Next Article in Journal
Thyroid Cancer Persistence in Patients with Unreliable Thyroglobulin Measurement: Circulating microRNA as Candidate Alternative Biomarkers
Next Article in Special Issue
Role of PARP Inhibitors in Cancer Immunotherapy: Potential Friends to Immune Activating Molecules and Foes to Immune Checkpoints
Previous Article in Journal
Chemosensitivity of 3D Pancreatic Cancer Organoids Is Not Affected by Transformation to 2D Culture or Switch to Physiological Culture Medium
Previous Article in Special Issue
Using Patient-Derived Xenograft (PDX) Models as a ‘Black Box’ to Identify More Applicable Patients for ADP-Ribose Polymerase Inhibitor (PARPi) Treatment in Ovarian Cancer: Searching for Novel Molecular and Clinical Biomarkers and Performing a Prospective Preclinical Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

BMN673 Is a PARP Inhibitor with Unique Radiosensitizing Properties: Mechanisms and Potential in Radiation Therapy

1
Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
2
Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
3
German Cancer Consortium (DKTK), Partner Site University Hospital Essen, German Cancer Research Center (DKFZ), 45147 Essen, Germany
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(22), 5619; https://doi.org/10.3390/cancers14225619
Submission received: 7 October 2022 / Revised: 10 November 2022 / Accepted: 13 November 2022 / Published: 16 November 2022

Abstract

:

Simple Summary

PARP inhibitors (PARPi) have broad applicability as monotherapy in tumors with HR defects. However, their combination with radiotherapy (RT) is less advanced owing to the rather modest radiosensitization typically achieved with most tested PARPi, including olaparib. Tumor resistance to PARPi or RT is a significant bottleneck in the clinic, and strategically designed combinations of PARPi and RT may offer approaches to overcome such resistance. Talazoparib (BMN673), as compared to other PARPi, is a superior radiosensitizer. In this review, we discuss our evolving understanding of the mechanistic underpinnings of BMN673 radiosensitization and outline the potential for combinations of BMN673 with RT to treat various forms of cancer.

Abstract

BMN673 is a relatively new PARP inhibitor (PARPi) that exhibits superior efficacy in vitro compared to olaparib and other clinically relevant PARPi. BMN673, similar to most clinical PARPi, inhibits the catalytic activities of PARP-1 and PARP-2 and shows impressive anticancer potential as monotherapy in several pre-clinical and clinical studies. Tumor resistance to PARPi poses a significant challenge in the clinic. Thus, combining PARPi with other treatment modalities, such as radiotherapy (RT), is being actively pursued to overcome such resistance. However, the modest to intermediate radiosensitization exerted by olaparib, rucaparib, and veliparib, limits the rationale and the scope of such combinations. The recently reported strong radiosensitizing potential of BMN673 forecasts a paradigm shift on this front. Evidence accumulates that BMN673 may radiosensitize via unique mechanisms causing profound shifts in the balance among DNA double-strand break (DSB) repair pathways. According to one of the emerging models, BMN673 strongly inhibits classical non-homologous end-joining (c-NHEJ) and increases reciprocally and profoundly DSB end-resection, enhancing error-prone DSB processing that robustly potentiates cell killing. In this review, we outline and summarize the work that helped to formulate this model of BMN673 action on DSB repair, analyze the causes of radiosensitization and discuss its potential as a radiosensitizer in the clinic. Finally, we highlight strategies for combining BMN673 with other inhibitors of DNA damage response for further improvements.

1. Introduction

Radiotherapy (RT) and chemotherapy are two main pillars of cancer treatment. As the primary treatment modality, RT contributes to about 40% of curative treatments for cancer [1,2]. Therapeutically exploiting DNA repair offers a potent means to target cancer cells [3,4]. PARP inhibitors (PARPi) are widely used chemotherapeutic agents in homologous recombination (HR) deficient tumors. The seminal finding of a synthetic lethal interaction between PARP-1 and HR defects [5,6] accelerated the development of highly specific, clinically relevant PARPi, including olaparib (AZD-2281), niraparib (MK-4827), veliparib (ABT888), rucaparib (AG-014699), and talazoparib (BMN673) [7,8,9]. Such clinical PARPi exclusively target PARP-1 and PARP-2 to execute their anti-cancer effects through a synthetically lethal interaction with HR defects. The idea of synthetic lethal interactions between the PARPi and HR defects has been exploited as an Achilles heel in the management of ovarian and breast cancers and has been extensively reviewed in the past [10,11,12,13,14,15,16,17].
The PARP family of proteins regulates several biochemical processes in a living cell by mediating a post-translational modification called poly- or mono-ADP-ribosylation (PARylation or MARylation). Where PARP-1, 2, 5a, and 5b are capable of modifying their targets through PARylation, the remaining PARP family members modify their targets through MARylation [18]. Among 17 reported members of the PARP family [19], only PARP-1, PARP-2, and PARP-3 were found to regulate DNA damage response and repair (Figure 1). However, the main target of PARPi is PARP-1, which regulates DSB repair through PARylation of various signaling proteins, including itself. PARP-1 is the critical protein involved in single-strand break repair (SSB) and alternative end joining (alt-EJ) [20]. Multiple studies extend the primary functions of PARP-1 in DNA repair and suggest a broader role for PARP-1 and PARylation in the complex network of multiple DNA repair mechanisms [21,22]. This has highlighted PARP-1 as a central molecule coordinating the overall cellular responses to genotoxic stress in general and DNA damage, in particular (DDR) [23].
Tumors lacking HR and therefore therapeutically treated with PARPi, frequently acquire resistance when treated for a prolonged period with such inhibitors [24]. The prime mechanisms behind development of PARPi resistance in tumors are restoring HR activity, losing PARP-1 activity, or upregulating alt-EJ repair functions [24]. Combining PARPi with other chemotherapeutics or RT may help in preventing or overcoming PARPi resistance, increasing thus the tumor response to therapy.
Combination with RT offers an attractive scenario for enhancing the efficacy of PARPi [25,26,27,28,29]. Several studies have shown potential for radiosensitization for several clinically relevant PARPi, however, most of them show only moderate radiosensitization [30,31,32,33,34,35,36,37,38] except for BMN673. BMN673 showed robust radiosensitization in cancer cells [39,40,41], while leaving normal cells almost unaffected [39]. BMN673 exerts distinct DSB repair modulatory activities and appears to offer surprisingly flexible drug treatment schedules that make it the “ideal” clinical radiosensitizer. The expansion of the clinical use of PARPi in the face of acquired resistance, rationalizes the active search for clinical strategies combining PARPi with other agents able to suppress, mitigate or delay resistance development. RT is a promising candidate along these lines. Here, we discuss the rationale and potential of combining PARPi and RT with particular emphasis on the ability of BMN673 to strongly radiosensitize tumor cells and xenografts, paving the way for clinical and pre-clinical tests.

2. PARP-1 Operates at the Heart of DNA Damage Response and DNA Repair

Exacerbation occasional deregulation, or suppression of the DNA repair machinery is a promising avenue for improvements in cancer treatment utilizing agents generating damage in the DNA. The genomic integrity of higher eukaryotes is continuously challenged by chemical and physical agents capable of introducing covalent DNA modifications [20,42]. Mammalian cells have evolved well-coordinated DNA damage response (DDR) signaling to protect genomic DNA [43,44,45,46,47,48]. DDR is particularly sensitive to DSBs and replication stress and orchestrates the engagement of DSB repair pathways, while coordinating at the same time their repair with cell cycle checkpoints. It is well-documented that PARP-1 is tightly associated with chromatin. PARP-1 is endowed with high detection sensitivity to DNA damage, particularly single strand breaks (SSBs) and DSBs, causing its rapid activation that generates large amounts of PAR-chains on several proteins, including itself [19,49,50]. PARP-1-mediated PARylation ensures the timely recruitment of DNA repair proteins to DSBs [22,51]. PARP-1 plays a crucial role as a DNA damage sensor in DDR activating crucial enzymes, operating as transducers and effectors of DDR signaling (Figure 2) [52].
In genetic studies, PARP-1 was originally identified as a key protein in the base excision repair (BER) pathway [53,54,55,56,57,58]. BER removes from DNA oxidative base damage, alkylation damage and abasic sites through the synchronized actions of DNA glycosylases, apurinic-apyrimidinic (AP) endonucleases, end-processing enzymes (polynucleotide kinase-phosphatase (PNKP) and flap endonuclease (FEN-1)), DNA polymerases and the DNA Ligase3/XRCC1 complex [59,60,61,62]. PARP-1 inhibition suppresses the oxidative stress response in human lens cells by inhibiting BER [63]. Several studies, on the other hand, suggest that PARP-1 is not essential for BER [22,54,64,65].
PARP-1 promptly detects single-strand breaks (SSBs) through direct interaction with the damaged DNA [22,66,67]. One of the crucial roles of PARP-1 in SSB repair is to recruit the Ligase 3/XRCC1 complex, which mediates DNA ligation. It has been demonstrated that the recruitment of XRCC1 to SSBs is PARP-1 and PARP-2 dependent [68,69] and that mutation in XRCC1 results in PARP-1 hyperactivity, which leads to exhaustion of the cellular NAD+, causing cell death. PARP-1 can also stimulate the gap-filling step and the final DNA ligation step of SSB repair, by intervening with the availability of ATP [70].
Recent studies have also suggested a role for PARP-1 in DSB repair pathways: classical non-homologous end joining (c-NHEJ), homologous recombination (HR), alternative end joining (alt-EJ), and single-strand annealing (SSA). The PARP-1 requirements for c-NHEJ function are less well characterized. It has been demonstrated that PARP-1 can PARylate the DNA-dependent protein kinase catalytic subunit (DNA-PKcs), which is the primary coordinator of c-NHEJ [71]. This study shows that PARylation of DNA-PKcs stimulates its kinase activity without requiring interaction with the KU70/80 heterodimer [71]. Moreover, it has been demonstrated that PARP-1 competes with the KU70/80 heterodimer on DSBs, thus negatively regulating c-NHEJ [72]. This suggests that PARP-1 may even shift DNA repair towards DNA end-resection-dependent pathways.
The role of PARP-1 in HR is somewhat complex, and earlier studies showed that PARP-1 is dispensable for HR. In PARP-depleted cells, HR was normal, and PARylation showed a little direct effect on HR [73]. Inhibition or depletion of PARP-1 resulted in spontaneous sister chromatid exchanges (SCEs). As SCEs are strongly dependent on HR, this result suggests that PARP-1 inhibits HR [74]. On the other hand, PARP-1 depletion failed to suppress gene conversion in an I-SceI-induced DSB in the specifically designed DR-GFP reporter cell line [39,73]. There is evidence that PARP-1 is required to efficiently detect DSBs, by interacting with MRE11 and the apical DDR kinase ATM [22,51]. These interactions implicate PARP-1 in DNA end-resection, the initial step of DSB-repair by HR. It was also shown that PARP-1 recruits MRE11 and RAD51, which restart stalled replication in a BRCA1/2-dependent manner [75]. The accumulation of replication protein A (RPA) at the generated single-stranded DNA tracts during DNA end-resection requires the activity of BRCA1. There is evidence that PARP-1 acts as a resection stimulatory factor also by interacting with BARD1, an auxiliary factor of BRCA1 that helps recruit BRCA1 to DSBs. PARP-1 activity is also detected at later HR steps, where the PARylated BRCA1 limits DNA end-resection by recruiting the RAP80 chromatin factor. The interaction with RAP80 stabilizes BRCA1 and suppresses HR, resulting in effective strand invasion and subsequent break repair [76]. Collectively, these observations suggest stimulatory and repressive functions for PARP1 in DNA end-resection.
PARP-1 has a well-established role in alt-EJ. Abrogation of PARP-1 activity by chemical inhibition or transcriptional inactivation reduces the frequency of alt-EJ-mediated chromosomal translocations, suggesting that PARP-1 is an alt-EJ factor [39,77,78,79]. However, in PARP-1 deficient genetic background, radiosensitization and DSB repair inhibition at high IR doses are robust mainly in Ku deficient cells [72]. This notion is supported by the avid PARP-1 recruitment at DSBs in KU deficient cells and the increased loading of MRN and XRCC1/LIG3 complex at DSBs [80]. In addition, there are reports attributing a role to PARP-1 in the recruitment of DNA polymerase θ (Pol θ), a molecule recently identified as a component of alt-EJ [81,82]. Advances in crystallography and the ongoing characterization of the functional network of PARP-1 interacting proteins, open new avenues for developing specific inhibitors to target PARP-1 activity, or to disrupt interactions with protein factors of alt-EJ.
Although the function of PARP-1 in SSA is scarcely documented, studies indicate a minor, indirect role for PARP-1 in this process. For example, it has been reported that deficiency of ERCC1, a component of the excision nuclease complex (ERCC1-XPF), activated during SSA, results in synthetic lethality after olaparib treatment [83]. Moreover, reports suggest that PARP-1 modulates the activity of FEN-1; a protein also involved in RAD52-mediated SSA [84,85]. However, as PARP-1, ERCC1-XPF and RAD52 are shown to be involved in multiple DNA repair pathways, this makes it difficult to establish specific roles of PARP-1 in SSA.

