Abstract
Clinical interest in poly(ADP-ribose) polymerase 1 (PARP-1) has increased over the past decade with the recognition of its roles in transcription regulation, DNA repair, epigenetic bookmarking, and chromatin restructuring. A number of PARP-1 inhibitors demonstrating clinical efficacy against tumors of various origins have emerged in recent years. These inhibitors have been essentially designed as NAD+ mimetics. However, because NAD+ is utilized by many enzymes other than PARP-1, NAD+ competitors tend to produce certain off-target effects. To overcome the limitation of NAD-like PARP-1 inhibitors, we have developed a new class of PARP-1 inhibitors that specifically targets the histone-dependent route of PARP-1 activation, a mechanism of activation that is unique to PARP-1. Novel histone-dependent inhibitors are highly specific for PARP-1 and demonstrate promising in vitro and in vivo efficacy against prostate and renal tumors. Our findings suggest that novel PARP-1 inhibitors have strong therapeutic potential for the treatment of urological tumors.
Keywords: PARP-1 inhibitors, NAD+, histone-dependent PARP-1 regulation, renal cell carcinoma, prostate cancer
Introduction
Poly(ADP-ribose) polymerase 1 (PARP-1) is an abundant and ubiquitous nuclear enzyme. When active, it captures NAD+ to assemble long and branching polymers of poly(ADP-ribose) (pADPr) covalently modifying itself and surrounding proteins [1]. One of the most common and well-studied activities of PARP-1 is its role in DNA repair [2, 3]. Inhibition of PARP-1 may be lethal if combined with a loss-of-function of certain DNA repair genes, including BRCA1, BRCA2, PTEN, ATM, CHEK2, FANCA, and several others. These genes are frequently mutated in various malignancies [4–8]. Thus, tumor cells harboring defects in DNA repair pathways can be selectively targeted with PARP-1 inhibitors. In addition to its DNA repair function, PARP-1 participates in many other nuclear processes, including ribosome biogenesis, regulation of chromatin, nuclear traffic, and epigenetic bookmarking [9–11]. PARP-1 is also involved in transcriptional regulation by promoting an ‘open’ chromatin conformation favoring transcriptional activation [12, 13] and by serving as a cofactor of different pro-tumorigenic transcription factors, most notably NF-κB and AP-1 [14, 15].
In prostate cancer (PC), the PARP-1 function is critical for transcriptional activity of the androgen receptor (AR) [16]. PARP-1 is recruited to sites of the AR transcriptional function; PARP-1 enzymatic activity is required for the AR-driven gene expression and subsequent proliferation of androgen-dependent and castration-resistant PC (CRPC) cells [16, 17]. PARP-1 inhibition results in attenuation of the AR transcriptional activity and, therefore, interferes with androgen-dependent and -independent modes of the AR activation, whereas conventional antiandrogens, such as abiraterone (Zytiga) and enzalutamide (Xtandi), target only androgen-mediated activation of the AR [18]. PARP-1 also enhances the stability and accumulation of hypoxia-inducible factor alpha subunits (HIF-1α and HIF-2α) [19, 20], arguably the most common factors in renal carcinogenesis [19–22]. Therefore, the inhibition of PARP-1 activity represents a powerful therapeutic strategy against tumors of various origins and has been a field of intense investigation over the last two decades.
The PARP-1 pathway is aberrantly activated in several types of tumors, including prostate and kidney cancers [23–27]. Our studies reveal that the expression levels of PARP-1 and pADPr, a marker of PARP-1 activity, are significantly elevated in PC cells, relative to those in normal prostate epithelial cells. In contrast, PC cells display a loss of poly(ADP-Ribose) glycohydrolase (PARG) [27], an enzymatic antagonist of PARP-1. Furthermore, the overall pADPr levels are significantly elevated in CRPC cells compared with androgen-dependent PC cells [16]. We have also found that levels of pADPr are significantly elevated in ccRCC cell lines compared to those in normal kidney epithelial cells. pADPr expression was also augmented in the specimens of primary ccRCC tumors compared with corresponding normal kidney tissue samples. Notably, severe misregulation of PARP-1 activity was observed in all examined metastatic ccRCC tumor specimens (unpublished results). Cancer cells sensitive to PARP-1 inhibitors display a loss of PARG expression and produce excessive amounts of pADPr [26, 28]. Increased accumulation of pADPr appears to be the best predictor of tumor sensitivity to PARP-1 inhibition [2, 26, 28].
