Purpose: Poly(ADP-ribose) polymerase (PARP) inhibitors are undergoing extensive clinical testing for their single-agent activity in homologous recombination (HR)-deficient tumors and ability to enhance the action of certain DNA-damaging agents. Compared with other PARP inhibitors in development, iniparib (4-iodo-3-nitrobenzamide) is notable for its simple structure and the reported ability of its intracellular metabolite 4-iodo-3-nitrosobenzamide to covalently inhibit PARP1 under cell-free conditions. The present preclinical studies were conducted to compare the actions iniparib with the more extensively characterized PARP inhibitors olaparib and veliparib.

Experimental Design: The abilities of iniparib, olaparib, and veliparib to (i) selectively induce apoptosis or inhibit colony formation in HR-deficient cell lines, (ii) selectively sensitize HR-proficient cells to topoisomerase I poisons, and (iii) inhibit formation of poly(ADP-ribose) polymer (pADPr) in intact cells were compared.

Results: Consistent with earlier reports, olaparib and veliparib selectively induced apoptosis and inhibited colony formation in cells lacking BRCA2 or ATM. Moreover, like earlier generation PARP inhibitors, olaparib and veliparib sensitized cells to the topoisomerase I poisons camptothecin and topotecan. Finally, olaparib and veliparib inhibited formation of pADPr in intact cells. In contrast, iniparib exhibited little or no ability to selectively kill HR-deficient cells, sensitize cells to topoisomerase I poisons, or inhibit pADPr formation in situ. In further experiments, iniparib also failed to sensitize cells to cisplatin, gemcitabine, or paclitaxel.

Conclusions: While iniparib kills normal and neoplastic cells at high (>40 μmol/L) concentrations, its effects are unlikely to reflect PARP inhibition and should not be used to guide decisions about other PARP inhibitors. Clin Cancer Res; 18(6); 1655–62. ©2012 AACR.

Translational Relevance

PARP inhibitors are currently undergoing extensive clinical testing. Recent negative results with one putative PARP inhibitor, iniparib, have raised concerns about the usefulness of inhibiting PARP to treat cancer. The present study shows that iniparib fails to exhibit hallmark features of PARP inhibitors, including ability to selectively kill homologous recombination–deficient cells or inhibit synthesis of pADPr in intact cells. These results raise questions about the classification of iniparib as a PARP inhibitor and encourage caution in making decisions about the utility of PARP inhibitors based on clinical trials of iniparib.

The concept of synthetic lethality centers on targeting 2 separate molecular pathways that are nonlethal when disrupted individually, but are lethal when inhibited simultaneously. This approach is increasingly being used to guide the development of targeted anticancer therapies (1, 2). For example, based on the observation that PARP inhibitors are selectively toxic to cells lacking homologous recombination (HR) proteins such as BRCA1, BRCA2, and ATM (3–5), PARP inhibitors are being tested in tumors harboring BRCA1 or BRCA2 mutations (2, 6, 7). Recent results have shown that the PARP inhibitor olaparib induces partial or complete remissions in 41% of advanced breast and 33% of recurrent ovarian cancers in BRCA1/2 mutation carriers (8, 9). In light of these promising results, there has been substantial effort to develop olaparib, veliparib, and a variety of other third-generation PARP inhibitors as antineoplastic agents (reviewed in refs. 2, 6, 7, 10, 11).

Among the PARP-directed agents currently under development, iniparib is the furthest along in clinical testing. A simple mimic of nicotinamide (Fig. 1), this agent was originally described as the prodrug of 4-iodo-3-nitrosobenzamide, an agent that covalently inhibits PARP1 by binding to its first zinc finger under cell-free conditions (12). Subsequent studies suggested that iniparib exhibits single-agent activity in triple-negative breast cancer lines and enhances the cytotoxicity of cisplatin and gemcitabine (13). A phase II clinical trial suggested that iniparib not only is well tolerated when administered with gemcitabine and carboplatin, but also increases the response rate of the gemcitabine/carboplatin regimen in patients with triple-negative breast cancer (14). A subsequent phase III trial, however, failed to reproduce these results, raising questions about the future of PARP inhibition as a therapeutic strategy (15).

Figure 1.

Structures of nicotinamide and the 3 drugs used in this study.

Figure 1.

Structures of nicotinamide and the 3 drugs used in this study.

