Targeting of PARP enzymes has emerged as an effective therapeutic strategy to selectively target cancer cells with deficiencies in homologous recombination signaling. Currently used to treat BRCA-mutated cancers, PARP inhibitors (PARPi) have demonstrated improved outcome in various cancer types as single agents. Ongoing efforts have seen the exploitation of PARPi combination therapies, boosting patient responses as a result of drug synergisms. Despite great successes using PARPi therapy, selecting those patients who will benefit from single agent or combination therapy remains one of the major challenges. Numerous reports have demonstrated that the presence of a BRCA mutation does not always result in synthetic lethality with PARPi therapy in treatment-naïve tumors. Cancer cells can also develop resistance to PARPi therapy. Hence, combination therapy may significantly affect the treatment outcomes. In this review, we discuss the development and utilization of PARPi in different cancer types from preclinical models to clinical trials, provide a current overview of the potential uses of PARP imaging agents in cancer therapy, and discuss the use of radiolabeled PARPi as radionuclide therapies.

Targeting DNA damage repair (DDR) signaling is a fast-expanding field for cancer therapy. DNA damage causes genomic instability in cells that require DDR for rescue. DDR signaling triggers activation of repair protein transcription, hence leading to overexpression of the related repair protein and activation of mechanisms for DNA repair (1). PARP is an essential and abundant DNA repair protein, which plays an important role in regulation of various DDR pathways, including base excision repair, homologous recombination (HR), classical and alternative nonhomologous end joining, nucleotide excision repair, microhomology-mediated end joining, maintenance of replication fork stability, and mismatch repair (2). In addition, PARP has been found overexpressed in various types of cancers, such as breast, ovarian, and oral cancers than their normal surrounding healthy tissues, making inhibition of PARP activity a promising strategy for cancer therapeutics by disrupting PARP functions, thereby impairing DDR pathways of cancer cells. PARP inhibition for the treatment of breast or ovarian cancers with BRAC1/2 mutations (deficiencies in the HR) is now a well-established approach. A PARP inhibitor (PARPi) is utilized to impair the process of single-strand break (SSB) repair, thereby converting SSB into double-strand breaks by stalling replication folk (1) or accelerating fork elongation (3), which requiring HR for repair during S phase (4). Therefore, if HR is defective for example, resulting from mutations in HR genes such as BRCA1 and 2, DSBs cannot be repaired, and the cell dies, a concept known as synthetic lethality. Over the past decade, four PARPi (olaparib, rucaparib, niraparib, talazoparib) have been FDA approved for clinical use as single agents, with veliparib expected to receive approval following promising phase III trial results (BROCADE3; NCT02163694). The ability of PARPi to impair DDR processes led to their employment in combination with chemotherapy and radiotherapy. Increasing attention is also being drawn to radiolabeled versions of PARPi that possibly allow direct visualization of PARP expression, quantification of PARPi biodistribution and tumor delivery, as well as target engagement, thereby potentially improve stratification of patients and enable the monitoring of treatment progress. In addition, radiolabeled PARPi-based radionuclide therapy agents are being proposed as therapies for PARP-expressing cancers. Here, we present an up-to-date understanding of PARP in DDR, the use of PARPis as mono- or combination therapies for cancer, and the potential of radiolabeled PARPi in clinical applications.

PARP enzymes form a 17-member group of mostly nuclear enzymes that share a conserved catalytic domain homologous to ADP-ribosyl-transferases diphtheria toxin (ARTD), which gives the PARP enzyme family its alternative name of ARTD-like enzymes (5). Upon DNA damage, members of the PARP family bind the DNA lesion, become catalytically active, and mediate ADP-ribosylation using NAD+ as substrate, leading to rapid recruitment of other DNA repair factors (6). PARP-1, PARP-2 and tankyrase (PARP-5A/5B) produce poly-ADP-ribose (PAR) modifications on target proteins, while other PARPs are responsible for generating mono-ADP-ribose and have potential cancer-related functions (7). The evidence to date suggests that only PARP-1, PARP-2, and PARP-3 are activated by DNA breaks and catalyze ADP-ribosylation after DNA damage, with PARP-1 responsible for 80%–90% of global PAR synthesis in response to DNA damaging agents (6). Apart from SSB recognition, recent studies showed that PARP-1 can also detect DSB rapidly, and plays an essential role in regulation of DNA end resection for DSB repair, with recruitment and activation of PARP-1 occurring within 0.1 μs after induction of DSBs (6).

PARP-1 is the most studied of the PARP enzyme family by far. Its structure comprises of three zinc finger domains for DNA binding, a BRCA-C-terminus domain (BRCT), a WGR (Trp-Gly-Arg) domain that contributes to, but is not required for, DNA interaction and catalytic activity regulation in response to DNA damage, and a catalytic domain consisting of a helical subdomain (HD) adjacent to a conserved ADP-ribosyl transferase fold (ART; ref. 8). Compared with PARP-1 (N-terminal region: >500 residues), PARP-2, and PARP-3 have more compact structures, with shorter N-terminal extensions (70 and 40 residues, respectively) from the WGR domain (Fig. 1A; ref. 9). PARP-1 requires the zinc finger domains for DNA break detection and binding (10), but PARP-2 and PARP-3 rely on their WGR domain for this function (2, 11).

Figure 1.

Allosteric regulation of PARP-1 and effects on PARP-1 allostery by PARPi. A, Schematic representation of multidomain organization of human PARP-1, PARP-2, and PARP-3. Zinc fingers (Zn1, Zn2, and Zn3); BRCT (BRCA C-terminus); WGR (Trp-Gly-Arg domain); HD (helical domain); ART (ADP-ribosyltransferase). B, Overall mechanism and allosteric regulation of PARP-1 (13). Upon DNA damage, the multidomains of PARP-1 quickly assemble on the DNA-damaged site. The binding of NAD+ to the catalytic domain promotes PARylation to produce PAR chains for the recruitment of target proteins (such as DNA ligase, XRCC1, etc.). Auto-PARylation leads to dissociation of PARP-1 from the DNA to allow binding of other repair proteins to the DNA; the HD domain regulates NAD+ access and catalytic activity of PARP-1 through a reversible substrate-blocking mechanism. In the absence of damaged DNA, the HD is in folded conformation and sterically blocks the substrate-binding site [the nicotinamide site (N) and the adenosine site (A)] to restrict the access of NAD+. The assembly of DNA-binding domains of PARP-1 on the DNA break interfaces with the HD domain, which promotes an unfolded HD conformation to restore access of NAD+ to the catalytic active site. The NAD+ binding to the catalytic site drives the HD equilibrium distribution to its unfolded state (purple arrow), thus strengthening the interaction between the regulatory domains and the DNA break. C, Impacts on PARP-1 allostery in DDR by PARPi binding (19). Three distinct types of PARPi based on their allosteric effects on the PARP-1/SSB DNA complex: Type I PARPi influence PARP-1 allostery and retain PARP-1 on DNA (in green), whereas type III PARPi perturb PARP-1 allostery and release PARP-1 from DNA (in red). Type II PARPi do not influence PARP-1 allostery (in black).

Figure 1.

Allosteric regulation of PARP-1 and effects on PARP-1 allostery by PARPi. A, Schematic representation of multidomain organization of human PARP-1, PARP-2, and PARP-3. Zinc fingers (Zn1, Zn2, and Zn3); BRCT (BRCA C-terminus); WGR (Trp-Gly-Arg domain); HD (helical domain); ART (ADP-ribosyltransferase). B, Overall mechanism and allosteric regulation of PARP-1 (13). Upon DNA damage, the multidomains of PARP-1 quickly assemble on the DNA-damaged site. The binding of NAD+ to the catalytic domain promotes PARylation to produce PAR chains for the recruitment of target proteins (such as DNA ligase, XRCC1, etc.). Auto-PARylation leads to dissociation of PARP-1 from the DNA to allow binding of other repair proteins to the DNA; the HD domain regulates NAD+ access and catalytic activity of PARP-1 through a reversible substrate-blocking mechanism. In the absence of damaged DNA, the HD is in folded conformation and sterically blocks the substrate-binding site [the nicotinamide site (N) and the adenosine site (A)] to restrict the access of NAD+. The assembly of DNA-binding domains of PARP-1 on the DNA break interfaces with the HD domain, which promotes an unfolded HD conformation to restore access of NAD+ to the catalytic active site. The NAD+ binding to the catalytic site drives the HD equilibrium distribution to its unfolded state (purple arrow), thus strengthening the interaction between the regulatory domains and the DNA break. C, Impacts on PARP-1 allostery in DDR by PARPi binding (19). Three distinct types of PARPi based on their allosteric effects on the PARP-1/SSB DNA complex: Type I PARPi influence PARP-1 allostery and retain PARP-1 on DNA (in green), whereas type III PARPi perturb PARP-1 allostery and release PARP-1 from DNA (in red). Type II PARPi do not influence PARP-1 allostery (in black).