PARP inhibitors: mechanisms of action
PARP-1 activity can be regulated by three mechanisms: 1) competitive binding with nicotinamide adenine dinucleotide (NAD) [1]; 2) disruption of the interaction of PARP-1 with histones [29]; and 3) obstruction of PARP-1 binding with DNA [27]. Pharmacological PARP-1 inhibitors have been designed as NAD+ competitors and represent various memes of nicotinamide pharmacophore [30–32]. However, because NAD+ is extremely abundant and ubiquitous in living cells and is utilized by many enzymes other than PARP-1, NAD+ competitors tend to produce off-target effects. Multiple studies show that NAD-like PARP-1 inhibitors can affect distant unrelated targets, showing inter-family polypharmacology (promiscuous activities at targets of different families) and intra-family polypharmacology (non-selective activities at targets of the same family) as well as multi-signaling polypharmacology (multi-signaling activities mediated by the same target) [33–35]. Examples of off-target effects include inhibitory activities against mono-ADP-ribosyl-transferases and sirtuins, which control vital metabolic processes [34, 36, 37]. Besides binding PARP-1, most of the clinically relevant inhibitors, including olaparib, ABT-888 (veliparib,) and rucaparib, also bind to other NAD-dependent enzymes, suggesting that these compounds lack specificity and have promiscuous inhibitory activity [34, 37]. The most common dose-limiting toxicities of PARP-1 inhibitors include anemia, neutropenia, and thrombocytopenia [38]. A small number of fatal cases of myelodysplastic syndrome and acute myeloid leukemia have been reported [39–42]. In addition to the risk of undesirable off-target interactions, the polypharmacologic effects of current NAD-like PARP-1 inhibitors may interfere with the interpretation of data obtained in basic biological research and clinical outcome studies.
Screening for novel histone-dependent PARP-1 inhibitors
To overcome the limitation of NAD-like PARP-1 inhibitors, we have developed a new class of PARP-1 inhibitors that specifically targets the histone-dependent route of PARP-1 activation, a mechanism of activation that is unique to PARP-1. To establish a screening platform, we designed a PARP-1 activation assay in a 384-well ELISA plate coated with histone H4 protein-activator [43]. PARP-1 reactions were performed in each well by incubating recombinant PARP-1 enzyme and NAD+ in the presence of a single small molecule compound or a positive and a negative control. We were able to detect compounds that could disrupt PARP-1 interaction with histone H4, compete with NAD+, or abolish the accumulation of poly (ADP)-ribose, the product of these reactions, by measuring the levels of pADPr, which were used as an indicator of PARP-1 activity [43]. Screening a ~50,000 member small-molecule library identified 903 small molecules which inhibit PARP-1 in a cell-free system. After eliminating redundancies that displayed negligible structural differences, we reanalyzed 639 selected compounds and confirmed that all strong positive hits were 100% reproducible. A total of 373 small molecules in this list inhibited PARP-1 with the same, or better efficacy than the commonly used NAD-like PARP-1 inhibitors.
To narrow down the list of small molecules for further analysis, we used a computational approach to eliminate molecules with a structural similarity to known biologically active molecules. We identified a group of 17 small molecules that showed no significant structural similarity to known inhibitors and NAD+. Moreover, these compounds had no obvious structural homologues among any components of eukaryotic enzymatic pathways. All these molecules showed a strong capacity to block PARP-1 interaction with histone H4 in a cell-free system. Structurally, these molecules could be split into two subgroups: the first contain N-methylpiperidin in its the core element (2-(N-methylpiperidin-1-yl)acetate or 2-(N-methylmorpholino)acetate or 2-(N-methylpyrrolidine-1-yl)acetate); the second contains 1,3-dioxolane-4-yl)methyl, with 1-((1,3-dioxolane-4-yl)methyl)piperidine or 1-((1,3-dioxolane-4-yl)methyl)N-methylmorpholino or 1-((1,3-dioxolane-4-yl)methyl) N-methylpyrrolidine as core elements [43]. Based on anti-PARP-1 activity, ease of synthesis, and structural analysis we ultimately selected a lead hit 5F02 [18, 30]. As it was confirmed by inosine-5′-monophosphate dehydrogenase (IMPDH) activity assay, 5F02 showed no significant reduction of NADH production in contrast to olaparib, which is a structural analogue of NAD+ and competes with NAD+ to reduce the production of NADH in the IMPDH reaction [30]. The results of this experiment indicate that non-NAD-like inhibitors like 5F02 do not affect metabolic pathways associated with NAD+.