3. Development of Clinically Relevant PARP Inhibitors

The discovery of synthetically lethal interactions between BRCA1/BRCA2 loss of function mutations and PARP inhibition paved the way to the development as a monotherapy of clinically relevant PAPRi with strong and specific activity. Olaparib (Lynparza®, developed by AstraZeneca, Cambridge, UK) was the first PARPi approved in 2014 by the European Medicines Agency (EMA) and by the Food and Drug Administration (FDA) in the United States for use as a single agent in the clinic. It showed a favorable response in patients with germline BRCA1/2-mutated advanced, recurrent ovarian cancer, and for maintenance treatment of adult patients with recurrent gynecologic cancers [86,87,88,89,90]. To date, three additional PARPi have been approved by the FDA. Rucaparib (Rubraca®, developed by Clovis Oncology, Boulder, CO, USA) was approved in 2016 for the treatment of ovarian cancer in patients with somatic and germline BRCA mutations, and in maintenance treatment settings [91,92]. Niraparib (Zejula™, developed by Tesaro, Waltham, MA, USA) was approved in 2017 for maintenance therapy of adult patients with recurrent gynecologic cancers, irrespective of BRCA mutations, or HR status [93]. Talazoparib (BMN673, Talzenna, developed by Pfizer, New York, NY, USA) was approved by the FDA (2018) and EMA (2019) for the treatment of germline BRCA-mutated, HER2-negative metastatic breast cancer [9,94]. Among all these clinical PARPi, BMN673 has shown superior efficacy in vitro [95] and may offer a significant clinical benefit as monotherapy or in combination with other anticancer agents.

4. Rationale for Combining PARP Inhibitors with Radiotherapy

RT has been one of the prime cancer treatment modalities [1,2]. While tumors occasionally develop radioresistance, compromising treatment efficacy, the likelihood of this development is lower than after treatment with chemotherapeutics. Altered DNA repair activity is the primary radioresistance mechanism, and targeting repair mechanisms with chemical inhibitors offers therefore attractive treatment possibilities. Such inhibitors may target specific pathways utilized by irradiated cells to repair DSBs. As described above, PARP-1 is involved in multiple DNA repair pathways, including the repair of SSBs and DSBs (Figure 2). Therefore, its inhibition is expected to cause radiosensitization. It is also relevant that elevated expression of PARP-1 is reported in several radioresistant tumors [35,96,97]. Combining, therefore, PARPi with RT may offer means to radiosensitize cells or to overcome radioresistance when it develops.
When PARPi are used as a monotherapy in HR deficient tumors for an extended period, resistance to PARPi develops by reduced PARP-1 levels [98,99], restoration of HR, or the acquisition of compensating mutations [100,101,102,103]. The overall tumor response rate to PARPi ranges from 30 to 50% [104], indicating that tumors either possess de novo resistance, or develop resistance later. Combining PARPi only during the course of RT may prevent the development of resistance owing to the short course of application (usually about 8 weeks). Additionally, PARPi may radiosensitize tumors with functional HR offering thus means to treat tumors irrespective of their HR status. Indeed, several pre-clinical and clinical studies have tested the efficacy of various PARPi-RT combinations [26]. Recently, radiolabeled PARP inhibitors have been developed and tested for targeted tumor therapy [105]. In a pre-clinical study, encouraging results have been recently reported in EGFR-mutated NSCLC, when niraparib was combined with radiation [106].

5. BMN673 Is a Superior Radiosensitizer

A few pre-clinical studies have shown the potential of BMN673 in combination with RT on multiple cancer cell lines and xenografts. BMN673 was shown to inhibit the proliferation of glioblastoma stem cells after exposure to low or high linear energy transfer radiations [107]. In 2018, we were the first to report the strong radiosensitizing potential of BMN673 (50 nM) in a battery of cancer cell lines originating from Rhabdoid tumors, Sarcomas, lung carcinomas, osteosarcomas, and colon cancer [39]. We found that only a short drug treatment time (~1 h) prior to RT was sufficient to achieve robust radiosensitization. Under similar experimental conditions, olaparib (3 µM), AG14361 (0.4 µM), and PJ34 (5 µM) showed only moderate radiosensitization. A potent radiosensitizing effect of BMN673 (200 nM) but not veliparib (1600 nM) was also reported in small cell lung carcinoma (SCLC) cell lines and Xenografts [40]. The non-small cell lung carcinoma (NSCLS) cell line, H460, was strongly radiosensitized after treatment with 2 nM BMN673, which could be further enhanced by treating cells with 5-Azacytidine [108]. Colorectal cancer cells and xenografts (BRAF wild type and mutant) could also be strongly radiosensitized by BMN673 (125 nM), whereas olaparib (250nM) and rucaparib (250 nM) exerted only a moderate radiosensitizing effect [109]. Highly radio- and chemotherapy-resistant chondrosarcoma cells were also shown to be strongly radiosensitized by BMN673 (5 nM) irrespective of their IDH (isocitrate dehydrogenase) mutation status [41]. Hepatocellular carcinoma cells, HepG2 and PLC/PRF/5, were also robustly radiosensitized with BMN673 (50 nM), whereas veliparib (10 µM) was ineffective [110]. A battery of melanoma cell lines was also strongly radiosensitized by BMN673 (50 nM). In the follow-up study, niraparib could not reach similar levels of radiosensitization, even at a concentration of 2500 nM [111].
One of the prominent features of BMN673, evident in the above-mentioned studies, is that it can be used at low nanomolar concentrations to achieve strong radiosensitization. Such concentrations are achievable in the blood of treated patients also ensuring in vivo radiosensitization [112]. No other clinical PARPi showed such efficacy and similar pharmacological properties, as pointed out earlier in this section. Not surprisingly, therefore, in pre-clinical and clinical settings, BMN673 showed robust antitumor activities at relatively low concentrations. For instance, olaparib was administered at 50 mg/kg, whereas BMN673 was used at a much lower dose of 0.3 mg/kg in pre-clinical studies (8). Similar trends were also noted in the clinical setting (protocols: NCT02032823, NCT02184195, NCT01945775, NCT02326844) [113,114,115]. Importantly, BMN673 as a monotherapy showed a manageable tolerability profile in breast cancer patients who participated in EMBRACA and ABRAZO studies [116,117].
Another critical property of BMN673 is the flexible drug treatment time and administration schedule required to achieve strong radiosensitization, at least in certain cell systems. Our results show that a one-hour treatment with BMN673 prior to RT exerted equally strong radiosensitization as continuous treatment during the course of incubation for colony formation, and that even administration several hours after IR radiosensitized cells with impressive efficacy [39]. This property, if proven general, will be very useful in the clinic, as it simplifies the effective combination of BMN673 treatment with radiation exposure in the hectic environment of a radiation therapy facility. Such administration-flexibility has not been reported for other radiosensitizers and requires in-depth analysis of its mechanistic underpinnings.
In addition, BMN673 specifically sensitized cancer cells to RT, whereas normal human cell lines were not sensitized [39], again a favorable parameter in the clinic, promising reduced normal tissue toxicity. The mechanistic underpinnings of this interesting observation require further in-depth investigations as well.
The above studies, in aggregate, highlight the strong potential of BMN673 as a promising radiosensitizer and warrant careful clinical trials to harness such potential in the clinic. To date, 49 clinical trials combining PARPi with RT are registered, which mainly involve testing of veliparib [118,119,120,121,122] or olaparib [123] (Table 1). However, only three of these trials, out of which two are still in the initial phase, involve testing of BMN673 (Table 2). Thus, clinical data supporting the radio-sensitization potential of BMN673 is currently lacking.
The following sections summarize the most prominent mechanisms underpinning the strong radiosensitization potential of BMN673.

6. Shift of Balance to Error-Prone DSB Repair Pathways Is Likely the Key to BMN673-Mediated Radiosensitization

Among the multiple lesions induced by IR in the DNA, DSBs are the most deleterious and are evolutionarily hardwired to provoke cellular responses affecting almost every aspect of the cellular metabolism that is known as DDR (see above) [42,124]. To maintain genome integrity, cells of higher eukaryotes utilize four major DSB repair pathways: HR, c-NHEJ, alt-EJ, and SSA, which have different cell cycle and homology dependencies and operate with different fidelity and kinetics [42,125]. By measuring the surrogate DSB marker, γ-H2AX, BMN673 was shown in numerous studies to increase foci load after IR and to inhibit DSB repair [39,40,126]. More specifically, our study revealed that BMN673 shifted the balance of DSB processing from c-NHEJ to error-prone repair pathways, such as alt-EJ, possibly explaining thus the observed radiosensitization [39]. Evidence supporting this postulate is summarized next.

6.1. BMN673 Increases DNA End-Resection after IR

DNA end-resection plays a pivotal role in DSB repair pathway choice. DSB repair pathways can be broadly classified as resection-dependent, or resection-independent. During resection an extended overhang of 3′-ssDNA is generated, which serves HR, alt-EJ, and SSA, while strongly antagonizing c-NHEJ [127,128]. Resected DNA ends are promptly coated and stabilized by RPA. In BMN673-treated cells exposed to low, clinically relevant IR doses, a 3- to 4-fold increase in RPA70 foci was observed, suggesting that a larger proportion of the induced DSBs was resected [39]. In this type of studies, it is important to carefully discriminate between increase in RPA foci numbers, which indicates an increase in the proportion of DSBs that undergo resection, and increase in focus intensity, which reflects increased resection at DSBs already shunted to resection dependent processing. Only the former result can be interpreted as a shift in balance from resection-independent to resection-dependent processing.
Another study also reported increase in RPA foci numbers in irradiated cells treated with BMN673 [129]. Notably, in contrast to BMN673, a study reported that olaparib decreased RPA70 foci formation and promoted c-NHEJ by facilitating DNA-PKcs phosphorylation [130]. This result was confirmed in a more recent study [81]. Thus, enhanced resection is a unique property of BMN673 and is likely to be the key factor of DSB repair pathway balance-disruption observed. It suggests that suppression of c-NHEJ may be associated with enhanced utilization of HR, a topic that is discussed next.