Close modal

Despite differences in DNA damage detection, PARP-1, PARP-2, and PARP-3 share allosteric regulatory activation mechanisms, where binding to damaged DNA leads to an increase of PARylation activity (12). Langelier and colleagues. demonstrated that the HD domain in PARP-1 can selectively and reversibly block or allow access of NAD+ or the NAD analog BAD upon DNA binding (13). The HD domain, in its native state, was found primarily in a folded conformation, sterically blocking the catalytic binding site. Upon DNA damage detection, the binding of NAD+ to the catalytic active site stabilizes the HD domain in its unfolded conformation and strengthens the interaction between the regulatory domains and DNA (Fig. 1B). This reversible blockage of the NAD+ binding site provides control of NAD+ access to the catalytic site, regulating the PARylation process.

Clinical PARPis compete for binding in the NAD+ binding pocket of the PARP enzymes, and are mostly selective for PARP-1 and PARP-2. However, different PARP inhibitors display markedly different levels of cytotoxicity, attributed to the ability of PARP inhibitors to prevent auto-PARylation of PARP, thereby preventing its dissociation from the broken DNA, thus stabilizing and trapping PARP on the DNA (14). This trapping leads to DSB formation by stalling and collapsing replication forks (15) or accelerating fork elongation (3). Hence, different degrees of trapping (talazoparib > niraparib ≥ olaparib = rucaparib > veliparib (16, 17)) lead to large differences in cellular toxicity. Although the trapping abilities of PARPi are dependent on catalytic inhibition and the resulting dissociation rates of PARPi (17, 18), reverse allostery effects also play a significant role. Molecular impacts of PARPi binding to PARP-1 corroborated by molecular dynamics simulation, hydrogen-exchange mass spectrometry and X-ray crystallography implied that different PARPi induce diverse degrees of structural deviations in the regulatory domains, thereby positively or negatively affecting the retention of PARP-1 trapped at the DNA damage site (19, 20). Three types of PARPi were discerned on the basis of their allosteric impacts on the PARP-1–DNA complex (19): type I, allosteric proretention on DNA, which induces HD conformational changes that reinforce contacts with the DNA break and result in slow release; type II, with no or mild allosteric effects; and type III, allosteric prorelease from DNA, which induce HD conformational changes opposite to type I inhibitors (Fig. 1C). These results have illuminated that the PARP-1 trapping induced by some PARPi, such as type II PARPi (talazoparib and olaparib) and type III PARPi (rucaparib and niraparib), does not rely on the allosteric effect, indicating that other factors may contribute to the trapping ability of these PARPi, and therefore further investigation is warranted.

PARPi as Monotherapy

The efficacy of PARPi monotherapy in HR deficient (HRD) cancers carrying BRCA1/2 mutations was first demonstrated in cancer cell lines, and later confirmed in patient-derived xenograft (PDX) models. Treatment with olaparib resulted in significant tumor graft regression (21, 22) in PDX tumors of BRCA1-mutant triple-negative breast cancer (TNBC). Other alterations, such as absence of PTEN or ATM, also cause sensitivity of TNBC (23) and small-cell lung cancer (SCLC; ref. 24) to talazoparib treatment in vivo, confirming the effectiveness of PARPi beyond germline BRCA1/2 mutation cancers.

A wealth of clinical data supports the use of PARPi for maintenance treatment in patients with platinum-sensitive high-grade serous ovarian cancer (HGSOC). In the randomized, placebo-controlled, double-blind phase III SOLO2 trial, patients with relapsed epithelial ovarian, fallopian tube, or primary peritoneal cancer carrying a BRCA1/2 mutation demonstrated a significantly longer median progression-free survival (PFS) under olaparib maintenance treatment than those receiving placebo (19.1 vs. 5.5 months; 95% confidence interval, 16.3–25.7 vs. 5.2–5.8; HR 0.3), without significant detrimental effects on health-related quality of life (HRQoL; refs. 25, 26).

The antitumor activity of olaparib was highlighted in the recent phase III SOLO3 (NCT02282020) and OlympiAD (27) trials, in germline BRCA-mutated ovarian cancer and HER2 breast cancer, respectively. Similarly, the ABRAZO study (28), and the latest EMBRACA trial, where patients underwent talazoparib monotherapy, showed a significant PFS advantage and higher objective response rate (ORR) over standard chemotherapy (8.6 vs. 5.6 months, 62.6% vs. 27.2%, respectively) with greater HRQoL in favor of talazoparib (29). Several recently reported trials including POLO and PROfound also demonstrated the benefits of PARPi (olaparib or rucaparib) over placebo or androgen receptor (AR)-directed therapy, respectively, in patients with metastatic pancreatic ductal adenocarcinoma (PDAC) harboring germline BRCA mutation (30) and in patients with castration-resistant prostate cancer (mCRPC) with confirmed HRD phenotype (31). Ongoing efforts to expand the use of PARPi to a wider population of patients with cancer in different settings beyond BRCA mutation will undoubtedly expand the potential use of PARPi as monotherapy, and several studies are currently underway (NCT03402841, NCT03286842, NCT02000622).

Heterogeneous responses to single-agent PARPi are frequently observed in both preclinical models and clinical practice, even in BRCA-mutated tumors, associated with complex intrinsic or acquired resistance. The mechanisms of PARPi resistance are well described in several recent reviews, with the majority related to restoration of HR capability (32). Rational combination treatment regimens thus become the next strategy to improve patient outcome, for example, using PARPi with chemotherapy, radiation, targeted drugs, or immunotherapy. The choice of partner must be based on the fundamental insights into the molecular pathways that drive tumorigenesis and therapy response. Strategies to optimize the use of PARPi as chemopotentiators or radiosensitizers are being actively pursued to overcome or prevent the development of treatment resistance in patients.

One approach is the combination of genotoxic agents and PARPi, where chemotherapy-induced DNA damage can increase the cellular reliance on DNA repair and hence sensitize tumors to PARPi, such as in ref. 33. Combination of PARPi and ionizing radiation (IR) is another effective strategy to sensitize tumors to radiotherapy, without overt signs of toxicity at least in mice, warranting further its evaluation in ongoing clinical trials (34), despite an early recognition of the limitation of this combinatorial approach in cells (35). Given the relationship between PARP and HR repair (HRR) regulatory pathways, it is not surprising to see the potential benefits of combination treatments between PARPi and DDR signaling targeting or otherwise genotoxic therapies. The increased reliance of cancer cells on alternative or specific DRR pathways provides an opportunity to pharmacologically induce HR deficiency, which in combination with PARPi achieves synergistic response. Disruption of, for example, checkpoint kinase 1 (CHK1) activity with PARP inhibition leads to the accumulation of DSB and induces apoptosis following treatment of PDX mouse models using olaparib/MK8776 (36) or olaparib/prexasertib (37) combinations. Tretinoin, used to treat acute promyelocytic leukemia patients, also acts as an effective HR disrupter and is capable of sensitizing BRCA-proficient TNBC to olaparib (38). Other studies involved the use of cyclic-dependent kinase (CDK) inhibitors to disrupt DNA damage checkpoint signaling and prevent DNA repair in neuroendocrine prostate cancer (NEPC; ref. 39) and TNBC PDX models (40). In addition, inhibition of bromodomain-containing protein (BRD4) disrupts DDR signaling by reducing DNA repair proteins levels, thereby sensitizing cancer cells to olaparib in preclinical models (41, 42). Treatment of mCRPC PDXs with an AR inhibitor enzalutamide induces downregulation of AR-regulated HRR gene expression and sensitizes to PARPi (43). An overview is presented in Table 1.

Table 1.

Combination therapies with PARPis in PDX models.