Close modal

Recent studies from our laboratory showed that veliparib and olaparib, which are active site-directed noncovalent PARP inhibitors, selectively kill HR-deficient ovarian and pancreatic cells by causing activation of the error-prone nonhomologous end-joining DNA repair pathway (16). In anticipation of experiments to determine whether covalent modification of PARP1 by 4-iodo-3-nitrosobenzamide kills in a similar fashion, we examined the cytotoxicity of iniparib in HR-deficient cells. Surprisingly, we observed that iniparib exhibited little selectivity for HR-deficient cells. Further studies failed to show the ability of iniparib to sensitize cells to topoisomerase I poisons or inhibit pADPr synthesis in situ, two other hallmarks of PARP inhibitors. Collectively, these studies argue against the possibility that iniparib is inducing cytotoxicity by inhibiting PARP.

Materials

Veliparib (ABT-888) was purchased from Enzo Life Sciences; olaparib (AZD2281), iniparib (BSI-201), and VE-821 were from ChemieTek; and camptothecin, gemcitabine, paclitaxel, propidium iodide (PI), etoposide, bovine serum albumin, and gelatin from Sigma-Aldrich. Topotecan was provided by the Drug Synthesis Branch of the National Cancer Institute (Bethesda, MD). AZD 7762 was a kind gift from L. Karnitz (Mayo Clinic, Rochester, MN). Polyclonal rabbit 96-10 anti-pADPr antiserum was raised as reported (17).

Cell culture

Mouse embryo fibroblasts (MEF) from wild-type or Atm−/− mice (Z. Lou, Mayo Clinic) were cultured in Dulbecco's Modified Eagle's Medium (DMEM; medium A). GM16666 and GM16667 human fibroblast lines from the Coriell Institute (Camden, NJ) were cultured in DMEM with 100 μg/mL hygromycin. PEO1 and PEO4 cells (18) from F. Couch (Mayo Clinic) were cultured in DMEM containing 100 μmol/L nonessential amino acids and 10 μg/mL insulin. SKOV3 cells (V. Shridhar, Mayo Clinic) were cultured in McCoy's 5A. All media contained 10% heat inactivated FBS, 40 units/mL penicillin G, 40 μg/mL streptomycin, and 1 mmol/L glutamine. Lines were genotyped shortly before acquisition and were reinitiated every 2 to 3 months from stocks that were cryopreserved immediately after receipt from the indicated sources.

Apoptosis assays

Cells plated at 5 × 104 cells per 60-mm dish were allowed to adhere for 24 hours, then treated with veliparib, olaparib, or iniparib in 0.1% (v/v) dimethyl sulfoxide for 4 days (olaparib) or 6 days (veliparib, iniparib) based on preliminary time course experiments. At the end of treatment, adherent cells were recovered by trypsinization, combined with cells in the supernatant, sedimented at 150 × g for 10 minutes, washed in ice-cold calcium- and magnesium-free Dulbecco's phosphate buffered saline (PBS), and fixed at 4°C by dropwise addition of ethanol to a final concentration of 50% (v/v). Cells were subsequently rehydrated in cold PBS, sedimented, resuspended in 300 μL 0.1% (w/v) sodium citrate containing 1 mg/mL RNase A, incubated for 15 minutes at 37°C, diluted with 300 μL 0.1% sodium citrate containing 100 μg/mL PI, incubated in the dark at 20°C for 15 minutes, and analyzed (20,000 events) on a FACSCanto II flow cytometer (Becton Dickinson) using excitation and emission wavelengths of 488 and 617 nm, respectively. After data were analyzed with Becton Dickinson CellQuest software, normalized apoptosis was calculated as (observed − baseline)/(100 − baseline) to correct for differences in basal apoptosis rates between cell lines. Results are representative of at least 3 independent experiments.

Colony-forming assays

Wild-type and Atm−/− MEFs were plated at 500 cells per dish in 60-mm dishes containing medium A, allowed to adhere overnight, and treated with the indicated agents continuously. Other cell lines were plated at 1,000 cells per plate (PEO1 and PEO4), 750 cells per plate (GM16666 and 16667), or 500 cells per plate (SKOV3) in their respective media, allowed to adhere for up to 14 hours, treated with the indicated agents for 48 hours (GM16666 and 16667) or continuously, stained with Coomassie Brilliant Blue, and scored for colony formation (≥50 cells) manually.

pADPr levels

Relative pADPr levels were assessed by quantitative fluorescence microscopy (modified from refs. 19, 20). In brief, SKOV3 cells grown on ethanol-washed coverslips were treated with the indicated agents for 4 hours before addition of 1 mmol/L methyl methanesulfonate (MMS) for 30 minutes to stimulate polymer formation. Following treatment, cells were fixed in −20°C methanol:acetone (70:30); incubated for 1 hour at 21°C in blocking buffer consisting of 1% (v/v) glycerol, 0.1% (w/v) gelatin from cold-water fish, 0.1% (w/v) bovine serum albumin, 5% (v/v) goat serum, and 0.4% (w/v) sodium azide in PBS; exposed to 96-10 anti-pADPr antiserum (1:500 dilution in blocking buffer) overnight at 4°C; washed; incubated for 1 hour with Alexa Fluor 568–conjugated goat anti-rabbit IgG (Invitrogen) in blocking buffer; washed; counterstained with 1 μg/mL Hoechst 33258 in PBS; and examined by confocal microscopy as described (20).