6.2. Key Questions Remain Unanswered Regarding the Effects of BMN673 on HR

HR is the only DSB repair pathway that restores the genome with high fidelity, becoming operational in the S -and G2-phases of the cell cycle. We recently reported that the contribution of HR rapidly increases to over 50% at doses between 0.1 and 2.0 Gy, while it becomes undetectable above 10 Gy [131]. For HR to initiate, the Rad51 recombinase, aided by the Rad51 paralogues, must displace RPA from ssDNA to form the Rad51 nucleoprotein filament that invades the sister chromatid [132,133,134].
Our results showed that BMN673 treatment caused a marked increase in Rad51 foci, suggesting elevated engagement of HR–as may be expected from the increased resection observed [39]. Similar findings were also reported by Caron M. et al. [129]. These results indicate that the contribution of HR increases in BMN673 treated cells, which contradicts the observed strong radiosensitization [39]. Indeed, increased engagement of an error-free DSB repair pathway is naturally expected to increase rather than decrease the survival of irradiated cells. During HR engagement analysis through RAD51 foci scoring, there are instances of combining outcomes at different endpoints, and the inference is made that increased RAD51 foci actually reflect futile HR, in the sense that it most likely remains incomplete; i.e., at some point the DNA ends are shunted for processing by error-prone repair pathways [135]. More work is urgently needed to quantitate the levels of productive and non-productive HR in BMN673-treated cells.
Another approach to functionally measure effects of various treatments on HR utilizes reporter cell lines with integrated constructs (e.g., DR-GFP) reporting processing by HR of an I-SceI-induced DSB [136]. Using such assays, it has been reported that olaparib decreases HR [81,137]. The same reporter assay failed in our hands to detect impact on HR after BMN673 treatment [39]. These results appear to contradict results with RAD51 foci formation and suggest peculiarities in the effect of BMN673 on HR that require more work for their elucidation.
When comparing results obtained by scoring RAD51 foci in irradiated cells with those obtained with reporter cell lines, it must be kept in mind that the latter assay measures effects on repair of an I-SceI-induced “clean” DSB [136]. It is likely that the damaged nucleotides typically present in IR-induced DSBs, generate different processing requirements that are differentially affected by BMN673. This aspect requires further investigations as well. It has been shown that HR preferentially repairs secondary replication induced DSBs after exposure to IR [138]. It would also be interesting to investigate how PARPi and in particular BMN673 modulate induction and repair by HR of such secondary, replication-induced DSBs.
Taken together, these results allow the tentative conclusion that BMN673 leaves HR unchanged at “clean” DSBs, but increases its engagement in IR-induced DSBs. It remains, however possible that in irradiated, BMN673-treated cells, a fraction of HR engagements is unproductive. Either way, the anticipated residual HR activity in BMN673 treated cells predicts benefits in cancer management from a combination of BMN673 with agents inhibiting HR, e.g., ATR inhibitors.

6.3. BMN673 Inhibits c-NHEJ

In mammals, c-NHEJ is the main repair pathway for DSB processing and operates with fast kinetics and efficiency, throughout the cell cycle [42,139,140,141]. Inhibition of c-NHEJ is shown to confer marked radiosensitivity in a wide range of cells [142,143,144]. We recently reported that at low doses of IR, c-NHEJ is partly suppressed by mechanisms that remain to be characterized, giving thus way to HR [131,145,146]. On the other hand, at high IR doses, HR is almost completely suppressed, leaving c-NHEJ as the clearly dominant DSB repair pathway [131,146].
BMN673 was found to inhibit c-NHEJ at high IR doses with efficiency similar to that achieved by DNA-PK inhibitors [39]. It is also highly relevant that BMN673 exerts radiosensitization equivalent to that of c-NHEJ deficient cells [39]. This result, although currently documented only in a few cell lines, is significant, as it is in line with the increase in resection observed, which will naturally suppress c-NHEJ.
Strikingly, in line with this conclusion, the “end-protector” 53BP1, an antagonist of BRCA1-CtIP-mediated end-resection, is suppressed by BMN673 at low IR doses [39]. This is in line with the observed suppression of c-NHEJ at high IR doses and predicts the increased engagement of BRCA1-CtIP-dependent HR at low IR doses. Since these experiments were carried out in the low-IR-dose region, they suggest that suppression of c-NHEJ occurs in a wide range of doses. Additional studies showed that BMN673 treatment strongly increased end-resection and decreased the accretion of 53BP1 and RIF1 proteins [129].
C-NHEJ is mechanistically suppressed at resected DNA ends owing to the low binding efficiency of Ku heterodimer on processed ends [147]. Thus, decreased 53BP1 accretion combined with extensive end-resection underpins the suppression of c-NHEJ by BMN673. It remains however open which factor of c-NHEJ is inhibited, or which resection factor is strengthened by BMN673 to mediate such profound shifts in DSB end-processing upon irradiation. Molecular characterization of this mechanism will not only advance our understanding of the mode of BMN673 action, but will also inform further search for PARPi with similar, or even improved characteristics. It is, however, relevant to point out that when PARPi are administered to HR-deficient cancers, a contribution of HR to DSB repair is a priori ruled out.
It is worth mentioning here that in contrast to combinations with IR, BMN673 treatment applied as a monotherapy resulted in elevated levels of 53BP1 foci [39,148,149,150], indicating enhanced c-NHEJ activity in this setting. More studies are warranted to elucidate the impact of BMN673 on c-NHEJ, either when used as a monotherapy or when combined with IR.

6.4. BMN673 Shunts DSBs to Error-Prone Alt-EJ

Alt-EJ is a collective term used to describe DSB processing that cannot be attributed to HR or c-NHEJ, and which engages as a backup when DSB ends are released owing to failures of these repair pathways [124]. Although alt-EJ may, in principle, also engage as a “first line” DSB repair pathway [151], when this happens, it is important to inquire first why these DSB-ends were “ignored” by mechanisms as efficient as c-NHEJ or HR. Although alt-EJ can, in principle, function on unresected DSB ends [152], it benefits from short-range resection and is therefore dependent on the CtIP/MRN complex. Other proteins implicated in alt-EJ include PARP-1 and Ligases1/3, which are involved in DSB sensing and ligation, respectively [80,153,154]. Polθ is a newcomer to alt-EJ but has raised enormous interest owing to its unusual properties that appear critical to the joining of resected DSB ends that have escaped HR or c-NHEJ. Thus, HR-deficient tumors strongly rely on the function of Polθ, in a PARP-1-dependent manner [155]. Polθ facilitates the annealing of resected 3′-tails, and extends one 3′ DNA-end using the annealing partner as a template to facilitate the final ligation step using the activity of either Lig1 or Lig3 [156].
Compared to c-NHEJ, alt-EJ operates with slower kinetics and lower efficiency, and with much greater error-proneness. Consequently, when DSBs are processed by alt-EJ, joining of “wrong”, unrelated DSB ends can occur, drastically increasing thus the probability of chromosomal translocation formation [157]. Indeed, several studies have implicated alt-EJ in chromosomal translocation formation [77,78,158,159,160].
Since PARP-1 plays a vital role in the DSB sensing step of alt-EJ, inhibition of PARP-1 using PARPi generally reduces the frequency of chromosomal translocations. Thus, PARPi such as PJ34, AG14361 and the clinically relevant olaparib, have shown to decrease translocations in irradiated cells [39,77,78,158,159]. By contrast, here again, BMN673 exerts the opposite effect, strongly promoting the formation of translocations after IR [39]. These findings support the notion that cells retreat to alt-EJ, as the ultimate backup, to process DSBs because canonical repair pathways, in this case most likely, predominantly c-NHEJ, failed. Of course, this happens at the very high price of increased incidence of chromosomal translocations. Why, here, BMN673 is again an outlier will require further studies.
Translocations are one of the critical drivers of oncogenesis and the culprits of cell death after exposure of cells to IR [161]. Our study showed that BMN673 treatment increases the frequency of translocations after IR in cancer but not in normal human cells [39], in line with the observed tumor-specific BMN673-mediated radiosensitization. Despite its error-proneness, alt-EJ remains an important mechanism of genome maintenance, because the severity of consequences of the only remaining alternative, a complete lack of DSB processing, are much more severe. Indeed, it appears that cells of higher eukaryotes, are wired to remove all DSBs from their genome using any combination of available DSB repair pathways, provided DNA ends are, or come, in close proximity. Indeed, this design may be aided by the increased chromatin mobility observed after the induction of DSBs [162].
A previous study tested alt-EJ using reporter cell lines and concluded that suppressing PARP-1-dependent alt-EJ by olaparib correlates positively to its radiosensitizing effect [33]. This observation, and our results of increased alt-EJ-mediated translocation formation in BMN673-treated cancer cells [39] suggests that BMN673-mediated radiosensitization can be further enhanced by targeting alt-EJ proteins other than PARP-1. It will be intriguing in this regard to examine, combinations of PARPi with readily available Polθ inhibitors that undergo clinical testing [163,164].

7. Speculations on the Mechanism of BMN673-Induced Radiosensitization

Retention of inhibited PARP on DNA lesions (the so-called PARP trapping) has been proposed as a key parameter of PAPRi-induced cytotoxicity underpinning the effects of monotherapy applications. PARPi-mediated inhibition of Parp1 auto-PARylation prevents its removal from DNA lesions and leads to replication fork collapse in S-phase cells, or to the suppression of other aspects of the DNA metabolism [95]. BMN63 has been reported as the most potent inducer of PARP-1 trapping, compared to other clinically relevant PARPi, and this property is often causally correlated to its increased cytotoxicity [95].
When analyzing the effects of PAPRi, it is important to address separately effects on cytotoxicity that is enhanced in HR deficient cells, and effects on radiosensitivity and DSB processing reviewed here, as they may be caused by different mechanisms. Thus, while it is possible that the enhanced trapping ability of BMN673 explains its toxicity in HR deficient cancers [8,11], it may not underpin, in its non-specific form, the notable radiosensitizing potential. It is relevant in this regard that the PARP trapping ability of clinical PARPi has been mostly demonstrated using DNA alkylating agents such as MMS that are likely to generate lesions strongly inhibiting DNA replication [40,95,165]. Similar forms of PARP1 trapping have not been reported for irradiated cells, and indeed our experiments failed to generate unequivocal evidence for trapping.
Can Parp1 trapping explain the above observed effects of BMN673 on DSB processing? The shift of DSB repair from c-NHEJ to HR and alt-EJ that requires extensive processing of DNA ends rules out general Parp1 trapping stopping all processing at the DSB end as a mechanism. Indeed, BMN673 mediated excessive resection and accretion of RPA70 and Rad51 after IR [39] is plausible only when DSB ends are not blocked by trapped PARP-1. Therefore, if Parp1 trapping underpins the above discussed DSB repair effects, it must be happening in a highly specific manner centering on c-NHEJ. We hypothesize that after BMN673 treatment, Parp1 inhibits the binding of Ku to DNA ends. Indeed, there is evidence that Parp1 precedes Ku at the DSB end [80], and it is therefore possible that the engagement of Ku is changed by trapped Parp1. Existing evidence suggests that this effect is observed both at high as well as at low doses of IR. We further hypothesize that the interaction of inhibited Parp1 is highly specific for Ku and that it leaves unchanged, or even facilitates all processes associated with DNA end resection.
It has been shown recently that p97 ATPase/segregase extracts BMN673-induced trapped PARP-1 from chromatin, a process that requires post-translational modification of PARP-1 by PIAS4-mediated SUMOylation and RNF4-mediated ubiquitination [166]. The chromatin remodeler ALC1 (amplified in liver cancer 1) could also remove inactive PARP1 through binding to readily PARylated chromatin prior to PARPi treatment. Moreover, ALC1 is implicated in the recruitment of Rad51, Ku70, RPA and MRE11 to DNA damage sites, thus acting upstream of DSB repair pathway choice after removing trapped PARP [167].
The above evidence in aggregate shows that BMN673 indeed traps Parp1 at the DSBs and provides clues on possible mechanisms to undo this trapping. It leaves however open the mechanism of how BMN673 specifically inhibits c-NHEJ and enhances in a directly compensating manner, resection-dependent DSB processing. The elucidation of the mechanisms underpinning these effects should be given high priority.
Figure 3 summarizes the postulated altered balance in DSB repair utilization after treatment of irradiated cells with BMN673, based on the assumption that it inhibits c-NHEJ. The color zones in the bars show the fraction of DSBs processed by each DSB repair pathway, except SSA that has not been sufficiently analyzed for BMN673 effects. It is drawn on the assumption that only very few DSBs remain unprocessed and that this fraction slightly increases with dose. It is also considering G2-phase cells to include effects on HR.
At low IR doses, c-NHEJ and HR repair similar fractions of DSBs, with alt-EJ having a small contribution [131,145,146]. BMN673 treatment at low IR doses inhibits c-NHEJ and shifts the balance towards HR and to alt-EJ that shows robust increases [39]. At high IR doses, HR is inherently suppressed and the majority of DSBs are processed by c-NHEJ, and to a much lesser extent by alt-EJ [131,146]. BMN673 treatment after exposure to high IR doses also inhibits c-NHEJ, but leaves now the majority of DSBs to be processed by alt-EJ [39].