Combination therapyAdditional targetTreatmentCancer typePublication
Genotoxic  BCL2 Olaparib + dacarbazine Uveal melanoma (74) 
   Olaparib + navitoclax Ewing sarcoma (75) 
  N/A Olaparib + chloroquine Ovarian (76) 
   Talazoparib + temozolomide SCLC (33) 
   Olaparib + trabectedin Ewing sarcoma (77) 
  N/A Olaparib + cisplatin Breast (78) 
   Niraparib + carboplatin + paclitaxel HGSOC (79) 
IR  N/A Olaparib + IR PDAC (80) 
   Olaparib + IR Renal cell carcinoma (81) 
   Talazoparib + IR SCLC (82) 
   Veliparib + IR Glioblastoma (83) 
Targeted DDR ATM Olaparib + AZD0156 Breast (78) 
  ATR Olaparib + AZD6738 or MK8776 HGSOC (36) 
  CHK1 Olaparib + prexasertib HGSOC (37) 
  CHK2 Talazoparib + prexasertib Gastric (84) 
  CDK2/CDK5 Olaparib + dinaciclib NEPC (39) 
  CDK12 Veliparib + dinaciclib TNBC (40) 
  LMO2 Olaparib + R-CHOP DLBCL (85) 
  PIN1 Olaparib + tretinoin TNBC (38) 
  DNA-PK Olaparib + AZD7658 Ovarian (86) 
   Olaparib + AZD7659 TNBC  
   Olaparib + AZD7660 NSCLC  
   Olaparib + AZD7661 Head and neck  
 Antioncogenic BRD4 Olaparib + AZD5153 PDAC (41) 
  BRD5 Olaparib + JQ1 PDAC (42) 
  BRD6 Talazoparib + JQ1 Breast (41) 
  MYCN Olaparib + danusertib NEPC (87) 
  AR Olaparib + enzalutamide CRPC (43) 
Combination therapyAdditional targetTreatmentCancer typePublication
Genotoxic  BCL2 Olaparib + dacarbazine Uveal melanoma (74) 
   Olaparib + navitoclax Ewing sarcoma (75) 
  N/A Olaparib + chloroquine Ovarian (76) 
   Talazoparib + temozolomide SCLC (33) 
   Olaparib + trabectedin Ewing sarcoma (77) 
  N/A Olaparib + cisplatin Breast (78) 
   Niraparib + carboplatin + paclitaxel HGSOC (79) 
IR  N/A Olaparib + IR PDAC (80) 
   Olaparib + IR Renal cell carcinoma (81) 
   Talazoparib + IR SCLC (82) 
   Veliparib + IR Glioblastoma (83) 
Targeted DDR ATM Olaparib + AZD0156 Breast (78) 
  ATR Olaparib + AZD6738 or MK8776 HGSOC (36) 
  CHK1 Olaparib + prexasertib HGSOC (37) 
  CHK2 Talazoparib + prexasertib Gastric (84) 
  CDK2/CDK5 Olaparib + dinaciclib NEPC (39) 
  CDK12 Veliparib + dinaciclib TNBC (40) 
  LMO2 Olaparib + R-CHOP DLBCL (85) 
  PIN1 Olaparib + tretinoin TNBC (38) 
  DNA-PK Olaparib + AZD7658 Ovarian (86) 
   Olaparib + AZD7659 TNBC  
   Olaparib + AZD7660 NSCLC  
   Olaparib + AZD7661 Head and neck  
 Antioncogenic BRD4 Olaparib + AZD5153 PDAC (41) 
  BRD5 Olaparib + JQ1 PDAC (42) 
  BRD6 Talazoparib + JQ1 Breast (41) 
  MYCN Olaparib + danusertib NEPC (87) 
  AR Olaparib + enzalutamide CRPC (43) 

Promising efficacy of PARPi with genotoxic agents was observed in preclinical models and successfully translated in various cancer types: The phase III VELIA study demonstrated the efficacy of concurrent administration of veliparib and carboplatin with paclitaxel (V/CP) followed by veliparib maintenance in advanced patients with HGSOC with and without germline BRCA mutation. A significant PFS advantage was observed in both BRCA-mutant (34.7 vs. 22 months, HR 0.44), HR defects cohorts (31.9 vs. 20.5 months), and the intention-to-treat population (23.5 vs. 17.3 months, HR 0.68) receiving veliparib versus placebo with chemotherapy (44). Similar efficacy was observed in patients with breast cancer in phase II and III studies (45, 46).

Other clinical studies showed improved patient outcomes for treatment regimens involving PARPi combined with targeted drugs, such as antiangiogenic and AR antagonists. The recent FDA approval of olaparib/bevacizumab combination therapy as a first-line maintenance treatment for patients with HGSOC regardless of their HRD phenotypes based on the PAOLA-1 trial (47) will undoubtedly encourage additional studies to investigate other combination pairs (such as ICON9; NCT03278717, NCT02502266). For example, the use of olaparib/cediranib showed appreciable benefits in patients with ovarian cancer (48). Other promising studies include the combination of abiraterone and olaparib in patients with mCRPC, irrespective of HRR status (49). Combination of immune checkpoint inhibitors with PARPi is under active investigation, with only a limited amount of clinical data available to date. The MEDIOLA trial showed initial promise in a subset of patients with ovarian cancer (platinum sensitive, germline BRCA-mutated) receiving this combination (50), but no improvement in ORR or PFS when a more heterogeneous patient population (without BRCA mutation) was considered (51). Notwithstanding, the preliminary outcomes supported the tolerability of this combination in patients and better patient selections in subsequent trials should help to create clinically meaningful responses (NCT02484404). An overview is presented in Table 2.

Table 2.

Selected clinical trials evaluating PARPis combinations.

TreatmentPhaseTrial nameStudy populationHRD testingNCTStatusPublication
Olaparib + carboplatin + paclitaxel II N/A High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma, irrespective of gBRCA1/2 status N/A NCT01081951 Active, not recruiting (88) 
Veliparib + carboplatin + paclitaxel III VELIA/GOG-3005 High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma stage III–IV, irrespective of gBRCA1/2 status BRACAnalysis or myChoice assay (Myriad Genetics), HRD score ≥ 33 NCT02470585 Active, not recruiting (44) 
Veliparib + carboplatin + paclitaxel II BROCADE HER2+ locally recurrent or metastatic breast cancer with gBRCA1/2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT01506609 Active, not recruiting (46) 
Veliparib + carboplatin + paclitaxel III BROCADE3 HER2 locally advanced or metastatic breast cancer with gBRCA1/2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT02163694 Active, not recruiting N/A 
Veliparib + carboplatin + paclitaxel III BrighTNess TNBC stage II–III (T1N1–2 or T2–4N0–2), irrespective of gBRCA1/2 status BRACAnalysis assay (Myriad Genetics), HRD score undetermined NCT02032277 Active, not recruiting (45) 
Veliparib + temozolomide II N/A SCLC sensitive or refractory to platinum-based chemotherapy, irrespective of gBRCA1/2 status N/A NCT01638546 Completed (89) 
Veliparib + cisplatin + etoposide II N/A SCLC with presence of extrathoracic metastatic disease, malignant pleural effusion, and bilateral or contralateral supraclavicular adenopathy, irrespective of gBRCA1/2 status N/A NCT01642251 Completed (90) 
Veliparib + cisplatin + gemcitabine II N/A Locally advanced and metastatic PDAC stage III–IV, with gBRCA1/2 and PALB2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT01585805 Active, not recruiting N/A 
Veliparib + FOLFIRI II N/A Locally advanced and metastatic PDAC stage IV, irrespective of gBRCA1/2 status N/A NCT02890355 Active, not recruiting N/A 
Olaparib + bevacizumab III PAOLA-1 High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma, irrespective of gBRCA1/2 status myChoice assay (Myriad Genetics), HRD score ≥ 42 NCT02477644 Active, not recruiting (47) 
Olaparib + cediranib I/II N/A High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma and TNBC, with gBRCA1/2 mutation BRACAnalysis assay (Myriad Genetics), HRD score undetermined NCT01116648 Active, not recruiting (48) 
Olaparib + abiraterone II N/A mCRPC, irrespective of gBRCA1/2 status N/A NCT01972217 Active, not recruiting (49) 
Olaparib + durvalumab + bevacizumab I/II MEDIOLA Advanced or metastatic solid tumor, irrespective of gBRCA1/2 status BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT02734004 Active, not recruiting (50) 
Olaparib + durvalumab + bevacizumab I/II N/A Advanced solid tumor or recurrent ovarian, triple-negative breast, lung, prostate, and colorectal cancers with gBRCA1/2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT02484404 Recruiting (51) 
TreatmentPhaseTrial nameStudy populationHRD testingNCTStatusPublication
Olaparib + carboplatin + paclitaxel II N/A High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma, irrespective of gBRCA1/2 status N/A NCT01081951 Active, not recruiting (88) 
Veliparib + carboplatin + paclitaxel III VELIA/GOG-3005 High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma stage III–IV, irrespective of gBRCA1/2 status BRACAnalysis or myChoice assay (Myriad Genetics), HRD score ≥ 33 NCT02470585 Active, not recruiting (44) 
Veliparib + carboplatin + paclitaxel II BROCADE HER2+ locally recurrent or metastatic breast cancer with gBRCA1/2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT01506609 Active, not recruiting (46) 
Veliparib + carboplatin + paclitaxel III BROCADE3 HER2 locally advanced or metastatic breast cancer with gBRCA1/2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT02163694 Active, not recruiting N/A 
Veliparib + carboplatin + paclitaxel III BrighTNess TNBC stage II–III (T1N1–2 or T2–4N0–2), irrespective of gBRCA1/2 status BRACAnalysis assay (Myriad Genetics), HRD score undetermined NCT02032277 Active, not recruiting (45) 
Veliparib + temozolomide II N/A SCLC sensitive or refractory to platinum-based chemotherapy, irrespective of gBRCA1/2 status N/A NCT01638546 Completed (89) 
Veliparib + cisplatin + etoposide II N/A SCLC with presence of extrathoracic metastatic disease, malignant pleural effusion, and bilateral or contralateral supraclavicular adenopathy, irrespective of gBRCA1/2 status N/A NCT01642251 Completed (90) 
Veliparib + cisplatin + gemcitabine II N/A Locally advanced and metastatic PDAC stage III–IV, with gBRCA1/2 and PALB2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT01585805 Active, not recruiting N/A 
Veliparib + FOLFIRI II N/A Locally advanced and metastatic PDAC stage IV, irrespective of gBRCA1/2 status N/A NCT02890355 Active, not recruiting N/A 
Olaparib + bevacizumab III PAOLA-1 High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma, irrespective of gBRCA1/2 status myChoice assay (Myriad Genetics), HRD score ≥ 42 NCT02477644 Active, not recruiting (47) 
Olaparib + cediranib I/II N/A High-grade serous epithelial ovarian, fallopian tube, or primary peritoneal carcinoma and TNBC, with gBRCA1/2 mutation BRACAnalysis assay (Myriad Genetics), HRD score undetermined NCT01116648 Active, not recruiting (48) 
Olaparib + abiraterone II N/A mCRPC, irrespective of gBRCA1/2 status N/A NCT01972217 Active, not recruiting (49) 
Olaparib + durvalumab + bevacizumab I/II MEDIOLA Advanced or metastatic solid tumor, irrespective of gBRCA1/2 status BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT02734004 Active, not recruiting (50) 
Olaparib + durvalumab + bevacizumab I/II N/A Advanced solid tumor or recurrent ovarian, triple-negative breast, lung, prostate, and colorectal cancers with gBRCA1/2 mutation BRACAnalysis or myChoice assay (Myriad Genetics), HRD score undetermined NCT02484404 Recruiting (51) 

