Next Article in Journal
Tumor BRCA Testing in High Grade Serous Carcinoma: Mutation Rates and Optimal Tissue Requirements
Next Article in Special Issue
Neuroendocrine and Aggressive-Variant Prostate Cancer
Previous Article in Journal
Exercise Reduces the Resumption of Tumor Growth and Proteolytic Pathways in the Skeletal Muscle of Mice Following Chemotherapy
Previous Article in Special Issue
Immunotherapy in Prostate Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of PARP Inhibitors in the Treatment of Metastatic Castration-Resistant Prostate Cancer

1
Deming Department of Medicine, Hematology-Oncology Section, Tulane University School of Medicine, New Orleans, LA 70112, USA
2
Tulane Cancer Center, New Orleans, LA 70112, USA
3
The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
*
Author to whom correspondence should be addressed.
Cancers 2020, 12(11), 3467; https://doi.org/10.3390/cancers12113467
Submission received: 8 October 2020 / Revised: 19 November 2020 / Accepted: 19 November 2020 / Published: 21 November 2020
(This article belongs to the Special Issue New Therapies for Prostate Cancer)

Abstract

:

Simple Summary

In recent years, the development of sequencing techniques to reveal the genomic information of prostate cancer tumors has allowed for the emergence of targeted therapies. Genomic aberrations in tumor cells have become popular due to the successful development of PARP inhibitors, which are particularly active in those tumors harboring DNA repair genomic defects. This review focuses on PARP inhibitors, two of which were approved for use by the US Food and Drug Administration in 2020 in metastatic castration-resistant prostate cancer. The article highlights the development of PARP inhibitors in the preclinical setting, summarizes the impactful clinical trials in the field, and discusses the need for continued research for further success in treating men with advanced prostate cancer.

Abstract

Metastatic castration-resistant prostate cancer (mCRPC) is an incurable malignancy with a poor prognosis. Up to 30% of patients with mCRPC have mutations in homologous recombination repair (HRR) genes. Poly (ADP-ribose) polymerase (PARP) inhibitors take advantage of HRR deficiency to kill tumor cells based on the concept of synthetic lethality. Several PARP inhibitors (PARPis) have been successful in various malignancies with HRR gene mutations including BRCA1/2, especially in breast cancer and ovarian cancer. More recently, olaparib and rucaparib were approved for mCRPC refractory to novel hormonal therapies, and other PARPis will likely follow. This article highlights the mechanism of action of PARPis at the cellular level, the preclinical data regarding a proposed mechanism of action and the effectiveness of PARPis in cancer cell lines and animal models. The article expands on the clinical development of PARPis in mCRPC, discusses potential biomarkers that may predict successful tumor control, and summarizes present and future clinical research on PARPis in the metastatic disease landscape.

1. Introduction

Metastatic castration-resistant prostate cancer (mCRPC) patients include men with distant metastases who have evidence of disease progression defined by either prostate-specific antigen (PSA) progression, new metastases, or clinical symptoms, despite a castrate level of testosterone [1]. While the success of androgen deprivation therapy (ADT) in suppressing tumor progression is almost universal, most patients invariably progress to castration-resistant prostate cancer. Exact mechanisms of progression to castration resistance remain unclear [2,3]. The current best hypothesis is the presence of a sustained androgen receptor signal, with several possible mechanisms leading up to this [3]. Although several therapeutic options have been developed for mCRPC, it remains incurable with a poor prognosis, with median survival for patients with mCRPC of approximately two to three years [4,5,6].
Analysis of whole-exome sequencing for mCRPC tumors has revealed a complex genomic landscape with variability among patients, but up to two-thirds of patients are found with genomic alterations in non-androgen-receptor-related pathways as possible therapeutic targets [2,7]. Germline and somatic mutations in homologous recombination repair (HRR) genes (including BRCA1, BRCA2, ATM, CHEK2 and others—expanded further in the following section) exist in 15–30% of cases, which increase in frequency during later stages of the disease [2,8,9,10]. Based on germline studies conducted in men with metastatic disease, DNA repair alterations are expected in approximately 12–17% of the time, more often including BRCA2, ATM, and CHEK2 genes, which is significantly higher than the incidence of men with localized prostate cancer or with family history of prostate cancer [8,11]. The high prevalence of germline mutations has led to the National Comprehensive Cancer Network recommendation of germline testing in all patients with metastatic disease [12]. However, clinical implications of germline and somatic HRR gene alterations, besides germline BRCA2 mutations, in prostate cancer remain uncertain due to limited studies, number of patients enrolled, and available approved treatments [13].
One actionable key protein involved in deoxyribonucleic acid (DNA) repair machinery is poly (ADP-ribose) polymerase (PARP). Targeted therapies against DNA repair proteins include the use of PARP inhibitors (PARPis), and the clinical development of these drugs has recently expanded to multiple tumor types. This article provides an overview of the mechanism of action and rationale for the use of PARPis alone or in combination with other therapies with an emphasis on studies leading to approval for use for mCRPC. The article summarizes the current data on putative predictive biomarkers and discusses present and future clinical trials with PARPis as they relate to mCRPC.

2. The Role of Cellular DNA Repair and Poly (ADP-Ribose) Polymerase

A complex system to repair DNA damage is in place to amend errors before cells replicate [14]. Major DNA repair pathways include direct repair, mismatch repair, base excision repair, nucleotide excision repair, and double-strand break recombination repair (which includes non-homologous end-joining and HRR) [15]. PARPs are nuclear enzymes involved in the enzymatic machinery for repairing DNA breaks, and in other roles including gene regulation, cell proliferation and cell death [16,17]. Since the original discovery of PARP-1 in 1963 [18], at least 18 PARPs have been identified [17].
PARPs attach poly (ADP-ribose) polymers to proteins, including to one another and to themselves with the ability to self-modify. When DNA becomes damaged, PARP-1 is recruited to the site of single-strand breaks, and it begins to cleave nicotinamide adenine dinucleotide (NAD+) into nicotinamide and ADP-ribose. PARP-1 catalyzes formation of ADP-ribose polymers (PARylation), which helps release the DNA from histones, and it also recruits and activates the base excision repair (BER) enzyme complex [19]. PARP-1 and PARP-2 both promote BER by two independent but intrinsically linked mechanisms, and disruption of both proteins leads to defective BER [20]. When BER is defective, homologous recombination (HR) may be able to rescue the damaged DNA and ensure proper DNA replication. HR is one of the most effective mechanisms to repair double-strand DNA breaks, which requires functional BRCA1 and BRCA2 proteins [21]. Other HR genes identified include PALB2, FANCI, FANCL, FANCC, RAD50, RAD51, RAD51C, RAD54L, ATM, ATR, CHEK1, and CHEK2 [22].
PARP inhibitors (PARPis) appear to work in two different ways: they compete with NAD+ at the catalytic site of PARPs to prevent PARylation, and they also trap PARP1/2 to damaged DNA to form cytotoxic PARP-DNA complexes [23]. Different PARPis were found to have varying potency of trapping PARP-DNA complexes not related to their catalytic inhibitory properties. PARP inhibition leads to stalling of replication forks, inducing double-strand DNA breaks and the need for HR [24,25]. Tumor cells with two mutated BRCA genes are unable to start HR, which ultimately leads to cell death, while normal cells still possess at least one functioning BRCA gene to begin HR to repair DNA and prevent apoptosis. Cells that have either BER deficiency or HR deficiency may survive; if both pathways are deficient, the cells die. This is the concept for using PARPis to selectively target malignancies with BRCA1/2 germline or somatic mutations, which is termed “synthetic lethality.” This model was supported by two landmark studies published in 2005–BRCA1/2 dysfunction sensitized tumor cells to PARPis in vitro, resulting in selective tumor cell death [26,27].