8. Summary and Conclusions

In the current review, we discussed the mechanisms underpinning the superior radiosensitizing potential of BMN673, as compared to other clinically relevant PARPi. In addition, we discussed the critical properties of BMN673 that pave the way for testing it in clinical settings as an adjuvant to RT. BMN673 inhibits c-NHEJ activity at low and high IR doses by promoting DSB end resection. As a result, resection-dependent pathways (HR and alt-EJ) show increased activity (Figure 3). This is evident by increased accretion of Rad51 foci and elevated frequency of translocations after irradiation, when cells were additionally treated with BMN673. Under such circumstances, alt-EJ supersedes HR resulting in error-prone repair and robust radiosensitization.
The potential of combining BMN673 with RT is evident from several ongoing pre-clinical studies, but the clinical evidence supporting this approach is scarce. The combination has the potential to generate a paradigm shift and to establish BMN673 as a promising radiosensitizer. Our in vitro results, as well as results from other pre-clinical studies (described in Section 5), make a strong argument in favor of such a combination. Furthermore, combining BMN673 with RT in the clinic may also pave the way to the treatment of HR proficient tumors using PARP inhibition.

Author Contributions

Conceptualization, A.S., X.L., G.I.; methodology, A.S., X.L., E.M., V.M., G.I., M.S.; software, A.S., X.L., E.M., V.M., G.I., M.S.; validation, A.S., X.L., E.M., V.M., G.I., M.S.; formal analysis, A.S., X.L., E.M., V.M., G.I., M.S.; resources, G.I., M.S.; data curation, A.S., X.L., E.M., V.M., G.I., M.S.; writing—original draft preparation, A.S., X.L., E.M., V.M., G.I., M.S.; writing—review and editing, A.S., X.L., E.M., G.I.; visualization, G.I., M.S.; supervision, G.I., M.S.; project administration, G.I., M.S.; funding acquisition, G.I., M.S. All authors have read and agreed to the published version of the manuscript.

Funding

Research activities in our lab are funded by grants from BMBF (02NUK037B, 02NUK043B, 02NUK054B) and by German Research Foundation [IL51.10, IL51.11, GRK1739] and by the German Federal Ministry for Economic Affairs [BMWi-50WB1836], as well as by DAAD Project #57515880.

Conflicts of Interest

Martin Stuschke: AstraZeneca (Advisory Board Function, Research and Clinical Trials), Bristol-Myers Squibb (Advisory Board Function), Sanofi-Aventis (Advisory Board Function) and Janssen-Cilag (Advisory Board Function). Other authors declare no conflict of interest.

