized controlled trial was then undertaken (NOVA), the results of which were the basis for the FDA approval of niraparib [24]. Five hundred fifty-three patients with recurrent, platinum-sensitive OC (with two or more prior platinum-based regimens) were randomized 2:1 to re- ceive niraparib or placebo as maintenance following a com- plete or partial response to the last platinum chemotherapy. A recent patent held by Myriad Genetics describes methods of assessing tumors for HR deficiency, which were used to group patients for the evaluation of response to niraparib (WO2016094391) [61]. Among germline BRCA mutation carriers, there was a significant prolongation in PFS in the niraparib group relative to placebo (median PFS 21.0 months vs. 5.5 months, HR 0.27, 95% CI 0.017-0.41, p < 0.0001).
There was a benefit among those without a germline BRCA mutation (median PFS 9.3 vs. 3.3 months, HR 0.45, 95% CI 0.34-0.61, p < 0.001) and in a prespecified analysis of the HRD-positive subgroup of the non-germline BRCA mutation group (median PFS 12.9 months vs. 3.8 months, HR 0.38, 95% CI 0.24-0.59, p < 0.0001). Patients with HRD-negative tumors were also found to have a prolonged PFS with niraparib relative to placebo (median PFS 6.9 months vs. 3.8 months, HR 0.58, 95% CI 0.36-0.92, p = 0.02). All patients

in the niraparib group experienced at least one AE, with a similar profile to other PARP inhibitors. Grade 3/4 AEs were primarily hematologic, including thrombocytopenia (34%), anemia (25%), and neutropenia (20%).
ONGOING TRIALS
Two additional trials are studying the potential role of niraparib in alternate clinical settings. QUADRA (NCT02354586) is a Phase II, single-arm trial investigating the efficacy of niraparib as treatment for patients with recur- rent, platinum-sensitive OC and BRCA-mutated or HRD- positive tumors. PRIMA (NCT02655016) is a Phase III RCT evaluating the use of niraparib as maintenance following a complete or partial response to frontline platinum-based chemotherapy in advanced stage, HRD-positive OC (Table 2). Niraparib is also being studied as combination therapy in two Phase I/II trials. ANANOVA (NCT02354131) is enroll- ing patients with recurrent, platinum-sensitive OC, with ran- domization to niraparib alone or in combination with bevaci- zumab. TOPACIO (NCT02657889) will administer escalat- ing doses of niraparib with pembrolizumab (a PD-1 inhibi- tor) in a heavily pretreated, platinum-resistant cohort.
VELIPARIB
Veliparib is a potent inhibitor of PARP1/2 that is not yet FDA-approved for use in OC, but has shown preliminary evidence of efficacy. In GOG 280, a Phase II, single-arm trial, veliparib 400mg bid was administered to 50 BRCA mu- tation carriers with recurrent OC and up to three prior lines of chemotherapy [62]. The ORR was 26%, with 35% of platinum-sensitive and 20% of platinum-resistant patients responding to therapy. Gastrointestinal AEs were similar to olaparib, but grade 3/4 hematologic AEs were infrequent (0- 2%). Other trials have investigated veliparib as combination therapy. A Phase I trial of veliparib plus whole abdominal radiation in patients with peritoneal carcinomatosis (includ- ing 18 OC patients) demonstrated tolerability but only a 3% ORR [63]. Veliparib combined with cyclophosphamide was evaluated in a randomized Phase II trial of patients with BRCA-mutated advanced solid tumors [64]. The addition of veliparib compared to cyclophosphamide alone did not im- prove the PR rate (11.8% vs. 19.4%, respectively) or median PFS (2.1 months vs. 2.3 months, respectively; p = 0.68). GOG 9923 was a Phase I trial that evaluated veliparib with frontline platinum-based chemotherapy (IV carboplatin and paclitaxel, or IV/IP cisplatin and paclitaxel), as well as bevacizumab [65]. Currently, in a Phase III, three-arm RCT of newly-diagnosed OC patients, carboplatin and paclitaxel are administered with or without veliparib therapy, followed by veliparib or placebo maintenance (GOG 3005, NCT02470585) (Table 2).