3. Preclinical Development of PARP Inhibitors

Olaparib (previously known as AZD2281 and KU-0059436) was discovered to be a potent orally bioavailable PARPi against BRCA-1/2 deficient cell lines. It was determined to have both good in vitro cellular potency (IC50 for PARP-1 inhibition of 6 nM) and in vivo efficacy because it led to 80% tumor inhibition when fed in combination with temozolomide to mice [22]. Olaparib also had in vitro activity against ataxia telangiectasia mutated (ATM)-deficient lymphoid tumors and in vivo activity in mice xenografted with ATM-deficient mantle cell lymphoma tumor [28,29].
Rucaparib (PF01367338 and AG014699) was selected from a group of several compounds tested for a clinical trial based on excellent chemosensitization, radiosensitization, aqueous solubility, and safety profile [30]. Rucaparib was cytotoxic to human cancer cell lines with mutated BRCA1/2 and was then found to be effective in a wide range of ovarian cancer cell lines (including homologous recombination pathway deficiencies not including BRCA1/2 mutations) alone and in combination with other agents [31,32].
Similarly, niraparib (MK-4827) [33], talazoparib (MDV3800 and BMN-673) [34], and veliparib (ABT-888) [35] moved forward to clinical trials based on potent in vitro activity against PARPs and successful oral bioavailability and in vivo activity in BRCA-deficient xenograft tumor murine models. Talazoparib was the most potent PARPi in vitro, about 100-fold more potent at trapping PARP-DNA complexes compared to both olaparib and rucaparib [36].
Research has shed some light on how PARPs are involved in prostate cancer and how PARPis are beneficial against prostate cancer, although the available literature is scant. The erythroblast transformation specific (ETS) gene fusion family is involved in the progression of a variety of cancers, and TMPRSS2:ERG is a prostate cancer-specific gene fusion; it has been noted that this gene fusion product interacts with PARP-1, and that olaparib was able to suppress ETS-positive (but not ETS-negative) prostate cancer cell invasion [37], yet no clinical data confirming this cellular effect is available. In fact, in a clinical trial with a subset of mCRPC patients who received veliparib and abiraterone, there was no difference in response rate between patients with or without ETS fusions [38]. The combination of rucaparib with radiation therapy was synergistic for prostate cancer cells expressing the TMPRSS2:ERG gene fusion, as these cells showed enhanced sensitivity towards rucaparib, which increased radiation response [39]. Schiewer et al. [40] demonstrated in prostate cancer cells that PARP-1 modulated both androgen-receptor function and response to DNA damage, suggesting its involvement in prostate cancer progression and maintenance of castration resistance; in this study, olaparib and veliparib successfully decreased androgen-receptor target gene expression and tumor growth using murine models and ex vivo prostate tumor cultures.

4. Clinical Development of Olaparib and Rucaparib in Prostate Cancer

4.1. Olaparib

The first clinical trial of olaparib (NCT00516373) included three patients with advanced prostate cancer, including one patient with a BRCA2 mutation who had a greater than 50% reduction in PSA level and resolution of bone metastases [41]. The open-label phase II trial (NCT01078662) by Kaufman et al. [42] included eight patients with mCRPC (one with BRCA1 mutation, seven with BRCA2 mutation) and a median of two prior therapies. Median total duration of olaparib treatment was 223.5 days, given at 400 mg twice daily. The one BRCA1 mutation patient and three of the seven BRCA2 mutation patients responded to the treatment. Two of the eight prostate cancer patients had stable disease that persisted for at least eight weeks. Median duration of response was 327 days, and median time to onset of response was 54.5 days. Progression-free survival (PFS) at 6 months was 62.5%, median overall survival was 18.4 months, and 50.0% of the patients were alive at 12 months.
Further encouraging efficacy data was reported in the open-label Phase II Trial of Olaparib in Patients with Advanced Castration Resistant Prostate Cancer (TOPARP-A trial, NCT01682772), which focused solely on mCRPC [43]. The primary endpoint was based on objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors (RECIST), PSA decline, or reduction in circulating tumor-cell count. The trial enrolled 50 patients, although one was lost to follow-up early and was not included in the later analysis. All 49 patients had previously received treatment with other agents, including docetaxel, abiraterone, enzalutamide, and cabazitaxel. Next-generation sequencing data revealed that 16 patients had tumor aberrations in DNA-repair genes, including BRCA1, BRCA2, ATM, FANCA, PALB2, HDAC2, and CHEK2. Median duration of olaparib was 12 weeks. Patients with these DNA-repair gene aberrations had a significantly better response to olaparib of 400 mg twice daily (14/16 compared to 2/33 biomarker-negative patients). All seven patients with BRCA2 loss had PSA levels decrease by over 50% from baseline, and the five who had measurable disease had radiologic partial response. Four of five patients with ATM mutations had response as well. The results in the TOPARP-A trial led the FDA in January 2016 to give olaparib a breakthrough therapy designation for mCRPC with BRCA1/2 or ATM mutations previously treated with a taxane-based chemotherapy and either enzalutamide or abiraterone.
On a continuum, TOPARB-B was an open-label phase II trial that included 98 mCRPC patients with known aberrations to 18 DNA-repair genes who received olaparib: 49 received 300 mg twice daily and 49 received 400 mg twice daily [44]. Overall, 43 of the 98 patients achieved a confirmed composite response. Patients with the BRCA1/2 mutation had the best response and longest median radiographic progression-free survival (rPFS) compared to patients with the 16 other gene mutations, but because olaparib did have an effect on these other mutations as well, this study supported the genomic stratification of mCRPC and olaparib’s potential in mCRPC patients with tumor gene aberrations besides the BRCA1/2 mutation.
These data leveraged conducting a confirmatory, phase III trial, PROfound (NCT02987543). This prospective, randomized, open-label study evaluated the efficacy and safety of 300 mg of olaparib twice daily versus 160 mg of enzalutamide daily or 1000 mg of abiraterone daily in 387 patients with mCRPC and 15 HRR gene alterations (BRCA1, BRCA2, ATM, BRIP1, BARD1, CDK12, CHEK1, CHEK2, FANCL, PALB2, PPP2R2A, RAD51B, RAD51C, RAD51D or RAD54L) [45]. The patients had all been treated previously with enzalutamide or abiraterone, and some patients had also been treated previously with taxane chemotherapy. Cohort A included 245 patients (162 received olaparib and 83 received the control treatment) with at least one alteration in BRCA1, BRCA2, or ATM, while cohort B included 142 patients (94 received olaparib and 48 received the control treatment) with the 12 other gene alterations. The primary endpoint was imaging-based PFS in cohort A. In cohort A, the olaparib group was better than the control group in terms of significantly increased median imaging-based PFS (7.4 vs. 3.6 months, hazard ratio [HR] 0.34; 95% confidence interval [CI], 0.25 to 0.47; p < 0.001), median overall survival (18.5 vs. 15.1 months, HR 0.64; 95% CI, 0.43 to 0.97; p = 0.02), objective response rate (33% vs. 2%, odds ratio 20.86; 95% CI, 4.18 to 379.18; p < 0.001), and median time to pain (HR 0.44; 95% CI, 0.22 to 0.91; p = 0.02). For cohorts A and B together, the olaparib group was also better than the control group based on median imaging-based PFS (5.3 vs. 3.5 months, HR 0.49; 95% CI, 0.38 to 0.63; p < 0.001), confirmed objective response rate (22% vs. 4%, odds ratio, 5.93; 95% CI, 2.01 to 25.40), free of pain progression at six months (85% vs. 75%, HR 0.64), estimated median overall survival (17.5 vs. 14.3 months, HR 0.67; 95% CI, 0.49 to 0.93), and PSA50 response (30% vs. 10%).
For cohort A, the ATM group demonstrated limited activity (62 olaparib patients with median rPFS 5.36 months, 95% CI, 3.61 to 6.21 months; vs. 24 control patients with median rPFS of 4.70 months, 95% CI, 1.84 to 7.26 months), as further discussed in a later section. The BRCA1 group had a small sample size (8 olaparib patients with median rPFS of 2.07 months, 95% CI, 1.38 to 5.52 months; vs. 5 control patients with median rPFS of 4.70 months, 95% CI, 1.71 to 3.71 months). For cohort B, interpreting results are limited by the relatively small sample size. Yet, promising findings were described in the RAD51B (4 olaparib patients with median rPFS of 10.89 months, 95% CI, 1.61 to 14.75; vs. 1 control patient with median rPFS of 1.77 months) and the RAD54L groups (3 olaparib patients with median rPFS of 7.20 months, 95% CI, 3.71 to 7.39; vs. 2 control patients with 2.41 months, 95% CI, 1.81 to 3.02 months).
In cohort A, more adverse events (AEs), including grade 3 or higher, were noted in the olaparib group compared to the control group in the PROfound trial, and in line with the known safety profile of PARP inhibitors [46,47]. While common AEs included anemia, nausea and decreased appetite, serious side effects associated with olaparib include the development of myelodysplastic syndrome, acute myeloid leukemia, and pneumonitis [48]. Thus, it is often helpful to obtain a complete blood count at baseline and then monthly to monitor for clinically significant changes.
This first biomarker-selected mCRPC study led the FDA in May 2020 to approve the use of olaparib for patients with mCRPC and HRR gene mutations who progressed despite previous treatment on enzalutamide or abiraterone [49]. Foundation-One was the approved companion diagnostic test, but other tissue and circulating tumor DNA assays are commercially available and future validation studies will clarify their role in identifying these biomarkers.
The efficacy of olaparib monotherapy in mCRPC patients without HRR mutations is under investigation. In a double-blinded, randomized, placebo-controlled phase II trial of mCRPC not required to have an HRR mutation (NCT0197221), 71 patients who received 300 mg of olaparib twice daily combined with 1000 mg of abiraterone daily were compared to 71 patients who received abiraterone and placebo [50]. Median rPFS was 13.8 months for olaparib and abiraterone compared to 8.2 months for abiraterone alone (HR 0.65, 95% CI, 0.44 to 0.97, p = 0.034), suggesting possible benefits for olaparib in mCRPC patients without HRR mutations. The ongoing phase III trial PROPEL (NCT03732820) builds on the success of the phase II trial that used the combination of olaparib and abiraterone in genomic unselected mCRPC patients. The primary outcome measure is rPFS, with expected completion in 2021.