Molecular imaging using PARP imaging agents could serve as a powerful tool for functional imaging of PARP in vivo. Tagging a PARPi with a radionuclide allows PET (11C, 18F) or SPECT (123I) imaging to potential clinical benefits: interrogation of PARP expression levels, PARPi distribution, tumor uptake, and drug-target engagement in a noninvasive manner, which may allow tumor diagnosis and staging, selection of patient subgroups suitable for PARP inhibition, monitor the treatment response, or gauge the emergence of resistance. If, on the other hand, the same chemical scaffold is radiolabeled with a therapeutic radionuclide, emitting alpha (211At), beta (131I), or Auger (123I) radiation, the resulting radionuclide therapy agent is capable of causing cell death by delivering IR to PARP-expressing tumor cells.

Over the past 15 years, numerous radiolabeled PARP agents have been developed (Fig. 2A) and undergone characterization in vitro and in animal models, with two compounds already evaluated in early-phase clinical trials (34, 52). [11C]PJ34 was the first radiolabeled PARPi, intended for PET imaging of tissue necrosis with upregulation of PARP-1 expression levels in a rat models of diabetes (53). The radiolabeled imaging agents [18F]BO (54), [18F]PARPi (55), [123/131I]PARPi (56, 57), [18F]PARPi-FL (58), and [64Cu]PARPi-DOTA (59), all shared structural similarity to olaparib, and most displayed high specificity toward PARP. Also of note is the development of [18F]olaparib, an 18F-labeled isotopologue of olaparib, with an identical chemical structure as the parent molecule, hence with identical pharmacokinetic and pharmacodynamic profiles in vivo (60). Among the olaparib-derived radiotracers, [18F]PARPi-FL, the dual-modality PARP imaging agent (optical/PET), was used to image drug distribution and drug-target engagement of different PARPis in different cancer models (61, 62), demonstrating the potential application of radiolabeled-PARPi in monitoring of drug distribution and drug-target engagement. Rucaparib-inspired radiolabeled compounds [18F]FE-LS-75 (without publicly available preclinical characterization; ref. 63), [125I]KX1 (64), [125I]KX-02–019 (65), and [18F]FTT (66) also show excellent PARP selectivity.

Figure 2.

Molecular structures and PARP-1 IC50 or inhibition constant (Ki) values of PARP-targeted PET imaging agents, and examples of their clinical studies. A, Molecular structures with radionuclide highlighted in red, and PARP-1 IC50 or inhibition constant (Ki) values of PARP-targeted PET imaging agents. B, Clinical [18F]FTT and [18F]FDG PET/CT images of a patient completed four cycles of carboplatin and paclitaxel (Image reproduced with permission from J Clin Invest, copyright 2018; ref. 66). A patient with omental metastases showed higher uptake of [18F]FTT than [18F]FDG prior (SUVmax: 5.1 vs. 2.0) and after (SUVmax: 7.8 vs. 3.4) chemotherapy. Yellow arrowheads indicate sites of disease. C, A 67-year-old patient with oropharyngeal squamous cell carcinoma (OPSCC) underwent [18F]PARPi PET/CT imaging (Image reproduced from Clinical Cancer Research; ref. 55). Maximum intensity projections at 30, 60, and 120 minutes post [18F]PARPi injection in the patient with OPSCC. At 120 minutes, uptake in the primary tumor was higher than lymph nodes (SUVmax: 4.1 vs. 3.6). Green arrow: primary tumor; Red arrow: metastatic lymph node.

Figure 2.

Molecular structures and PARP-1 IC50 or inhibition constant (Ki) values of PARP-targeted PET imaging agents, and examples of their clinical studies. A, Molecular structures with radionuclide highlighted in red, and PARP-1 IC50 or inhibition constant (Ki) values of PARP-targeted PET imaging agents. B, Clinical [18F]FTT and [18F]FDG PET/CT images of a patient completed four cycles of carboplatin and paclitaxel (Image reproduced with permission from J Clin Invest, copyright 2018; ref. 66). A patient with omental metastases showed higher uptake of [18F]FTT than [18F]FDG prior (SUVmax: 5.1 vs. 2.0) and after (SUVmax: 7.8 vs. 3.4) chemotherapy. Yellow arrowheads indicate sites of disease. C, A 67-year-old patient with oropharyngeal squamous cell carcinoma (OPSCC) underwent [18F]PARPi PET/CT imaging (Image reproduced from Clinical Cancer Research; ref. 55). Maximum intensity projections at 30, 60, and 120 minutes post [18F]PARPi injection in the patient with OPSCC. At 120 minutes, uptake in the primary tumor was higher than lymph nodes (SUVmax: 4.1 vs. 3.6). Green arrow: primary tumor; Red arrow: metastatic lymph node.

Close modal

Moving beyond preclinical models, both [18F]FTT (66, 67) and [18F]PARPi (55, 68) have been evaluated in different types of cancers in clinical practice. [18F]FTT is currently the subject of phase I trials in patients with ovarian cancer (Fig 2B; ref. 66), and also able to measure PARP-1 expression in different subtypes of patients with breast cancer (estrogen receptor positive, HER2+ and triple negative; ref. 67). [18F]PARPi is currently also in phase I trials but for head and neck cancer (Fig. 2C; ref. 55). Furthermore, recent clinical data have shown that the utility of [18F]PARPi can be potentially expanded to image brain cancer in patients for treatment monitoring (68). In addition to applying radiolabeled-PARPi in patients for cancer detection, a fluorescent-labeled PARPi, PARPi-FL, was also subjected to clinical studies for the detection of oral carcinoma, and potentially serve as the surgical-margin assessment of epithelial cancer of the upper intestinal tract (69). These translational studies established the encouraging potential of PARP imaging agents in cancer detection for clinical practice.

Although these agents showed promising tumor uptake, they display different affinity and selectivity for PARP-1 and other isoforms, with a range of IC50 values (2.3–200 nmol/L) as a proxy for PARP affinity (Fig. 2A). Their selectivity toward other PARP family enzymes is not always fully characterized. Combined results indicate that small structural modifications may have significant impact on the target specificity of the compound. The Reiner group showed, for example, that altering the iodination position on the aromatic ring and the length of the linker between the iodinated-aromatic ring and the olaparib core resulted in very different IC50 values for PARP-1 (57). Similarly, recently reported on the effects of structure modification on tumor uptake and target specificity of the olaparib-derived radiolabeled PARPi [18F]olaparib and [18F]AZD2461 (70).