TALAZOPARIB
Talazoparib is a PARP inhibitor with antitumor activity at significantly lower concentrations than other drugs in this class [66]. A Phase I trial that included 23 patients with OC, of whom 17 had germline BRCA mutations, established a maximum tolerable dose of 1mg daily. In an expansion co-
hort with 12 BRCA-mutated OC patients, there was an ORR of 42% [67]. Toxicities were somewhat milder than other PARP inhibitors, with grade 1/2 nausea and fatigue each occurring in less than 40% of patients, and grade 3/4 AEs limited to anemia (23%), thrombocytopenia (18%), and neu- tropenia (10%). Talazoparib was investigated together with either temozolomide or irinotecan in a Phase I dose- escalation trial, which demonstrated partial responses in 4/7 (57%) patients with platinum-resistant OC [68]. Interest- ingly, response was correlated with an HRD score of 42 or greater as assessed by next-generation sequencing of tumors. Grade 3/4 AEs were mostly hematologic, and this combina- tion was generally well-tolerated. Phase II trials were initi- ated to assess talazoparib monotherapy or in combination with temozolomide in OC patients with prior PARP inhibi- tion treatment, but are no longer open.
CONCLUSION
In an era of molecular therapy, PARP inhibitors have emerged as an exciting new option for patients with ad- vanced stage, recurrent ovarian cancer. Ongoing clinical tri- als and the completion of novel translational objectives di- rected at predicting response will continue to inform patient selection. Although both drug resistance and high cost of therapy remain targets for improvement, PARP inhibitors, both as single agents and in combination with other drugs, represent a new treatment paradigm.
CURRENT & FUTURE DEVELOPMENTS
Predicting PARP Responsiveness
With a multitude of ongoing studies in different patient populations and clinical settings, it is likely that the indica- tions for PARP inhibitors in OC will continue to expand. Yet the question of accurately predicting who will benefit most from this therapy remains unanswered. Germline and so- matic BRCA1/2 mutations have thus far been used to predict, to some extent, who may respond to PARP inhibition; how- ever, more sensitive and specific biomarkers are needed. HRD assays are currently being developed as a means of assessing genomic instability characteristic of the “BRCAness” phenotype, by determining the extent of LOH, Telomeric Allelic Imbalance (TAI), and large-scale state transitions in DNA [69-71]. Myriad Genetics has developed the MyChoice HR deficiency assays, which incorporates all three of these HRD scores [72] (WO2016094391) [61] (Ta- ble 4) [77-84]. Foundation Medicine, together with Clovis, has utilized a LOH threshold of 14% to define “LOH-high” patients within the BRCA wild-type subgroup in ARIEL2 (WO2015108986) [57]. A high correlation with response to rucaparib was noted, and a planned post hoc analysis has already “refined” this cutoff to be more predictive of im- proved PFS with rucaparib [73].
Other recent patents reflect an ongoing effort to identify biomarkers to predict either response to or resistance to PARP inhibitors. Richardson and colleagues developed a Global Chromosomal Aberration score (GCAS) which incorporates Copy Number Alterations (CNA) in addition to detecting

Table 4. Recent Patents on PARP Inhibitor Use as Combination Therapy and the Development of Novel Biomarker Assays.

Target Patent No. Patent title Assignee Pub. Year Ref.

PARP inhibitors as combination ther- apy WO2017031445 Combination therapy for cancer treatment Merrimack Pharmaceuticals, Inc. 2017 [77]
WO2012016876 Therapeutic combination comprising a PARP-1 inhibitor and an anti-neoplastic agent Nerviano Medical Sciences S.R.L. 2013 [78]
WO2017077326 Combination of an inhibitor of PARP with an inhibitor of GSK-3 or DOT1L King’s College London 2017 [79]

Development of biomarker assays WO2016018089 Novel biomarker for predicting sensitiv- ity to PARP inhibitor, and use thereof The Asan Foundation 2016 [80]
US20130224312 Methods and materials for assessing responsiveness to PARP inhibitors and platinating agents Mayo Foundation for Medical Education and Research 2013 [81]
WO2014138101 Gene signature to predict homologous recombination (HR) deficient cancer Board of Regents, The University of Texas System 2014 [82]
WO2013182645 Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors INSERM 2015 [83]
WO2014205105 Biomarkers of response to inhibition of poly-ADP ribose polymerase (PARP) in cancer The Regents of the University of California 2016 [84]