Given that inhibiting PARP-1 in a cell-free system does not warrant activity of a compound in cell-based assays, we tested whether the new compounds were capable of inhibiting PARP-1 activity in a cell-based system. All 17 molecules identified by our screen potently inhibited PARP-1 in a panel of human prostate and ccRCC cell lines [30]. Dose-dependent decreases in clonogenic survival and cell viability were observed upon treatment of prostate and kidney cancer cells with histone-dependent PARP-1 inhibitors [18, 30].
Pre-clinical evaluation of the efficacy of histone-dependent PARP-1 inhibitors against ccRCC and prostate cancer
Notably, the mechanism of action of histone-dependent PARP-1 inhibitors is completely different from that of the “classical” NAD-like PARP-1 inhibitors. NAD-like PARP-1 inhibitors mediate their antitumor effect through two general mechanisms: (i) catalytic inhibition of PARP-1 and (ii) locking or ‘trapping’ PARP-1 on damaged DNA [44]. The results of our experiments are in agreement with these findings indicating that NAD-like inhibitors stabilize binding of PARP-1 to the activator histone H4 arresting PARP-1-activator complex in a transient conformation. In contrast, histone-dependent inhibitors disrupt PARP-1 binding to histone H4 and exclude PARP-1 from functional complexes [18, 30, 45]. Our experimental data support this premise, demonstrating that histone-dependent inhibitors suppress PARP-1-mediated transcription more potently than NAD-like inhibitors (unpublished results). Indeed, the expression of vascular endothelial growth factor (VEGF) was markedly suppressed in patient-derived PNX0010 ccRCC cells treated with 5F02, whereas treatment with olaparib had minimal effect on the expression of VEGF (unpublished results). Treatment with 5F02 inhibited the AR transcriptional activity and prostate-specific antigen (PSA) expression in LNCaP PC cells expressing either the wild-type AR or constitutively active AR splice variant, AR-V7 [18]. In contrast, treatment with the NAD-competing inhibitor olaparib and antiandrogen enzalutamide produced only negligible inhibitory effect on the AR transcriptional activity in LNCaP cells expressing AR-V7. Furthermore, 5F02 exerted significantly higher inhibitory effect on viability of LNCaP cells expressing AR-V7 compared with olaparib and enzalutamide. Collectively, our data demonstrate that unlike NAD-like PARP-1 inhibitors and antiandrogens that target androgen-mediated activation of the AR, histone-dependent PARP-1 inhibitors suppress the AR transcriptional function and, therefore, may be effective against both androgen-dependent and -independent routes of the AR activation. Furthermore, 5F02 demonstrated potent antitumor activity against true castration-resistant AR-negative PC-3 and DU-145 PC cells [18, 30].
In light of these encouraging in vitro data, we tested the antitumor activity of 5F02 using PNX0010 renal cell carcinoma and castration-resistant AR-negative PC-3 PC xenograft animal models. Animals treated with 5F02 showed a significantly stronger inhibition of tumor growth relative to control animals and animals treated with the classical PARP-1 inhibitor olaparib [30]. Moreover, 5F02 demonstrated superior in vivo antitumor activity compared with clinically relevant anticancer drugs, i.e., docetaxel for prostate xenograft tumors and sunitinib for ccRCC xenograft tumors [34].
Conclusions
Clinical interest in PARP-1 has increased over the past decade with the recognition of its roles in transcription regulation, DNA repair, epigenetic bookmarking, and chromatin restructuring. Currently, over 50 clinical studies are being carried out to evaluate PARP-1 inhibitors for the treatment of solid and hematological malignancies. Given the initial promising results, the efforts are now focused on finding more effective and specific PARP-1 inhibitors. Our group was the first to identify the agents that specifically target the histone-dependent route of PARP-1 activation, a mechanism of activation that is unique to PARP-1. Greater specificity and selectivity of new inhibitors compared with the conventional NAD-like PARP-1 inhibitors is expected. The new histone-dependent PARP-1 inhibitors demonstrate higher efficacy in in vitro and in vivo settings against prostate and renal tumors compared to the classical NAD-like PARP-1 inhibitors. Taken together, our findings suggest that histone-dependent PARP-1 inhibitors have strong therapeutic potential for the treatment of urological tumors.
Funding
This work was supported in part by the Department of Defense Grants PC160049 (to AVT) and KC170127 (to VMK) and the National Institutes of Health Grants R03 CA212949 (to VMK), R03 CA235060 (to VMK), and R03 CA216173 (to PM).