4.2. Rucaparib

Rucaparib was granted accelerated approval by the FDA in May 2020 for treatment of mCRPC with a deleterious germline or somatic BRCA mutation previously treated with androgen-receptor-directed therapy and a taxane-based therapy, based on data from TRITON-2 [51]. The Trial of Rucaparib in Prostate Indications (TRITON)-2 is an open-label phase II trial (NCT02952534) evaluating 600 mg of rucaparib twice daily (with gonadotropin-releasing hormone [GnRH] analogue or prior bilateral orchiectomy) in mCRPC patients who progressed on androgen-deprivation therapy and one prior taxane-based chemotherapy with a deleterious germline or somatic alteration in BRCA1, BRCA2, ATM, BARD1, BRIP1, CDK12, CHEK2, FANCA, NBN, PALB2, RAD51, RAD51B, RAD51C, RAD51D, or RAD54L. Of 62 BRCA-mutated patients, 27 had a confirmed ORR, and 15 of these 27 had a response duration of at least six months. TRITON-3 is an actively recruiting, randomized, open-label phase III trial (NCT02975934) studying rucaparib 600 mg twice daily versus either abiraterone, enzalutamide, or docetaxel in patients with mCRPC and a deleterious germline or somatic mutation in BRCA1, BRCA2, or ATM that progressed on androgen-receptor signaling-directed therapy, building on the success of TRITON-2.

5. Other PARP Inhibitors in Prostate Cancer

Niraparib is being evaluated in the open-label phase II trial GALAHAD (NCT02854436) in mCRPC patients with gene alterations in BRCA1, BRCA2, ATM, FANCA, PALB2, CHEK2, BRIP1, or HDAC2 who progressed despite androgen-receptor-targeted therapy and taxane-based chemotherapy [52]. At the 2019 European Society for Medical Oncology (ESMO) Congress, data presented on 81 patients (46 BRCA and 35 non-BRCA) showed better performance for BRCA patients, who had a 41% objective response rate, 63% complete response rate, median rPFS of 8.2 months, and overall survival of 12.6 months; for non-BRCA patients, the numbers were 9%, 16%, 5.3 months, and 14.0 months, respectively. This led the FDA in October 2019 to give niraparib a breakthrough designation as therapy for BRCA1/2-mutant positive mCRPC [53].
One milligram of talazoparib daily is being tested in the open-label phase II trial TALAPRO-1 (NCT03148795) in patients with mCRPC with mutations in ATM, ATR, BRCA1, BRCA2, CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2, or RAD51C who progressed despite androgen-receptor-targeted therapy and taxane-based chemotherapy [54]. Preliminary data for 43 patients (20 BRCA1/2, 14 ATM, 2 PALB2, 7 other) showed an overall response rate of 25.6% (13.5–41.2), with an ORR of 50% (27.2–72.8) in the BRCA1/2 subgroup and 7.1% (0.2–33.9) in the ATM subgroup.

6. Looking into the Future: Potential Biomarkers of Response to PARP Inhibitors, Cautious Optimism, and Ongoing Clinical Trials