The great potential of radiolabeled PARPi has now also expanded to the use of therapeutic radionuclides: 123I (Auger electron therapy; ref. 71), 131I (high energy beta particles; ref. 72), and 211At (alpha radiation; ref. 73). These PARPi-based radionuclide therapy agents have shown some efficacy in brain cancer and neuroblastoma models with little toxicity to normal tissues, making use of the high ionizing potential and short ranges of these particle emitters for cancer treatment (Fig. 3AC).

Figure 3.

PARP-targeted therapeutic agents and their preclinical evaluations. A,In vivo [123I]MAPi SPECT/CT imaging and [123I]MAPi efficacy (Image reproduced from Clinical Cancer Research; ref. 71). Molecular structure of [123I]MAPi; SPECT/CT images of orthotopic glioblastoma model were taken at 18 hours after injecting with [123I]MAPi (T = tumor, Cl = clearing organs), showing specific tumor uptake of [123I]MAPi; Kaplan–Meier survival curves of vehicle or [123I]MAPi (single injection: 0.37–1.11 MBq) treated mice. P < 0.01. [123I]MAPi-treated mice showed improved survival compared with the vehicle-treated mice; cytology staining and MRI imaging of brain tissues of untreated mouse at week 3, vehicle treated mouse at week 7 and [123I]MAPi treated mouse at 14 week after tumor implantation. B,In vivo therapeutic effect of [131I]PARPi in subcutaneous glioblastoma mouse models (This work was originally published in JNM, © SNMMI; ref. 72). Molecular structure of [131I]PARPi; Tumor growth at week 0 and week 2 of the mice treated with PBS, [127I]PARPi or [131I]PARPi. The treatment with [131I]PARPi suppressed tumor growth P < 0.01. C,In vivo comparison of single and fractional dosing regimens of [211At]MM4 in IMR-05 xenograft models (Image reproduced with permission from Molecular Cancer Therapeutics; ref. 73). Molecular structure of [211At]MM4; tumor growth and Kaplan–Meier curves for IMR-05 tumor-bearing mice treated with single high dose of 1,480 kBq versus a fractionated dose of 370 kBq twice weekly for a cumulative dose of 1,480 kBq; Immunofluorescence staining for PARP-1 in tumor sections harvested from IMR-05 tumor-bearing mice treated with 370 kBq of [211At]MM4 at 24, 48, and 72 hours after treatment, showing upregulation of PARP-1 compared with the control.

Figure 3.

PARP-targeted therapeutic agents and their preclinical evaluations. A,In vivo [123I]MAPi SPECT/CT imaging and [123I]MAPi efficacy (Image reproduced from Clinical Cancer Research; ref. 71). Molecular structure of [123I]MAPi; SPECT/CT images of orthotopic glioblastoma model were taken at 18 hours after injecting with [123I]MAPi (T = tumor, Cl = clearing organs), showing specific tumor uptake of [123I]MAPi; Kaplan–Meier survival curves of vehicle or [123I]MAPi (single injection: 0.37–1.11 MBq) treated mice. P < 0.01. [123I]MAPi-treated mice showed improved survival compared with the vehicle-treated mice; cytology staining and MRI imaging of brain tissues of untreated mouse at week 3, vehicle treated mouse at week 7 and [123I]MAPi treated mouse at 14 week after tumor implantation. B,In vivo therapeutic effect of [131I]PARPi in subcutaneous glioblastoma mouse models (This work was originally published in JNM, © SNMMI; ref. 72). Molecular structure of [131I]PARPi; Tumor growth at week 0 and week 2 of the mice treated with PBS, [127I]PARPi or [131I]PARPi. The treatment with [131I]PARPi suppressed tumor growth P < 0.01. C,In vivo comparison of single and fractional dosing regimens of [211At]MM4 in IMR-05 xenograft models (Image reproduced with permission from Molecular Cancer Therapeutics; ref. 73). Molecular structure of [211At]MM4; tumor growth and Kaplan–Meier curves for IMR-05 tumor-bearing mice treated with single high dose of 1,480 kBq versus a fractionated dose of 370 kBq twice weekly for a cumulative dose of 1,480 kBq; Immunofluorescence staining for PARP-1 in tumor sections harvested from IMR-05 tumor-bearing mice treated with 370 kBq of [211At]MM4 at 24, 48, and 72 hours after treatment, showing upregulation of PARP-1 compared with the control.

Close modal

To date, PARPi-derived radiolabeled compounds are based on either olaparib-like structures or rucaparib-like structures. Given that small variations in structure can lead to large differences in PARP IC50 values and trapping, it may not be assumed that all radiolabeled variants are similar in PARP trapping ability, or elicit the same allosteric effects under the same circumstances. Olaparib and rucaparib have a comparable ability for trapping PARP-1 on DNA via catalytic inhibition, yet their allosteric effects differ significantly, and may even be different for other PARP isoforms. These variances may result in diverse degrees of target engagement and PARP trapping, including PARPi trapping. This may in turn influence tumor versus normal tissue uptake, affecting imaging contrast and toxicity, of radionuclide imaging and therapy. Therefore, target specificity and PARP trapping ability of PARPi or their radiolabeled analog, and target expression should be taken into account in PARP-targeting approaches. For example, high accumulation of 18F-PARPi in healthy tissues such as lymph nodes, bone marrow, liver, and spleen may induce higher toxicity to these organs due to high physiologic expression of PARP-1 and PARP-2 (55). Careful consideration is needed in the interpretation of PARPi-derived imaging agents, which cannot necessarily predict the precise behavior of analogous, but nonidentical radionuclide therapy compounds. This can of course be overcome by using thera(g)nostic compounds, which allow imaging patient dosimetry calculation and therapy response, as demonstrated by the recent study using [123I]PARPi in glioblastoma models (Fig. 3A; ref. 71).

The discovery and development of PARPi in BRCA-mutant cancers is arguably the most elegant example of synthetic lethality to target tumor suppression gene loss in clinical practice. PARPi therapy can be of considerable benefit to a wide variety of patients with cancer as a monotherapy and can synergize with appropriately selected drugs to improve treatment outcome. However, before the full potential of mono- and combination therapies may be achieved in clinical practice, it is imperative to recognize the limitations currently faced in the PARP inhibition in cancer therapy. Attention should be paid to identification and validation of predictive biomarkers to predict synthetic lethality, how distinct molecular mechanisms of action of different PARPi, off-target effects due to high physiologic PARP-expressing healthy organs, tumor heterogeneity, and genetic modifications in patients may lead to unexpected treatment responses. Genetic testing, for example, BRACAnalysis or myChoice test (Myriad Genetics) to select those patients who will benefit from PARPi treatment has been implemented in many clinical trials, and the use of diagnostic PARPi-based PET imaging may serve as a noninvasive method to monitor patient treatment response or study the degree of drug engagement in tumor (based on PARP-1 expression levels) at any time during the treatment cycle. Proof-of-concept studies using imaging agents to study PARP levels will add value to the interpretation of clinical data. The emergence of PARP-targeting radionuclide therapy represents another treatment strategy that warrants further investigation to explore its potential.

C.Y. Chan reports grants from Pancreatic Cancer UK and CRUK through the Oxford Institute for Radiation Oncology during the conduct of the study. K.V. Tan reports grants from University of Hong Kong during the conduct of the study. B. Cornelissen reports grants from Pancreatic Cancer UK, CRUK, and other from Theragnostics Ltd during the conduct of the study; in addition, B. Cornelissen has a patent licensed (application number 1804924.7).

This work was supported by Pancreatic Cancer U.K.; CRUK through the Oxford Institute for Radiation Oncology (for B. Cornelissen), and in part by Seed Fund for Basic Research of University of Hong Kong (for K.V. Tan and C.Y. Chan).