Footnotes
Conflict of interest
None
References
- [1].D’Amours D, Desnoyers S, D’Silva I, Poirier GG. Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem J. 1999;342 (Pt 2):249–68. [PMC free article] [PubMed] [Google Scholar]
- [2].Swindall AF, Stanley JA, Yang ES. PARP-1: Friend or Foe of DNA Damage and Repair in Tumorigenesis? Cancers. 2013;5:943–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Pascal JM. The comings and goings of PARP-1 in response to DNA damage. DNA Repair (Amst). 2018;71:177–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434:917–21. [DOI] [PubMed] [Google Scholar]
- [5].Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434:913–7. [DOI] [PubMed] [Google Scholar]
- [6].Mendes-Pereira AM, Martin SA, Brough R, McCarthy A, Taylor JR, Kim JS, et al. Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO molecular medicine. 2009;1:315–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, et al. Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res. 2006;66:8109–15. [DOI] [PubMed] [Google Scholar]
- [8].Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N Engl J Med. 2015;373:1697–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Thomas C, Tulin AV. Poly-ADP-ribose polymerase: machinery for nuclear processes. Molecular aspects of medicine. 2013;34:1124–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Kim MY, Mauro S, Gevry N, Lis JT, Kraus WL. NAD+-dependent modulation of chromatin structure and transcription by nucleosome binding properties of PARP-1. Cell. 2004;119:803–14. [DOI] [PubMed] [Google Scholar]
- [11].Weaver AN, Yang ES. Beyond DNA Repair: Additional Functions of PARP-1 in Cancer. Front Oncol. 2013;3:290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Moses RE, O’Malley BW. DNA transcription and repair: a confluence. J Biol Chem. 2012;287:23266–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Krishnakumar R, Gamble MJ, Frizzell KM, Berrocal JG, Kininis M, Kraus WL. Reciprocal binding of PARP-1 and histone H1 at promoters specifies transcriptional outcomes. Science. 2008;319:819–21. [DOI] [PubMed] [Google Scholar]
- [14].Aguilar-Quesada R, Munoz-Gamez JA, Martin-Oliva D, Peralta-Leal A, Quiles-Perez R, Rodriguez-Vargas JM, et al. Modulation of transcription by PARP-1: consequences in carcinogenesis and inflammation. Current medicinal chemistry. 2007;14:1179–87. [DOI] [PubMed] [Google Scholar]
- [15].Castri P, Lee YJ, Ponzio T, Maric D, Spatz M, Bembry J, et al. Poly(ADP-ribose) polymerase-1 and its cleavage products differentially modulate cellular protection through NF-kappaB-dependent signaling. Biochim Biophys Acta. 2014;1843:640–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Schiewer MJ, Goodwin JF, Han S, Brenner JC, Augello MA, Dean JL, et al. Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov. 2012;2:1134–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Schiewer MJ, Knudsen KE. Transcriptional roles of PARP1 in cancer. Mol Cancer Res. 2014;12:1069–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Karpova Y, Wu C, Divan A, McDonnell ME, Hewlett E, Makhov P, et al. Non-NAD-like PARP-1 inhibitors in prostate cancer treatment. Biochem Pharmacol. 2019;167:149–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Elser M, Borsig L, Hassa PO, Erener S, Messner S, Valovka T, et al. Poly(ADP-ribose) polymerase 1 promotes tumor cell survival by coactivating hypoxia-inducible factor-1-dependent gene expression. Mol Cancer Res. 2008;6:282–90. [DOI] [PubMed] [Google Scholar]
- [20].Gonzalez-Flores A, Aguilar-Quesada R, Siles E, Pozo S, Rodriguez-Lara MI, Lopez-Jimenez L, et al. Interaction between PARP-1 and HIF-2alpha in the hypoxic response. Oncogene. 2014;33:891–8. [DOI] [PubMed] [Google Scholar]
- [21].Gudas LJ, Fu L, Minton DR, Mongan NP, Nanus DM. The role of HIF1alpha in renal cell carcinoma tumorigenesis. Journal of molecular medicine. 2014;92:825–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Baldewijns MM, van Vlodrop IJ, Vermeulen PB, Soetekouw PM, van Engeland M, de Bruine AP. VHL and HIF signalling in renal cell carcinogenesis. J Pathol. 2010;221:125–38. [DOI] [PubMed] [Google Scholar]
- [23].Wu W, Zhu H, Liang Y, Kong Z, Duan X, Li S, et al. Expression of PARP-1 and its active polymer PAR in prostate cancer and benign prostatic hyperplasia in Chinese patients. Int Urol Nephrol. 2014;46:1345–9. [DOI] [PubMed] [Google Scholar]
- [24].Salemi M, Galia A, Fraggetta F, La Corte C, Pepe P, La Vignera S, et al. Poly (ADP-ribose) polymerase 1 protein expression in normal and neoplastic prostatic tissue. Eur J Histochem. 2013;57:e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Yun EJ, Lin CJ, Dang A, Hernandez E, Guo J, Chen WM, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Gottipati P, Vischioni B, Schultz N, Solomons J, Bryant HE, Djureinovic T, et al. Poly(ADP-ribose) polymerase is hyperactivated in homologous recombination-defective cells. Cancer Res. 2010;70:5389–98. [DOI] [PubMed] [Google Scholar]
- [27].Kirsanov KI, Kotova E, Makhov P, Golovine K, Lesovaya EA, Kolenko VM, et al. Minor grove binding ligands disrupt PARP-1 activation pathways. Oncotarget. 2014;5:428–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Tulin A Re-evaluating PARP1 inhibitor in cancer. Nature biotechnology. 2011;29:1078–9. [DOI] [PubMed] [Google Scholar]
- [29].Pinnola A, Naumova N, Shah M, Tulin AV. Nucleosomal core histones mediate dynamic regulation of poly(ADP-ribose) polymerase 1 protein binding to chromatin and induction of its enzymatic activity. J Biol Chem. 2007;282:32511–9. [DOI] [PubMed] [Google Scholar]
- [30].Thomas C, Ji Y, Lodhi N, Kotova E, Pinnola AD, Golovine K, et al. Non-NAD-Like poly(ADP-Ribose) Polymerase-1 Inhibitors effectively Eliminate Cancer in vivo. EBioMedicine. 2016;13:90–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Javle M, Curtin NJ. The potential for poly (ADP-ribose) polymerase inhibitors in cancer therapy. Ther Adv Med Oncol. 2011;3:257–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Murai J, Huang SY, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Ekblad T, Camaioni E, Schuler H, Macchiarulo A. PARP inhibitors: polypharmacology versus selective inhibition. The FEBS journal. 2013;280:3563–75. [DOI] [PubMed] [Google Scholar]
- [34].Liscio P, Camaioni E, Carotti A, Pellicciari R, Macchiarulo A. From polypharmacology to target specificity: the case of PARP inhibitors. Curr Top Med Chem. 2013;13:2939–54. [DOI] [PubMed] [Google Scholar]
- [35].Passeri D, Camaioni E, Liscio P, Sabbatini P, Ferri M, Carotti A, et al. Concepts and Molecular Aspects in the Polypharmacology of PARP-1 Inhibitors. ChemMedChem. 2016;11:1219–26. [DOI] [PubMed] [Google Scholar]
- [36].Mangerich A, Burkle A. How to kill tumor cells with inhibitors of poly(ADP-ribosyl)ation. Int J Cancer. 2011;128:251–65. [DOI] [PubMed] [Google Scholar]
- [37].Wahlberg E, Karlberg T, Kouznetsova E, Markova N, Macchiarulo A, Thorsell AG, et al. Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitors. Nature biotechnology. 2012;30:283–8. [DOI] [PubMed] [Google Scholar]
- [38].Zhou JX, Feng LJ, Zhang X. Risk of severe hematologic toxicities in cancer patients treated with PARP inhibitors: a meta-analysis of randomized controlled trials. Drug Des Devel Ther. 2017;11:3009–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Sonnenblick A, de Azambuja E, Azim HA Jr., Piccart M. An update on PARP inhibitors--moving to the adjuvant setting. Nat Rev Clin Oncol. 2015;12:27–41. [DOI] [PubMed] [Google Scholar]
- [40].Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial. Lancet. 2010;376:235–44. [DOI] [PubMed] [Google Scholar]
- [41].Wagner LM. Profile of veliparib and its potential in the treatment of solid tumors. OncoTargets and therapy. 2015;8:1931–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390:1949–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Kotova E, Pinnola AD, Tulin AV. Small-molecule collection and high-throughput colorimetric assay to identify PARP1 inhibitors. Methods Mol Biol. 2011;780:491–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Mateo J, Lord CJ, Serra V, Tutt A, Balmana J, Castroviejo-Bermejo M, et al. A decade of clinical development of PARP inhibitors in perspective. Ann Oncol. 2019;30:1437–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Nieborowska-Skorska M, Maifrede S, Ye M, Toma M, Hewlett E, Gordon J, et al. Non-NAD-like PARP1 inhibitor enhanced synthetic lethal effect of NAD-like PARP inhibitors against BRCA1-deficient leukemia. Leuk Lymphoma. 2019;60:1098–101. [DOI] [PMC free article] [PubMed] [Google Scholar]