Inability of iniparib to selectively kill HR-deficient cells

Previous studies have identified important cellular effects of PARP inhibitors, including selective toxicity in HR-deficient cells (3–5, 16, 21, 22), synergistic cytotoxicity when combined with topoisomerase I poisons (20, 23–27), and ability to inhibit pADPr formation in cells with damaged DNA. In the present study, we compared 3 of the agents currently undergoing clinical testing (Fig. 1) in assays of these effects.

To compare the ability of these agents to selectively induce apoptosis in HR-deficient cells, BRCA2-deficient PEO1 human ovarian cancer cells and their BRCA2-revertant PEO4 counterparts (18, 21) were incubated with olaparib, veliparib, or iniparib, then stained with PI and subjected to flow cytometry. As depicted in Fig. 2A and summarized in Fig. 2B, PEO1 cells were much more sensitive to olaparib and veliparib than the PEO4 cells. In contrast, iniparib exhibited much less selectivity (Fig. 2A and B, right). Similar results were observed when the cells were stained with Hoechst 33258 and examined for apoptotic morphologic changes (Supplementary Fig. S1).

Figure 2.

Effects of veliparib, olaparib, and iniparib on PEO1 and PEO4 cells. A, PEO1 and PEO4 treated with diluent, 20 μmol/L veliparib (left), 5 μmol/L olaparib (middle), or 80 μmol/L iniparib (right) were stained with PI and subjected to flow microfluorimetry. B, summary of results obtained in (A) and additional concentrations. Inset in B, whole-cell lysates were subjected to immunoblotting with antibody to BRCA2 and, as a loading control, PARP1. C, effects of various concentrations of veliparib (left), olaparib (middle), and iniparib (right) on colony formation. Error bars in these and subsequent colony-forming assays, ±SD of triplicate aliquots. All results are representative of 3 or more independent experiments.

Figure 2.

Effects of veliparib, olaparib, and iniparib on PEO1 and PEO4 cells. A, PEO1 and PEO4 treated with diluent, 20 μmol/L veliparib (left), 5 μmol/L olaparib (middle), or 80 μmol/L iniparib (right) were stained with PI and subjected to flow microfluorimetry. B, summary of results obtained in (A) and additional concentrations. Inset in B, whole-cell lysates were subjected to immunoblotting with antibody to BRCA2 and, as a loading control, PARP1. C, effects of various concentrations of veliparib (left), olaparib (middle), and iniparib (right) on colony formation. Error bars in these and subsequent colony-forming assays, ±SD of triplicate aliquots. All results are representative of 3 or more independent experiments.

Close modal

In further experiments, antiproliferative effects of the 3 agents were compared in colony-forming assays. This assay likewise showed that veliparib and olaparib exhibited selectivity for the BRCA2-deficient PEO1 cells (Fig. 2C, left and middle), whereas iniparib exhibited no selectivity (Fig. 2C, right).

To ensure that these observations were not unique to PEO1 and PEO4 cells, we also examined the effects of the 3 agents in ATM-deficient GM16666 and ATM-restored GM16667 fibroblasts. Once again, veliparib and olaparib exhibited selectivity for the HR-deficient cells (Fig. 3A and B, left and middle), whereas iniparib exhibited very little selectivity (Fig. 3A and B, right). Similar results were also observed in Atm−/− mouse fibroblasts compared with their wild-type counterparts (Fig. 3C).

Figure 3.

Effects of veliparib, olaparib, and iniparib on paired GM16666 (ATM deficient) and GM16667 (ATM restored) human fibroblasts. A, cells treated with diluent or the indicated concentrations of veliparib (left), olaparib (middle) or iniparib (right) were stained with PI and examined by flow microfluorimetry as in Fig. 2A. Inset in A, whole cell lysates were subjected to immunoblotting for ATM and, as a loading control, heat shock protein 90β (HSP90β). B, effects of veliparib (left), olaparib (middle) and iniparib (right) on colony formation. C, Atm/ and wild-type MEFs examined by colony-forming assays.

Figure 3.