LOH, TAI, and LST (US20170037478) [74]. Another patent describes the detection of a Single Nucleotide Polymorphism (SNP) in the PARP1 gene that may confer resistance or sensi- tization to PARP inhibition (WO2016073298) [75]. Mach describes a novel imaging method whereby radiolabeled PARP1 inhibitors are combined with Positron Emission To- mographic (PET) imaging to detect tumoral PARP activity and the efficacy of PARP inhibition, as a guide for drug dos- ing (WO2015103526) [76].
Resistance to PARP Inhibition
Another challenge to overcome is acquired PARP resis- tance after exposure to drug. Numerous mechanisms have been identified for inherent or acquired resistance. One is the up regulation of genes encoding p-glycoprotein efflux pumps, which over time decrease intracellular drug levels [85]. In a murine breast cancer model, a PARP inhibitor (AZD2281) was administered with initial tumor shrinkage that then plateaued, correlating with a significant increase in the expression of p-glycoprotein efflux pumps. Co- administration of p-glycoprotein inhibitor, tariquidar, re- versed this resistance.
Restoration of HR can also be responsible for the de- velopment of resistance to PARP inhibitors, through a secondary mutation in a protein critical to the HR pathway. In tumors harboring a BRCA1/2 mutation, a new somatic mutation or deletion can restore the open reading frame of the gene, allowing for translation of a functional BRCA protein [86]. Similarly, in ARIEL2 Part 1, secondary somatic mutations in RAD51C and RAD51D- both integral to HR- were associated with resistance to PARP inhibition [87]. The genetic profile of pre-treatment tumor samples
genetic profile of pre-treatment tumor samples was com- pared to that of post-treatment biopsies in 12 patients who progressed on therapy. Five of these patients had a delete- rious mutation in RAD51C or RAD51D on the initial bi- opsy, with post-progression biopsy revealing a secondary mutation that restored the open reading frame of the corre- sponding gene. Loss of function of 53bp1, a protein in- volved in NHEJ, may restore HR pathway function and has been correlated with PARP inhibitor resistance in BRCA1- mutated tumors [86].
Expression of Schlafen 11 (SLFN11) has recently been identified as a marker of PARP inhibitor resistance in a manner distinct from the previously discussed mechanisms. In two separate analyses of the publically-available Genom- ics of Drug Sensitivity in Cancer (GDSC) dataset and the National Cancer Institute (NCI)-60 genomic database, SLFN11 was highly correlated with sensitivity to olaparib, rucaparib, and veliparib, as well as talazoparib [88, 89]. Mu- rai et al conducted a series of experiments suggesting that, instead of altering drug penetration or restoring HR activity, SLFN11 induces prolonged S-phase arrest and induces apop- tosis in cells treated with talazoparib, thus conferring sensi- tivity [89]. In vitro assays using genetic knockout or knock- down of SLFN11 demonstrate a significant increase in resis- tance to PARP inhibitors in small cell lung cancer [88, 90], as well as in cell lines derived from prostate cancer, leuke- mia, and Ewing’s sarcoma that exhibit high baseline expres- sion of SLFN11 [89]. These data are supported by patient- derived xenograft models, demonstrating that high tumor expression of SLFN11 (assessed by immunohistochemical staining) was correlated with significant tumor regression in numerous cancer types [88-90]. Interestingly, Lok et al. de-

tected a bimodal distribution in SLFN11 expression in treat- ment-naïve patient tumor samples, suggesting the potential exploration of SLFN11 as a predictive biomarker for PARP inhibitor sensitivity [88].
Currently, methods are being developed in an attempt to reverse resistance to PARP inhibitors. Regarding tumors affected by low SLFN11 expression, Murai et al. demon- strated that inhibition of ATR, a mediator of the DNA dam- age response upon which SLFN11-deficient cells rely for survival, can reverse resistance to PARP inhibitors [89]. In vitro studies have demonstrated that histone deacetylase in- hibitors enhance sensitivity of breast cancer cells to PARP inhibition [91]. An inhibitor of heat shock protein 90 (HSP90), which prevents ubiquitin-proteosome degradation of many oncogenes, has shown synergy with PARP inhibitor administration in a preclinical model of PARP inhibitor- resistant ovarian cancer [92]. Other pharmacologic methods for reversing PARP inhibitor resistance are being investi- gated, targeting proteins such as cyclin-dependent kinase 1 and BARD1 that interact closely with BRCA proteins [93].
Cost Effectiveness
for cost reduction, while maintaining patient access to this exciting class of drugs.
CONSENT FOR PUBLICATION
Not applicable.
CONFLICT OF INTEREST
Ramez N. Eskander has received speaker compensation from AZ Oncology, Genentech, and Clovis Oncology.
Kristin N. Taylor has no conflict of interest related to the content of this article.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin 2017; 67(1): 7-30.
McMeekin DS, Tillmanns T, Chaudry T, Gold M, Johnson G, Walker J, et al. Timing isn’t everything: An analysis of when to start salvage chemotherapy in ovarian cancer. Gynecol Oncol 2004;