BRCA1 and BRCA2 are logical candidates to be biomarkers of response to PARPis, based on the current knowledge of DNA damage repair with wild-type BRCA1 and BRCA2 part of the complex in homologous recombination to fix double-strand breaks [55]. However, recent data have sparked debate over just how predictive BRCA1/2-mutated cancers can be to PARPis, especially in non-BRCA-associated cancer types (cancers not including breast, ovary, prostate, or pancreatic cancer) [56]. Response to PARPis in different BRCA1/2-associated cancers varies widely, and BRCA1/2-mutations are not synonymous with HRR deficiency, as other secondary somatic mutations may restore or bypass BRCA function [56,57].
Cyclin-dependent kinase 12 (CDK12) phosphorylates the C-terminal domain of RNA polymerase II, which ensures several functions, including optimal transcription elongation, translation of a subset of human protein-coding genes, and maintenance of genomic stability [58]. A genome-wide synthetic lethal screen involving ovarian cancer cell lines and olaparib determined that CDK12 deficiency may confer sensitivity to PARPis [59]. However, as more clinical data become available, CDK12 mutations in prostate cancer appear to minimally respond to PARPis. In the TRITON2 trial, CDK12 mutated mCRPC patients had dismal response rates to rucaparib (0/10 with a radiographic response, 1/15 with a PSA response), and this cohort was discontinued [60]. Antonarakis et al. [61] revealed early results of a multi-institution retrospective study of 60 men with CDK12-altered prostate cancers that showed 0 of the 11 who received PARPis (10 olaparib, 1 rucaparib) had a PSA response.
ATM (ataxia-telangiectasia mutated) is a phosphatidylinositol-3 related kinase involved in DNA double-strand break repair that generates signaling networks for DNA repair proteins [62]. Response rates to PARPis in ATM-deficient tumor cell lines were seen in chronic lymphocytic leukemia [28], gastric cancer [63], and mantle cell lymphoma [64]. However, other studies revealed ATM deficiency may not be enough to fully sensitize these cells to PARP inhibition; experiments showed olaparib given alone to ATM-deficient cancer cells induced only a cytostatic state, while there are emerging data suggesting that olaparib combined with an ATR (ATM- and RAD3-related) inhibitor provides an additional cytotoxic effect [65,66]. A study using prostate cancer cell lines agreed that ATM loss may not respond to PARPis, but they did respond well to an ATR inhibitor [67].
In the PROFOUND trial, the hazard ratio for progression or death of mCRPC patients with ATM mutation was 1.04 (95% CI of 0.61–1.87) [45]. In the TRITON2 trial, only 2 of 19 mCRPC patients with ATM mutation receiving rucaparib had a radiographic response and 2 of 49 patients had a PSA response [60]. Taken together, cancers with ATM aberrations may rarely respond to PARPis, and the response is, in general, more limited compared with the activity in BRCA1/2 tumors.
Other DNA repair genes such as PALB2, FANCA and the RAD51 family are currently being evaluated in several of the above-mentioned trials, although the number of patients with these mutations enrolled is relatively low. Preclinical data using cell lines have suggested synergy of PARPis with these impaired DNA repair proteins, such as FANCA [68], RAD51C [69], or MRE11 [70]. Limited clinical data exist to make definitive conclusions about the effectiveness of PARPis in patients with these mutations [71]. Notably, prostate cancer patients with PALB2 mutations receiving different PARPis have demonstrated antitumor activity. More robust prospective studies must be done to better determine the reliability of these biomarkers for PARPis in mCRPC.
Overall success of PARPis in mCRPC must still be interpreted cautiously, as the response to patients harboring various mutations is still variable, and eventually patients experience disease progression after prolonged administration of PARPis. Multiple proposed mechanisms of PARPi resistance include restoration of HRR, DNA replication fork protection, reversion mutations, epigenetic modifications, and restoration of PARylation [72]. Resistance to PARPis was already proposed and demonstrated very early in the preclinical setting using a PARPi-resistant pancreatic cancer cell line with the intragenic deletion of c.6174delT of BRCA2 [73]. Because of these possibilities, identifying patients with mCRPC who may develop resistance to PARPis would be helpful. For example, Quigley et al. [74] detected BRCA2 reversion mutations associated with olaparib and talazoparib resistance in mCRPC patients through analysis of circulating cell-free DNA. There continue to be several active clinical trials for mCRPC patients at various stages involving different PARPis, either as monotherapy (Table 1) or combined with other therapies to enhance success (Table 2). These trials aim to further improve upon the progress so far with PARPis against this incurable malignancy.

7. Conclusions

The success of PARPis in treating cancer points to the importance of understanding the molecular phenotype of mCRPC, the therapeutic implications of genomic information, and the potential of precision oncology. Olaparib and rucaparib are now available for mCRPC, and other PARPis are likely to be approved soon, based on several ongoing studies. More studies are required to determine the full benefit of these agents, including use in earlier stages of the disease, identification of further predictive biomarkers and evaluation of synergism when combined with other agents.