Effects of veliparib, olaparib, and iniparib on paired GM16666 (ATM deficient) and GM16667 (ATM restored) human fibroblasts. A, cells treated with diluent or the indicated concentrations of veliparib (left), olaparib (middle) or iniparib (right) were stained with PI and examined by flow microfluorimetry as in Fig. 2A. Inset in A, whole cell lysates were subjected to immunoblotting for ATM and, as a loading control, heat shock protein 90β (HSP90β). B, effects of veliparib (left), olaparib (middle) and iniparib (right) on colony formation. C, Atm/ and wild-type MEFs examined by colony-forming assays.

Close modal

Failure of iniparib to synergize with topoisomerase I poisons

Another hallmark of PARP inhibitors is their ability to synergize with topoisomerase I poisons (20, 23–27). To avoid the potential confounding effect of P-glycoprotein, which is constitutively expressed at low levels in rodent cells (28) and has been reported to affect uptake of topotecan (29–31), experiments in MEFs used camptothecin. At submicromolar concentrations that were themselves nontoxic, veliparib and olaparib enhanced the sensitivity of wild-type MEFs to camptothecin (Fig. 4A). In contrast, 100-fold higher iniparib concentrations, which were just at the point of inhibiting colony formation by themselves, had no discernible effect on camptothecin sensitivity (Fig. 4B). When topotecan, which is used to treat epithelial ovarian cancer (32, 33), was administered to SKOV3 cells, veliparib and olaparib likewise enhanced the cytotoxicity of the topoisomerase I poison whereas iniparib did not (Fig. 4C and data not shown).

Figure 4.

Inability of iniparib to sensitize cells to topoisomerase I poisons. MEFs (A and B) or SKOV3 cells (C) were treated continuously with the indicated concentrations of camptothecin (A and B) or topotecan (C) in the presence of 100 nmol/L veliparib, 100 nmol/L olaparib, or 10 μmol/L iniparib, then examined for colony formation.

Figure 4.

Inability of iniparib to sensitize cells to topoisomerase I poisons. MEFs (A and B) or SKOV3 cells (C) were treated continuously with the indicated concentrations of camptothecin (A and B) or topotecan (C) in the presence of 100 nmol/L veliparib, 100 nmol/L olaparib, or 10 μmol/L iniparib, then examined for colony formation.

Close modal

Further effects of iniparib in combination

In view of the inability of iniparib to sensitize cells to topoisomerase I poisons, we also examined the ability of iniparib to sensitize SKOV3 cells to a number of other classes of agents with which it is being combined in the clinic (www.clinicaltrials.gov). In these experiments, iniparib failed to sensitize cells to cisplatin (Fig. 5A). In contrast, sensitization by the ATR inhibitor VE-821 was readily detected (Fig. 5B) as previously reported (34), indicating that sensitization by iniparib could have been observed if present. Likewise, iniparib failed to sensitize to gemcitabine (Fig. 5C) even though sensitization by the checkpoint kinase inhibitor AZD 7762 (35) was readily demonstrated (Fig. 5D). We also failed to observe sensitization of SKOV3 cells to paclitaxel (Fig. 5E). In contrast, iniparib slightly but reproducibly sensitized SKOV3 cells to etoposide (Fig. 5F).

Figure 5.

Effects of iniparib in combination with cisplatin, gemcitabine, paclitaxel, or etoposide. SKOV3 cells were treated continuously with the indicated concentrations of cisplatin (A), gemcitabine (C), paclitaxel (E), or etoposide (F) in the presence of diluent or 10 μmol/L iniparib and examined for colony formation. In panels B and D, the ATR inhibitor VE-821 (1 μmol/L), which is known to sensitize other cells to cisplatin (34), or the Chk1 inhibitor AZD 7762 (100 nmol/L), which is known to sensitize to gemcitabine (35), was substituted for iniparib as positive controls. Error bars in each panel, ±SD of triplicate samples.

Figure 5.

Effects of iniparib in combination with cisplatin, gemcitabine, paclitaxel, or etoposide. SKOV3 cells were treated continuously with the indicated concentrations of cisplatin (A), gemcitabine (C), paclitaxel (E), or etoposide (F) in the presence of diluent or 10 μmol/L iniparib and examined for colony formation. In panels B and D, the ATR inhibitor VE-821 (1 μmol/L), which is known to sensitize other cells to cisplatin (34), or the Chk1 inhibitor AZD 7762 (100 nmol/L), which is known to sensitize to gemcitabine (35), was substituted for iniparib as positive controls. Error bars in each panel, ±SD of triplicate samples.