Because of space limitations, we regret that many primary and historical published works on the topic have not been cited in this review.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Min
A
,
Im
S-A
. 
PARP inhibitors as therapeutics: beyond modulation of PARylation
.
Cancers
2020
;
12
:
394
.
2.
Pascal
JM
. 
The comings and goings of PARP-1 in response to DNA damage
.
DNA Repair
2018
;
71
:
177
82
.
3.
Maya-Mendoza
A
,
Moudry
P
,
Merchut-Maya
JM
,
Lee
M
,
Strauss
R
,
Bartek
J
. 
High speed of fork progression induces DNA replication stress and genomic instability
.
Nature
2018
;
559
:
279
84
.
4.
Cortés‐Ledesma
F
,
Aguilera
A
. 
Double‐strand breaks arising by replication through a nick are repaired by cohesin‐dependent sister‐chromatid exchange
.
EMBO Rep
2006
;
7
:
919
26
.
5.
Palazzo
L
,
Mikoč
A
,
Ahel
I
. 
ADP‐ribosylation: new facets of an ancient modification
.
FEBS J
2017
;
284
:
2932
46
.
6.
Caron
M-C
,
Sharma
AK
,
O'Sullivan
J
,
Myler
LR
,
Ferreira
MT
,
Rodrigue
A
, et al
Poly (ADP-ribose) polymerase-1 antagonizes DNA resection at double-strand breaks
.
Nat Commun
2019
;
10
:
2954
.
7.
Vyas
S
,
Chang
P
. 
New PARP targets for cancer therapy
.
Nat Rev Cancer
2014
;
14
:
502
9
.
8.
Langelier
M-F
,
Planck
JL
,
Roy
S
,
Pascal
JM
. 
Structural basis for DNA damage–dependent poly (ADP-ribosyl) ation by human PARP-1
.
Science
2012
;
336
:
728
32
.
9.
Riccio
AA
,
Cingolani
G
,
Pascal
JM
. 
PARP-2 domain requirements for DNA damage-dependent activation and localization to sites of DNA damage
.
Nucleic Acids Res
2016
;
44
:
1691
702
.
10.
Gagné
J-P
,
Rouleau
M
,
Poirier
GG
. 
PARP-1 activation—bringing the pieces together
.
Science
2012
;
336
:
678
9
.
11.
Obaji
E
,
Haikarainen
T
,
Lehtiö
L
. 
Structural basis for DNA break recognition by ARTD2/PARP2
.
Nucleic Acids Res
2018
;
46
:
12154
65
.
12.
Langelier
M-F
,
Riccio
AA
,
Pascal
JM
. 
PARP-2 and PARP-3 are selectively activated by 5′ phosphorylated DNA breaks through an allosteric regulatory mechanism shared with PARP-1
.
Nucleic Acids Res
2014
;
42
:
7762
75
.
13.
Langelier
M-F
,
Zandarashvili
L
,
Aguiar
PM
,
Black
BE
,
Pascal
JM
. 
NAD+ analog reveals PARP-1 substrate-blocking mechanism and allosteric communication from catalytic center to DNA-binding domains
.
Nat Commun
2018
;
9
:
844
.
14.
Murai
J
,
Pommier
Y
. 
PARP trapping beyond homologous recombination and platinum sensitivity in cancers
.
Annu Rev Cancer Biol
2019
;
3
:
131
50
.
15.
Prasad
CB
,
Prasad
SB
,
Yadav
SS
,
Pandey
LK
,
Singh
S
,
Pradhan
S
, et al
Olaparib modulates DNA repair efficiency, sensitizes cervical cancer cells to cisplatin and exhibits anti-metastatic property
.
Sci Rep
2017
;
7
:
12876
.
16.
Murai
J
,
Shar-yin
NH
,
Das
BB
,
Renaud
A
,
Zhang
Y
,
Doroshow
JH
, et al
Trapping of PARP1 and PARP2 by clinical PARP inhibitors
.
Cancer Res
2012
;
72
:
5588
99
.
17.
Murai
J
,
Huang
S-YN
,
Renaud
A
,
Zhang
Y
,
Ji
J
,
Takeda
S
, et al
Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib
.
Mol Cancer Ther
2014
;
13
:
433
43
.
18.
Hopkins
TA
,
Shi
Y
,
Rodriguez
LE
,
Solomon
LR
,
Donawho
CK
,
DiGiammarino
EL
, et al
Mechanistic dissection of PARP1 trapping and the impact on in vivo tolerability and efficacy of PARP inhibitors
.
Mol Cancer Res
2015
;
13
:
1465
77
.
19.
Zandarashvili
L
,
Langelier
M-F
,
Velagapudi
UK
,
Hancock
MA
,
Steffen
JD
,
Billur
R
, et al
Structural basis for allosteric PARP-1 retention on DNA breaks
.
Science
2020
;
368
:
eaax6367
.
20.
Kumar
C
,
PTV
L
,
Arunachalam
A
. 
A mechanistic approach to understand the allosteric reverse signaling by selective and trapping poly (ADP-ribose) polymerase 1 (PARP-1) inhibitors
.
J Biomol Struct Dyn
2020
;
38
:
2482
92
.
21.
Ter Brugge
P
,
Kristel
P
,
Van Der Burg
E
,
Boon
U
,
De Maaker
M
,
Lips
E
, et al
Mechanisms of therapy resistance in patient-derived xenograft models of BRCA1-deficient breast cancer
.
J Natl Cancer Inst
2016
;
108
:
djw148
.
22.
Park
HS
,
Lee
JD
,
Kim
JY
,
Park
S
,
Kim
JH
,
Han
HJ
, et al
Establishment of chemosensitivity tests in triple-negative and BRCA-mutated breast cancer patient-derived xenograft models
.
PLoS One
2019
;
14
:
e0225082
.
23.
Evans
KW
,
Yuca
E
,
Akcakanat
A
,
Scott
SM
,
Arango
NP
,
Zheng
X
, et al
A population of heterogeneous breast cancer patient-derived xenografts demonstrate broad activity of PARP inhibitor in BRCA1/2 wild-type tumors
.
Clin Cancer Res
2017
;
23
:
6468
77
.
24.
Stewart
CA
,
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
:
28575
87
.
25.
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
:
1274
84
.
26.
Friedlander
M
,
Gebski
V
,
Gibbs
E
,
Davies
L
,
Bloomfield
R
,
Hilpert
F
, et al
Health-related quality of life and patient-centred outcomes with olaparib maintenance after chemotherapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT Ov-21): a placebo-controlled, phase 3 randomised trial
.
Lancet Oncol
2018
;
19
:
1126
34
.
27.
Robson
M
,
Im
S-A
,
Senkus
E
,
Xu
B
,
Domchek
SM
,
Masuda
N
, et al
Olaparib for metastatic breast cancer in patients with a germline BRCA mutation
.
N Engl J Med
2017
;
377
:
523
33
.
28.
Turner
NC
,
Telli
ML
,
Rugo
HS
,
Mailliez
A
,
Ettl
J
,
Grischke
E-M
, et al
A phase II study of talazoparib after platinum or cytotoxic nonplatinum regimens in patients with advanced breast cancer and germline BRCA1/2 mutations (ABRAZO)
.
Clin Cancer Res
2019
;
25
:
2717
24
.
29.
Litton
JK
,
Rugo
HS
,
Ettl
J
,
Hurvitz
SA
,
Gonçalves
A
,
Lee
K-H
, et al
Talazoparib in patients with advanced breast cancer and a germline BRCA mutation
.
N Engl J Med
2018
;
379
:
753
63
.
30.
Golan
T
,
Hammel
P
,
Reni
M
,
Van Cutsem
E
,
Macarulla
T
,
Hall
MJ
, et al
Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer
.
N Engl J Med
2019
;
381
:
317
27
.
31.
de Bono
J
,
Mateo
J
,
Fizazi
K
,
Saad
F
,
Shore
N
,
Sandhu
S
, et al
Olaparib for metastatic castration-resistant prostate cancer
.
N Engl J Med
2020
;
382
:
2091
102
.
32.
Noordermeer
SM
,
van Attikum
H
. 
PARP inhibitor resistance: a tug-of-war in BRCA-mutated cells
.
Trends Cell Biol
2019
;
29
:
820
34
.
33.
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
:
523
35
.
34.
Jannetti
SA
,
Zeglis
BM
,
Zalutsky
MR
,
Reiner
T
. 
Poly (ADP-Ribose) polymerase (PARP) inhibitors and radiation therapy