Author Contributions

Writing—original draft preparation, A.J.; writing—review and editing, A.J., O.S., P.C.B., C.J.P.; supervision, P.C.B., C.J.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Saad, F.; Hotte, S.J. Guidelines for the management of castrate-resistant prostate cancer. Can. Urol. Assoc. J. 2010, 4, 380–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Robinson, D.; Van Allen, E.M.; Wu, Y.M.; Schultz, N.; Lonigro, R.J.; Mosquera, J.M.; Montgomery, B.; Taplin, M.E.; Pritchard, C.C.; Attard, G.; et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015, 161, 1215–1228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Huang, Y.; Jiang, X.; Liang, X.; Jiang, G. Molecular and cellular mechanisms of castration resistant prostate cancer. Oncol. Lett. 2018, 15, 6063–6076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ryan, C.J.; Smith, M.R.; Fizazi, K.; Saad, F.; Mulders, P.F.; Sternberg, C.N.; Miller, K.; Logothetis, C.J.; Shore, N.D.; Small, E.J.; et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): Final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2015, 16, 152–160. [Google Scholar] [CrossRef]
  5. Beer, T.M.; Armstrong, A.J.; Rathkopf, D.; Loriot, Y.; Sternberg, C.N.; Higano, C.S.; Iversen, P.; Evans, C.P.; Kim, C.S.; Kimura, G.; et al. Enzalutamide in Men with Chemotherapy-naïve Metastatic Castration-resistant Prostate Cancer: Extended Analysis of the Phase 3 PREVAIL Study. Eur. Urol. 2017, 71, 151–154. [Google Scholar] [CrossRef] [Green Version]
  6. Mehtälä, J.; Zong, J.; Vassilev, Z.; Brobert, G.; Gabarró, M.S.; Stattin, P.; Khanfir, H. Overall survival and second primary malignancies in men with metastatic prostate cancer. PLoS ONE 2020, 15, e0227552. [Google Scholar] [CrossRef] [Green Version]
  7. van Dessel, L.F.; van Riet, J.; Smits, M.; Zhu, Y.; Hamberg, P.; van der Heijden, M.S.; Bergman, A.M.; van Oort, I.M.; de Wit, R.; Voest, E.E.; et al. The genomic landscape of metastatic castration-resistant prostate cancers reveals multiple distinct genotypes with potential clinical impact. Nat. Commun. 2019, 10, 5251. [Google Scholar] [CrossRef] [Green Version]
  8. Pritchard, C.C.; Mateo, J.; Walsh, M.F.; De Sarkar, N.; Abida, W.; Beltran, H.; Garofalo, A.; Gulati, R.; Carreira, S.; Eeles, R.; et al. Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer. N. Engl. J. Med. 2016, 375, 443–453. [Google Scholar] [CrossRef]
  9. Marshall, C.H.; Fu, W.; Wang, H.; Baras, A.S.; Lotan, T.L.; Antonarakis, E.S. Prevalence of DNA repair gene mutations in localized prostate cancer according to clinical and pathologic features: Association of Gleason score and tumor stage. Prostate Cancer Prostatic Dis. 2019, 22, 59–65. [Google Scholar] [CrossRef]
  10. Abida, W.; Armenia, J.; Gopalan, A.; Brennan, R.; Walsh, M.; Barron, D.; Danila, D.; Rathkopf, D.; Morris, M.; Slovin, S.; et al. Prospective Genomic Profiling of Prostate Cancer Across Disease States Reveals Germline and Somatic Alterations That May Affect Clinical Decision Making. JCO Precis. Oncol. 2017, 2017. [Google Scholar] [CrossRef]
  11. Nicolosi, P.; Ledet, E.; Yang, S.; Michalski, S.; Freschi, B.; O’Leary, E.; Esplin, E.D.; Nussbaum, R.L.; Sartor, O. Prevalence of Germline Variants in Prostate Cancer and Implications for Current Genetic Testing Guidelines. JAMA Oncol. 2019, 5, 523–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Mohler, J.L.; Antonarakis, E.S.; Armstrong, A.J.; D’Amico, A.V.; Davis, B.J.; Dorff, T.; Eastham, J.A.; Enke, C.A.; Farrington, T.A.; Higano, C.S.; et al. Prostate Cancer, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2019, 17, 479–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lozano, R.; Castro, E.; Aragón, I.M.; Cendón, Y.; Cattrini, C.; López-Casas, P.P.; Olmos, D. Genetic aberrations in DNA repair pathways: A cornerstone of precision oncology in prostate cancer. Br. J. Cancer 2020. [Google Scholar] [CrossRef] [PubMed]
  14. Hoeijmakers, J.H. Genome maintenance mechanisms for preventing cancer. Nature 2001, 411, 366–374. [Google Scholar] [CrossRef] [PubMed]
  15. Bernstein, C.; Bernstein, H.; Payne, C.M.; Garewal, H. DNA repair/pro-apoptotic dual-role proteins in five major DNA repair pathways: Fail-safe protection against carcinogenesis. Mutat. Res. 2002, 511, 145–178. [Google Scholar] [CrossRef]
  16. Amé, J.C.; Spenlehauer, C.; de Murcia, G. The PARP superfamily. Bioessays 2004, 26, 882–893. [Google Scholar] [CrossRef] [PubMed]
  17. Morales, J.; Li, L.; Fattah, F.J.; Dong, Y.; Bey, E.A.; Patel, M.; Gao, J.; Boothman, D.A. Review of poly (ADP-ribose) polymerase (PARP) mechanisms of action and rationale for targeting in cancer and other diseases. Crit. Rev. Eukaryot. Gene Expr. 2014, 24, 15–28. [Google Scholar] [CrossRef] [Green Version]
  18. Chambon, P.; Weill, J.D.; Mandel, P. Nicotinamide mononucleotide activation of new DNA-dependent polyadenylic acid synthesizing nuclear enzyme. Biochem. Biophys. Res. Commun. 1963, 11, 39–43. [Google Scholar] [CrossRef]
  19. Gupte, R.; Liu, Z.; Kraus, W.L. PARPs and ADP-ribosylation: Recent advances linking molecular functions to biological outcomes. Genes Dev. 2017, 31, 101–126. [Google Scholar] [CrossRef] [Green Version]
  20. Ronson, G.E.; Piberger, A.L.; Higgs, M.R.; Olsen, A.L.; Stewart, G.S.; McHugh, P.J.; Petermann, E.; Lakin, N.D. PARP1 and PARP2 stabilise replication forks at base excision repair intermediates through Fbh1-dependent Rad51 regulation. Nat. Commun. 2018, 9, 746. [Google Scholar] [CrossRef] [Green Version]
  21. Powell, S.N.; Kachnic, L.A. Roles of BRCA1 and BRCA2 in homologous recombination, DNA replication fidelity and the cellular response to ionizing radiation. Oncogene 2003, 22, 5784–5791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Konstantinopoulos, P.A.; Ceccaldi, R.; Shapiro, G.I.; D’Andrea, A.D. Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. Cancer Discov. 2015, 5, 1137–1154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Murai, J.; Huang, S.Y.; Das, B.B.; Renaud, A.; Zhang, Y.; Doroshow, J.H.; Ji, J.; Takeda, S.; Pommier, Y. Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors. Cancer Res. 2012, 72, 5588–5599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Dziadkowiec, K.N.; Gąsiorowska, E.; Nowak-Markwitz, E.; Jankowska, A. PARP inhibitors: Review of mechanisms of action and BRCA1/2 mutation targeting. Prz. Menopauzalny 2016, 15, 215–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Wang, H.; Zhang, S.; Song, L.; Qu, M.; Zou, Z. Synergistic lethality between PARP-trapping and alantolactone-induced oxidative DNA damage in homologous recombination-proficient cancer cells. Oncogene 2020, 39, 2905–2920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Farmer, H.; McCabe, N.; Lord, C.J.; Tutt, A.N.; Johnson, D.A.; Richardson, T.B.; Santarosa, M.; Dillon, K.J.; Hickson, I.; Knights, C.; et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005, 434, 917–921. [Google Scholar] [CrossRef]