Close modal

Failure of iniparib to inhibit pADPr synthesis

In view of the limited selectively of iniparib for HR-deficient cells (Figs. 2 and 3) and inability of iniparib to sensitize to topoisomerase I poisons (Fig. 4), we examined the ability of iniparib to inhibit PARP in situ. For these studies polymer levels were determined by quantitative fluorescence microscopy, a method that is widely used and is more quantifiable than immunoblotting (27, 36). Treatment with veliparib or olaparib caused a dose-dependent decrease in DNA damage–induced pADPr levels (Fig. 6). In contrast, iniparib had no detectable effect on polymer formation in situ at concentrations up to 100 μmol/L.

Figure 6.

Effects of veliparib (Veli), olaparib (Ola), and iniparib (Ini) on pADPr synthesis. A, following a 4-hour treatment with the indicated agent, SKOV3 cells were treated with MMS in the continued presence of the small molecule inhibitor, fixed, stained for pADPr and examined by confocal microscopy. B, mean fluorescence of pADPr in nuclei after treatment of cells with diluent (open bar) or 1 mmol/L MMS (closed bars) in the presence of diluent or the indicated agent at 1, 10, and 100 μmol/L. Error bars, ±SEM for 30 nuclei.

Figure 6.

Effects of veliparib (Veli), olaparib (Ola), and iniparib (Ini) on pADPr synthesis. A, following a 4-hour treatment with the indicated agent, SKOV3 cells were treated with MMS in the continued presence of the small molecule inhibitor, fixed, stained for pADPr and examined by confocal microscopy. B, mean fluorescence of pADPr in nuclei after treatment of cells with diluent (open bar) or 1 mmol/L MMS (closed bars) in the presence of diluent or the indicated agent at 1, 10, and 100 μmol/L. Error bars, ±SEM for 30 nuclei.

Close modal

Previous studies have identified a number of biological properties of PARP inhibitors, including selective toxicity in HR-deficient cells, ability to synergize with topoisomerase I poisons, and, by definition, ability to inhibit pADPr synthesis in intact cells. Iniparib was originally described as a prodrug of a covalent PARP inhibitor (12, 37) and more recently as a small molecule cytotoxic with some PARP inhibitory activity (14). This agent displayed promising activity in combination with gemcitabine and cisplatin in phase II trials in triple-negative breast cancer (14) and ovarian cancer (38, 39) as well as anecdotal activity against BRCA2-deficient pancreatic cancer (40). Interestingly, iniparib has not been previously compared head-to-head with other PARP inhibitors in preclinical studies. Results of the present study, however, indicate that iniparib (in contrast to olaparib and veliparib) fails to exhibit much selectivity for HR-deficient cells (Figs. 2 and 3), fails to sensitize to topoisomerase I poisons (Fig. 4), and fails to inhibit pADPr synthesis in intact cells (Fig. 6). These results have important implications for current interpretations of iniparib clinical trials.

Earlier studies showed that PARP knockdown or PARP inhibition is selectively toxic to cells with HR defects (2, 6, 7). More recent studies have suggested that this selective toxicity stems from the ability of active site-directed noncovalent PARP inhibitors to tip the balance toward error-prone nonhomologous end-joining in HR-deficient cells (16). In anticipation of studies that would determine whether the intracellular metabolite 4-iodo-3-nitrosobenzamide, a putative covalent inhibitor of PARP1, acts in a similar fashion, we examined cells lacking BRCA2 (Fig. 2) or ATM (Fig. 3). In both cases, iniparib at concentrations exceeding 40 μmol/L killed cells but failed to show the anticipated selectivity for the HR-deficient cells. This lack of selectivity, which was demonstrated using assays for both apoptosis and colony formation, immediately distinguishes iniparib from other widely studied PARP inhibitors such as veliparib, olaparib, and MK-4827 (Figs. 2 and 3; ref. 20).

A wide variety of PARP inhibitors have also been shown to selectively enhance the cytotoxicity of topoisomerase I poisons (20, 23–27). This sensitization was readily detected with submicromolar concentrations of olaparib and veliparib (Fig. 4). In contrast, iniparib failed to sensitize multiple cell lines to either camptothecin or topotecan, again indicating a substantial difference in behavior compared with bona fide PARP inhibitors. Conversely, iniparib sensitized cells to etoposide (Fig. 5F), albeit modestly, whereas other PARP inhibitors failed to sensitize cells to this agent (20, 23, 41), again distinguishing iniparib from the PARP inhibitors.

Earlier studies indicated that iniparib can inhibit pADPr synthesis when glutathione levels in cells are depleted with buthionine sulfoximine (42). In contrast, we were unable to detect inhibition of pADPr synthesis in cells containing endogenous levels of glutathione (Fig. 6). This failure to inhibit pADPr synthesis presumably accounts for the inability of iniparib to selectively kill HR-deficient cells or synergize with topoisomerase I poisons.