References

  1. Begg, A.C.; Stewart, F.A.; Vens, C. Strategies to improve radiotherapy with targeted drugs. Nat. Rev. Cancer 2011, 11, 239–253. [Google Scholar] [CrossRef] [PubMed]
  2. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.-W. Cancer and Radiation Therapy: Current Advances and Future Directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Jackson, S.P.; Helleday, T. DNA REPAIR. Drugging DNA repair. Science 2016, 352, 1178–1179. [Google Scholar] [CrossRef]
  4. Nickoloff, J.A.; Jones, D.; Lee, S.-H.; Williamson, E.A.; Hromas, R. Drugging the Cancers Addicted to DNA Repair. JNCI J. Natl. Cancer Inst. 2017, 109, djx059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. 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] [PubMed]
  6. Farmer, H.; McCabe, N.; Lord, C.J.; Tutt, A.N.J.; 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]
  7. Wang, B.; Chu, D.; Feng, Y.; Shen, Y.; Aoyagi-Scharber, M.; Post, L.E. Discovery and Characterization of (8S,9R)-5-Fluoro-8-(4-fluorophenyl)-9-(1-methyl-1H-1,2,4-triazol-5-yl)-2,7,8,9-tetrahydro-3H-pyrido[4,3,2-de]phthalazin-3-one (BMN 673, Talazoparib), a Novel, Highly Potent, and Orally Efficacious Poly(ADP-ribose) Polymerase-1/2 Inhibitor, as an Anticancer Agent. J. Med. Chem. 2016, 59, 335–357. [Google Scholar]
  8. 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] [Green Version]
  9. Hoy, S.M. Talazoparib: First Global Approval. Drugs 2018, 78, 1939–1946. [Google Scholar] [CrossRef]
  10. Thomas, H. The underlying mechanism for the PARP and BRCA synthetic lethality: Clearing up the misunderstandings. Mol. Oncol. 2011, 5, 387–393. [Google Scholar]
  11. Lord, C.J.; Ashworth, A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017, 355, 1152–1158. [Google Scholar] [CrossRef] [PubMed]
  12. Ashworth, A.; Lord, C.J. Synthetic lethal therapies for cancer: What’s next after PARP inhibitors? Nat. Rev. Clin. Oncol. 2018, 15, 564–576. [Google Scholar] [CrossRef] [PubMed]
  13. Topatana, W.; Juengpanich, S.; Li, S.; Cao, J.; Hu, J.; Lee, J.; Suliyanto, K.; Ma, D.; Zhang, B.; Chen, M.; et al. Advances in synthetic lethality for cancer therapy: Cellular mechanism and clinical translation. J. Hematol. Oncol. 2020, 13, 1–22. [Google Scholar] [CrossRef] [PubMed]
  14. Xie, T.; Dickson, K.-A.; Yee, C.; Ma, Y.; Ford, C.E.; Bowden, N.A.; Marsh, D.J. Targeting Homologous Recombination Deficiency in Ovarian Cancer with PARP Inhibitors: Synthetic Lethal Strategies That Impact Overall Survival. Cancers 2022, 14, 4621. [Google Scholar] [CrossRef] [PubMed]
  15. O’Neil, N.J.; Bailey, M.L.; Hieter, P. Synthetic lethality and cancer. Nat. Rev. Genet. 2017, 18, 613–623. [Google Scholar] [CrossRef]
  16. Huang, A.; Garraway, L.A.; Ashworth, A.; Weber, B. Synthetic lethality as an engine for cancer drug target discovery. Nat. Rev. Drug Discov. 2019, 19, 23–38. [Google Scholar] [CrossRef]
  17. Lord, C.J.; Tutt, A.N.; Ashworth, A. Synthetic Lethality and Cancer Therapy: Lessons Learned from the Development of PARP Inhibitors. Annu. Rev. Med. 2015, 66, 455–470. [Google Scholar] [CrossRef]
  18. Zhu, H.; Tang, Y.-D.; Zhan, G.; Su, C.; Zheng, C. The Critical Role of PARPs in Regulating Innate Immune Responses. Front. Immunol. 2021, 12, 712556. [Google Scholar] [CrossRef]
  19. Gibson, B.A.; Kraus, W.L. New insights into the molecular and cellular functions of poly(ADP-ribose) and PARPs. Nat. Rev. Mol. Cell Biol. 2012, 13, 411–424. [Google Scholar] [CrossRef]
  20. Mladenov, E.; Iliakis, G. Induction and repair of DNA double strand breaks: The increasing spectrum of non-homologous end joining pathways. Mutat. Res. Mol. Mech. Mutagen. 2011, 711, 61–72. [Google Scholar] [CrossRef]
  21. Pazzaglia, S.; Pioli, C. Multifaceted Role of PARP-1 in DNA Repair and Inflammation: Pathological and Therapeutic Implications in Cancer and Non-Cancer Diseases. Cells 2019, 9, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Chaudhuri, A.R.; Nussenzweig, A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cell Biol. 2017, 18, 610–621. [Google Scholar] [CrossRef] [PubMed]
  23. Hall, M.; Benafif, S. An update on PARP inhibitors for the treatment of cancer. Onco Targets Ther. 2015, 8, 519–528. [Google Scholar] [CrossRef] [Green Version]
  24. Du, Y.; Yamaguchi, H.; Hsu, J.L.; Hung, M.-C. PARP inhibitors as precision medicine for cancer treatment. Natl. Sci. Rev. 2017, 4, 576–592. [Google Scholar] [CrossRef] [Green Version]
  25. Angel, M.; Zarba, M.; Sade, J.P. PARP inhibitors as a radiosensitizer: A future promising approach in prostate cancer? Ecancermedicalscience 2021, 15, ed118. [Google Scholar] [CrossRef] [PubMed]
  26. Barcellini, A.; Loap, P.; Murata, K.; Villa, R.; Kirova, Y.; Okonogi, N.; Orlandi, E. PARP Inhibitors in Combination with Radiotherapy: To Do or Not to Do? Cancers 2021, 13, 5380. [Google Scholar] [CrossRef] [PubMed]
  27. Knelson, E.H.; Patel, S.A.; Sands, J.M. PARP Inhibitors in Small-Cell Lung Cancer: Rational Combinations to Improve Responses. Cancers 2021, 13, 727. [Google Scholar] [CrossRef]
  28. Lin, L.L.; Lakomy, D.S.; Ning, M.S.; Simpkins, F.; Jhingran, A. Combining novel agents with radiotherapy for gynecologic malignancies: Beyond the era of cisplatin. Int. J. Gynecol. Cancer Off. J. Int. Gynecol. Cancer Soc. 2020, 30, 409–423. [Google Scholar] [CrossRef] [Green Version]
  29. Lesueur, P.; Chevalier, F.; Austry, J.-B.; Waissi, W.; Burckel, H.; Noël, G.; Habrand, J.-L.; Saintigny, Y.; Joly, F. Poly-(ADP-ribose)-polymerase inhibitors as radiosensitizers: A systematic review of pre-clinical and clinical human studies. Oncotarget 2017, 8, 69105–69124. [Google Scholar] [CrossRef] [Green Version]
  30. 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. 2004, 58, 410–419. [Google Scholar] [CrossRef] [PubMed]
  31. Dungey, F.A.; Löser, D.A.; Chalmers, A.J. Replication-Dependent Radiosensitization of Human Glioma Cells by Inhibition of Poly(ADP-Ribose) Polymerase: Mechanisms and Therapeutic Potential. Int. J. Radiat. Oncol. 2008, 72, 1188–1197. [Google Scholar] [CrossRef] [PubMed]
  32. Karnak, D.; Engelke, C.G.; Parsels, L.A.; Kausar, T.; Wei, D.; Robertson, J.R.; Marsh, K.B.; Davis, M.A.; Zhao, L.; Maybaum, J.; et al. Combined Inhibition of Wee1 and PARP1/2 for Radiosensitization in Pancreatic Cancer. Clin. Cancer Res. 2014, 20, 5085–5096. [Google Scholar] [CrossRef]
  33. Kötter, A.; Cornils, K.; Borgmann, K.; Dahm-Daphi, J.; Petersen, C.; Dikomey, E.; Mansour, W.Y. Inhibition of PARP1-dependent end-joining contributes to Olaparib-mediated radiosensitization in tumor cells. Mol. Oncol. 2014, 8, 1616–1625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Albert, J.M.; Cao, C.; Kim, K.W.; Willey, C.D.; Geng, L.; Xiao, D.; Wang, H.; Sandler, A.; Johnson, D.H.; Colevas, A.D.; et al. Inhibition of Poly(ADP-Ribose) Polymerase Enhances Cell Death and Improves Tumor Growth Delay in Irradiated Lung Cancer Models. Clin. Cancer Res. 2007, 13, 3033–3042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Liu, C.; Gross, N.; Li, Y.; Li, G.; Wang, Z.; Zhong, S.; Li, Y.; Hu, G. PARP inhibitor Olaparib increases the sensitization to radiotherapy in FaDu cells. J. Cell. Mol. Med. 2020, 24, 2444–2450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Michmerhuizen, A.R.; Pesch, A.M.; Moubadder, L.; Chandler, B.C.; Wilder-Romans, K.; Cameron, M.; Olsen, E.; Thomas, D.G.; Zhang, A.; Hirsh, N.; et al. PARP1 Inhibition Radiosensitizes Models of Inflammatory Breast Cancer to Ionizing Radiation. Mol. Cancer Ther. 2019, 18, 2063–2073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Nile, D.L.; Rae, C.; Hyndman, I.J.; Gaze, M.N.; Mairs, R.J. An evaluation in vitro of PARP-1 inhibitors, rucaparib and olaparib, as radiosensitisers for the treatment of neuroblastoma. BMC Cancer 2016, 16, 1–13. [Google Scholar] [CrossRef] [Green Version]
  38. Senra, J.M.; Telfer, B.A.; Cherry, K.E.; McCrudden, C.M.; Hirst, D.G.; O’Connor, M.J.; Wedge, S.R.; Stratford, I.J. Inhibition of PARP-1 by Olaparib (AZD2281) Increases the Radiosensitivity of a Lung Tumor Xenograft. Mol. Cancer Ther. 2011, 10, 1949–1958. [Google Scholar] [CrossRef] [Green Version]
  39. Soni, A.; Li, F.; Wang, Y.; Grabos, M.; Krieger, L.M.; Chaudhary, S.; Hasan, M.S.M.; Ahmed, M.; Coleman, C.N.; Teicher, B.A.; et al. Inhibition of Parp1 by BMN673 Effectively Sensitizes Cells to Radiotherapy by Upsetting the Balance of Repair Pathways Processing DNA Double-Strand Breaks. Mol. Cancer Ther. 2018, 17, 2206–2216. [Google Scholar] [CrossRef] [Green Version]
  40. Laird, J.H.; Lok, B.H.; Ma, J.; Bell, A.; de Stanchina, E.; Poirier, J.T.; Rudin, C.M. Talazoparib Is a Potent Radiosensitizer in Small Cell Lung Cancer Cell Lines and Xenografts. Clin. Cancer Res. 2018, 24, 5143–5152. [Google Scholar] [CrossRef] [Green Version]
  41. Venneker, S.; Kruisselbrink, A.B.; Bruijn, I.H.B.-D.; de Jong, Y.; van Wijnen, A.J.; Danen, E.H.; Bovée, J.V. Inhibition of PARP Sensitizes Chondrosarcoma Cell Lines to Chemo- and Radiotherapy Irrespective of the IDH1 or IDH2 Mutation Status. Cancers 2019, 11, 1918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Mladenov, E.; Magin, S.; Soni, A.; Iliakis, G. DNA double-strand-break repair in higher eukaryotes and its role in genomic instability and cancer: Cell cycle and proliferation-dependent regulation. Semin. Cancer Biol. 2016, 37–38, 51–64. [Google Scholar] [CrossRef] [PubMed]
  43. Trovesi, C.; Manfrini, N.; Falcettoni, M.; Longhese, M.P. Regulation of the DNA Damage Response by Cyclin-Dependent Kinases. J. Mol. Biol. 2013, 425, 4756–4766. [Google Scholar] [CrossRef] [PubMed]
  44. Brinkmann, K.; Schell, M.; Hoppe, T.; Kashkar, H. Regulation of the DNA damage response by ubiquitin conjugation. Front. Genet. 2015, 6, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. O’Connor, M.J. Targeting the DNA Damage Response in Cancer. Mol. Cell 2015, 60, 547–560. [Google Scholar] [CrossRef] [Green Version]
  46. Blackford, A.N.; Jackson, S.P. ATM, ATR, and DNA-PK: The Trinity at the Heart of the DNA Damage Response. Mol. Cell 2017, 66, 801–817. [Google Scholar] [CrossRef] [Green Version]
  47. Gourley, C.; Balmaña, J.; Ledermann, J.A.; Serra, V.; Dent, R.; Loibl, S.; Pujade-Lauraine, E.; Boulton, S.J. Moving From Poly (ADP-Ribose) Polymerase Inhibition to Targeting DNA Repair and DNA Damage Response in Cancer Therapy. J. Clin. Oncol. 2019, 37, 2257–2269. [Google Scholar] [CrossRef] [Green Version]
  48. Lans, H.; Hoeijmakers, J.H.J.; Vermeulen, W.; Marteijn, J.A. The DNA damage response to transcription stress. Nat. Rev. Mol. Cell Biol. 2019, 20, 766–784. [Google Scholar] [CrossRef]
  49. Perina, D.; Mikoč, A.; Ahel, J.; Ćetković, H.; Žaja, R.; Ahel, I. Distribution of protein poly(ADP-ribosyl)ation systems across all domains of life. DNA Repair 2014, 23, 4–16. [Google Scholar] [CrossRef] [Green Version]
  50. Wei, H.; Yu, X. Functions of PARylation in DNA Damage Repair Pathways. Genom. Proteom. Bioinform. 2016, 14, 131–139. [Google Scholar] [CrossRef] [Green Version]
  51. Wang, Z.; Wang, F.; Tang, T.; Guo, C. The role of PARP1 in the DNA damage response and its application in tumor therapy. Front. Med. 2012, 6, 156–164. [Google Scholar] [CrossRef] [PubMed]
  52. de Murcia, G.; Menissier de Murcia, J. Poly(ADP-ribose) polymerase: A molecular nick-sensor. Trends Biochem. Sci. 1994, 19, 172–173. [Google Scholar] [CrossRef]