  27. Bryant, H.E.; Schultz, N.; Thomas, H.D.; Parker, K.M.; Flower, D.; Lopez, E.; Kyle, S.; Meuth, M.; Curtin, N.J.; Helleday, T. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005, 434, 913–917. [Google Scholar] [CrossRef]
  28. Weston, V.J.; Oldreive, C.E.; Skowronska, A.; Oscier, D.G.; Pratt, G.; Dyer, M.J.; Smith, G.; Powell, J.E.; Rudzki, Z.; Kearns, P.; et al. The PARP inhibitor olaparib induces significant killing of ATM-deficient lymphoid tumor cells in vitro and in vivo. Blood 2010, 116, 4578–4587. [Google Scholar] [CrossRef] [Green Version]
  29. Williamson, C.T.; Muzik, H.; Turhan, A.G.; Zamò, A.; O’Connor, M.J.; Bebb, D.G.; Lees-Miller, S.P. ATM deficiency sensitizes mantle cell lymphoma cells to poly(ADP-ribose) polymerase-1 inhibitors. Mol. Cancer Ther. 2010, 9, 347–357. [Google Scholar] [CrossRef] [Green Version]
  30. Thomas, H.D.; Calabrese, C.R.; Batey, M.A.; Canan, S.; Hostomsky, Z.; Kyle, S.; Maegley, K.A.; Newell, D.R.; Skalitzky, D.; Wang, L.Z.; et al. Preclinical selection of a novel poly(ADP-ribose) polymerase inhibitor for clinical trial. Mol. Cancer Ther. 2007, 6, 945–956. [Google Scholar] [CrossRef] [Green Version]
  31. Drew, Y.; Mulligan, E.A.; Vong, W.T.; Thomas, H.D.; Kahn, S.; Kyle, S.; Mukhopadhyay, A.; Los, G.; Hostomsky, Z.; Plummer, E.R.; et al. Therapeutic potential of poly(ADP-ribose) polymerase inhibitor AG014699 in human cancers with mutated or methylated BRCA1 or BRCA2. J. Natl. Cancer Inst. 2011, 103, 334–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ihnen, M.; zu Eulenburg, C.; Kolarova, T.; Qi, J.W.; Manivong, K.; Chalukya, M.; Dering, J.; Anderson, L.; Ginther, C.; Meuter, A.; et al. Therapeutic potential of the poly(ADP-ribose) polymerase inhibitor rucaparib for the treatment of sporadic human ovarian cancer. Mol. Cancer Ther. 2013, 12, 1002–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Jones, P.; Altamura, S.; Boueres, J.; Ferrigno, F.; Fonsi, M.; Giomini, C.; Lamartina, S.; Monteagudo, E.; Ontoria, J.M.; Orsale, M.V.; et al. Discovery of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): A novel oral poly(ADP-ribose)polymerase (PARP) inhibitor efficacious in BRCA-1 and -2 mutant tumors. J. Med. Chem. 2009, 52, 7170–7185. [Google Scholar] [CrossRef] [PubMed]
  34. Shen, Y.; Rehman, F.L.; Feng, Y.; Boshuizen, J.; Bajrami, I.; Elliott, R.; Wang, B.; Lord, C.J.; Post, L.E.; Ashworth, A. 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, 5003–5015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Donawho, C.K.; Luo, Y.; Penning, T.D.; Bauch, J.L.; Bouska, J.J.; Bontcheva-Diaz, V.D.; Cox, B.F.; DeWeese, T.L.; Dillehay, L.E.; Ferguson, D.C.; et al. ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin. Cancer Res. 2007, 13, 2728–2737. [Google Scholar] [CrossRef] [Green Version]
  36. Murai, J.; Huang, S.Y.; Renaud, A.; Zhang, Y.; Ji, J.; Takeda, S.; Morris, J.; Teicher, B.; Doroshow, J.H.; Pommier, Y. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol. Cancer Ther. 2014, 13, 433–443. [Google Scholar] [CrossRef] [Green Version]
  37. Brenner, J.C.; Ateeq, B.; Li, Y.; Yocum, A.K.; Cao, Q.; Asangani, I.A.; Patel, S.; Wang, X.; Liang, H.; Yu, J.; et al. Mechanistic rationale for inhibition of poly(ADP-ribose) polymerase in ETS gene fusion-positive prostate cancer. Cancer Cell 2011, 19, 664–678. [Google Scholar] [CrossRef] [Green Version]
  38. Hussain, M.; Daignault-Newton, S.; Twardowski, P.W.; Albany, C.; Stein, M.N.; Kunju, L.P.; Siddiqui, J.; Wu, Y.M.; Robinson, D.; Lonigro, R.J.; et al. Targeting Androgen Receptor and DNA Repair in Metastatic Castration-Resistant Prostate Cancer: Results From NCI 9012. J. Clin. Oncol. 2018, 36, 991–999. [Google Scholar] [CrossRef]
  39. Chatterjee, P.; Choudhary, G.S.; Sharma, A.; Singh, K.; Heston, W.D.; Ciezki, J.; Klein, E.A.; Almasan, A. PARP inhibition sensitizes to low dose-rate radiation TMPRSS2-ERG fusion gene-expressing and PTEN-deficient prostate cancer cells. PLoS ONE 2013, 8, e60408. [Google Scholar] [CrossRef]
  40. Schiewer, M.J.; Goodwin, J.F.; Han, S.; Brenner, J.C.; Augello, M.A.; Dean, J.L.; Liu, F.; Planck, J.L.; Ravindranathan, P.; Chinnaiyan, A.M.; et al. Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov. 2012, 2, 1134–1149. [Google Scholar] [CrossRef] [Green Version]
  41. Fong, P.C.; Boss, D.S.; Yap, T.A.; Tutt, A.; Wu, P.; Mergui-Roelvink, M.; Mortimer, P.; Swaisland, H.; Lau, A.; O’Connor, M.J.; et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 2009, 361, 123–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kaufman, B.; Shapira-Frommer, R.; Schmutzler, R.K.; Audeh, M.W.; Friedlander, M.; Balmaña, J.; Mitchell, G.; Fried, G.; Stemmer, S.M.; Hubert, A.; et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J. Clin. Oncol. 2015, 33, 244–250. [Google Scholar] [CrossRef] [PubMed]
  43. Mateo, J.; Carreira, S.; Sandhu, S.; Miranda, S.; Mossop, H.; Perez-Lopez, R.; Nava Rodrigues, D.; Robinson, D.; Omlin, A.; Tunariu, N.; et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N. Engl. J. Med. 2015, 373, 1697–1708. [Google Scholar] [CrossRef] [PubMed]
  44. Mateo, J.; Porta, N.; Bianchini, D.; McGovern, U.; Elliott, T.; Jones, R.; Syndikus, I.; Ralph, C.; Jain, S.; Varughese, M.; et al. Olaparib in patients with metastatic castration-resistant prostate cancer with DNA repair gene aberrations (TOPARP-B): A multicentre, open-label, randomised, phase 2 trial. Lancet Oncol. 2020, 21, 162–174. [Google Scholar] [CrossRef]
  45. de Bono, J.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020. [Google Scholar] [CrossRef]
  46. Montemorano, L.; Lightfoot, M.D.; Bixel, K. Role of Olaparib as Maintenance Treatment for Ovarian Cancer: The Evidence to Date. OncoTargets Ther. 2019, 12, 11497–11506. [Google Scholar] [CrossRef] [Green Version]
  47. Caulfield, S.E.; Davis, C.C.; Byers, K.F. Olaparib: A Novel Therapy for Metastatic Breast Cancer in Patients With a BRCA1/2 Mutation. J. Adv. Pract. Oncol. 2019, 10, 167–174. [Google Scholar]
  48. Ringley, J.T.; Moore, D.C.; Patel, J.; Rose, M.S. Poly (ADP-ribose) Polymerase Inhibitors in the Management of Ovarian Cancer: A Drug Class Review. Pharm. Ther. 2018, 43, 549–556. [Google Scholar]
  49. U.S. Food and Drug Administration. FDA Approves Olaparib for HRR Gene-Mutated Metastatic Castration-Resistant Prostate Cancer. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-olaparib-hrr-gene-mutated-metastatic-castration-resistant-prostate-cancer (accessed on 27 June 2020).
  50. Clarke, N.; Wiechno, P.; Alekseev, B.; Sala, N.; Jones, R.; Kocak, I.; Chiuri, V.E.; Jassem, J.; Fléchon, A.; Redfern, C.; et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2018, 19, 975–986. [Google Scholar] [CrossRef]
  51. U.S. Food and Drug Administration. FDA Grants Accelerated Approval to Rucaparib for BRCA-Mutated Metastatic Castration-Resistant Prostate Cancer. Available online: https://www.fda.gov/drugs/fda-grants-accelerated-approval-rucaparib-brca-mutated-metastatic-castration-resistant-prostate (accessed on 27 June 2020).