Although iniparib does not seem to be exerting its effects through inhibition of pADPr synthesis, this agent clearly is cytotoxic to a variety of cell lines at concentrations above 40 μmol/L (Figs. 2 and 3). While the mechanism of this cytotoxicity was not explored in the present study, structural similarity of iniparib to nicotinamide (Fig. 1) raises the possibility that the cytotoxic effects of iniparib reflect the inhibition of one or more enzymes that bind the nicotinamide derivative NAD+, including possibly glyceraldehyde-3-phosphate dehydrogenase (GAPDH) or sirtuins (42), rather than primary effects on PARP.

In further experiments, the effect of combining iniparib with other chemotherapeutic agents was examined. Current or recently completed trials (www.clinicaltrials.gov) have paired iniparib with paclitaxel (triple-negative breast cancer), carboplatin + gemcitabine (triple-negative breast cancer, ovarian cancer, non–small cell lung cancer), or carboplatin + paclitaxel (uterine carcinosarcoma). Accordingly, we specifically focused on the effects of combining iniparib with these classes of agents. Iniparib failed to sensitize SKOV3 cells to cisplatin, gemcitabine, or paclitaxel (Fig. 5A, C, and E), whereas other enzyme inhibitors caused readily detectable sensitization (Fig. 5B and D). Even though carboplatin was not used in these experiments because of its lower solubility and potency in vitro, results with carboplatin would likely be similar to our observations with cisplatin because of the identical mechanism of action of these agents (43). It is possible that different results might be obtained in different cell lines with different genetic and epigenetic changes. Nonetheless, our observation that iniparib has limited impact on sensitivity of SKOV3 cells to platinating agents, taxanes or gemcitabine might be important for interpreting results of recently completed and ongoing clinical trials of iniparib in combination with these agents.

In view of the marked differences between iniparib and other PARP inhibitors described here, it is important that clinical results obtained with iniparib not be allowed to unduly influence development of bona fide PARP inhibitors. In particular, the recent disclosure that results of the phase III iniparib trial in triple-negative breast cancer were negative (15, 44) should not be interpreted to mean that bona fide PARP inhibitors will also fail to exhibit activity in this disease, as it is unlikely that iniparib inhibited PARP in this trial. In a similar fashion, if iniparib exhibits clinical activity in a different setting (38–40), these positive findings should also be extrapolated to bona fide PARP inhibitors extremely cautiously, if at all, because of the failure of iniparib to exhibit the properties of a PARP inhibitor as documented in the present study.

No potential conflicts of interest were disclosed.

Study design: A.G. Patel, S.H. Kaufmann

Experiments and interpretation of data: A.G. Patel, S.B. De Lorenzo, K.S. Flatten

Critical reagents: G.G. Poirier

Writing and editing the manuscript: A.G. Patel, S.H. Kaufmann, S.B. De Lorenzo, K.S. Flatten, G.G. Poirier

The authors thank Larry Karnitz, Zhenkun Lou, Viji Shridhar, and Fergus Couch for reagents; Deb Strauss for secretarial assistance; and Matt Goetz and Lynn Hartmann for provocative discussions.

This work was supported in part by P50 CA136393 (S.H. Kaufmann and G.G. Poirier). A.G. Patel was supported by T32 GM072474 and a predoctoral fellowship from the Mayo Foundation.