.
Front Pharmacol
2020
;
11
:
170
.
35.
Chalmers
A
,
Johnston
P
,
Woodcock
M
,
Joiner
M
,
Marples
B
. 
PARP-1, PARP-2, and the cellular response to low doses of ionizing radiation
.
Int J Radiat Oncol Biol Phys
2004
;
58
:
410
9
.
36.
Kim
H
,
George
E
,
Ragland
RL
,
Rafail
S
,
Zhang
R
,
Krepler
C
, et al
Targeting the ATR/CHK1 axis with PARP inhibition results in tumor regression in BRCA-mutant ovarian cancer models
.
Clin Cancer Res
2017
;
23
:
3097
108
.
37.
Parmar
K
,
Kochupurakkal
BS
,
Lazaro
J-B
,
Wang
ZC
,
Palakurthi
S
,
Kirschmeier
PT
, et al
The CHK1 inhibitor prexasertib exhibits monotherapy activity in high-grade serous ovarian cancer models and sensitizes to PARP inhibition
.
Clin Cancer Res
2019
;
25
:
6127
40
.
38.
Luo
M-L
,
Zheng
F
,
Chen
W
,
Liang
Z-M
,
Chandramouly
G
,
Tan
J
, et al
Inactivation of the prolyl isomerase Pin1 sensitizes BRCA1-proficient breast cancer to PARP inhibition
.
Cancer Res
2020
;
80
:
3033
45
.
39.
Liu
B
,
Li
L
,
Yang
G
,
Geng
C
,
Luo
Y
,
Wu
W
, et al
PARP inhibition suppresses GR–MYCN–CDK5–RB1–E2F1 signaling and neuroendocrine differentiation in castration-resistant prostate cancer
.
Clin Cancer Res
2019
;
25
:
6839
51
.
40.
Johnson
SF
,
Cruz
C
,
Greifenberg
AK
,
Dust
S
,
Stover
DG
,
Chi
D
, et al
CDK12 inhibition reverses de novo and acquired PARP inhibitor resistance in BRCA wild-type and mutated models of triple-negative breast cancer
.
Cell Rep
2016
;
17
:
2367
81
.
41.
Sun
C
,
Yin
J
,
Fang
Y
,
Chen
J
,
Jeong
KJ
,
Chen
X
, et al
BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency
.
Cancer Cell
2018
;
33
:
401
16.e8
.
42.
Miller
AL
,
Fehling
SC
,
Garcia
PL
,
Gamblin
TL
,
Council
LN
,
van Waardenburg
RC
, et al
The BET inhibitor JQ1 attenuates double-strand break repair and sensitizes models of pancreatic ductal adenocarcinoma to PARP inhibitors
.
EBioMedicine
2019
;
44
:
419
30
.
43.
Li
L
,
Karanika
S
,
Yang
G
,
Wang
J
,
Park
S
,
Broom
B
, et al
Enzalutamide-induced “BRCAness” and PARP inhibition is a synthetic lethal therapy for castration-resistant prostate cancer
.
Sci Signal
2017
;
10
:
eaam7479
.
44.
Coleman
RL
,
Fleming
GF
,
Brady
MF
,
Swisher
EM
,
Steffensen
KD
,
Friedlander
M
, et al
Veliparib with first-line chemotherapy and as maintenance therapy in ovarian cancer
.
N Engl J Med
2019
;
381
:
2403
15
.
45.
Loibl
S
,
O'Shaughnessy
J
,
Untch
M
,
Sikov
WM
,
Rugo
HS
,
McKee
MD
, et al
Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): a randomised, phase 3 trial
.
Lancet Oncol
2018
;
19
:
497
509
.
46.
Han
H
,
Diéras
V
,
Robson
M
,
Palácová
M
,
Marcom
P
,
Jager
A
, et al
Veliparib with temozolomide or carboplatin/paclitaxel versus placebo with carboplatin/paclitaxel in patients with BRCA1/2 locally recurrent/metastatic breast cancer: randomized phase II study
.
Ann Oncol
2018
;
29
:
154
61
.
47.
Ray-Coquard
I
,
Pautier
P
,
Pignata
S
,
Pérol
D
,
González-Martín
A
,
Berger
R
, et al
Olaparib plus bevacizumab as first-line maintenance in ovarian cancer
.
N Engl J Med
2019
;
381
:
2416
28
.
48.
Liu
J
,
Barry
W
,
Birrer
M
,
Lee
J-M
,
Buckanovich
R
,
Fleming
G
, et al
Overall survival and updated progression-free survival outcomes in a randomized phase II study of combination cediranib and olaparib versus olaparib in relapsed platinum-sensitive ovarian cancer
.
Ann Oncol
2019
;
30
:
551
7
.
49.
Clarke
N
,
Wiechno
P
,
Alekseev
B
,
Sala
N
,
Jones
R
,
Kocak
I
, 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
86
.
50.
Drew
Y
,
De Jonge
M
,
Hong
S
,
Park
Y
,
Wolfer
A
,
Brown
J
, et al
An open-label, phase II basket study of olaparib and durvalumab (MEDIOLA): results in germline BRCA-mutated (gBRCAm) platinum-sensitive relapsed (PSR) ovarian cancer (OC)
.
Gynecol Oncol
2018
;
149
:
246
7
.
51.
Lampert
EJ
,
Zimmer
AS
,
Padget
MR
,
Cimino-Mathews
A
,
Nair
JR
,
Liu
Y
, et al
Combination of PARP inhibitor olaparib, and PD-L1 inhibitor durvalumab, in recurrent ovarian cancer: a proof-of-concept phase 2 study
.
Clin Cancer Res
2020
;
26
:
4268
79
.
52.
Carney
B
,
Kossatz
S
,
Reiner
T
. 
Molecular imaging of PARP
.
J Nucl Med
2017
;
58
:
1025
30
.
53.
Tu
Z
,
Chu
W
,
Zhang
J
,
Dence
CS
,
Welch
MJ
,
Mach
RH
. 
Synthesis and in vivo evaluation of [11C] PJ34, a potential radiotracer for imaging the role of PARP-1 in necrosis
.
Nucl Med Biol
2005
;
32
:
437
43
.
54.
Reiner
T
,
Lacy
J
,
Keliher
EJ
,
Yang
KS
,
Ullal
A
,
Kohler
RH
, et al
Imaging therapeutic PARP inhibition in vivo through bioorthogonally developed companion imaging agents
.
Neoplasia
2012
;
14
:
169
77
.
55.
Schöder
HM
,
França
PDDS
,
Nakajima
R
,
Burnazi
EM
,
Roberts
S
,
Brand
C
, et al
Safety and feasibility of PARP1/2 imaging with 18F-PARPi in patients with head and neck cancer
.
Clin Cancer Res
2020
;
26
:
3110
6
.
56.
Zmuda
F
,
Malviya
G
,
Blair
A
,
Boyd
M
,
Chalmers
AJ
,
Sutherland
A
, et al
Synthesis and evaluation of a radioiodinated tracer with specificity for poly (ADP-ribose) polymerase-1 (PARP-1) in vivo
.
J Med Chem
2015
;
58
:
8683
93
.
57.
Salinas
B
,
Irwin
CP
,
Kossatz
S
,
Bolaender
A
,
Chiosis
G
,
Pillarsetty
N
, et al
Radioiodinated PARP1 tracers for glioblastoma imaging
.
EJNMMI Res
2015
;
5
:
123
.
58.
Carlucci
G
,
Carney
B
,
Brand
C
,
Kossatz
S
,
Irwin
CP
,
Carlin
SD
, et al
Dual-modality optical/PET imaging of PARP1 in glioblastoma
.
Mol Imaging Biol
2015
;
17
:
848
55
.
59.
Huang
T
,
Hu
P
,
Banizs
AB
,
He
J
. 
Initial evaluation of Cu-64 labeled PARPi-DOTA PET imaging in mice with mesothelioma
.
Bioorg Med Chem Lett
2017
;
27
:
3472
6
.
60.
Wilson
TC
,
Xavier
M-A
,
Knight
J
,
Verhoog
S
,
Torres
JB
,
Mosley
M
, et al
PET Imaging of PARP Expression Using 18F-Olaparib
.
J Nucl Med
2019
;
60
:
504
10
.
61.
Carney
B
,
Kossatz
S
,
Lok
BH
,
Schneeberger
V
,
Gangangari
KK
,
Pillarsetty
NVK
, et al
Target engagement imaging of PARP inhibitors in small-cell lung cancer
.
Nat Commun
2018
;
9
:
176
.
62.
Kossatz
S
,
Carney
B
,
Farley
C
,
Weber
WA
,
Drain
CM
,
Reiner
T
. 
Direct imaging of drug distribution and target engagement of the PARP inhibitor rucaparib
.
J Nucl Med
2018
;
59
:
1316
20
.
63.
Riss
PJ
,
Soskic
V
,
Schrattenholz
A
,
Roesch
F
. 
Synthesis and radiosynthesis of N5‐[18F] fluoroethyl‐pirenzepine and its metabolite N5‐[18F] fluoroethyl‐LS 75