  53. Schreiber, V.; Amé, J.-C.; Dollé, P.; Schultz, I.; Rinaldi, B.; Fraulob, V.; Ménissier-De Murcia, J.; de Murcia, G. Poly(ADP-ribose) Polymerase-2 (PARP-2) Is Required for Efficient Base Excision DNA Repair in Association with PARP-1 and XRCC1. J. Biol. Chem. 2002, 277, 23028–23036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Allinson, S.L.; Dianova, I.I.; Dianov, G.L. Poly(ADP-ribose) polymerase in base excision repair: Always engaged, but not essential for DNA damage processing. Acta Biochim. Pol. 2003, 50, 169–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Le Page, F.; Schreiber, V.; Dherin, C.; de Murcia, G.; Boiteux, S. Poly(ADP-ribose) Polymerase-1 (PARP-1) Is Required in Murine Cell Lines for Base Excision Repair of Oxidative DNA Damage in the Absence of DNA Polymerase ß. J. Biol. Chem. 2003, 278, 18471–18477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Sukhanova, M.; Khodyreva, S.; Lavrik, O. Poly(ADP-ribose) polymerase 1 regulates activity of DNA polymerase b in long patch base excision repair. Mutat. Res. /Fundam. Mol. Mech. Mutagen. 2010, 685, 80–89. [Google Scholar] [CrossRef] [PubMed]
  57. Horton, J.K.; Stefanick, D.F.; Prasad, R.; Gassman, N.R.; Kedar, P.S.; Wilson, S.H. Base Excision Repair Defects Invoke Hypersensitivity to PARP Inhibition. Mol. Cancer Res. 2014, 12, 1128–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Lavrik, O.I. PARPs’ impact on base excision DNA repair. DNA Repair 2020, 93, 102911. [Google Scholar] [CrossRef]
  59. Seeberg, E.; Eide, L.; Bjoras, M. The base excision repair pathway. Trends Biochem. Sci. 1995, 20, 391–397. [Google Scholar] [CrossRef]
  60. Grasso, S.; Tell, G. Base excision repair in Archaea: Back to the future in DNA repair. DNA Repair 2014, 21, 148–157. [Google Scholar] [CrossRef]
  61. Kennedy, E.; Caffrey, P.J.; Delaney, S. Initiating base excision repair in chromatin. DNA Repair 2018, 71, 87–92. [Google Scholar] [CrossRef] [PubMed]
  62. Mullins, E.; Rodriguez, A.; Bradley, N.P.; Eichman, B.F. Emerging Roles of DNA Glycosylases and the Base Excision Repair Pathway. Trends Biochem. Sci. 2019, 44, 765–781. [Google Scholar] [CrossRef] [PubMed]
  63. Smith, A.J.; Ball, S.S.; Bowater, R.P.; Wormstone, I.M. PARP-1 inhibition influences the oxidative stress response of the human lens. Redox Biol. 2016, 8, 354–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ström, C.E.; Johansson, F.; Uhlen, M.; Szigyarto, C.A.-K.; Erixon, K.; Helleday, T. Poly (ADP-ribose) polymerase (PARP) is not involved in base excision repair but PARP inhibition traps a single-strand intermediate. Nucleic Acids Res. 2010, 39, 3166–3175. [Google Scholar] [CrossRef]
  65. Reynolds, P.; Cooper, S.; Lomax, M.; O’Neill, P. Disruption of PARP1 function inhibits base excision repair of a sub-set of DNA lesions. Nucleic Acids Res. 2015, 43, 4028–4038. [Google Scholar] [CrossRef] [Green Version]
  66. Satoh, M.S.; Lindahl, T. Role of poly(ADP-ribose) formation in DNA repair. Nature 1992, 356, 356–358. [Google Scholar] [CrossRef]
  67. Beck, C.; Robert, I.; Reina-San-Martin, B.; Schreiber, V.; Dantzer, F. Poly(ADP-ribose) polymerases in double-strand break repair: Focus on PARP1, PARP2 and PARP3. Exp. Cell Res. 2014, 329, 18–25. [Google Scholar] [CrossRef]
  68. Marintchev, A.; Robertson, A.; Dimitriadis, E.K.; Prasad, R.; Wilson, S.H.; Mullen, G.P. Domain specific interaction in the XRCC1-DNA polymerase beta complex. Nucleic Acids Res. 2000, 28, 2049–2059. [Google Scholar] [CrossRef] [Green Version]
  69. El-Khamisy, S.F.; Masutani, M.; Suzuki, H.; Caldecott, K.W. A requirement for PARP-1 for the assembly or stability of XRCC1 nuclear foci at sites of oxidative DNA damage. Nucleic Acids Res. 2003, 31, 5526–5533. [Google Scholar] [CrossRef] [Green Version]
  70. Oei, S.L.; Ziegler, M. ATP for the DNA Ligation Step in Base Excision Repair Is Generated from Poly(ADP-ribose). J. Biol. Chem. 2000, 275, 23234–23239. [Google Scholar] [CrossRef] [Green Version]
  71. Ruscetti, T.; Lehnert, B.E.; Halbrook, J.; Le Trong, H.; Hoekstra, M.F.; Chen, D.J.; Peterson, S.R. Stimulation of the DNA-dependent Protein Kinase by Poly(ADP-Ribose) Polymerase. J. Biol. Chem. 1998, 273, 14461–14467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Wang, M.; Wu, W.; Wu, W.; Rosidi, B.; Zhang, L.; Wang, H.; Iliakis, G. PARP-1 and Ku compete for repair of DNA double strand breaks by distinct NHEJ pathways. Nucleic Acids Res. 2006, 34, 6170–6182. [Google Scholar] [CrossRef] [PubMed]
  73. Schultz, N.; Lopez, E.; Saleh-Gohari, N.; Helleday, T. Poly(ADP-ribose) polymerase (PARP-1) has a controlling role in homologous recombination. Nucleic Acids Res. 2003, 31, 4959–4964. [Google Scholar] [CrossRef] [PubMed]
  74. Oikawa, A.; Tohda, H.; Kanai, M.; Miwa, M.; Sugimura, T. Inhibitors of poly(adenosine diphosphate ribose) polymerase induce sister chromatid exchanges. Biochem. Biophys. Res. Commun. 1980, 97, 1311–1316. [Google Scholar] [CrossRef]
  75. Zhang, F.; Shi, J.; Bian, C.; Yu, X. Poly(ADP-Ribose) Mediates the BRCA2-Dependent Early DNA Damage Response. Cell Rep. 2015, 13, 678–689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Hu, Y.; Petit, S.A.; Ficarro, S.B.; Toomire, K.J.; Xie, A.; Lim, E.; Cao, S.A.; Park, E.; Eck, M.J.; Scully, R.; et al. PARP1-Driven Poly-ADP-Ribosylation Regulates BRCA1 Function in Homologous Recombination–Mediated DNA Repair. Cancer Discov. 2014, 4, 1430–1447. [Google Scholar] [CrossRef] [Green Version]
  77. Soni, A.; Siemann, M.; Grabos, M.; Murmann, T.; Pantelias, G.E.; Iliakis, G. Requirement for Parp-1 and DNA ligases 1 or 3 but not of Xrcc1 in chromosomal translocation formation by backup end joining. Nucleic Acids Res. 2014, 42, 6380–6392. [Google Scholar] [CrossRef] [Green Version]
  78. Soni, A.; Siemann, M.; Pantelias, G.E.; Iliakis, G. Marked cell cycle-dependent contribution of alternative end joining to formation of chromosome translocations by stochastically induced DNA double strand breaks in human cells. Mutat. Res. 2015, 793, 2–8. [Google Scholar] [CrossRef]
  79. Dueva, R.; Iliakis, G. Alternative pathways of non-homologous end joining (NHEJ) in genomic instability and cancer. Transl. Cancer Res. 2013, 2, 163–177. [Google Scholar]
  80. Yang, G.; Liu, C.; Chen, S.-H.; Kassab, M.A.; Hoff, J.D.; Walter, N.G.; Yu, X. Super-resolution imaging identifies PARP1 and the Ku complex acting as DNA double-strand break sensors. Nucleic Acids Res. 2018, 46, 3446–3457. [Google Scholar] [CrossRef] [Green Version]
  81. Luedeman, M.E.; Stroik, S.; Feng, W.; Luthman, A.J.; Gupta, G.P.; Ramsden, D.A. Poly(ADP) ribose polymerase promotes DNA polymerase theta-mediated end joining by activation of end resection. Nat. Commun. 2022, 13, 1–10. [Google Scholar] [CrossRef]
  82. Ceccaldi, R.; Rondinelli, B.; D’Andrea, A.D. Repair Pathway Choices and Consequences at the Double-Strand Break. Trends Cell Biol. 2015, 26, 52–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Mesquita, K.A.; Alabdullah, M.; Griffin, M.; Toss, M.S.; Fatah, T.M.A.; Alblihy, A.; Moseley, P.; Chan, S.; Rakha, E.A.; Madhusudan, S. ERCC1-XPF deficiency is a predictor of olaparib induced synthetic lethality and platinum sensitivity in epithelial ovarian cancers. Gynecol. Oncol. 2019, 153, 416–424. [Google Scholar] [CrossRef] [PubMed]
  84. Trenner, A.; Sartori, A.A. Harnessing DNA Double-Strand Break Repair for Cancer Treatment. Front. Oncol. 2019, 9, 1388. [Google Scholar] [CrossRef] [Green Version]
  85. Vu, T.V.; Das, S.; Nguyen, C.C.; Kim, J.; Kim, J.Y. Single-strand annealing: Molecular mechanisms and potential applications in CRISPR-Cas-based precision genome editing. Biotechnol. J. 2022, 17, e2100413. [Google Scholar] [CrossRef]
  86. 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]
  87. Shen, Y.; Aoyagi-Scharber, M.; Wang, B. Trapping Poly(ADP-Ribose) Polymerase. J. Pharmacol. Exp. Ther. 2015, 353, 446–457. [Google Scholar] [CrossRef] [Green Version]
  88. Domchek, S.M.; Aghajanian, C.; Shapira-Frommer, R.; Schmutzler, R.K.; Audeh, M.W.; Friedlander, M.; Balmaña, J.; Mitchell, G.; Fried, G.; Stemmer, S.M.; et al. Efficacy and safety of olaparib monotherapy in germline BRCA1/2 mutation carriers with advanced ovarian cancer and three or more lines of prior therapy. Gynecol. Oncol. 2016, 140, 199–203. [Google Scholar] [CrossRef] [Green Version]
  89. Sisay, M.; Edessa, D. PARP inhibitors as potential therapeutic agents for various cancers: Focus on niraparib and its first global approval for maintenance therapy of gynecologic cancers. Gynecol. Oncol. Res. Pract. 2017, 4, 1–13. [Google Scholar] [CrossRef] [Green Version]
  90. Yoshihama, T.; Kuroda, Y.; Chiyoda, T.; Takahashi, M.; Yoshimura, T.; Saotome, K.; Nanki, Y.; Sakai, K.; Kobayashi, Y.; Yamagami, W.; et al. Efficacy and safety of olaparib maintenance monotherapy for Japanese patients with platinum-sensitive relapsed ovarian, fallopian tube, and primary peritoneal cancer. Int. J. Clin. Oncol. 2022, 27, 1644–1650. [Google Scholar] [CrossRef]
  91. Dockery, L.; Gunderson, C.; Moore, K. Rucaparib: The past, present, and future of a newly approved PARP inhibitor for ovarian cancer. Onco Targets Ther. 2017, 10, 3029–3037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Swisher, E.M.; Lin, K.K.; Oza, A.M.; Scott, C.L.; Giordano, H.; Sun, J.; Konecny, G.E.; Coleman, R.L.; Tinker, A.V.; O’Malley, D.M.; et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): An international, multicentre, open-label, phase 2 trial. Lancet Oncol. 2017, 18, 75–87. [Google Scholar] [CrossRef] [Green Version]
  93. Deeks, E.D. Olaparib: First global approval. Drugs 2015, 75, 231–240. [Google Scholar] [CrossRef] [PubMed]
  94. Eskiler, G.G. Talazoparib to treat BRCA-positive breast cancer. Drugs Today 2019, 55, 459–467. [Google Scholar] [CrossRef]
  95. Murai, J.; Huang, S.-Y.N.; 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]
  96. Kupczyk, P.; Simiczyjew, A.; Marczuk, J.; Dratkiewicz, E.; Beberok, A.; Rok, J.; Pieniazek, M.; Biecek, P.; Nevozhay, D.; Slowikowski, B.; et al. PARP1 as a Marker of an Aggressive Clinical Phenotype in Cutaneous Melanoma—A Clinical and an In Vitro Study. Cells 2021, 10, 286. [Google Scholar] [CrossRef]
  97. Raleigh, D.; Ahmed, K.M.; Zhang, H.; Ziaee, S.; Park, C.C. PARP-1 modulates β1-integrin/NF-κB-mediated radioresistance in human breast cancer. J. Cancer Ther. Res. 2016, 5, 1. [Google Scholar] [CrossRef] [Green Version]
  98. Liu, X.; Han, E.K.; Anderson, M.; Shi, Y.; Semizarov, D.; Wang, G.; McGonigal, T.; Roberts, L.; Lasko, L.; Palma, J.; et al. Acquired Resistance to Combination Treatment with Temozolomide and ABT-888 Is Mediated by Both Base Excision Repair and Homologous Recombination DNA Repair Pathways. Mol. Cancer Res. 2009, 7, 1686–1692. [Google Scholar] [CrossRef] [Green Version]
  99. Volinia, S.; Galasso, M.; Sana, M.E.; Wise, T.F.; Palatini, J.; Huebner, K.; Croce, C.M. Breast cancer signatures for invasiveness and prognosis defined by deep sequencing of microRNA. Proc. Natl. Acad. Sci. USA 2012, 109, 3024–3029. [Google Scholar] [CrossRef] [Green Version]
  100. Barber, L.J.; Sandhu, S.; Chen, L.; Campbell, J.; Kozarewa, I.; Fenwick, K.; Assiotis, I.; Rodrigues, D.N.; Reis-Filho, J.S.; Moreno, V.; et al. Secondary mutations in BRCA2 associated with clinical resistance to a PARP inhibitor. J. Pathol. 2012, 229, 422–429. [Google Scholar] [CrossRef]
  101. Kim, Y.; Kim, A.; Sharip, A.; Sharip, A.; Jiang, J.; Yang, Q.; Xie, Y. Reverse the Resistance to PARP Inhibitors. Int. J. Biol. Sci. 2017, 13, 198–208. [Google Scholar] [CrossRef] [PubMed]
  102. Montoni, A.; Robu, M.; Pouliot, E.; Shah, G.M. Resistance to PARP-Inhibitors in Cancer Therapy. Front. Pharmacol. 2013, 4, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Giudice, E.; Gentile, M.; Salutari, V.; Ricci, C.; Musacchio, L.; Carbone, M.V.; Ghizzoni, V.; Camarda, F.; Tronconi, F.; Nero, C.; et al. PARP Inhibitors Resistance: Mechanisms and Perspectives. Cancers 2022, 14, 1420. [Google Scholar] [CrossRef] [PubMed]