  52. Smith, M.R.; Sandhu, S.K.; Kelly, W.K.; Scher, H.I.; Efstathiou, E.; Lara, P.N.; Yu, E.Y.; George, D.J.; Chi, K.N.; Saad, F.; et al. Pre-specified interim analysis of GALAHAD: A phase II study of niraparib in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) and biallelic DNA-repair gene defects (DRD). Ann. Oncol. 2019, 30, v884–v885. [Google Scholar] [CrossRef]
  53. Johnson & Johnson. Janssen Announces U.S. FDA Breakthrough Therapy Designation Granted for Niraparib for the Treatment of Metastatic Castration-Resistant Prostate Cancer. Available online: https://www.jnj.com/janssen-announces-u-s-fda-breakthrough-therapy-designation-granted-for-niraparib-for-the-treatment-of-metastatic-castration-resistant-prostate-cancer#_edn1 (accessed on 27 June 2020).
  54. De Bono, J.S.; Mehra, N.; Higano, C.S.; Saad, F.; Buttigliero, C.; Mata, M.; Chen, H.-C.; Healy, C.G.; Paccagnella, M.L.; Czibere, A. TALAPRO-1: A phase II study of talazoparib (TALA) in men with DNA damage repair mutations (DDRmut) and metastatic castration-resistant prostate cancer (mCRPC)—First interim analysis (IA). J. Clin. Oncol. 2020, 38 suppl. 119. [Google Scholar] [CrossRef]
  55. Ganguly, B.; Dolfi, S.C.; Rodriguez-Rodriguez, L.; Ganesan, S.; Hirshfield, K.M. Role of Biomarkers in the Development of PARP Inhibitors. Biomark. Cancer 2016, 8, 15–25. [Google Scholar] [CrossRef] [PubMed]
  56. Jonsson, P.; Bandlamudi, C.; Cheng, M.L.; Srinivasan, P.; Chavan, S.S.; Friedman, N.D.; Rosen, E.Y.; Richards, A.L.; Bouvier, N.; Selcuklu, S.D.; et al. Tumour lineage shapes BRCA-mediated phenotypes. Nature 2019, 571, 576–579. [Google Scholar] [CrossRef] [PubMed]
  57. Curtin, N.J.; Drew, Y.; Sharma-Saha, S. Why BRCA mutations are not tumour-agnostic biomarkers for PARP inhibitor therapy. Nat. Rev. Clin. Oncol. 2019, 16, 725–726. [Google Scholar] [CrossRef]
  58. Wu, Y.M.; Cieślik, M.; Lonigro, R.J.; Vats, P.; Reimers, M.A.; Cao, X.; Ning, Y.; Wang, L.; Kunju, L.P.; de Sarkar, N.; et al. Inactivation of CDK12 Delineates a Distinct Immunogenic Class of Advanced Prostate Cancer. Cell 2018, 173, 1770–1782.e1714. [Google Scholar] [CrossRef] [Green Version]
  59. Bajrami, I.; Frankum, J.R.; Konde, A.; Miller, R.E.; Rehman, F.L.; Brough, R.; Campbell, J.; Sims, D.; Rafiq, R.; Hooper, S.; et al. Genome-wide profiling of genetic synthetic lethality identifies CDK12 as a novel determinant of PARP1/2 inhibitor sensitivity. Cancer Res. 2014, 74, 287–297. [Google Scholar] [CrossRef] [Green Version]
  60. Abida, W.; Campbell, D.; Patnaik, A.; Shapiro, J.D.; Sautois, B.; Vogelzang, N.J.; Voog, E.G.; Bryce, A.H.; McDermott, R.; Ricci, F.; et al. Non-BRCA DNA Damage Repair Gene Alterations and Response to the PARP Inhibitor Rucaparib in Metastatic Castration-Resistant Prostate Cancer: Analysis From the Phase II TRITON2 Study. Clin. Cancer Res. 2020. [Google Scholar] [CrossRef] [Green Version]
  61. Antonarakis, E.S.; Isaacsson Velho, P.; Fu, W.; Wang, H.; Agarwal, N.; Santos, V.S.; Maughan, B.L.; Pili, R.; Adra, N.; Sternberg, C.N. CDK12-altered prostate cancer: Clinical features and therapeutic outcomes to standard systemic therapies, poly (ADP-ribose) polymerase inhibitors, and PD-1 inhibitors. JCO Precis. Oncol. 2020, 4, 370–381. [Google Scholar] [CrossRef]
  62. Shiloh, Y.; Ziv, Y. The ATM protein kinase: Regulating the cellular response to genotoxic stress, and more. Nat. Rev. Mol. Cell Biol. 2013, 14, 197–210. [Google Scholar] [CrossRef]
  63. Kubota, E.; Williamson, C.T.; Ye, R.; Elegbede, A.; Peterson, L.; Lees-Miller, S.P.; Bebb, D.G. Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines. Cell Cycle 2014, 13, 2129–2137. [Google Scholar] [CrossRef] [Green Version]
  64. Williamson, C.T.; Kubota, E.; Hamill, J.D.; Klimowicz, A.; Ye, R.; Muzik, H.; Dean, M.; Tu, L.; Gilley, D.; Magliocco, A.M.; et al. Enhanced cytotoxicity of PARP inhibition in mantle cell lymphoma harbouring mutations in both ATM and p53. EMBO Mol. Med. 2012, 4, 515–527. [Google Scholar] [CrossRef] [PubMed]
  65. Jette, N.R.; Radhamani, S.; Arthur, G.; Ye, R.; Goutam, S.; Bolyos, A.; Petersen, L.F.; Bose, P.; Bebb, D.G.; Lees-Miller, S.P. Combined poly-ADP ribose polymerase and ataxia-telangiectasia mutated/Rad3-related inhibition targets ataxia-telangiectasia mutated-deficient lung cancer cells. Br. J. Cancer 2019, 121, 600–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Jette, N.R.; Radhamani, S.; Ye, R.; Yu, Y.; Arthur, G.; Goutam, S.; Bismar, T.A.; Kumar, M.; Bose, P.; Yip, S. ATM-deficient lung, prostate and pancreatic cancer cells are acutely sensitive to the combination of olaparib and the ATR inhibitor AZD6738. Genome Instab. Dis. 2020, 1, 197–205. [Google Scholar] [CrossRef] [Green Version]
  67. Rafiei, S.; Fitzpatrick, K.; Liu, D.; Cai, M.Y.; Elmarakeby, H.A.; Park, J.; Ricker, C.; Kochupurakkal, B.S.; Choudhury, A.D.; Hahn, W.C.; et al. ATM Loss Confers Greater Sensitivity to ATR Inhibition than PARP Inhibition in Prostate Cancer. Cancer Res. 2020. [Google Scholar] [CrossRef] [Green Version]
  68. Wilkes, D.C.; Sailer, V.; Xue, H.; Cheng, H.; Collins, C.C.; Gleave, M.; Wang, Y.; Demichelis, F.; Beltran, H.; Rubin, M.A.; et al. A germline FANCA alteration that is associated with increased sensitivity to DNA damaging agents. Cold Spring Harb. Mol. Case Stud. 2017, 3. [Google Scholar] [CrossRef] [Green Version]
  69. Min, A.; Im, S.A.; Yoon, Y.K.; Song, S.H.; Nam, H.J.; Hur, H.S.; Kim, H.P.; Lee, K.H.; Han, S.W.; Oh, D.Y.; et al. RAD51C-deficient cancer cells are highly sensitive to the PARP inhibitor olaparib. Mol. Cancer Ther. 2013, 12, 865–877. [Google Scholar] [CrossRef] [Green Version]
  70. Koppensteiner, R.; Samartzis, E.P.; Noske, A.; von Teichman, A.; Dedes, I.; Gwerder, M.; Imesch, P.; Ikenberg, K.; Moch, H.; Fink, D.; et al. Effect of MRE11 loss on PARP-inhibitor sensitivity in endometrial cancer in vitro. PLoS ONE 2014, 9, e100041. [Google Scholar] [CrossRef]
  71. Bryce, A.H.; Sartor, O.; de Bono, J. DNA Repair and Prostate Cancer: A Field Ripe for Harvest. Eur. Urol. 2020. [Google Scholar] [CrossRef]
  72. Li, H.; Liu, Z.Y.; Wu, N.; Chen, Y.C.; Cheng, Q.; Wang, J. PARP inhibitor resistance: The underlying mechanisms and clinical implications. Mol. Cancer 2020, 19, 107. [Google Scholar] [CrossRef]
  73. Edwards, S.L.; Brough, R.; Lord, C.J.; Natrajan, R.; Vatcheva, R.; Levine, D.A.; Boyd, J.; Reis-Filho, J.S.; Ashworth, A. Resistance to therapy caused by intragenic deletion in BRCA2. Nature 2008, 451, 1111–1115. [Google Scholar] [CrossRef]
  74. Quigley, D.; Alumkal, J.J.; Wyatt, A.W.; Kothari, V.; Foye, A.; Lloyd, P.; Aggarwal, R.; Kim, W.; Lu, E.; Schwartzman, J.; et al. Analysis of Circulating Cell-Free DNA Identifies Multiclonal Heterogeneity of BRCA2 Reversion Mutations Associated with Resistance to PARP Inhibitors. Cancer Discov. 2017, 7, 999–1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Active, recruiting, and planned trials involving castration resistant metastatic prostate cancer and PARP inhibitors as monotherapy.