Although there are compelling data demonstrating the ef- ficacy of PARP inhibitors in various disease settings, the cost of these novel agents is important to consider. An early analysis evaluated the use of olaparib as maintenance in re-

95(1): 157-64.
Friedman LS, Ostermeyer EA, Szabo CI, Dowd P, Lynch ED, Rowell SE, et al. Confirmation of BRCA1 by analysis of germline mutations linked to breast and ovarian cancer in ten families. Nat Genet 1994; 8(4): 399-404.

current, platinum-sensitive OC [94]. Three scenarios were considered following completion of platinum-based chemo- therapy: Olaparib therapy until progression (global olaparib); BRCA1/2 mutation testing, followed by olaparib until pro-
gression for mutation carriers; or observation. Assuming a
Miki Y, Swensen J, Shattuck-Eidens D, Futreal PA, Harshman K,
Tavtigian S, et al. A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 1994; 266(5182): 66-
71.
Wooster R, Bignell G, Lancaster J, Swift S, Seal S, Mangion J,
et al. Identification of the breast cancer susceptibility gene BRCA2.

14% mutation rate, and a superior PFS in BRCA mutation carriers, global olaparib was found to be the least cost-

Nature 199; 378(6559): 789-92.
Ramus SJ, Gayther SA. The contribution of BRCA1 and BRCA2 to ovarian cancer. Mol Oncol 2009; 3(2): 138-50.

effective strategy, with an Incremental Cost-Effectiveness
Ratio (ICER) of $234,128 per progression-free year-of-life
saved (PF-YLS) compared to $193,442 in the BRCA testing strategy group. In order to achieve an ICER of $100,000 per
Alsop K, Fereday S, Meldrum C, deFazio A, Emmanuel C, George J, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: A report from the Australian Ovarian Cancer Study Group. J Clin

PF-YLS (the cost-effectiveness threshold), the price of ola- parib would need to be decreased by more than 50%, to
$3000 per month. A similar analysis compared olaparib

Oncol 2012; 30(21): 2654-63.
Venkitaraman AR. Cancer suppression by the chromosome custodians, BRCA1 and BRCA2. Science 2014; 343(6178): 1470-
5.

maintenance to observation in two separate decisions mod- els: one for germline BRCA mutations carriers, and another for those with sporadic OC [95]. Similarly, olaparib was not found to be cost-effective in either population, with an ICER of $258,864 per PF-YLS in the BRCA patients, compared to
Lee J, Ledermann JA, Kohn EC. PARP inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann Oncol 2014; 25(1): 32-40.
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(22):

$600,552 in wild-type BRCA patients.
A more recent study evaluated olaparib in the treatment setting, relative to intravenous agents (platinum chemother- apy, non-platinum chemotherapy, and bevacizumab-based regimens) [96]. Again, olaparib was not cost-effective and was

3785-90.
De Murcia JM, Niedergang C, Trucco C, Ricoul M, Dutrillaux B, Mark M, et al. Requirement of poly(ADP-ribose) polymerase in recovery from DNA damage in mice and in cells. Proc Natl Acad Sci 1997; 94(14): 7303-7.
Caldecott KW. Single-strand break repair and genetic disease. Nat Rev Genet 2008; 9(8): 619-31.

greater than five times more costly than platinum chemother-
Satoh MS, Lindahl T. Role of poly(ADP-ribose) formation in DNA

apy, which was considered to be the most cost-effective treat- ment. An additional model included all three FDA-approved PARP inhibitors (olaparib, rucaparib, and niraparib) as main-

repair. Nature 1992; 356(6367): 356-8.
Pommier Y, O'Connor MJ, De Bono J. Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci Transl Med 2016; 8(362): 362ps17.

tenance therapy in three populations: germline BRCA mutation
Murai J. Targeting DNA repair and replication stress in the

tumors, somatic HRD tumors, and wild-type tumors [97]. None of the individual drugs was found to be cost-effective in any tumor type, with ICERs ranging two-to five-fold that of the cost-effectiveness threshold of $100,000 per PF-LYS.

treatment of ovarian cancer. Int J Clin Oncol 2017; 22(4): 619-28. Farmer H, McCabe N, Lord CJ, Tutt ANJ, Johnson DA, Richardson TB, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005; 434(7035): 917-21.