  104. Yap, T.A.; Plummer, R.; Azad, N.S.; Helleday, T. The DNA Damaging Revolution: PARP Inhibitors and Beyond. Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 185–195. [Google Scholar] [CrossRef]
  105. Jannetti, S.A.; Zeglis, B.M.; Zalutsky, M.R.; Reiner, T. Poly(ADP-Ribose)Polymerase (PARP) Inhibitors and Radiation Therapy. Front. Pharmacol. 2020, 11, 170. [Google Scholar] [CrossRef] [Green Version]
  106. Zhang, N.; Gao, Y.; Zeng, Z.; Luo, Y.; Jiang, X.; Zhang, J.; Li, J.; Zhang, J.; Gong, Y.; Xie, C. PARP inhibitor niraparib as a radiosensitizer promotes antitumor immunity of radiotherapy in EGFR-mutated non-small cell lung cancer. Clin. Transl. Oncol. Off. Publ. Fed. Span. Oncol. Soc. Natl. Cancer Inst. Mex. 2021, 23, 1827–1837. [Google Scholar] [CrossRef]
  107. Lesueur, P.; Chevalier, F.; El-Habr, E.A.; Junier, M.-P.; Chneiweiss, H.; Castera, L.; Müller, E.; Stefan, D.; Saintigny, Y. Radiosensitization Effect of Talazoparib, a Parp Inhibitor, on Glioblastoma Stem Cells Exposed to Low and High Linear Energy Transfer Radiation. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef]
  108. Abbotts, R.; Topper, M.J.; Biondi, C.; Fontaine, D.; Goswami, R.; Stojanovic, L.; Choi, E.Y.; McLaughlin, L.; Kogan, A.A.; Xia, L.; et al. DNA methyltransferase inhibitors induce a BRCAness phenotype that sensitizes NSCLC to PARP inhibitor and ionizing radiation. Proc. Natl. Acad. Sci. USA 2019, 116, 22609–22618. [Google Scholar] [CrossRef]
  109. Carter, R.; Cheraghchi-Bashi, A.; Westhorpe, A.; Yu, S.; Shanneik, Y.; Seraia, E.; Ouaret, D.; Inoue, Y.; Koch, C.; Wilding, J.; et al. Identification of anticancer drugs to radiosensitise BRAF-wild-type and mutant colorectal cancer. Cancer Biol. Med. 2019, 16, 234–246. [Google Scholar]
  110. Gerossier, L.; Dubois, A.; Paturel, A.; Fares, N.; Cohen, D.; Merle, P.; Lachuer, J.; Wierinckx, A.; Saintigny, P.; Bancel, B.; et al. PARP inhibitors and radiation potentiate liver cell death in vitro. Do hepatocellular carcinomas have an achilles’ heel? Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101553. [Google Scholar] [CrossRef]
  111. Jonuscheit, S.; Jost, T.; Gajdošová, F.; Wrobel, M.; Hecht, M.; Fietkau, R.; Distel, L. PARP Inhibitors Talazoparib and Niraparib Sensitize Melanoma Cells to Ionizing Radiation. Genes 2021, 12, 849. [Google Scholar] [CrossRef] [PubMed]
  112. de Bono, J.S.; Mina, L.A.; Gonzalez, M.; Curtin, N.J.; Wang, E.; Henshaw, J.W.; Chadha, M.; Sachdev, J.C.; Matei, D.; Jameson, G.S.; et al. First-in-human trial of novel oral PARP inhibtior BMN673 in patients with solid tumors. J. Clin. Oncol. 2013, 31, 2580. [Google Scholar] [CrossRef]
  113. Carney, B.; Kossatz, S.; Lok, B.H.; Schneeberger, V.; Gangangari, K.K.; Pillarsetty, N.V.K.; Weber, W.A.; Rudin, C.M.; Poirier, J.T.; Reiner, T. Target engagement imaging of PARP inhibitors in small-cell lung cancer. Nat. Commun. 2018, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  114. Loap, P.; Loirat, D.; Berger, F.; Ricci, F.; Vincent-Salomon, A.; Ezzili, C.; Mosseri, V.; Fourquet, A.; Ezzalfani, M.; Kirova, Y. Combination of Olaparib and Radiation Therapy for Triple Negative Breast Cancer: Preliminary Results of the RADIOPARP Phase 1 Trial. Int. J. Radiat. Oncol. 2020, 109, 436–440. [Google Scholar] [CrossRef] [PubMed]
  115. Boussios, S.; Abson, C.; Moschetta, M.; Rassy, E.; Karathanasi, A.; Bhat, T.; Ghumman, F.; Sheriff, M.; Pavlidis, N. Poly (ADP-Ribose) Polymerase Inhibitors: Talazoparib in Ovarian Cancer and Beyond. Drugs RD 2020, 20, 55–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Litton, J.K.; Rugo, H.S.; Ettl, J.; Hurvitz, S.A.; Gonçalves, A.; Lee, K.-H.; Fehrenbacher, L.; Yerushalmi, R.; Mina, L.A.; Martin, M.; et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. N. Engl. J. Med. 2018, 379, 753–763. [Google Scholar] [CrossRef] [PubMed]
  117. Ettl, J.; Quek, R.G.W.; Lee, K.H.; Rugo, H.S.; Hurvitz, S.; Gonçalves, A.; Fehrenbacher, L.; Yerushalmi, R.; Mina, L.A.; Martin, M.; et al. Quality of life with talazoparib versus physician’s choice of chemotherapy in patients with advanced breast cancer and germline BRCA1/2 mutation: Patient-reported outcomes from the EMBRACA phase III trial. Ann. Oncol. 2018, 29, 1939–1947. [Google Scholar] [CrossRef]
  118. Jagsi, R.; Griffith, K.A.; Bellon, J.R.; Woodward, W.; Horton, J.K.; Ho, A.; Feng, F.Y.; Speers, C.; Overmoyer, B.; Sabel, M.; et al. Concurrent Veliparib With Chest Wall and Nodal Radiotherapy in Patients With Inflammatory or Locoregionally Recurrent Breast Cancer: The TBCRC 024 Phase I Multicenter Study. J. Clin. Oncol. 2018, 36, 1317–1322. [Google Scholar] [CrossRef]
  119. Czito, B.G.; Deming, D.A.; Jameson, G.S.; Mulcahy, M.F.; Vaghefi, H.; Dudley, M.W.; Holen, K.D.; DeLuca, A.; Mittapalli, R.K.; Munasinghe, W.; et al. Safety and tolerability of veliparib combined with capecitabine plus radiotherapy in patients with locally advanced rectal cancer: A phase 1b study. Lancet Gastroenterol. Hepatol. 2017, 2, 418–426. [Google Scholar] [CrossRef]
  120. Chabot, P.; Hsia, T.C.; Ryu, J.S.; Gorbunova, V.; Belda-Iniesta, C.; Ball, D.; Kio, E.; Mehta, M.; Papp, K.; Qin, Q.; et al. Veliparib in combination with whole-brain radiation therapy for patients with brain metastases from non-small cell lung cancer: Results of a randomized, global, placebo-controlled study. J. Neurooncol. 2017, 131, 105–115. [Google Scholar] [CrossRef] [Green Version]
  121. Baxter, P.A.; Su, J.M.; Onar-Thomas, A.; Billups, C.A.; Li, X.-N.; Poussaint, T.Y.; Smith, E.R.; Thompson, P.; Adesina, A.; Ansell, P.; et al. A phase I/II study of veliparib (ABT-888) with radiation and temozolomide in newly diagnosed diffuse pontine glioma: A Pediatric Brain Tumor Consortium study. Neuro-Oncology 2020, 22, 875–885. [Google Scholar] [CrossRef] [PubMed]
  122. Sim, H.W.; McDonald, K.L.; Lwin, Z.; Barnes, E.H.; Rosenthal, M.; Foote, M.C.; Koh, E.S.; Back, M.; Wheeler, H.; Sulman, E.P.; et al. A randomized phase II trial of veliparib, radiotherapy, and temozolomide in patients with unmethylated MGMT glioblastoma: The VERTU study. Neuro-Oncology 2021, 23, 1736–1749. [Google Scholar] [CrossRef] [PubMed]
  123. Karam, S.D.; Reddy, K.; Blatchford, P.J.; Waxweiler, T.; DeLouize, A.M.; Oweida, A.; Somerset, H.; Marshall, C.; Young, C.; Davies, K.D.; et al. Final Report of a Phase I Trial of Olaparib with Cetuximab and Radiation for Heavy Smoker Patients with Locally Advanced Head and Neck Cancer. Clin. Cancer Res. 2018, 24, 4949–4959. [Google Scholar] [CrossRef] [Green Version]
  124. Ciccia, A.; Elledge, S.J. The DNA Damage Response: Making It Safe to Play with Knives. Mol. Cell 2010, 40, 179–204. [Google Scholar] [CrossRef]
  125. Mladenova, V.; Mladenov, E.; Stuschke, M.; Iliakis, G. DNA Damage Clustering after Ionizing Radiation and Consequences in the Processing of Chromatin Breaks. Molecules 2022, 27, 1540. [Google Scholar] [CrossRef]
  126. Lourenco, L.H.M.; Jiang, Y.; Drobnitzky, N.; Green, M.; Cahill, F.; Patel, A.; Shanneik, Y.; Moore, J.; Ryan, A.J. PARP Inhibition Combined With Thoracic Irradiation Exacerbates Esophageal and Skin Toxicity in C57BL6 Mice. Int. J. Radiat. Oncol. 2018, 100, 767–775. [Google Scholar] [CrossRef] [PubMed]
  127. Symington, L.S.; Gautier, J. Double-Strand Break End Resection and Repair Pathway Choice. Annu. Rev. Genet. 2011, 45, 247–271. [Google Scholar] [CrossRef] [PubMed]
  128. Zhao, F.; Kim, W.; Kloeber, J.A.; Lou, Z. DNA end resection and its role in DNA replication and DSB repair choice in mammalian cells. Exp. Mol. Med. 2020, 52, 1705–1714. [Google Scholar] [CrossRef]
  129. Caron, M.-C.; Sharma, A.K.; O’Sullivan, J.; Myler, L.R.; Ferreira, M.T.; Rodrigue, A.; Coulombe, Y.; Ethier, C.; Gagné, J.-P.; Langelier, M.-F.; et al. Poly(ADP-ribose) polymerase-1 antagonizes DNA resection at double-strand breaks. Nat. Commun. 2019, 10, 1–16. [Google Scholar] [CrossRef] [Green Version]
  130. Kong, E.A.Y.; Xu, C.; Sun, X.; Sun, H.; Zhao, X.; He, N.; Ji, K.; Wang, Q.; Du, L.; Wang, J.; et al. BLM helicase inhibition synergizes with PARP inhibition to improve the radiosensitivity of olaparib resistant non-small cell lung cancer cells by inhibiting homologous recombination repair. Cancer Biol. Med. 2021, 18, 1–32. [Google Scholar] [CrossRef]
  131. Mladenov, E.; Staudt, C.; Soni, A.; Murmann-Konda, T.; Siemann-Loekes, M.; Iliakis, G. Strong suppression of gene conversion with increasing DNA double-strand break load delimited by 53BP1 and RAD52. Nucleic Acids Res. 2019, 48, 1905–1924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Albala, J.S.; Thelen, M.P.; Prange, C.; Fan, W.; Christensen, M.; Thompson, L.H.; Lennon, G.G. Identification of a novel human RAD51 homolog, RAD51B. Genomics 1997, 48, 476–479. [Google Scholar] [CrossRef] [PubMed]
  133. Thacker, J. The RAD51 gene family, genetic instability and cancer. Cancer Lett. 2005, 219, 125–135. [Google Scholar] [CrossRef] [PubMed]
  134. So, A.; Dardillac, E.; Muhammad, A.; Chailleux, C.; Sesma-Sanz, L.; Ragu, S.; Le Cam, E.; Canitrot, Y.; Masson, J.Y.; Dupaigne, P.; et al. RAD51 protects against nonconservative DNA double-strand break repair through a nonenzymatic function. Nucleic Acids Res. 2022, 50, 2651–2666. [Google Scholar] [CrossRef]
  135. Kijas, A.W.; Lim, Y.C.; Bolderson, E.; Cerosaletti, K.; Gatei, M.; Jakob, B.; Tobias, F.; Taucher-Scholz, G.; Gueven, N.; Oakley, G.; et al. ATM-dependent phosphorylation of MRE11 controls extent of resection during homology directed repair by signalling through Exonuclease 1. Nucleic Acids Res. 2015, 43, 8352–8367. [Google Scholar] [CrossRef] [Green Version]
  136. Gunn, A.; Stark, J.M. I-SceI-based assays to examine distinct repair outcomes of mammalian chromosomal double strand breaks. Methods Mol. Biol. (Clifton N.J.) 2012, 920, 379–391. [Google Scholar]
  137. Jelinic, P.; Levine, D.A. New Insights into PARP Inhibitors’ Effect on Cell Cycle and Homology-Directed DNA Damage Repair. Mol. Cancer Ther. 2014, 13, 1645–1654. [Google Scholar] [CrossRef] [Green Version]
  138. Groth, P.; Orta, M.L.; Elvers, I.; Majumder, M.M.; Lagerqvist, A.; Helleday, T. Homologous recombination repairs secondary replication induced DNA double-strand breaks after ionizing radiation. Nucleic Acids Res. 2012, 40, 6585–6594. [Google Scholar] [CrossRef] [Green Version]
  139. Rothkamm, K.; Kruger, I.; Thompson, L.H.; Lobrich, M. Pathways of DNA Double-Strand Break Repair during the Mammalian Cell Cycle. Mol. Cell. Biol. 2003, 23, 5706–5715. [Google Scholar] [CrossRef] [Green Version]
  140. Feng, W.; Simpson, D.A.; Cho, J.-E.; Carvajal-Garcia, J.; Smith, C.M.; Headley, K.M.; Hathaway, N.; Ramsden, D.A.; Gupta, G.P. Marker-free quantification of repair pathway utilization at Cas9-induced double-strand breaks. Nucleic Acids Res. 2021, 49, 5095–5105. [Google Scholar] [CrossRef]
  141. Brinkman, E.K.; Chen, T.; de Haas, M.; Holland, H.A.; Akhtar, W.; van Steensel, B. Kinetics and Fidelity of the Repair of Cas9-Induced Double-Strand DNA Breaks. Mol. Cell 2018, 70, 801–813.e806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Sun, Q.; Guo, Y.; Liu, X.; Czauderna, F.; Carr, M.I.; Zenke, F.T.; Blaukat, A.; Vassilev, L.T. Therapeutic Implications of p53 Status on Cancer Cell Fate Following Exposure to Ionizing Radiation and the DNA-PK Inhibitor M3814. Mol. Cancer Res. 2019, 17, 2457–2468. [Google Scholar] [CrossRef] [Green Version]
  143. Dickreuter, E.; Eke, I.; Krause, M.; Borgmann, K.; van Vugt, M.A.; Cordes, N. Targeting of beta1 integrins impairs DNA repair for radiosensitization of head and neck cancer cells. Oncogene 2016, 35, 1353–1362. [Google Scholar] [CrossRef] [PubMed]
  144. Chughtai, A.A.; Pannhausen, J.; Dinger, P.; Wirtz, J.; Knüchel, R.; Gaisa, N.T.; Eble, M.J.; Rose, M. Effective Radiosensitization of Bladder Cancer Cells by Pharmacological Inhibition of DNA-PK and ATR. Biomedicines 2022, 10, 1277. [Google Scholar] [CrossRef] [PubMed]