Table 1. Active, recruiting, and planned trials involving castration resistant metastatic prostate cancer and PARP inhibitors as monotherapy.
Trial NumberNotable CharacteristicsPhaseInterventionPrimary Outcome Measures
Olaparib
NCT03263650Aggressive variant prostate cancer
Prior cabazitaxel, carboplatin, and prednisone
IIOlaparib PFS
NCT03434158mCRPC with HRR defects
Prior docetaxel
IIOlaparibRadiographic PFS
NCT02987543
(PROfound)
mCRPC with HRR defects
Prior abiraterone or enzalutamide
IIIOlaparib versus enzalutamide or abirateroneChange in radiographic PFS
Rucaparib
NCT02952534 (TRITON2)mCRPC with HRR deficiencyIIRucaparib ORR and PSA response
NCT03442556mCRPC with no prior platinum chemotherapyIIRucaparib maintenance after induction carboplatin and docetaxelRadiographic PFS
NCT04171700Advanced prostate cancer with HRR deficiency besides BRCA1/2IIRucaparibOverall response rate
NCT02975934 (TRITON3)mCRPC with HRR deficiencyIIIRucaparib versus abiraterone, enzalutamide, or docetaxelRadiographic PFS
Niraparib
NCT02854436 (GALAHAD)mCRPC
Prior taxane and androgen receptor-targeted therapy
IINiraparibObjective response rate
NCT04288687mCRPC
Prior platinum-based chemotherapy
IINiraparibRadiographic PFS
Talazoparib
NCT03148795 (TALAPRO-1)mCRPC with HRR deficiency
Prior taxane and novel hormonal therapy
IITalazoparibObjective response rate
Note: HRR: homologous recombination repair; mCRPC: metastatic castration-resistant prostate cancer; PFS: progression-free survival; PSA: prostate-specific antigen.
Table 2. Active, recruiting, and planned trials involving castration resistant metastatic prostate cancer and PARP inhibitors in combination with other agents.
Table 2. Active, recruiting, and planned trials involving castration resistant metastatic prostate cancer and PARP inhibitors in combination with other agents.
Trial NumberNotable CharacteristicsPhaseInterventionPrimary Outcome Measures
Olaparib
NCT02861573mCRPCIOlaparib with pembrolizumab in one cohort; total of four cohorts with all cohorts receiving pembrolizumabPSA response, adverse events, objective response rate
NCT03205176mCRPCIOlaparib with AZD513 (reversible BRD4 inhibitor)Dose-limiting toxicity
NCT03874884mCRPC
Prior abiraterone and/or enzalutamide
IOlaparib with 177Lutetium-prostate- specific membrane antigenDose-limiting toxicity, maximum-tolerated dose
NCT02484404mCRPC
Prior enzalutamide/abiraterone or chemotherapy containing docetaxel
I/IIOlaparib with cediranib and MEDI4736 (PD-L1 inhibitor) in different combinationsSafety, overall response rate
NCT02769962mCRPC
Prior enzalutamide or abiraterone or chemotherapy containing docetaxel
I/IIOlaparib with camptothecinOverall response rate
NCT03317392mCRPC with metastases to the boneI/IIOlaparib with radium Ra 223 dichlorideMaximum tolerated dose and radiographic PFS
NCT04556617mCRPC with homologous recombination repair defectsI/IIOlaparib with PLX2853 (BRD4 inhibitor)Disease response, dose-limiting toxicities, treatment emergent adverse events
NCT01972217mCRPC
Prior chemotherapy containing docetaxel
IIOlaparib with abirateroneSafety and tolerability, median radiographic PFS
NCT02893917mCRPCPrevious taxane therapyIIOlaparib with cediranib versus olaparib aloneRadiographic PFS
NCT03012321mCRPC with DNA damage repair defects and no prior chemotherapy or new hormonal agentsIIAbiraterone versus olaparib versus abiraterone with olaparibObjective PFS
NCT03516812mCRPC
Prior abiraterone and/or enzalutamide
IIOlaparib with testosteronePSA response
NCT03787680mCRPCIIOlaparib with AZD6738 (ATR inhibitor)Change in radiographic response or PSA
NCT03732820mCRPC with no prior cytotoxic chemotherapy or new hormonal agentsIIIOlaparib with abiraterone versus placebo with abirateroneRadiographic PFS
NCT03834519mCRPC
Prior abiraterone or enzalutamide, and docetaxel
IIIOlaparib with pembrolizumab versus abiraterone versus enzalutamideOverall survival, radiographic PFS
Rucaparib
NCT04179396mCRPCIRucaparib with enzalutamide or rucaparib with abirateronePharmacokinetics, adverse events
NCT03338790mCRPCI/IIRucaparib with nivolumab in one cohort; total of three cohorts all receiving nivolumabORR, PSA response rate
NCT03572478mCRPC
Prior abiraterone or enzalutamide
I/IIRucaparib with nivolumab, in combination and as monotherapiesDose-limiting toxicity, T cell inflammation in the tumor
NCT03840200mCRPC
Prior second-generation androgen receptor targeted therapy
I/IIRucaparib with ipatasertibPSA response, dose-limiting toxicity, maximum-tolerated dose, adverse events
NCT04253262mCRPC
Prior abiraterone, enzalutamide, and/or apalutamide
I/IIRucaparib with copanlisibMaximum-tolerated dose, response
NCT04455750mCRPC with no prior therapy while in mCRPC stateIIIRucaparib with enzalutamideRadiographic PFS, overall survival
Niraparib
NCT03076203mCRPC
At least one prior line androgen receptor-targeted therapy or androgen biosynthesis inhibitor
INiraparib with radium Ra 223 dichlorideMaximum-tolerated dose
NCT03431350mCRPC
One or two previous lines of novel androgen receptor-targeted therapy
I/IINiraparib with cetrelimab or with abirateroneToxicity, objective response rate, adverse events, pharmacokinetics
NCT03748641mCRPC with no prior systemic therapy in the mCRPC settingIIINiraparib with abiraterone versus placebo with abirateroneRadiographic PFS
Talazoparib
NCT04019327mCRPC
Progression on at least one second generation hormonal agent
I/IITalazoparib with temozolomideAdverse events, overall response rates
NCT03330405mCRPC with BRCA or ATM gene defectI/IITalazoparib with avelumabDose-limiting toxicity, overall response
NCT04052204mCRPC with DNA damage response defectsI/IITalazoparib with avelumab and bempegaldesleukinDose-limiting toxicity, soft tissue response
NCT03395197
(TALAPRO-2)
mCRPC without prior systemic treatmentIIITalazoparib with enzalutamide versus placebo with enzalutamideRadiographic PFS
Veliparib
NCT01576172mCRPC
Up to two prior chemotherapy regimens
IIVeliparib with abiraterone versus abiraterone alonePSA response
Note: mCRPC: metastatic castration-resistant prostate cancer; PARP: poly (ADP-ribose) polymerase; PFS: progression-free survival; PSA: prostate-specific antigen.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jang, A.; Sartor, O.; Barata, P.C.; Paller, C.J. Therapeutic Potential of PARP Inhibitors in the Treatment of Metastatic Castration-Resistant Prostate Cancer. Cancers 2020, 12, 3467. https://doi.org/10.3390/cancers12113467

AMA Style

Jang A, Sartor O, Barata PC, Paller CJ. Therapeutic Potential of PARP Inhibitors in the Treatment of Metastatic Castration-Resistant Prostate Cancer. Cancers. 2020; 12(11):3467. https://doi.org/10.3390/cancers12113467

Chicago/Turabian Style

Jang, Albert, Oliver Sartor, Pedro C. Barata, and Channing J. Paller. 2020. "Therapeutic Potential of PARP Inhibitors in the Treatment of Metastatic Castration-Resistant Prostate Cancer" Cancers 12, no. 11: 3467. https://doi.org/10.3390/cancers12113467

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop