PARP inhibitors have been approved by the FDA for use in the treatment of patients with ovarian, breast, pancreatic, and prostate cancers. PARP inhibitors show diverse suppressive effects on PARP family members and PARP-DNA trapping potency. These properties are associated with distinct safety/efficacy profiles. Here, we report the nonclinical characteristics of venadaparib (also known as IDX-1197 or NOV140101), a novel potent PARP inhibitor. The physiochemical properties of venadaparib were analyzed. Furthermore, the efficacy of venadaparib against PARP enzymes, PAR formation, and PARP trapping activities, and growth inhibition of cell lines with BRCA mutations were evaluated. Ex vivo and in vivo models were also established to study pharmacokinetics/pharmacodynamics, efficacy, and toxicity. Venadaparib specifically inhibits PARP-1 and -2 enzymes. Oral administration of venadaparib HCl at doses above 12.5 mg/kg significantly reduced tumor growth in the OV_065 patient-derived xenograft model. Intratumoral PARP inhibition remained at over 90% until 24 hours after dosing. Venadaparib had wider safety margins than olaparib. Notably, venadaparib showed favorable physicochemical properties and superior anticancer effects in homologous recombination-deficient in vitro and in vivo models with improved safety profiles. Our results suggest the possibility of venadaparib as a next-generation PARP inhibitor. On the basis of these findings, phase Ib/IIa studies on the efficacy and safety of venadaparib have been initiated.

This article is featured in Highlights of This Issue, p. 289

Endogenous and exogenous mutagens consistently damage the DNA in normal cells (1). The DNA is protected by robust DNA repair and damage-bypass mechanisms (2). Nevertheless, as DNA repair systems are not perfect, genetic and epigenetic alterations can accumulate in the DNA (3, 4). Function-altering mutations and continuous genomic instability have been described as characteristics that induce tumor pathogenesis and progression (5). In addition, inherited or somatically acquired genetic alterations in DNA repair genes may promote tumorigenesis (3). A combination of genetic and clinical evaluation is generally used to determine the best course of action for patients (6). Therefore, genetic testing is commonly conducted to make treatment decisions for patients with cancer (7).

PARP is an enzyme that plays a key role in DNA repair, transcription, apoptosis, and immune function (8). PARP-1 recruits synthetic DNA enzymes to activate DNA recovery (9). PARP-2 maintains genomic stability and repairs DNA single-strand breaks (SSB; ref. 10). Both PARP-1 and PARP-2 share regulatory mechanisms in DNA repair systems but differ in the mechanism by which they bind to DNA breaks (11). Thus, inhibition of PARP-1/2 results in DNA SSBs and promotes the conversion of SSBs to double-strand breaks (DSB), which induces synthetic lethality in cancer cells due to the lack of homologous recombination (HR; ref. 12). As such, BRCA1- and BRCA2-defective tumors are intrinsically sensitive to PARP inhibition (13). The PARP–BRCA interaction is an example of a successful synthetically lethal approach. To date, four PARP inhibitors (olaparib, rucaparib, niraparib, and talazoparib) have been approved by the FDA for various indications (14).

However, 54% of patients treated with olaparib have been reported to experience adverse events (AE) of ≥grade 3 as per the Common Terminology Criteria for Adverse Events (15). The most frequent grade 3 or worse treatment-emergent AEs are hematologic toxicities such as anemia, thrombocytopenia, and neutropenia (16–18). The common nonhematologic AEs associated with PARP inhibitors are fatigue, nausea, and vomiting (15). The difference in hematologic toxicities among PARP inhibitors may be related to their action on PARP-3 (19). PARP-3 inhibition damages non-homologous end joining DNA repair system, which increases apoptosis of hematopoietic stem cells and multipotent progenitor cells. In addition, inhibition of PARP-5A, also known as tankyrase-1, leads to the loss of bone density with increased osteoclasts and reversible intestinal toxicity (20, 21).

Another important factor that regulates the efficacy and safety of PARP inhibitors is in their ability to trap PARP. Until the PARP inhibitor dissociates from the active site, PARP-1 and PARP-2 are effectively trapped on the DNA (22). As PARP trapping is mediated by the presence of PARP, it is mechanistically dependent on PAR formation or enzymatic inhibition. PARP trapping contributes to the variation in toxicity among PARP inhibitors (23). Therefore, it is important to identify PARP inhibitors with a balanced PARP trapping activity for maximal therapeutic advantages.

We had synthesized a series of derivatives based on the chemical structure of phthalazine; in our in-house studies, venadaparib (code name: IDX-1197, NOV140101) was the most prominent during the discovery phase. Hence, in this study, we evaluated the physicochemical properties, efficacy, and safety of venadaparib, a novel and highly selective PARP inhibitor.

Molecular docking to PARP-1

All applications in the molecular docking simulation were provided in the Maestro module of Schrödinger Suite 2020-3. The X-ray crystal structure of the PARP-1/olaparib complex was acquired from the Protein Data Bank (PDB ID: 5DS3; ref. 24). The receptor grid was generated in a 20 × 20 × 20 Å space region centered at the coligand of the complex structure. Thereafter, energy-minimized three-dimensional structure of venadaparib was docked with default values in the SP mode using Glide. The protein–ligand interactions were analyzed using Discovery Studio Modeling Environment v4.026 (BIOVIA) and the docking models were displayed using PyMOL v2.0.47 (Schrödinger Suite).

Enzymatic assay against recombinant PARP enzymes

The effects of venadaparib on recombinant human PARP enzymes (PARP-1, PARP-2, PARP-3, TNKS-1, TNKS-2, PARP-6, PARP-7, PARP-8, PARP-10, PARP-11, PARP-12, PARP-14, and PARP-15) were determined using in vitro enzymatic assays performed by BPS Bioscience. Venadaparib was examined at concentrations ranging from 0.000005 to 10 μmol/L. Luminescent outputs were measured using a Synergy 2 microplate reader (BioTek Instruments). IC50 was determined using Prism 9 (GraphPad Software).

In vitro PAR assay

Venadaparib and olaparib were synthesized at research laboratories of Ildong Pharmaceutical. Experimental details for the synthesis of venadaparib were described in U.S. patent application publication (publication no. US 2021/0323946 A1; example 12, Supplementary Fig. S1; ref. 25). Rucaparib (catalog no. S1098; Selleck Chemicals), niraparib (catalog no. CT-MK4827; ChemieTek), talazoparib (catalog no. S7048; Selleck Chemicals), and veliparib (catalog no. S1004; Selleck Chemicals) were purchased from commercial vendor, respectively. All compounds were dissolved in DMSO. HeLa cells (Korea Cell Line Bank, Seoul, Republic of Korea) were treated with the above compounds at doses ranging from 0.01 to 100 nmol/L (final concentration of less than 1% DMSO). HeLa cells were then treated with hydrogen peroxide to induce DNA damage. The luminescence signals were detected in the HeLa cells using a PAR antibody on a Synergy H4 hybrid microplate reader (BioTek Instruments) and normalized to the number of cells per plate. The value of the DMSO-only control group was calculated to be 100%. Measurements for each concentration of the test substances were calculated as percentages relative to those of the control group.

PARP trapping assay

The effects of venadaparib on the DNA-binding activities of the recombinant human PARP-1 enzyme were determined using an in vitro enzymatic assay performed by BPS Bioscience. The PARPtrap assay kit (catalog no. 80584; BPS Bioscience) was used for the assay. Venadaparib, olaparib, rucaparib, niraparib, talazoparib, and veliparib were examined at concentrations ranging from 0.000026 to 5 μmol/L. After enzymatic reactions according to the manufacturer's instructions, fluorescent signals were measured using an M1000 microplate reader (Tecan Infinite) with an excitation wavelength of 485 nm and an emission wavelength of 528 nm.

Colony formation assay

Stock solutions of the test molecules were serially diluted separately with DMSO for use in three ovarian cancer cell lines (IGROV-1, SK-OV-3, and OVCAR-3), three breast cancer cell lines (MDA-MB-453, HCC1569, and MX-1), one pancreatic cancer cell line (CAPAN-1), and one gastric cancer cell line (KATOIII) at varying treatment concentrations (Supplementary Table S1). The cells were treated such that the final concentration of DMSO, which was used as the solvent to dissolve the compounds, was less than 1%. The cells were treated with the compounds (venadaparib: 0.03 to 50,000 nmol/L or olaparib: 0.64 to 50,000 nmol/L) and maintained at 37°C in a 5% CO2 incubator. They were left undisturbed for 11 to 30 days (depending on the characteristics of each cell type on PARP inhibitory effect) until colonies appeared. Thereafter, the colonies were stained with a mixture of crystal violet and glutaraldehyde, and the colonies were counted. The test results were calculated as the percentage of the colony number at each concentration of the compound relative to that of the control group (100%), in which only the solvent was added.

Animal studies

Animal studies were conducted in various sites. All studies were performed in accordance with the guidelines established by the U.S. NIH Animal Research and Care and approved by the Institutional Animal Care and Use Committee of each site. A pharmacokinetic study and an animal study using CAPAN-1 cell line–derived xenograft (CDX) models were carried out in the research laboratories of Ildong Pharmaceutical. Animal studies using MX-1 CDX and the OV_065 (high-grade serous ovarian cancer, germline BRCA1 mutation; p.Glu1257Glyfs*p) patient-derived xenograft (PDX) models were performed by Crown Bioscience Inc. and by Asan Medical Center (Seoul, Republic of Korea), respectively. The chronic toxicity study in rats was conducted at Covance Laboratories.

Xenograft models

For the OV_065 PDX model, OV_065 cancer tissue was implanted less than 1 mm into the subcutaneous pocket of the right flank of athymic nude mice (female, 6 weeks old, total 40 mice, n = 8/group). To establish the CDX model, BALB/c nude mice (female, 7–8 weeks old) were inoculated subcutaneously in the right flank region with MX-1 (5 × 106 cells/mouse, total 50 mice, n = 10/group) or CAPAN-1 (1 × 107 cells/mouse, total 48 mice, n = 8/group). Tumor size was assessed twice a week as (length × width2)/2 using calipers. When the mean tumor size reached a volume of 100–250 mm3, the mice were assigned to each study group in such a way that intergroup differences in mean tumor size were minimized. The mice were orally administrated venadaparib HCl (12.5–50 mg/kg to the OV_065 and MX-1 models, 50–200 mg/kg to the CAPAN-1 model) or olaparib (50–100 mg/kg). Morbidity and mortality were checked once daily. To compare tumor growth trends among the test groups, tumor growth inhibition (TGI; %) was calculated as 100 − (100 × (T − T0)/(C − C0)); T = mean tumor volume of the drug treatment group on the final day; T0 = mean tumor volume of the drug treatment group at the start of treatment; C = mean tumor volume of the vehicle control group on the final day; C0 = mean tumor volume of the vehicle control group at the start of treatment. Body weight was measured twice a week, and mice were euthanized when their tumor volume reached over 2,000 mm3 or they lost over 20% of their body weight.

Pharmacokinetic/pharmacodynamic studies

Female Institute of Cancer Research (ICR) mice (18–22 g, 6–7 weeks old) were used to evaluate the pharmacokinetic properties of venadaparib. Nine mice received a single 20 mg/kg intravenous dose of venadaparib HCl via the tail vein. Six fasted mice received a single 60 mg/kg oral dose of venadaparib HCl. Food was supplied 4 hours after dosing. Blood was collected at the following time schedule: intravenous group: 5, 10, 15, 30, 60, 120, and 240 minutes after dosing; oral group: 15, 30, 60, 120, and 240 minutes after dosing. The plasma concentrations of venadaparib were determined using LC/MS-MS (Agilent 1290 Infinity; Agilent Technologies), after protein precipitation with acetonitrile containing an internal standard and centrifugation at 13,000 rpm for 5 minutes at 4°C.

The OV_065 PDX model was used for the pharmacokinetic/pharmacodynamic study. The mice were divided into four groups (n = 4/group), treated with venadaparib HCl once daily (12.5 mg/kg) for 7 days, and then euthanized at 4, 7, and 24 hours after dosing. One vehicle control group, treated only with distilled water, was euthanized 4 hours after administration. Following euthanasia using CO2 gas at the indicated timepoints, plasma and tumor samples were collected and shipped to the research laboratories of Ildong Pharmaceutical, and stored at −70°C. The concentrations of venadaparib in the plasma and tumor tissues were determined using the method described above. To examine the pharmacodynamic activity of venadaparib, we measured intratumoral PAR levels using an HT PARP in vivo pharmacodynamic assay II kit (catalog no. 4520-096-K; R&D Systems).

Four-week toxicity and toxicokinetic study in rats

In the 4-week toxicity and toxicokinetic (TK) studies, Wistar Han rats (male and female, n = 10/sex/group for toxicity groups, 3 or 6/sex/group for TK groups) were administered vehicle or venadaparib HCl at 15, 30, or 60 mg/kg (as venadaparib) daily; recovery animals (n = 5/sex/group) were treated alongside toxicity groups followed by a 4-week off-dose period. We determined hemoglobin level, hematocrit ratio, erythrocyte, neutrophil, platelet, and absolute reticulocyte counts to evaluate hematologic values.

Ex vivo bone marrow toxicity

The experiment for bone marrow toxicity of venadaparib was performed at Stemcell Technologies. Bone marrow–derived hematopoietic progenitor cells from three lots of human bone marrow mononuclear cells were incubated with venadaparib (0.000169–30 μmol/L) or a control compound, 5-fluorouracil (F6627; Sigma-Aldrich), in MethoCult GF H84434 (catalog no. 84434; Stemcell Technologies) at 37°C in a 5% CO2 incubator. After 14 days, the hematopoietic colonies were assessed and scored by trained personnel. The mean colony number was calculated for triplicate cultures under each condition and normalized to the solvent controls. Standard t tests were performed to compare the solvent controls for each test compound. Because of the potential subjectivity of colony enumeration, a P value of less than 0.01 was considered significant.

Off-target panel assay

The selectivity profile of venadaparib on diverse panels covering 88 kinds of enzymes and receptors involved in the cardiovascular, respiratory, and nervous systems was performed at Eurofins Panlabs. Screening was performed using either a radioligand binding assay or an enzymatic assay. Venadaparib was tested at 10 μmol/L in duplicate, and results showing relative inhibition higher than 50% were considered to be significant. Relative inhibition ranging from 25% to 50% was indicative of weak to moderate effects, and results showing a relative inhibition lower than 25% were not considered significant.

Statistical analysis

The results are presented as mean ± SD and processed using GraphPad Prism 9. All results represent the average of at least three independent experiments. Unless otherwise stated, statistical comparisons between the control and each treatment group were performed using a one-way or two-way analysis of variance followed by Dunnett test. Statistical significance was set at P < 0.05.

Data availability statement

The data generated in this study are available upon request from the corresponding author.

Physicochemical properties

Venadaparib exhibited appropriate drug-likeness, adhering to Egan's and Pfizer's 3/75 rule (26, 27). The binding mode of venadaparib to PARP-1 and its physicochemical properties are presented in Fig. 1.

Figure 1.

Molecular docking modeling of the PARP-1/venadaparib or olaparib complex and the physicochemical properties of venadaparib. A, Chemical structure of PARP inhibitors. B, Venadaparib interaction with key amino acid residues of PARP-1 is represented using a green stick compared with olaparib in orange. Hydrogen bonds are shown as blue dashed lines. The protein–ligand interactions were analyzed using Discovery Studio Modeling Environment v4.026, and the docking models are displayed using PyMOL v2.0.47. C, Physicochemical properties of venadaparib. TPSA, topological polar surface area.

Figure 1.

Molecular docking modeling of the PARP-1/venadaparib or olaparib complex and the physicochemical properties of venadaparib. A, Chemical structure of PARP inhibitors. B, Venadaparib interaction with key amino acid residues of PARP-1 is represented using a green stick compared with olaparib in orange. Hydrogen bonds are shown as blue dashed lines. The protein–ligand interactions were analyzed using Discovery Studio Modeling Environment v4.026, and the docking models are displayed using PyMOL v2.0.47. C, Physicochemical properties of venadaparib. TPSA, topological polar surface area.

Close modal

Efficacy of venadaparib in the enzymatic assay against recombinant PARP enzymes

The enzyme-inhibiting activities of venadaparib were tested to assess its selectivity for recombinant PARP enzymes. As shown in Table 1A, venadaparib selectively inhibited PARP-1/2 with IC50 values of 0.8 and 3 nmol/L, respectively. The IC50 values determined for venadaparib against other PARP enzymes were 780 nmol/L for PARP-3 and 3,200 nmol/L (62% inhibition at 10 μmol/L) for TNKS-2; venadaparib did not affect TNKS-1 and PARP-6/7/8/10/11/12/14/15 up to 10,000 nmol/L. The data revealed that venadaparib is highly selective for PARP-1/2.

Table 1.

Inhibitory effects of venadaparib on PARP enzymes, PAR formation in HeLa cells, and PARP trapping potency.

(A) IC50 values indicated that venadaparib has selectivity for PARP-1 and -2.
IC50 (nmol/L)
EnzymeVenadaparibOlaparib
PARP-1 0.8 0.6 
PARP-2 0.5 
PARP-3 ∼780 14 
TNKS-1 >10,000  
TNKS-2 ∼3,200  
PARP-6 >10,000 750 
PARP-7 >10,000 410 
PARP-8 >10,000 1,650 
PARP-10 >10,000 540 
PARP-11 >10,000  
PARP-12 >10,000 1,100 
PARP-14 >10,000  
PARP-15 >10,000 6,000 
(B) EC50 values indicated that venadaparib potently inhibited PAR formation and PARP trapping activity unlike the other PARP inhibitors. 
 EC50 (nmol/L) 
Compound PAR formation PARP trapping 
Venadaparib 0.5 2.2 
Olaparib 0.7 7.3 
Rucaparib 1.9 6.4 
Niraparib 5.6 118.0 
Talazoparib 0.7 1.9 
Veliparib 4.5 57.7 
(A) IC50 values indicated that venadaparib has selectivity for PARP-1 and -2.
IC50 (nmol/L)
EnzymeVenadaparibOlaparib
PARP-1 0.8 0.6 
PARP-2 0.5 
PARP-3 ∼780 14 
TNKS-1 >10,000  
TNKS-2 ∼3,200  
PARP-6 >10,000 750 
PARP-7 >10,000 410 
PARP-8 >10,000 1,650 
PARP-10 >10,000 540 
PARP-11 >10,000  
PARP-12 >10,000 1,100 
PARP-14 >10,000  
PARP-15 >10,000 6,000 
(B) EC50 values indicated that venadaparib potently inhibited PAR formation and PARP trapping activity unlike the other PARP inhibitors. 
 EC50 (nmol/L) 
Compound PAR formation PARP trapping 
Venadaparib 0.5 2.2 
Olaparib 0.7 7.3 
Rucaparib 1.9 6.4 
Niraparib 5.6 118.0 
Talazoparib 0.7 1.9 
Veliparib 4.5 57.7 

Abbreviation: EC50, half-maximal effective concentration.

Activities of venadaparib on PAR formation and PARP trapping

The PAR formation inhibitory activity of venadaparib was assessed by the cellular PARP-1–inhibiting activity of venadaparib. As shown in Table 1B, venadaparib effectively inhibited the synthesis of PAR at an EC50 of 0.5 nmol/L measured in a HeLa cell line where DNA damage was induced. This value was similar to that of olaparib (EC50 = 0.7 nmol/L) and talazoparib (EC50 = 0.7 nmol/L), whereas rucaparib, niraparib, and veliparib showed relatively low activities (EC50 values of 1.9, 5.6, and 4.5 nmol/L, respectively).

An in vitro enzymatic assay using fluorescence polarization showed that venadaparib (EC50 = 2.2 nmol/L) has a trapping activity similar to that of talazoparib (EC50 = 1.9 nmol/L). The ability of venadaparib to trap PARP was higher than that of other PARP inhibitors (EC50 = 6.4–118.0 nmol/L; Table 1B).

Growth inhibitory effect of venadaparib in cancer cell lines with BRCA alteration

In the colony formation assay using genetically well-annotated cancer cell lines with or without mutations in the BRCA genes, venadaparib showed better inhibitory activity of colony formation than that of olaparib in cancers with mutations in BRCA1 and BRCA2; both compounds showed a decrease in efficacy against cell lines with wild-type BRCA genes (Table 2).

Table 2.

Effects of venadaparib and olaparib on colony formation in a panel of cancer cell lines.

Genetic alterationIC50 (nmol/L)
Cell lineBRCA1BRCA2VenadaparibOlaparib
IGROV-1 Mutation — 11.5 938.8 
MDA-MB-453 Deletion — 2.6 434.6 
HCC1569 — Mutation 0.8 356.1 
OVCAR-3 — Deletion 5.8 506.9 
MX-1 Deletion Mutation 14.4 611.2 
CAPAN-1 Amplification Mutation 129.7 773.8 
KATOIII — — 1,544.0 2,433.0 
SK-OV-3 — — 1,784.0 2,627.0 
Genetic alterationIC50 (nmol/L)
Cell lineBRCA1BRCA2VenadaparibOlaparib
IGROV-1 Mutation — 11.5 938.8 
MDA-MB-453 Deletion — 2.6 434.6 
HCC1569 — Mutation 0.8 356.1 
OVCAR-3 — Deletion 5.8 506.9 
MX-1 Deletion Mutation 14.4 611.2 
CAPAN-1 Amplification Mutation 129.7 773.8 
KATOIII — — 1,544.0 2,433.0 
SK-OV-3 — — 1,784.0 2,627.0 

Note: Cancer cell lines that had different pathogenic BRCA gene alterations were treated with various concentrations of venadaparib or olaparib. Colony formation units were determined by staining with a mixture of crystal violet and glutaraldehyde. Venadaparib was potent against BRCA-altered cancer cells and showed potent efficacy than olaparib.

Pharmacokinetics/pharmacodynamics and tumor growth inhibitory effects of venadaparib in animal models

The pharmacokinetic characteristics of venadaparib were determined in healthy female ICR mice (Fig. 2A). Venadaparib has a high steady-state volume of distribution (Vss = 5.28 L/kg) and a terminal elimination half-life of 0.90 hours. Venadaparib was rapidly absorbed, and the maximum plasma concentration was reached within 0.25 hours after dosing. The oral bioavailability of the solution formulation was 70.63%.

Figure 2.

In vivo pharmacokinetics/pharmacodynamics and efficacy of venadaparib. A, The pharmacokinetic characteristics of venadaparib were determined in healthy female ICR mice. B, Time-dependent plasma/tumor concentrations and correlated intratumoral PAR inhibition were measured by administering venadaparib HCl 12.5 mg/kg in an OV_065 PDX model. Venadaparib exhibited PAR inhibition over 90% until 24 hours after dosing. C, In the OV_065 PDX model (athymic nude mice), venadaparib HCl ranging from 12.5 to 50 mg/kg and olaparib 50 mg/kg were administered for 63 days (orally, once daily). D,BALB/c nude mice implanted with MX-1 cells were treated with venadaparib HCl at 12.5–50 mg/kg for 23 days (orally, once daily). E, In the CAPAN-1 model (BALB/c nude mice), venadaparib HCl doses ranged from 50 to 200 mg/kg for 28 days (orally, once daily). Tumor volume and body weight were measured twice weekly. Dose-dependent tumor regression was observed in venadaparib-treated groups. All treatments had no effect on mouse body weight. Data are presented as mean ± SD. *** indicates statistically significant differences from the vehicle control group at P < 0.001.

Figure 2.

In vivo pharmacokinetics/pharmacodynamics and efficacy of venadaparib. A, The pharmacokinetic characteristics of venadaparib were determined in healthy female ICR mice. B, Time-dependent plasma/tumor concentrations and correlated intratumoral PAR inhibition were measured by administering venadaparib HCl 12.5 mg/kg in an OV_065 PDX model. Venadaparib exhibited PAR inhibition over 90% until 24 hours after dosing. C, In the OV_065 PDX model (athymic nude mice), venadaparib HCl ranging from 12.5 to 50 mg/kg and olaparib 50 mg/kg were administered for 63 days (orally, once daily). D,BALB/c nude mice implanted with MX-1 cells were treated with venadaparib HCl at 12.5–50 mg/kg for 23 days (orally, once daily). E, In the CAPAN-1 model (BALB/c nude mice), venadaparib HCl doses ranged from 50 to 200 mg/kg for 28 days (orally, once daily). Tumor volume and body weight were measured twice weekly. Dose-dependent tumor regression was observed in venadaparib-treated groups. All treatments had no effect on mouse body weight. Data are presented as mean ± SD. *** indicates statistically significant differences from the vehicle control group at P < 0.001.

Close modal

Oral administration of venadaparib HCl at a dose of 12.5 mg/kg considerably reduced tumor PAR levels by over 90% until 24 hours after dosing in the OV_065 PDX model. Drug concentrations remained over 100 ng/g at 24 hours after administration in the tumor, whereas it was close to being below the lower limit of quantification (1 ng/mL) in the plasma (Fig. 2B). Treatment with venadaparib HCl induced a significant tumor regression (Fig. 2C). Treatment with venadaparib HCl at doses of 12.5, 25, and 50 mg/kg and olaparib at 50 mg/kg showed TGI rates of 131.0%, 132.7%, 135.2%, and 118.2%, respectively. Tumors completely disappeared in 50% of animals in the group treated with 50 mg/kg venadaparib.

In the MX-1 CDX model, the inhibitory effect of venadaparib on tumor growth was assessed using TGI. The TGI of groups treated with venadaparib HCl at doses of 12.5, 25, and 50 mg/kg was 43.8%, 62.9%, and 71.0%, respectively. The group treated with 12.5 mg/kg venadaparib HCl (TGI = 43.8%) had similar tumor growth patterns as the group treated with 100 mg/kg olaparib (TGI = 41.7%; Fig. 2D). In the CAPAN-1 CDX model, the TGI was 62.2%, 82.5%, and 110.7% in the groups treated with venadaparib HCl at 50, 100, and 200 mg/kg, respectively (Fig. 2E). Venadaparib regulated tumor growth in a dose-dependent manner. The group treated with 100 mg/kg venadaparib HCl (TGI = 82.5%) seemed to show higher TGI than the group treated with 100 mg/kg olaparib (TGI = 36.0%). The group treated with 200 mg/kg venadaparib HCl (TGI = 110.7%) presented the highest antitumor effect. None of the dosing groups had test drug-related deaths or any other general symptoms throughout the study period.

Four-week toxicity and TK of venadaparib in rats

The 4-week toxicity and TK of venadaparib were investigated in male and female Wistar Han rats by dosing at 15, 30, or 60 mg/kg daily for 4 weeks via oral gavage. No unscheduled deaths or abnormal changes in clinical signs were observed during the study. The no-observed-adverse-effect-level (NOAEL) of venadaparib was 30 mg/kg/day in rats. In the hematologic examination following 4 weeks of repeated administration, significant decreases in erythrocyte count, hemoglobin level, and hematocrit ratios were observed at a dose of 60 mg/kg in males and ≥15 mg/kg in females (Table 3A). None of the groups dosed with venadaparib showed a decline in neutrophil/platelet counts or in absolute reticulocyte counts (immature red blood cells), except females treated with 60 mg/kg. The platelet counts increased in males at 60 mg/kg and females at 15 mg/kg or more. This response had resolved by the recovery period (Supplementary Table S2). Reductions in red cell mass, related to erythrocyte count, hemoglobin level, and hematocrit ratio at 15 and 30 mg/kg in female rats did not influence the NOAEL as a complete recovery (no significant difference from the control group; Supplementary Table S2) was made after the 4-week recovery period. In the clinical chemistry, venadaparib did not cause an increase in creatinine, aspartate aminotransferase, and alanine aminotransferase levels in both sexes following 28 days of repeated oral administration (Supplementary Table S2). The TK characteristics of venadaparib are presented in Table 3B.

Table 3.

Effects on hematologic values and toxicokinetics of venadaparib in the 4-week toxicity study using Wistar Han rats.

A
Dose (mg/kg)0153060
SexMaleFemaleMaleFemaleMaleFemaleMaleFemale
Erythrocyte (106/μL) 8.9 ± 0.4 8.5 ± 0.5 8.8 ± 0.6 7.7 ± 0.4** 8.3 ± 0.7 6.9 ± 0.5** 6.1 ± 0.6** 5.3 ± 0.4** 
Hemoglobin (g/dL) 16.4 ± 0.5 15.8 ± 0.6 16.6 ± 0.7 14.3 ± 0.8** 15.6 ± 1.2 13.1 ± 1.1** 11.4 ± 1.5** 9.9 ± 0.8** 
Hematocrit ratio (%) 53.1 ± 2.6 49.6 ± 2.2 52.4 ± 3.3 45.7 ± 2.5** 49.6 ± 3.7 41.6 ± 3.2** 36.2 ± 4.7** 31.5 ± 2.7** 
Neutrophils (103/μL) 1.1 ± 0.2 0.7 ± 0.1 0.8 ± 0.2 0.6 ± 0.1 1.0 ± 0.2 0.5 ± 0.1 1.2 ± 0.5 0.7 ± 0.4 
Platelets (103/μL) 845.5 ± 155.4 740.4 ± 91.8 927.8 ± 156.1 1,004.0 ± 156.9** 990.6 ± 203.4 1,146.8 ± 168.7** 1,698.8 ± 192.3** 1,553.5 ± 205.4** 
Absolute reticulocytes (103/μL) 205.7 ± 54.1 213.2 ± 41.8 170.1 ± 40.0 194.9 ± 28.7 199.3 ± 38.7 201.5 ± 40.7 184.6 ± 62.1 171.3 ± 34.4* 
A
Dose (mg/kg)0153060
SexMaleFemaleMaleFemaleMaleFemaleMaleFemale
Erythrocyte (106/μL) 8.9 ± 0.4 8.5 ± 0.5 8.8 ± 0.6 7.7 ± 0.4** 8.3 ± 0.7 6.9 ± 0.5** 6.1 ± 0.6** 5.3 ± 0.4** 
Hemoglobin (g/dL) 16.4 ± 0.5 15.8 ± 0.6 16.6 ± 0.7 14.3 ± 0.8** 15.6 ± 1.2 13.1 ± 1.1** 11.4 ± 1.5** 9.9 ± 0.8** 
Hematocrit ratio (%) 53.1 ± 2.6 49.6 ± 2.2 52.4 ± 3.3 45.7 ± 2.5** 49.6 ± 3.7 41.6 ± 3.2** 36.2 ± 4.7** 31.5 ± 2.7** 
Neutrophils (103/μL) 1.1 ± 0.2 0.7 ± 0.1 0.8 ± 0.2 0.6 ± 0.1 1.0 ± 0.2 0.5 ± 0.1 1.2 ± 0.5 0.7 ± 0.4 
Platelets (103/μL) 845.5 ± 155.4 740.4 ± 91.8 927.8 ± 156.1 1,004.0 ± 156.9** 990.6 ± 203.4 1,146.8 ± 168.7** 1,698.8 ± 192.3** 1,553.5 ± 205.4** 
Absolute reticulocytes (103/μL) 205.7 ± 54.1 213.2 ± 41.8 170.1 ± 40.0 194.9 ± 28.7 199.3 ± 38.7 201.5 ± 40.7 184.6 ± 62.1 171.3 ± 34.4* 
B
Dose (mg/kg)153060
Day128128128
SexMaleFemaleMaleFemaleMaleFemaleMaleFemaleMaleFemaleMaleFemale
Cmax (ng/mL) 602 2,940 952 1,020 2,550 5,710 2,750 2,720 6,950 14,400 4,530 8,120 
AUC0–24 h (μg·h/mL) 1.27 6.08 3.14 4.54 7.36 19.40 11.60 10.70 19.00 42.50 22.00 40.80 
R — — 2.48 0.75 — — 1.58 0.55 — — 1.16 0.96 
B
Dose (mg/kg)153060
Day128128128
SexMaleFemaleMaleFemaleMaleFemaleMaleFemaleMaleFemaleMaleFemale
Cmax (ng/mL) 602 2,940 952 1,020 2,550 5,710 2,750 2,720 6,950 14,400 4,530 8,120 
AUC0–24 h (μg·h/mL) 1.27 6.08 3.14 4.54 7.36 19.40 11.60 10.70 19.00 42.50 22.00 40.80 
R — — 2.48 0.75 — — 1.58 0.55 — — 1.16 0.96 

Note: The 4-week toxicity and TK of venadaparib were investigated in male and female Wistar Han rats by dosing at 15, 30, or 60 mg/kg (as venadaparib) daily for 4 weeks via oral gavage. In a hematologic examination following 4 weeks of repeated administration, a significant decrease in erythrocyte count, hemoglobin level, and hematocrit ratio was observed at a dose of 60 mg/kg in males and ≥15 mg/kg in females. Data are presented as mean ± SD. * and ** indicate statistically significant differences from the control group at P < 0.05 and 0.01, respectively.

Abbreviation: R, accumulation ratio.

Ex vivo bone marrow toxicity of venadaparib

We assessed bone marrow toxicity through continuous incubation for 14 days with venadaparib at various concentrations (0.000169, 0.000508, 0.00152, 0.00457, 0.0137, 0.041, 0.12, 0.37, 1.11, 3.33, 10, and 30 μmol/L) on three lots of human bone marrow–derived stem and progenitor cells for erythroid and myeloid colony proliferation in MethoCult GF H84434 (Fig. 3A). The average IC50 of venadaparib in total colony-forming cells was 0.1006 μmol/L.

Figure 3.

Bone marrow toxicity and in vitro off-target screening of venadaparib. A, Bone marrow colony formation data, calculated from three independent cultures and normalized to solvent controls, are presented as mean ± SD. The IC50 and IC90 values were 0.1006 and 8.993 μmol/L, respectively. B, The selectivity profile of venadaparib was determined using 88 kinds of enzyme/receptor panels. Venadaparib at 10 μmol/L did not induce significant responses and showed weak to moderate responses on monoamine oxidase MAO-B, acetylcholinesterase, adrenergic alpha 2A, and 5-hydroxytryptamine1A. IC90, 90% of the maximal inhibitory concentration.

Figure 3.

Bone marrow toxicity and in vitro off-target screening of venadaparib. A, Bone marrow colony formation data, calculated from three independent cultures and normalized to solvent controls, are presented as mean ± SD. The IC50 and IC90 values were 0.1006 and 8.993 μmol/L, respectively. B, The selectivity profile of venadaparib was determined using 88 kinds of enzyme/receptor panels. Venadaparib at 10 μmol/L did not induce significant responses and showed weak to moderate responses on monoamine oxidase MAO-B, acetylcholinesterase, adrenergic alpha 2A, and 5-hydroxytryptamine1A. IC90, 90% of the maximal inhibitory concentration.

Close modal

Evaluation of venadaparib in an off-target panel assay

In the secondary pharmacology test, venadaparib at 10 μmol/L did not show significant off-target activity (defined as ≥50% inhibition) in any of the enzymes and receptors tested. In addition, venadaparib weakly to moderately inhibited monoamine oxidase MAO-B (37% inhibition), acetylcholinesterase (37% inhibition), adrenergic alpha 2A (25% inhibition), and 5-hydroxytryptamine1A (25% inhibition). These data revealed that venadaparib has a low potential to cause undesirable pharmacodynamic effects on physiologic functions (Fig. 3B).

Venadaparib is a novel and selective PARP inhibitor currently in clinical development. To the best of our knowledge, this is the first report on the physicochemical and nonclinical pharmacologic effects of venadaparib. The results showed that venadaparib has potent anticancer activities in both in vitro and in vivo models with favorable physicochemical and pharmacokinetic properties, as well as a toxicologic profile.

The remarkable characteristics of venadaparib include its high solubility, extensive tumor distribution, potent antitumor effects, and broad tolerability. Venadaparib exhibited appropriate physicochemical properties by adhering to Egan's and Pfizer's 3/75 rule (26, 27). These characteristics resulted in promising pharmacologic and toxicologic aspects, as discussed below.

PARP-1 and -2 are enzymes that repair DNA damage and maintain genomic stability. Inhibition of PARP-1 and -2 has shown promising antitumor effects, whereas undesirable inhibition of PARP-3 or -5 can induce adverse effects, including hematotoxicity and bone loss (9, 10, 20, 21). Enzymatic assays using various PARPs showed that venadaparib specifically affects PARP-1 and -2 (PARP-1 IC50 = 0.8 nmol/L; PARP-2 IC50 = 3 nmol/L) compared with olaparib (PARP-1 IC50 = 0.6 nmol/L; PARP-2 IC50 = 0.5 nmol/L; PARP-3 IC50 = 14 nmol/L). In addition, it has been reported that approved PARP inhibitors suppress other PARP enzymes as well as PARP-1 and -2 (talazoparib: PARP-3 IC50 = 62.8 nmol/L, TNKS-2 IC50 = 108 nmol/L, rucaparib: PARP-3 IC50 = 512 nmol/L; TNKS-1 IC50 = 144 nmol/L; ref. 28). Inhibition of PAR formation and trapping on the DNA/PARP complex are the major mechanisms that lead to tumor cell death by PARP inhibitors (22). PARP inhibitors interfere with the repair of SSBs by trapping the DNA/PARP complex, resulting in DNA DSBs, which require an alternative pathway for repair (14, 29). The ability of venadaparib to inhibit PAR formation (EC50 = 0.5 nmol/L) was comparable with that of olaparib (EC50 = 0.7 nmol/L). The PARP trapping activity of venadaparib (EC50 = 2.2 nmol/L) was demonstrated to be similar to that of talazoparib (EC50 = 1.9 nmol/L) and more potent than that of other PARP inhibitors (EC50 values ranged from 6.4 to 118.0 nmol/L).

Germline mutations in HR-related genes such as BRCA1 and BRCA2 are critical for the accumulation of DNA damage, inducing DNA DSBs (30). BRCA-deficient tumor cells diminished the ability to rescue DNA DSBs by HR, and in the absence of an alternative repair mechanism, such a deficiency leads to cell death (31). Inhibition of PARP, which is a known therapeutic strategy for BRCA1/2-deficient cancer, shows high sensitivity against cancers that have lost the ability to repair DNA breaks through the HR-related pathway (13). It was observed that venadaparib has approximately 40–440 times potent antitumor potency than olaparib in BRCA-mutated tumor cell lines. This inhibitory effect vanishes in tumor cells with wild-type BRCA. In other words, venadaparib is highly selective for BRCA-mutated cancer cells over BRCA wild-type cancer cell lines compared with olaparib. In particular, we confirmed that PARP inhibitor–induced tumor cell death depends on pathogenic BRCA mutations in CAPAN-1 cells. Although CAPAN-1 cells have a BRCA2 mutation, tumor growth inhibition by venadaparib was weakened due to the amplification of wild-type BRCA1.

Here, venadaparib was characterized by a high steady-state volume of distribution (Vss = 5.28 L/kg). After a single oral dose of 60 mg/kg venadaparib HCl, venadaparib was rapidly absorbed and cleared from the blood. Our preliminary studies indicate that venadaparib is a potential substrate of efflux transporters (i.e., MDR1, BCRP). However, the venadaparib concentration in the tumor remained high (over 100 ng/g) even at 24 hours after dosing with only 12.5 mg/kg venadaparib HCl in the OV_065 PDX model. These distribution characteristics may be due to the basicity of venadaparib originating from the secondary amine, which was designed to enhance tumor penetration. Inhibition of intratumoral PAR remained over 90% until 24 hours after administration of 12.5 mg/kg venadaparib HCl. Moreover, the tumor:plasma partition coefficient (i.e., |$ {K}_{p,tumor:plasma} = \ \frac{{AU{C}_{tumor}}}{{AU{C}_{plasma}}}$|⁠) of venadaparib was 10.0, suggesting that venadaparib tends to more concentrate in tumor tissue than olaparib and niraparib (i.e., 0.33 for olaparib, and 5.19 for niraparib in the OVC135 PDX model; ref. 32). Notably, in the OV_065 PDX model, we observed complete tumor regression by venadaparib HCl, administered orally for a maximum of 63 days (50 mg/kg, once daily) in female mice. This result is consistent with a previous study finding that maintaining the PAR inhibition levels above 90% for 24 hours is important to completely eliminate the tumor (33).

The tumor regressive effects of venadaparib were demonstrated in other xenograft models using MX-1 and CAPAN-1. The data consistently indicated that venadaparib exhibited significant tumor growth-suppressive effects in a dose-dependent manner and had greater antitumor potency than olaparib. There were no abnormal clinical signs or body weight changes during the process.

Another compelling characteristic of venadaparib is its refined toxicity profile. It has been reported that the NOAEL of olaparib after 4 weeks of daily repeated oral dosing in rats is 15 mg/kg/day (33). Following 4 weeks of dosing at the NOAEL, the exposure (AUC0–24 h) was 1.057 and 6.267 μg·h/mL in male and female rats, respectively. Safety margins from exposure to the effective dosage of olaparib (80 mg/kg, AUC0-inf = 14.408 and 12.574 μg·h/mL in males and females, respectively) were calculated to be 0.07 and 0.5 times. In the rat 28-day repeat dose toxicity study of talazoparib, the highest nonseverely toxic dose was 0.05 mg/kg/day, (NOAEL is not determined); Cmax = 0.0195 μg/mL, AUC = 0.148 μg·h/mL at 0.05 mg/kg, once daily on day 28 (34). Talazoparib showed superior anticancer effect in the MX-1 xenograft model at 0.33 mg/kg dose; however, the dose seems to be intolerable to mice as the AUC0–6 h of talazoparib was in range of 0.075–128 μg·h/mL in mice at 0.15 mg/kg dose (35). In the 4-week TK study of venadaparib, drug exposure (AUC0–24 h) at the NOAEL (30 mg/kg/day) was 11.60 and 10.70 μg·h/mL in male and female rats, respectively. Assuming the linear pharmacokinetics in mice, 100 mg/kg venadaparib in mice was estimated to be equivalent to NOAEL in female rats. Considering the safety margin of olaparib, 200 mg/kg venadaparib could have the same safety margin for rodents.