Moving forward, it will be important to identify opportunities
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(7035): 913-7.
Tomkinson, A.E., Rassool, F. Targeting abnormal DNA repair in therapy-resistant breast and pancreatic cancers. US20160051517 (2016).
Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, Friedlander M, Balmaña J, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 2015; 33(3): 244-50.
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(15): 1382-92.
Pujade-Lauraine E, Ledermann JA, Selle F, Gebski V, Penson RT, Oza AM, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): A double-blind, randomised, placebo-controlled, Phase 3 trial. Lancet Oncol 2017; 18(9): 1274- 84.
Kristeleit R, Shapiro GI, Burris HA, Oza AM, LoRusso P, Patel MR, et al. A Phase I-II study of the oral PARP inhibitor rucaparib in patients with germline BRCA1/2 -mutated ovarian carcinoma or other solid tumors. Clin Cancer Res 2017; 23(15); 4095-4106.
Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, Sun J, 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.
Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med 2016; 375(22): 2154-64.
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(2): 123-34.
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(15): 2512-9.
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(9737): 245-51.
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(9): 852-61.
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(4): 372-9.
Matulonis UA, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Olaparib maintenance therapy in patients with platinum-sensitive, relapsed serous ovarian cancer and a BRCA mutation: Overall survival adjusted for postprogression poly(adenosine diphosphate ribose) polymerase inhibitor therapy. Cancer 2016; 122(12): 1844-52.
Scholl, T., Pruss, D., Hendrickson, B.C. Method of identifying genomic rearrangements. US6895337 (2005).
Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: A preplanned retrospective analysis of outcomes by BRCA status in a randomised Phase 2 trial. Lancet Oncol 2014; 15(8): 852-61.
Dougherty BA, Lai Z, Hodgson DR, Orr MCM, Hawryluk M, Sun J, et al. Biological and clinical evidence for somatic mutations in BRCA1 and BRCA2 as predictive markers for olaparib response in high-grade serous ovarian cancers in the maintenance setting. Oncotarget. 2017; 8(27): 43653-61.
Mateo J, Moreno V, Gupta A, Kaye SB, Dean E, Middleton MR, et al. An adaptive study to determine the optimal dose of the tablet formulation of the PARP inhibitor olaparib. Target Oncol 2016; 11(3): 401-15.
Pujade-Lauraine E, Ledermann JA, Penson RiT, Oza AM, Korach J, Huzarski T, et al. Treatment with olaparib monotherapy in the maintenance setting significantly improves progression-free survival in patients with platinum-sensitive relapsed ovarian cancer: Results from the Phase III SOLO2 study. Society of Gynecologic Oncology - 48th Annual Meeting, National Harbor, USA (2017).
Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): A randomised, double-blind, placebo-controlled, Phase 3 trial. Lancet 2017; 390(10106): 1949-61.
Ledermann JA, Lortholary A, Penson RT, Gibbs E, Provencher DM, Bruchim I, et al. Adverse events (AEs) with maintenance olaparib tablets in patients (pts) with BRCA-mutated (BRCAm) platinum-sensitive relapsed serous ovarian cancer (PSR SOC): Phase III SOLO2 trial. J Clin Oncol 2017; 35.
Ledermann JA, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, et al. Quality of life during olaparib maintenance therapy in platinum-sensitive relapsed serous ovarian cancer. Br J Cancer 2016; 115(11): 1313-20.
Friedlander M, Gebski V, Gibbs E, Bloomfield R, Hilpert F, Wenzel L, et al. Health-related quality of life (HRQOL) and patient-centered outcomes with maintenance olaparib compared with placebo following chemotherapy in patients with germline (g) BRCA-mutated (m) platinum-sensitive relapsed serous ovarian cancer (PSR SOC): SOLO2 Phase III trial. J Clin Oncol 2017; 35.
Rivkin S, Iriarte D, Sloan H, Wiseman C, Moon J. Phae Ib/II with expansion of patients at the MTD study of olaparib plus weekly (metronomic) carboplatin and paclitaxel in relapsed ovarian cancer patients. J Clin Oncol 2014; 32(5s).
Lee J-M, Hays JL, Annunziata CM, Noonan AM, Minasian L, Zujewski JA, et al. Phase I/Ib study of Olaparib and Carboplatin in BRCA1 or BRCA2 mutation-associated breast or ovarian cancer with biomarker analyses. J Natl Cancer Inst 2014; 106(6): dju089.
Oza AM, Cibula D, Benzaquen AO, Poole C, Mathijssen RHJ, Sonke GS, et al. Olaparib combined with chemotherapy for recurrent platinum-sensitive ovarian cancer: A randomised Phase 2 trial. Lancet Oncol 2015; 16(1): 87-97.
Dean E, Middleton MR, Pwint T, Swaisland H, Carmichael J, Goodege-Kunwar P, et al. Phase I study to assess the safety and tolerability of olaparib in combination with bevacizumab in patients with advanced solid tumours. Br J Cancer 2012; 106(3): 468-74.
Liu JF, Tolaney SM, Birrer M, Fleming GF, Buss MK, Dahlberg SE, et al. A Phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti- angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur J Cancer 2013; 49(14): 2972-8.
Liu JF, Barry WT, Birrer M, Lee J-M, Buckanovich RJ, Fleming GF, et al. Combination cediranib and olaparib versus olaparib alone for women with recurrent platinum-sensitive ovarian cancer: A randomised phase 2 study. Lancet Oncol 2014; 15(11): 1207-14.
Lee J-M, Cimino-Mathews A, Peer CJ, Zimmer A, Lipkowitz S, Annunziata CM, et al. Safety and clinical activity of the programmed death-ligand 1 inhibitor durvalumab in combination with poly (adp-ribose) polymerase inhibitor olaparib or vascular endothelial growth factor receptor 1-3 inhibitor cediranib in women’s cancers: a dose-escalation, Phase I study. J Clin Oncol 2017; 35(19): 2193-202.
Webber, S., Canon-Koch, S., Tikhe, J., Thoresen, L. Tricyclic inhibitors of poly(ADP-ribose) polymerases. US20060009517 (2005).
Helleday, T. Use of RNAI inhibiting PARP activity for the manufacture of a medicament for the treatment of cancer. Use of RNAI inhibiting PARP activity for the manufacture of a medicament for the treatment of cancer. US20070179160 (2007).
Morrison, N., Martin, B., Smith, G.C.M., Jackson, S.P., Loh, V., Cockcroft, X.L. Phthalazinone derivatives. US20120010204 (2012).
Ashworth, A., Jackson, S.P., Martin, N.M.B., Smith, G.C.M., O’Connor, M.J. DNA damage repair inhibitors for treatment of cancer. US20060142231 (2005).
Martin, N.M.B., Smith, G.C.M., Jackson, S.P., Loh, V., Cockcroft, X.L., Matthews, I. Phthalazinone derivatives. US20050059663 (2005).