  145. Soni, A.; Murmann-Konda, T.; Siemann-Loekes, M.; Pantelias, G.E.; Iliakis, G. Chromosome breaks generated by low doses of ionizing radiation in G2-phase are processed exclusively by gene conversion. DNA Repair 2020, 89, 102828. [Google Scholar] [CrossRef]
  146. Murmann-Konda, T.; Soni, A.; Stuschke, M.; Iliakis, G. Analysis of chromatid-break-repair detects a homologous recombination to non-homologous end-joining switch with increasing load of DNA double-strand breaks. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2021, 867, 503372. [Google Scholar] [CrossRef]
  147. Waters, C.A.; Strande, N.T.; Wyatt, D.W.; Pryor, J.M.; Ramsden, D.A. Nonhomologous end joining: A good solution for bad ends. DNA Repair 2014, 17, 39–51. [Google Scholar] [CrossRef] [Green Version]
  148. Wu, S.; Gao, F.; Zheng, S.; Zhang, C.; Martinez-Ledesma, E.; Ezhilarasan, R.; Ding, J.; Li, X.; Feng, N.; Multani, A.; et al. EGFR Amplification Induces Increased DNA Damage Response and Renders Selective Sensitivity to Talazoparib (PARP Inhibitor) in Glioblastoma. Clin. Cancer Res. 2020, 26, 1395–1407. [Google Scholar] [CrossRef]
  149. Bellare, G.P.; Saha, B.; Patro, B.S. Targeting autophagy reverses de novo resistance in homologous recombination repair proficient breast cancers to PARP inhibition. Br. J. Cancer 2021, 124, 1260–1274. [Google Scholar] [CrossRef]
  150. Pai Bellare, G.; Sankar Patro, B. Resveratrol sensitizes breast cancer to PARP inhibitor, talazoparib through dual inhibition of AKT and autophagy flux. Biochem. Pharmacol. 2022, 199, 115024. [Google Scholar] [CrossRef]
  151. Oh, S.; Harvey, A.; Zimbric, J.; Wang, Y.; Nguyen, T.; Jackson, P.J.; Hendrickson, E.A. DNA ligase III and DNA ligase IV carry out genetically distinct forms of end joining in human somatic cells. DNA Repair 2014, 21, 97–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Mansour, W.Y.; Rhein, T.; Dahm-Daphi, J. The alternative end-joining pathway for repair of DNA double-strand breaks requires PARP1 but is not dependent upon microhomologies. Nucleic Acids Res. 2010, 38, 6065–6077. [Google Scholar] [CrossRef] [PubMed]
  153. Kent, T.; Chandramouly, G.; McDevitt, S.M.; Ozdemir, A.Y.; Pomerantz, R.T. Mechanism of microhomology-mediated end-joining promoted by human DNA polymerase theta. Nat. Struct. Mol. Biol. 2015, 22, 230–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Masani, S.; Han, L.; Meek, K.; Yu, K. Redundant function of DNA ligase 1 and 3 in alternative end-joining during immunoglobulin class switch recombination. Proc. Natl. Acad. Sci. USA 2016, 113, 1261–1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Ceccaldi, R.; Liu, J.C.; Amunugama, R.; Hajdu, I.; Primack, B.; Petalcorin, M.I.R.; O’Connor, K.W.; Konstantinopoulos, P.A.; Elledge, S.J.; Boulton, S.J.; et al. Homologous-recombination-deficient tumours are dependent on Pol[thgr]-mediated repair. Nature 2015, 518, 258–262. [Google Scholar] [CrossRef]
  156. Wyatt, D.W.; Feng, W.; Conlin, M.P.; Yousefzadeh, M.J.; Roberts, S.A.; Mieczkowski, P.; Wood, R.D.; Gupta, G.P.; Ramsden, D.A. Essential Roles for Polymerase theta-Mediated End Joining in the Repair of Chromosome Breaks. Mol. Cell 2016, 63, 662–673. [Google Scholar] [CrossRef] [Green Version]
  157. Iliakis, G.; Murmann, T.; Soni, A. Alternative end-joining repair pathways are the ultimate backup for abrogated classical non-homologous end-joining and homologous recombination repair: Implications for the formation of chromosome translocations. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2015, 793, 166–175. [Google Scholar] [CrossRef]
  158. Schipler, A.; Mladenova, V.; Soni, A.; Nikolov, V.; Saha, J.; Mladenov, E.; Iliakis, G. Chromosome thripsis by DNA double strand break clusters causes enhanced cell lethality, chromosomal translocations and 53BP1-recruitment. Nucleic Acids Res. 2016, 44, 7673–7690. [Google Scholar] [CrossRef] [Green Version]
  159. Wray, J.; Williamson, E.A.; Singh, S.B.; Wu, Y.; Cogle, C.; Weinstock, D.M.; Zhang, Y.; Lee, S.-H.; Zhou, D.; Shao, L.; et al. PARP1 is required for chromosomal translocations. Blood 2013, 121, 4359–4365. [Google Scholar] [CrossRef] [Green Version]
  160. Bai, W.; Zhu, G.; Xu, J.; Chen, P.; Meng, F.; Xue, H.; Chen, C.; Dong, J. The 3′-flap endonuclease XPF-ERCC1 promotes alternative end joining and chromosomal translocation during B cell class switching. Cell Rep. 2021, 36, 109756. [Google Scholar] [CrossRef]
  161. Bunting, S.F.; Nussenzweig, A. End-joining, translocations and cancer. Nature Reviews. Cancer 2013, 13, 443–454. [Google Scholar] [PubMed] [Green Version]
  162. Soutoglou, E.; Misteli, T. Mobility and immobility of chromatin in transcription and genome stability. Curr. Opin. Genet. Dev. 2007, 17, 435–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Zatreanu, D.; Robinson, H.M.R.; Alkhatib, O.; Boursier, M.; Finch, H.; Geo, L.; Grande, D.; Grinkevich, V.; Heald, R.A.; Langdon, S.; et al. Polθ inhibitors elicit BRCA-gene synthetic lethality and target PARP inhibitor resistance. Nat. Commun. 2021, 12, 1–15. [Google Scholar] [CrossRef] [PubMed]
  164. Zhou, J.; Gelot, C.; Pantelidou, C.; Li, A.; Yücel, H.; Davis, R.E.; Färkkilä, A.; Kochupurakkal, B.; Syed, A.; Shapiro, G.I.; et al. A first-in-class polymerase theta inhibitor selectively targets homologous-recombination-deficient tumors. Nat. Cancer 2021, 2, 598–610. [Google Scholar] [CrossRef]
  165. Hopkins, T.A.; Shi, Y.; Rodriguez, L.E.; Solomon, L.R.; Donawho, C.K.; DiGiammarino, E.L.; Panchal, S.C.; Wilsbacher, J.L.; Gao, W.; Olson, A.M.; et al. Mechanistic Dissection of PARP1 Trapping and the Impact on In Vivo Tolerability and Efficacy of PARP Inhibitors. Mol. Cancer Res. 2015, 13, 1465–1477. [Google Scholar] [CrossRef] [PubMed]
  166. Krastev, D.B.; Li, S.; Sun, Y.; Wicks, A.J.; Hoslett, G.; Weekes, D.; Badder, L.M.; Knight, E.G.; Marlow, R.; Pardo, M.C.; et al. The ubiquitin-dependent ATPase p97 removes cytotoxic trapped PARP1 from chromatin. Nature 2022, 24, 62–73. [Google Scholar] [CrossRef]
  167. Juhász, S.; Smith, R.; Schauer, T.; Spekhardt, D.; Mamar, H.; Zentout, S.; Chapuis, C.; Huet, S.; Timinszky, G. The chromatin remodeler ALC1 underlies resistance to PARP inhibitor treatment. Sci. Adv. 2020, 6, eabb8626. [Google Scholar] [CrossRef]
Figure 1. Domain structure of human PARP-1, PARP-2, and PARP-3 proteins. The domain locations within the full-length proteins were extracted from the uniport database by using the following entries for PARP-1 (P09874), PARP-2 (Q9UGN5), and PARP-3 (Q9Y6F1). Main protein domains involved in DNA-binding, auto-modification, and catalytic functions are indicated. ZnFI: zinc finger I domain, ZnFII: zinc finger II domain, ZnFIII: zinc finger III domain, BRCT: BRCA1 C-terminal domain, WGR: Trp-Gly-Arg domain, HD: Helical domain, ART: (ADP-ribosyl) transferase domain. ZnFI and ZnFII help to recognize and bind to DNA damage sites, while ZnFIII activates the enzyme upon DNA binding. The BRCT domain of PARP-1 is required to localize XRCC1 and XRCC1-complexed DNA repair factors to DNA damage sites.
Figure 1. Domain structure of human PARP-1, PARP-2, and PARP-3 proteins. The domain locations within the full-length proteins were extracted from the uniport database by using the following entries for PARP-1 (P09874), PARP-2 (Q9UGN5), and PARP-3 (Q9Y6F1). Main protein domains involved in DNA-binding, auto-modification, and catalytic functions are indicated. ZnFI: zinc finger I domain, ZnFII: zinc finger II domain, ZnFIII: zinc finger III domain, BRCT: BRCA1 C-terminal domain, WGR: Trp-Gly-Arg domain, HD: Helical domain, ART: (ADP-ribosyl) transferase domain. ZnFI and ZnFII help to recognize and bind to DNA damage sites, while ZnFIII activates the enzyme upon DNA binding. The BRCT domain of PARP-1 is required to localize XRCC1 and XRCC1-complexed DNA repair factors to DNA damage sites.
Cancers 14 05619 g001
Figure 2. A central and critical role of PARP-1 in DNA repair. PARP-1 is the prime target of clinical PARPi and mediates multiple DNA repair processes (Please refer to Section 2 for details).
Figure 2. A central and critical role of PARP-1 in DNA repair. PARP-1 is the prime target of clinical PARPi and mediates multiple DNA repair processes (Please refer to Section 2 for details).
Cancers 14 05619 g002
Figure 3. A model of altered DSB repair pathway balance at low and high IR doses in BMN673-treated cells. See text for details.
Figure 3. A model of altered DSB repair pathway balance at low and high IR doses in BMN673-treated cells. See text for details.
Cancers 14 05619 g003
Table 1. Published Clinical Studies on PARPi combined with RT.
Table 1. Published Clinical Studies on PARPi combined with RT.
Trial PhaseDisease SettingHR StatusTreatmentsEligible Enrolled PatientsEfficacyRef.
IInflammatory or loco-regionally recurrent breast cancerN/ART + veliparib303-year OS-56.6%, PFS -50%[118]
ILocally advanced rectal cancerN/ART + capecitabine + veliparib32Tumor downstaging after surgery-71%; pCR-29%[119]
IIBrain metastases from NSCLCN/AWBRT +/− veliparibRandomization (1:1:1);WBRT alone[120]
103 in 50mg veliparib arm, 102 in 200 mg veliparib arm, 102 in control armMedian OS: 185 days
WBRT+50 mg veliparib
Median OS: 209 days (p = 0.933)
WBRT+200 mg veliparib
Median OS: 209 days (p = 0.909)
I/IIDiffuse pontine gliomasN/ART + veliparib65Phase I[121]
PR(0%) SD-91.7% PD(8.3%)
Phase II
PR(13.2%) SD-71.7% PD-9.4%
IIUnmethyated MGMT glioblastomaN/ART+ temozolomide +/− veliparibRandomization (2:1)Without veliparib[122]
84 in veliparib arm and 41 in control armPFS at 6 months-31%. Median OS: 12.8months
With veliparib
PFS at 6 months (46%). Median OS: 12.7 months
IHead and neck squamous cell carcinomaN/Aolaparib + RT + cetuximab15Median OS: 37 months[123]
2-year OS (72%), PFS (63%)
N/A: not applicable; RT: radiotherapy; OS: overall survival; PFS: progression free survival; pCR: pathological complete response; WBRT: whole brain radiation therapy; PR: partial response; SD: stable disease; PD: progressive disease.
Table 2. Registered clinical studies of BMN673 in combination with RT.
Table 2. Registered clinical studies of BMN673 in combination with RT.
Trial PhaseDisease SettingHR StatusTreatmentsTrial StatusIdentifier
IExtensive-stage small cell lung cancerN/ABMN673 + low dose consolidative thoracic RTRecruitingNCT04170946
ILocally recurrent gynecologic cancersN/ABMN673 + RTRecruitingNCT03968406
IIMetastatic triple negative breast cancergBRCA 1/2 negativeBMN673 + Atezolizumab + SBRTRecruitingNCT04690855
N/A: not applicable; RT: radiotherapy; gBRCA: germline Breast cancer gene 1/2; SBRT: Stereotactic body radiation therapy.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Soni, A.; Lin, X.; Mladenov, E.; Mladenova, V.; Stuschke, M.; Iliakis, G. BMN673 Is a PARP Inhibitor with Unique Radiosensitizing Properties: Mechanisms and Potential in Radiation Therapy. Cancers 2022, 14, 5619. https://doi.org/10.3390/cancers14225619

AMA Style

Soni A, Lin X, Mladenov E, Mladenova V, Stuschke M, Iliakis G. BMN673 Is a PARP Inhibitor with Unique Radiosensitizing Properties: Mechanisms and Potential in Radiation Therapy. Cancers. 2022; 14(22):5619. https://doi.org/10.3390/cancers14225619

Chicago/Turabian Style

Soni, Aashish, Xixi Lin, Emil Mladenov, Veronika Mladenova, Martin Stuschke, and George Iliakis. 2022. "BMN673 Is a PARP Inhibitor with Unique Radiosensitizing Properties: Mechanisms and Potential in Radiation Therapy" Cancers 14, no. 22: 5619. https://doi.org/10.3390/cancers14225619

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