PARP inhibitors disrupt cell division and are therefore expected to have the greatest effect on rapidly dividing cells, as is common in the bone marrow, lymphoid, skin, and gastrointestinal tract tissue (36). Venadaparib also showed mild changes in hematology values after 4 weeks of repeated dosing up to 30 mg/kg; however, it was resolved during the off-dose recovery period. To further study the hematologic toxicity of venadaparib, we measured erythroid and myeloid colony proliferation using human bone marrow–derived stem and progenitor cells. Taken together, venadaparib inhibited colony growth of bone marrow–derived hematopoietic progenitor cells with an IC50 of 0.1006 μmol/L, which is 200-fold greater than the EC50 value of venadaparib in the inhibition of PAR formation. Hopkins and colleagues suggested that the PARP-trapping activities associated with greater cytotoxic potency may limit the therapeutic advantages of PARP inhibitors (23). It was verified that the resolution between PAR inhibition and cytotoxicity covers about a 100-fold gap for olaparib, rucaparib, and veliparib, but less than a 2-fold difference for talazoparib. This result could help reduce the incidence of adverse drug reactions that characterize and identify secondary pharmacology profiling of drug candidates (37). There were no significant off-target activities of venadaparib in the secondary pharmacology test at a concentration of 10 μmol/L. These results suggest that venadaparib is a novel PARP inhibitor with a lower possibility of unexpected adverse effects in the range of pharmacologically active doses.

In this study, we evaluated the nonclinical properties of venadaparib using in vitro/ex vivo systems and animal models. Our results revealed that venadaparib has improved efficacy and safety; further investigation will be necessary in the clinical phase as an alternative class of PARP inhibitors in humans.

BRCA1 and BRCA2 are crucial for HR, and thus, it is expected that patients with a defective DNA damage repair system may also respond to PARP inhibitors (38). Although olaparib was approved for patients with HR repair gene-mutated metastatic castration-resistant prostate cancer, the clinical application is still limited mainly to patients with BRCA-mutated/platinum-sensitive cancers. A more potent PARP inhibitor with a tolerable safety profile is needed to overcome the restricted boundary of using PARP inhibitors. Our results showed that venadaparib, a novel and selective PARP inhibitor, has potent antitumor potency in multiple in vitro and in vivo models and a balanced trapping effect. Venadaparib also demonstrated favorable pharmacokinetic properties in the nonclinical studies. All these features indicate it is suitable for clinical trials. A phase I study evaluating the safety and tolerability of venadaparib in patients with advanced solid tumors (NCT03317743) has been completed. Therein, venadaparib showed a tolerable safety profile and a potential clinical benefit with a 1-day dosing schedule in patients with advanced solid tumors that progressed after standard-of-care therapy (39). Venadaparib is also being investigated in a Ib/IIa study in patients with HR repair genes–mutated solid tumors including patients previously treated with a PARP inhibitor (NCT4174716), and in those with advanced gastric cancer treated in combination with XELOX or irinotecan (NCT04725994). Hence, this study, highlighting the role of venadaparib as a PARP inhibitor, can further support the outcomes of these trials.

M. Lee reports grants from National-OncoVenture and personal fees from Ildong Pharmaceutical during the conduct of the study. I.-G. Je reports grants from National-OncoVenture during the conduct of the study. J.E. Kim reports grants from National-OncoVenture and personal fees from Ildong Pharmaceutical during the conduct of the study. Y. Yoo reports grants from National-OncoVenture during the conduct of the study. J.-H. Lim reports grants from National-OncoVenture, Grant during the conduct of the study. E. Jang reports grants from National-OncoVenture during the conduct of the study. Y. Lee reports personal fees from Ildong Pharmaceutical during the conduct of the study. D.K. Song reports personal fees from Ildong Pharmaceutical during the conduct of the study. A.-N. Moon reports personal fees from Ildong Pharmaceutical during the conduct of the study. J.-A. Kim reports personal fees from Ildong Pharmaceutical during the conduct of the study. J. Jeong reports grants from National-OncoVenture during the conduct of the study. J.-T. Park reports grants from National-OncoVenture during the conduct of the study. J.W. Lee reports grants from National-OncoVenture during the conduct of the study. J.-H. Yang reports grants from National-OncoVenture during the conduct of the study. C.-H. Hong reports grants from National-OncoVenture during the conduct of the study. S.-Y. Park reports grants from National-OncoVenture during the conduct of the study. N.-S. Baek reports grants from Ministry of Health and Welfare Korea during the conduct of the study. S. Lee reports grants from Ministry of Health and Welfare Korea during the conduct of the study. K.S. Ha is an employee of the Idience Inc. S. Choi reports grants from National-OncoVenture during the conduct of the study. W.S. Lee reports grants from National-OncoVenture during the conduct of the study. No disclosures were reported by the other author.

M. Lee: Conceptualization, writing–original draft, project administration. I.-G. Je: Formal analysis, investigation, visualization, methodology, writing–original draft. J.E. Kim: Formal analysis, validation, investigation. Y. Yoo: Formal analysis, investigation. J.-H. Lim: Formal analysis, investigation. E. Jang: Formal analysis, investigation. Y. Lee: Conceptualization. D.K. Song: Investigation. A.-N. Moon: Investigation. J.-A. Kim: Investigation. J. Jeong: Investigation. J.-T. Park: Resources, data curation, project administration. J.W. Lee: Data curation, project administration. J.-H. Yang: Investigation. C.-H. Hong: Investigation. S.-Y. Park: Investigation. Y.-W. Park: Funding acquisition. N.S. Baek: Funding acquisition, project administration. S. Lee: Funding acquisition. K.S. Ha: Writing–review and editing. S. Choi: Supervision. W.S. Lee: Supervision.

This study was conducted with the National-OncoVenture supported by the National Cancer Center, designated by the Ministry of Health and Welfare Korea (HI17C2196).