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

1.
Kaelin
WG
 Jr
. 
The concept of synthetic lethality in the context of anticancer therapy
.
Nat Rev Cancer
2005
;
5
:
689
98
.
2.
Rehman
FL
,
Lord
CJ
,
Ashworth
A
. 
Synthetic lethal approaches to breast cancer therapy
.
Nat Rev Clin Oncol
2010
;
7
:
718
24
.
3.
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
.
4.
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
.
5.
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
.
6.
Rouleau
M
,
Patel
A
,
Hendzel
MJ
,
Kaufmann
SH
,
Poirier
GG
. 
PARP inhibition: PARP1 and beyond
.
Nat Rev Cancer
2010
;
10
:
293
301
.
7.
Javle
M
,
Curtin
NJ
. 
The role of PARP in DNA repair and its therapeutic exploitation
.
Br J Cancer
2011
;
105
:
1114
22
.
8.
Audeh
MW
,
Carmichael
J
,
Penson
RT
,
Friedlander
M
,
Powell
B
,
Bell-McGuinn
KM
, et al
Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial
.
Lancet
2010
;
376
:
245
51
.
9.
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
.
10.
Penning
TD
. 
Small-molecule PARP modulators–current status and future therapeutic potential
.
Curr Opin Drug Discov Devel
2010
;
13
:
577
86
.
11.
Calvert
H
,
Azzariti
A
. 
The clinical development of inhibitors of poly(ADP-ribose) polymerase
.
Ann Oncol
2011
;
22
Suppl 1
:
i53
9
.
12.
Mendeleyev
J
,
Kirsten
E
,
Hakam
A
,
Buki
KG
,
Kun
E
. 
Potential chemotherapeutic activity of 4-iodo-3-nitrobenzamide. Metabolic reduction to the 3-nitroso derivative and induction of cell death in tumor cells in culture
.
Biochem Pharmacol
1995
;
50
:
705
14
.
13.
Ossovskaya
V
,
Li
L
,
Broude
EV
. 
BSI-201 enhances the activity of multiple classes of cytotoxic agaents and irradiation in triple negative breast cancer [abstract]
. In:
Proceedings of the 100th Annual Meeting of the American Association for Cancer Research; 2009 Apr 18–22
;
Denver, CO. Philadelphia (PA)
:
AACR
; 
2009
.
Abstract nr 5552
.
14.
O'Shaughnessy
J
,
Osborne
C
,
Pippen
JE
,
Yoffe
M
,
Patt
D
,
Rocha
C
, et al
Iniparib plus chemotherapy in metastatic triple-negative breast cancer
.
N Engl J Med
2011
;
364
:
205
14
.
15.
Guha
M
. 
PARP inhibitors stumble in breast cancer
.
Nat Biotechnol
2011
;
29
:
373
4
.
16.
Patel
A
,
Sarkaria
J
,
Kaufmann
SH
. 
Nonhomologous end-joining drives PARP inhibitor synthetic lethality in homologous recombination-deficient cells
.
Proc Natl Acad Sci U S A
2011
;
108
:
3406
11
.
17.
Affar
EB
,
Duriez
PJ
,
Shah
RG
,
Sallmann
FR
,
Bourassa
S
,
Kupper
JH
, et al
Immunodot blot method for the detection of poly(ADP-ribose) synthesized in vitro and in vivo
.
Anal Biochem
1998
;
259
:
280
3
.
18.
Langdon
SP
,
Lawrie
SS
,
Hay
FG
,
Hawkes
MM
,
McDonald
A
,
Hayward
IP
, et al
Characterization and properties of nine human ovarian adenocarcinoma cell lines
.
Cancer Res
1988
;
48
:
6166
72
.
19.
Haince
JF
,
McDonald
D
,
Rodrigue
A
,
Dery
U
,
Masson
JY
,
Hendzel
MJ
, et al
PARP1-dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites
.
J Biol Chem
2008
;
283
:
1197
208
.
20.
Patel
AG
,
Flatten
KS
,
Schneider
PA
,
Dai
NT
,
McDonald
JS
,
Poirier
GG
, et al
Enhanced killing of cancer cells by poly(ADP-ribose) polymerase inhibitors and topoisomerase I inhibitors reflects poisoning of both enzymes
.
J Biol Chem
2012
;
287
:
4198
210
.
21.
Sakai
W
,
Swisher
EM
,
Karlan
BY
,
Agarwal
MK
,
Higgins
J
,
Friedman
C
, et al
Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers
.
Nature
2008
;
451
:
1116
20
.
22.
Edwards
SL
,
Brough
R
,
Lord
CJ
,
Natrajan
R
,
Vatcheva
R
,
Levine
DA
, et al
Resistance to therapy caused by intragenic deletion in BRCA2
.
Nature
2008
;
451
:
1111
5
.
23.
Bowman
KJ
,
Newell
DR
,
Calvert
AH
,
Curtin
NJ
. 
Differential effects of the poly (ADP-ribose) polymerase (PARP) inhibitor NU1025 on topoisomerase I and II inhibitor cytotoxicity in L1210 cells in vitro