Menear, K., Ottridge, A., Londesbrough, D., Hallett, M., Mulholland, K., Pittam, J. Phthalazinone derivative. US20090270617 (2009).
Martin, N.M.B., Smith, G.C.M., White, C., Newton, R., Douglas, D., Eversley, P. Phthalazinone derivatives. US20020183325 (2002).
Martin, N.M.B., Smith, G.C.M., Jackson, S.P., Loh, V., Cockcroft, X.L., Matthews, I. Phthalazinone derivatives. US20080200469 (2008).
Etter, J. High dosage strength tablets of rucaparib. WO2016028689 (2016).
Jones, P., Ontoria, J., Scarpelli, R., Schultz-Fademrecht, C. Amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors. US8071623 (2011).
Lin, K. Use of PARP inhibitors to treat breast or ovarian cancer patients showing a loss of heterozygosity. WO2015108986 (2015).
Swisher E. BRCA1 and RAD51C promoter hypermethylation confer sensitivity to the PARP inhibitor rucaparib in patients with relapsed, platinum sensitive ovarian carcinoma in ARIEL2 Part 1. Society of Gynecologic Oncology - 48th Annual Meeting, National Harbor, USA (2017).
Konecny G, Oza AM, Tinker AV, Coleman RL, O’Malley DM, Maloney L, et al. Rucaparib in patients with relapsed, primary platinum-sensitive high-grade ovarian carcinoma with germline or somatic BRCA mutations: Integrated summary of efficacy and safety from the Phase 2 study ARIEL2 (NCT01891344). Society of Gynecologic Oncology - 48th Annual Meeting, National Harbor, USA (2017).
Sandhu SK, Schelman WR, Wilding G, Moreno V, Baird RD, Miranda S, et al. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: A Phase 1 dose-escalation trial. Lancet Oncol 2013; 14(9): 882-92.
Wilcoxen, K., Lanchbury, J., Gutin, A., Timms, K.M., Abkevich, V., Wang, Y. Methods and materials for predicting response to niraparib. WO2016094391 (2016).
Coleman RL, Sill MW, Bell-McGuinn K, Aghajanian C, Gray HJ, Tewari KS, et al. A Phase II evaluation of the potent, highly selective PARP inhibitor veliparib in the treatment of persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer in patients who carry a germline BRCA1 or BRCA2 mutation - An NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol 2015; 137(3): 386-91.
Reiss KA, Herman JM, Armstrong D, Zahurak M, Fyles A, Brade A, et al. A final report of a phase I study of veliparib (ABT-888) in combination with Low-Dose Fractionated Whole Abdominal Radiation Therapy (LDFWAR) in patients with advanced solid malignancies and peritoneal carcinomatosis with a dose escalation in ovarian and fallopian tube cancers. Gynecol Oncol 2017; 144(3): 486-90.
Kummar S, Oza AM, Fleming GF, Sullivan DM, Gandara DR, Naughton MJ, et al. Randomized trial of oral cyclophosphamide and veliparib in high-grade serous ovarian, primary peritoneal, or fallopian tube cancers, or BRCA-mutant ovarian cancer. Clin Cancer Res 2015; 21(7): 1574-82.
Bell-McGuinn KM, Brady W, Schilder R, P F, Moore K, Walker J, et al. A phase I study of continuous veliparib in combination with IV carboplatin/paclitaxel or IV/IP paclitaxel/cisplatin and bevacizumab in newly diagnosed patients with previously untreated epithelial ovarian, fallopian tube, or primary peritoneal cancer: An. J Clin Oncol 2015; 33(suppl; abstr 5507).
Shen Y, Rehman FL, Feng Y, Boshuizen J, Bajrami I, Elliott R, et al. BMN 673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency. Clin Cancer Res 2013; 19(18): 5003-15.
De Bono J, Ramanathan RK, Mina L, Chugh R, Glaspy J, Rafii S, et al. Phase I, dose-escalation, two-part trial of the parp inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers. Cancer Discov 2017; 7(6): 620-9.
Wainberg ZA, Hecht DW, Konecny GE, Goldman J, Sadeghi S, Chmielowski B, et al. Safety and efficacy results from a Phase I dose-escalation trial of the PARP inhibitor talazoparib in combination with either temozoomide or irinotecan in patients with advanced malignancies. Cancer Res 2016; 76(14 Suppl): CT011.