.
J Labelled Comp Radiopharm
2009
;
52
:
576
9
.
64.
Makvandi
M
,
Xu
K
,
Lieberman
BP
,
Anderson
R-C
,
Effron
SS
,
Winters
HD
, et al
A radiotracer strategy to quantify PARP-1 expression in vivo provides a biomarker that can enable patient selection for PARP inhibitor therapy
.
Cancer Res
2016
;
76
:
4516
24
.
65.
Anderson
R-C
,
Makvandi
M
,
Xu
K
,
Lieberman
BP
,
Zeng
C
,
Pryma
DA
, et al
Iodinated benzimidazole PARP radiotracer for evaluating PARP1/2 expression in vitro and in vivo
.
Nucl Med Biol
2016
;
43
:
752
8
.
66.
Makvandi
M
,
Pantel
A
,
Schwartz
L
,
Schubert
E
,
Xu
K
,
Hsieh
C-J
, et al
A PET imaging agent for evaluating PARP-1 expression in ovarian cancer
.
J Clin Invest
2018
;
128
:
2116
26
.
67.
McDonald
ES
,
Doot
RK
,
Pantel
AR
,
Farwell
MD
,
Mach
RH
,
Maxwell
KN
, et al
Positron emission tomography imaging of poly–(adenosine diphosphate–ribose) polymerase 1 expression in breast cancer: a nonrandomized clinical trial
.
JAMA Oncol
2020
;
6
:
921
3
.
68.
Young
RJ
,
de Souza Franca
PD
,
Pirovano
G
,
Piotrowski
AF
,
Nicklin
PJ
,
Riedl
CC
, et al
Preclinical and first-in-human-brain-cancer applications of [18F] poly-(ADP-ribose) polymerase inhibitor PET/MR
.
medRxiv
2020
.
69.
Kossatz
S
,
Pirovano
G
,
França
PDDS
,
Strome
AL
,
Sunny
SP
,
Zanoni
DK
, et al
Validation of the use of a fluorescent PARP1 inhibitor for the detection of oral, oropharyngeal and oesophageal epithelial cancers
.
Nat Biomed Eng
2020
;
4
:
272
85
.
70.
Guibbal
F
,
Hopkins
SL
,
Pacelli
A
,
Isenegger
PG
,
Mosley
M
,
Torres
JB
, et al
[18F] AZD2461, an insight on difference in PARP binding profiles for DNA damage response PET imaging
.
Mol Imaging Biol
2020
;
22
:
1226
34
.
71.
Pirovano
G
,
Jannetti
SA
,
Carter
LM
,
Sadique
A
,
Kossatz
S
,
Guru
N
, et al
Targeted brain tumor radiotherapy using an auger emitter
.
Clin Cancer Res
2020
;
26
:
2871
81
.
72.
Jannetti
SA
,
Carlucci
G
,
Carney
B
,
Kossatz
S
,
Shenker
L
,
Carter
LM
, et al
PARP-1–targeted radiotherapy in mouse models of glioblastoma
.
J Nucl Med
2018
;
59
:
1225
33
.
73.
Makvandi
M
,
Lee
H
,
Puentes
LN
,
Reilly
SW
,
Rathi
KS
,
Weng
C-C
, et al
Targeting PARP-1 with alpha-particles is potently cytotoxic to human neuroblastoma in preclinical models
.
Mol Cancer Ther
2019
;
18
:
1195
204
.
74.
De Koning
L
,
Decaudin
D
,
El Botty
R
,
Nicolas
A
,
Carita
G
,
Schuller
M
, et al
PARP inhibition increases the response to chemotherapy in uveal melanoma
.
Cancers
2019
;
11
:
751
.
75.
Heisey
DA
,
Lochmann
TL
,
Floros
KV
,
Coon
CM
,
Powell
KM
,
Jacob
S
, et al
The ewing family of tumors relies on BCL-2 and BCL-XL to escape PARP inhibitor toxicity
.
Clin Cancer Res
2019
;
25
:
1664
75
.
76.
Santiago‐O'Farrill
JM
,
Weroha
SJ
,
Hou
X
,
Oberg
AL
,
Heinzen
EP
,
Maurer
MJ
, et al
Poly (adenosine diphosphate ribose) polymerase inhibitors induce autophagy‐mediated drug resistance in ovarian cancer cells, xenografts, and patient‐derived xenograft models
.
Cancer
2020
;
126
:
894
907
.
77.
Ordóñez
JL
,
Amaral
AT
,
Carcaboso
AM
,
Herrero-Martín
D
,
del Carmen García-Macías
M
,
Sevillano
V
, et al
The PARP inhibitor olaparib enhances the sensitivity of Ewing sarcoma to trabectedin
.
Oncotarget
2015
;
6
:
18875
90
.
78.
Cruz
C
,
Castroviejo-Bermejo
M
,
Gutiérrez-Enríquez
S
,
Llop-Guevara
A
,
Ibrahim
Y
,
Gris-Oliver
A
, et al
RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer
.
Ann Oncol
2018
;
29
:
1203
10
.
79.
AlHilli
MM
,
Becker
MA
,
Weroha
SJ
,
Flatten
KS
,
Hurley
RM
,
Harrell
MI
, et al
In vivo anti-tumor activity of the PARP inhibitor niraparib in homologous recombination deficient and proficient ovarian carcinoma
.
Gynecol Oncol
2016
;
143
:
379
88
.
80.
Lohse
I
,
Kumareswaran
R
,
Cao
P
,
Pitcher
B
,
Gallinger
S
,
Bristow
RG
, et al
Effects of combined treatment with ionizing radiation and the PARP inhibitor olaparib in BRCA mutant and wild type patient-derived pancreatic cancer xenografts
.
PLoS One
2016
;
11
:
e0167272
.
81.
Yun
E-J
,
Lin
C-J
,
Dang
A
,
Hernandez
E
,
Guo
J
,
Chen
W-M
, et al
Downregulation of human DAB2IP gene expression in renal cell carcinoma results in resistance to ionizing radiation
.
Clin Cancer Res
2019
;
25
:
4542
51
.
82.
Laird
JH
,
Lok
BH
,
Ma
J
,
Bell
A
,
de Stanchina
E
,
Poirier
JT
, et al
Talazoparib is a potent radiosensitizer in small cell lung cancer cell lines and xenografts
.
Clin Cancer Res
2018
;
24
:
5143
52
.
83.
Jue
TR
,
Nozue
K
,
Lester
AJ
,
Joshi
S
,
Schroder
LB
,
Whittaker
SP
, et al
Veliparib in combination with radiotherapy for the treatment of MGMT unmethylated glioblastoma
.
J Transl Med
2017
;
15
:
61
.
84.
Yin
Y
,
Shen
Q
,
Zhang
P
,
Tao
R
,
Chang
W
,
Li
R
, et al
Chk1 inhibition potentiates the therapeutic efficacy of PARP inhibitor BMN673 in gastric cancer
.
Am J Cancer Res
2017
;
7
:
473
83
.
85.
Parvin
S
,
Ramirez-Labrada
A
,
Aumann
S
,
Lu
X
,
Weich
N
,
Santiago
G
, et al
LMO2 confers synthetic lethality to PARP inhibition in DLBCL
.
Cancer Cell
2019
;
36
:
237
49.e6
.
86.
Fok
JH
,
Ramos-Montoya
A
,
Vazquez-Chantada
M
,
Wijnhoven
PW
,
Follia
V
,
James
N
, et al
AZD7648 is a potent and selective DNA-PK inhibitor that enhances radiation, chemotherapy and olaparib activity
.
Nat Commun
2019
;
10
:
5065
.
87.
Zhang
W
,
Liu
B
,
Wu
W
,
Li
L
,
Broom
BM
,
Basourakos
SP
, et al
Targeting the MYCN–PARP–DNA damage response pathway in neuroendocrine prostate cancer
.
Clin Cancer Res
2018
;
24
:
696
707
.
88.
Oza
AM
,
Cibula
D
,
Benzaquen
AO
,
Poole
C
,
Mathijssen
RH
,
Sonke
GS
, et al
Olaparib combined with chemotherapy for recurrent platinum-sensitive ovarian cancer: a randomised phase 2 trial
.
Lancet Oncol
2015
;
16
:
87
97
.
89.
Pietanza
MC
,
Waqar
SN
,
Krug
LM
,
Dowlati
A
,
Hann
CL
,
Chiappori
A
, et al
Randomized, double-blind, phase II study of temozolomide in combination with either veliparib or placebo in patients with relapsed-sensitive or refractory small-cell lung cancer
.
J Clin Oncol
2018
;
36
:
2386
94
.
90.
Owonikoko
TK
,
Dahlberg
SE
,
Sica
GL
,
Wagner
LI
,
Wade
JL
 III
,
Srkalovic
G
, et al
Randomized phase II trial of cisplatin and etoposide in combination with veliparib or placebo for extensive-stage small-cell lung cancer: ECOG-ACRIN 2511 study
.
J Clin Oncol
2019
;
37
:
222
9
.