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

1.
Stratton
MR
,
Campbell
PJ
,
Futreal
PA
.
The cancer genome
.
Nature
2009
;
458
:
719
24
.
2.
Chatterjee
N
,
Walker
GC
.
Mechanisms of DNA damage, repair, and mutagenesis
.
Environ Mol Mutagen
2017
;
58
:
235
63
.
3.
Lahtz
C
,
Pfeifer
GP
.
Epigenetic changes of DNA repair genes in cancer
.
J Mol Cell Biol
2011
;
3
:
51
8
.
4.
Takeshima
H
,
Ushijima
T
.
Accumulation of genetic and epigenetic alterations in normal cells and cancer risk
.
NPJ Precis Oncol
2019
;
3
:
7
.
5.
Hanahan
D
,
Weinberg
RA
.
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
6.
Aronson
SJ
,
Rehm
HL
.
Building the foundation for genomics in precision medicine
.
Nature
2015
;
526
:
336
42
.
7.
U.S. Food and Drug Administration
.
List of cleared or approved companion diagnostic devices (in vitro and imaging tools)
;
2021
.
Available from:
https://www.fda.gov/medical-devices/in-vitro-diagnostics/list-cleared-or-approved-companion-diagnostic-devices-in-vitro-and-imaging-tools.
8.
Krishnakumar
R
,
Kraus
WL
.
The PARP side of the nucleus: molecular actions, physiological outcomes, and clinical targets
.
Mol Cell
2010
;
39
:
8
24
.
9.
Rouleau
M
,
Patel
A
,
Hendzel
MJ
,
Kaufmann
SH
,
Poirier
GG
.
PARP inhibition: PARP1 and beyond
.
Nat Rev Cancer
2010
;
10
:
293
301
.
10.
Schreiber
V
,
Amé
JC
,
Dollé
P
,
Schultz
I
,
Rinaldi
B
,
Fraulob
V
, et al
.
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
36
.
11.
Langelier
MF
,
Riccio
AA
,
Pascal
JM
.
PARP-2 and PARP-3 are selectively activated by 5' phosphorylated DNA breaks through an allosteric regulatory mechanism shared with PARP-1
.
Nucleic Acids Res
2014
;
42
:
7762
75
.
12.
Zheng
F
,
Zhang
Y
,
Chen
S
,
Weng
X
,
Rao
Y
,
Fang
H
.
Mechanism and current progress of Poly ADP-ribose polymerase (PARP) inhibitors in the treatment of ovarian cancer
.
Biomed Pharmacother
2020
;
123
:
109661
.
13.
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
.
14.
Helleday
T
.
The underlying mechanism for the PARP and BRCA synthetic lethality: clearing up the misunderstandings
.
Mol Oncol
2011
;
5
:
387
93
.
15.
Kaufman
B
,
Shapira-Frommer
R
,
Schmutzler
RK
,
Audeh
MW
,
Friedlander
M
,
Balmana
J
, et al
.
Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation
.
J Clin Oncol
2015
;
33
:
244
50
.
16.
Swisher
EM
,
Lin
KK
,
Oza
AM
,
Scott
CL
,
Giordano
H
,
Sun
J
, et al
.
Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial
.
Lancet Oncol
2017
;
18
:
75
87
.
17.
Mirza
MR
,
Monk
BJ
,
Herrstedt
J
,
Oza
AM
,
Mahner
S
,
Redondo
A
, et al
.
Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer
.
N Engl J Med
2016
;
375
:
2154
64
.
18.
Litton
JK
,
Rugo
HS
,
Ettl
J
,
Hurvitz
SA
,
Goncalves
A
,
Lee
KH
, et al
.
Talazoparib in patients with advanced breast cancer and a germline BRCA mutation
.
N Engl J Med
2018
;
379
:
753
63
.
19.
Oplustil O'Connor
L
,
Rulten
SL
,
Cranston
AN
,
Odedra
R
,
Brown
H
,
Jaspers
JE
, et al
.
The PARP inhibitor AZD2461 provides insights into the role of PARP3 inhibition for both synthetic lethality and tolerability with chemotherapy in preclinical models
.
Cancer Res
2016
;
76
:
6084
94
.
20.
Mukai
T
,
Fujita
S
,
M Y
.
Tankyrase (PARP5) inhibition induces bone loss through accumulation of its substrate SH3BP2
.
Cells
2019
;
8
:
195
.
21.
Zhong
Y
,
Katavolos
P
,
Nguyen
T
,
Lau
T
,
Boggs
J
,
Sambrone
A
, et al
.
Tankyrase inhibition causes reversible intestinal toxicity in mice with a therapeutic index < 1
.
Toxicol Pathol
2016
;
44
:
267
78
.
22.
Pommier
Y
,
O'Connor
MJ
,
de Bono
J
.
Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action
.
Sci Transl Med
2016
;
8
:
362ps17
.
23.
Hopkins
TA
,
Ainsworth
WB
,
Ellis
PA
,
Donawho
CK
,
DiGiammarino
EL
,
Panchal
SC
, et al
.
PARP1 trapping by PARP inhibitors drives cytotoxicity in both cancer cells and healthy bone marrow
.
Mol Cancer Res
2019
;
17
:
409
19
.
24.
Dawicki-McKenna
JM
,
Langelier
MF
,
DeNizio
JE
,
Riccio
AA
,
Cao
CD
,
Karch
KR
, et al
.
PARP-1 activation requires local unfolding of an autoinhibitory domain
.
Mol Cell
2015
;
60
:
755
68
.
25.
Ryu
K
,
Lee
S
,
Seo
H
,
Yang
H
,
Seong
W
,
Yoon
J
.
Process for preparing a phthalazinone derivative and intermediates thereof
;
2021
.
Available from:
https://patents.google.com/patent/US20210323946A1/en.
26.
Egan
WJ
,
Merz
KM
Jr
,
Baldwin
JJ
.
Prediction of drug absorption using multivariate statistics
.
J Med Chem
2000
;
43
:
3867
77
.
27.
Hughes
JD
,
Blagg
J
,
Price
DA
,
Bailey
S
,
Decrescenzo
GA
,
Devraj
RV
, et al
.
Physiochemical drug properties associated with in vivo toxicological outcomes
.
Bioorg Med Chem Lett
2008
;
18
:
4872
5
.
28.
Thorsell
AG
,
Ekblad
T
,
Karlberg
T
,
Low
M
,
Pinto
AF
,
Tresaugues
L
, et al
.
Structural basis for potency and promiscuity in poly(ADP-ribose) polymerase (PARP) and tankyrase inhibitors
.
J Med Chem
2017
;
60
:
1262
71
.
29.
O'Connor
MJ
.
Targeting the DNA damage response in cancer
.
Mol Cell
2015
;
60
:
547
60
.
30.
Brown
JS
,
O'Carrigan
B
,
Jackson
SP
,
Yap
TA
.
Targeting DNA repair in cancer: beyond PARP inhibitors
.
Cancer Discov
2017
;
7
:
20
37
.
31.
Nowsheen
S
,
Yang
ES
.
The intersection between DNA damage response and cell death pathways
.
Exp Oncol
2012
;
34
:
243
54
.
32.
Sun
K
,
Mikule
K
,
Wang
Z
,
Poon
G
,
Vaidyanathan
A
,
Smith
G
, et al
.
A comparative pharmacokinetic study of PARP inhibitors demonstrates favorable properties for niraparib efficacy in preclinical tumor models
.
Oncotarget
2018
;
9
:
37080
96
.
33.
U.S. Food and Drug Administration
.
206162Orig1s000 Pharmacology review(s)
;
2014
.
Available from:
https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206162Orig1s000PharmR.pdf.
34.
European Medicines Agency
.
Assessment report Talzenna
;
2019
.
Available from:
https://www.ema.europa.eu/en/documents/assessment-report/talzenna-epar-public-assessment-report_en.pdf.
35.
Shen
Y
,
Rehman
FL
,
Feng
Y
,
Boshuizen
J
,
Bajrami
I
,
Elliott
R
, et al
.
BMN 673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency
.
Clin Cancer Res
2013
;
19
:
5003
15
.
36.
Weber GF.
DNA damaging drugs
. In:
Weber GF, Molecular therapies of cancer
.
Switzerland
:
Springer International Publishing
;
2015
. p.
9
112
.
37.
Bowes
J
,
Brown
AJ
,
Hamon
J
,
Jarolimek
W
,
Sridhar
A
,
Waldron
G
, et al
.
Reducing safety-related drug attrition: the use of in vitro pharmacological profiling
.
Nat Rev Drug Discov
2012
;
11
:
909
22
.
38.
Risdon
EN
,
Chau
CH
,
Price
DK
,
Sartor
O
,
Figg
WD
.
PARP inhibitors and prostate cancer: to infinity and beyond BRCA
.
Oncologist
2021
;
26
:
e115
29
.
39.
Kim
YM
,
Bae
K-S
,
Baek
NS
,
Kim
K
,
Lee
WS
,
Roh
E-J
, et al
.
First-in-human dose-finding study of venadaparib (IDX-1197), a potent and selective PARP inhibitor, in patients with advanced solid tumors
.
J Clin Oncol
39
:
15s
,
2021
(
suppl; abstr 3107
).
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.