Abkevich V, Timms KM, Hennessy BT, Potter J, Carey MS, Meyer LA, et al. Patterns of genomic loss of heterozygosity predict
homologous recombination repair defects in epithelial ovarian cancer. Br J Cancer 2012; 107(10): 1776-82.
Birkbak NJ, Wang ZC, Kim JY, Eklund AC, Li Q, Tian R, et al. Telomeric allelic imbalance indicates defective DNA repair and sensitivity to DNA-damaging agents. Cancer Discov 2012; 2(4): 366-75.
Popova T, Manie E, Rieunier G, Caux-Moncoutier V, Tirapo C, Dubois T, et al. Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation. Cancer Res 2012; 72(21): 5454-62.
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(6): 475.
Coleman RL, Swisher EM, Oza AM, Scott CL, Giordano H, Lin KK, et al. Refinement of prespecified cutoff for genomic loss of heterozygosity (LOH) in ARIEL2 part 1: A Phase II study of rucaparib in patients (pts) with high grade ovarian carcinoma (HGOC). J Clin Oncol 2016; 34 (suppl; abstr 5540).
Richardson, A.L., Wang, Z.C., Silver, D.P., Szallasi, Z., Birkbak, N.J., Eklund, A.C. Methods for predicting anti-cancer response. US20170037478 (2017).
Tawbi, H., Pancoska, P., Benos, P., Romkes, M., Sedgewick, A., Abecassis, I. PARP genomic variants conferring resistance and sensitization to chemotherapy under inhibition. WO2016073298 (2016).
Mach, R., Chu, W., Zhou, D., Michel, L., Chen, D. Radiolabeled tracers for poly (ADP-ribose) polymerase-1 (PARP-1), methods and uses therefor. WO2015103526 (2015).
Blanchetter, S., Drummond, D., Fitzgerald, J., Moyo, V. Combination therapy for cancer treatment. WO2017031445 (2017).
Ciavolella, A., Montagnoli, A., Pesenti, E. Therapeutic combination comprising a PARP-1 inhibitor and an anti-neoplastic agent. WO2012016876 (2012).
So, C.W.E. Combination of an inhibitor of PARP with an inhibitor of GSK-3 or DOT1L. WO2017077326 (2017).
Kim, T.W., Jin, D.H., Hong, S.W., Moon, J.H., Skin, J.S., Hwang, I.Y., et al. Novel biomarker for predicting sensitivity to parp inhibitor, and use thereof. WO2016018089 (2016).
Kaufmann, S.H., Patel, A. Methods and materials for assessing responsiveness to PARP inhibitors and platinating agents. US20130224312 (2013).
Lin, C.J., Peng, G., Lin, S.Y., Mills, G. Gene signature to predict Homologous Recombination (HR) deficient cancer. WO2014138101 (2014).
Stern, M.H., Manie, E., Popova, T. Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors. WO2013182645 (2013).
Slamon, D., Finn, R., Conklin, D. Biomarkers of response to inhibition of poly-ADP ribose polymerase (PARP) in cancer. WO2014205105 (2014).
Rottenberg S, Jaspers JE, Kersbergen A, Van der Burg E, Nygren AOH, Zander SAL, et al. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. Proc Natl Acad Sci 2008; 105(44): 17079-84.
Lord CJ, Ashworth A. Mechanisms of resistance to therapies targeting BRCA-mutant cancers. Nat Med 2013; 19(11): 1381-8.
Kondrashova O, Nguyen M, Shield-Artin K, Tinker A V, Teng NNH, Harrell MI, et al. Secondary somatic mutations restoring RAD51C and RAD51D associated with acquired resistance to the parp inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov 2017; 7(9): 984-98.
Lok BH, Gardner EE, Schneeberger VE, Ni A, Desmeules P, Rekhtman N, et al. PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with temozolomide in small cell lung cancer. Clin Cancer Res 2017; 23(2): 523-35.
Murai J, Feng Y, Yu GK, Ru Y, Tang SW, Shen Y, et al. Resistance to PARP inhibitors by SLFN11 inactivation can be overcome by ATR inhibition. Oncotarget 2016; 7(47): 76534-50.
Allison Stewart C, Tong P, Cardnell RJ, Sen T, Li L, Gay CM, et al. Dynamic variations in Epithelial-to-Mesenchymal Transition (EMT), ATM, and SLFN11 govern response to PARP inhibitors and cisplatin in small cell lung cancer. Oncotarget 2017; 8(17): 28575-87.

Ha K, Fiskus W, Choi DS, Bhaskara S, Cerchietti L, Devaraj SGT, et al. Histone deacetylase inhibitor treatment induces 'BRCAness' and synergistic lethality with PARP inhibitor and cisplatin against human triple negative breast cancer cells. Oncotarget 2014; 5(14): 5637-50.
Konstantinopoulos P, Palakurthi S, Zeng Q, Zhou S, Liu JF, Ivanova E. In vivo synergism between PARP-inhibitor olaparib and HSP90-inhibitor AT13387 in high grade serous ovarian cancer patient derived xenografts. J Clin Oncol 2016; 34 (Suppl; abstr 17045).
Kim Y, Kim A, Sharip A, Sharip A, Jiang J, Yang Q, et al. Reverse the resistance to PARP inhibitors. Int J Biol Sci 2017; 13(2): 198- 208.
Secord AA, Barnett JC, Ledermann JA, Peterson BL, Myers ER, Havrilesky LJ. Cost-effectiveness of BRCA1 and BRCA2 mutation
testing to target PARP inhibitor use in platinum-sensitive recurrent ovarian cancer. Int J Gynecol Cancer 2013; 23(5): 846-52.
Smith HJ, Walters Haygood CL, Arend RC, Leath CA, Straughn JM. PARP inhibitor maintenance therapy for patients with platinum-sensitive recurrent ovarian cancer: A cost-effectiveness analysis. Gynecol Oncol 2015; 139(1): 59-62.
Wolford J, Bai J, Eskander R, Keller R, Minion L, Chan J. Evaluating the cost-effectiveness of current FDA-approved PARP ihibitors for the treatment of recurrent ovarian cancer. J Clin Oncol 2017; 35(Suppl; abstr 5516).
Liu A, Cohen J, Walsh C, Holschneider C, Sinno A. A cost- effectiveness analysis of three PARP inhibitors for maintenance therapy in platinum-sensitive recurrent ovarian cancer. Western Association of Gynecologic Oncology - 46th Annual Meeting, Palm Springs, USA (2017).AZD-9574