.
Br J Cancer
2001
;
84
:
106
12
.
24.
Calabrese
CR
,
Almassy
R
,
Barton
S
,
Batey
MA
,
Calvert
AH
,
Canan-Koch
S
, et al
Anticancer chemosensitization and radiosensitization by the novel poly(ADP-ribose) polymerase-1 inhibitor AG14361
.
J Natl Cancer Inst
2004
;
96
:
56
67
.
25.
Smith
LM
,
Willmore
E
,
Austin
CA
,
Curtin
NJ
. 
The novel poly(ADP-Ribose) polymerase inhibitor, AG14361, sensitizes cells to topoisomerase I poisons by increasing the persistence of DNA strand breaks
.
Clin Cancer Res
2005
;
11
:
8449
57
.
26.
Thomas
HD
,
Calabrese
CR
,
Batey
MA
,
Canan
S
,
Hostomsky
Z
,
Kyle
S
, et al
Preclinical selection of a novel poly(ADP-ribose) polymerase inhibitor for clinical trial
.
Mol Cancer Ther
2007
;
6
:
945
56
.
27.
Zhang
YW
,
Regairaz
M
,
Seiler
JA
,
Agama
KK
,
Doroshow
JH
,
Pommier
Y
. 
Poly(ADP-ribose) polymerase and XPF-ERCC1 participate in distinct pathways for the repair of topoisomerase I-induced DNA damage in mammalian cells
.
Nucleic Acids Res
2011
;
39
:
3607
20
.
28.
Kartner
N
,
Evernden-Porelle
D
,
Bradley
G
,
Ling
V
. 
Detection of P-glycoprotein in multidrug-resistant cell lines by monoclonal antibodies
.
Nature
1985
;
316
:
820
3
.
29.
Chen
AY
,
Yu
C
,
Potmesil
M
,
Wall
ME
,
Wani
MC
,
Liu
LF
. 
Camptothecin overcomes mdrl-mediated resistance in human KB carcinoma cells
.
Cancer Res
1991
;
51
:
6039
44
.
30.
Hendricks
CB
,
Rowinsky
EK
,
Grochow
LB
,
Donehower
RC
,
Kaufmann
SH
. 
Effect of P-glycoprotein expression on the accumulation and cytotoxicity of topotecan (SK&F 104864), a new camptothecin analogue
.
Cancer Res
1992
;
52
:
2268
78
.
31.
Rasheed
ZA
,
Rubin
EH
. 
Mechanisms of resistance to topoisomerase I-targeting drugs
.
Oncogene
2003
;
22
:
7296
304
.
32.
Armstrong
DK
. 
Topotecan dosing guidelines in ovarian cancer: reduction and management of hematologic toxicity
.
Oncologist
2004
;
9
:
33
42
.
33.
Markman
M
. 
Topotecan as second-line therapy for ovarian cancer: dosage versus toxicity
.
Oncologist
2005
;
10
:
695
7
.
34.
Reaper
PM
,
Griffiths
MR
,
Long
JM
,
Charrier
JD
,
Maccormick
S
,
Charlton
PA
, et al
Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR
.
Nat Chem Biol
2011
;
7
:
428
30
.
35.
Zabludoff
SD
,
Deng
C
,
Grondine
MR
,
Sheehy
AM
,
Ashwell
S
,
Caleb
BL
, et al
AZD7762, a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies
.
Mol Cancer Ther
2008
;
7
:
2955
66
.
36.
Haince
JF
,
Poirier
GG
,
Kirkland
JB
. 
Nonisotopic methods for determination of poly(ADP-ribose) levels and detection of poly(ADP-ribose) polymerase
.
Curr Protoc Cell Biol
2004
;
Chapter 18:Unit18.7
.
37.
Rice
WG
,
Hillyer
CD
,
Harten
B
,
Schaeffer
CA
,
Dorminy
M
,
Lackey
DA
 III
, et al
Induction of endonuclease-mediated apoptosis in tumor cells by C-nitroso-substituted ligands of poly(ADP-ribose) polymerase
.
Proc Natl Acad Sci U S A
1992
;
89
:
7703
7
.
38.
Birrer
MJ
,
Konstantinopoulos
P
,
Penson
RT
,
Roche
M
,
Ambrosio
A
,
Stallings
TE
, et al
A phase II trial of iniparib (BSI-201) in combination with gemcitabine/carboplatin (GC) in patients with platinum-resistant recurrent ovarian cancer
.
J Clin Oncol
29
: 
2011
(
suppl; abstr 5005
).
39.
Penson
RT
,
Whalen
C
,
Lasonde
B
,
Krasner
CN
,
Konstantinopoulos
P
,
Stallings
TE
, et al
A phase II trial of iniparib (BSI-201) in combination with gemcitabine/carboplatin (GC) in patients with platinum-sensitive recurrent ovarian cancer
.
J Clin Oncol
29
: 
2011
(
suppl; abstr 5004
).
40.
Fogelman
DR
,
Wolff
RA
,
Kopetz
S
,
Javle
M
,
Bradley
C
,
Mok
I
, et al
Evidence for the efficacy of Iniparib, a PARP-1 inhibitor, in BRCA2-associated pancreatic cancer
.
Anticancer Res
2011
;
31
:
1417
20
.
41.
Huehls
AM
,
Wagner
JM
,
Huntoon
CJ
,
Geng
L
,
Erlichman
C
,
Patel
AG
, et al
Poly(ADP-Ribose) polymerase inhibition synergizes with 5-fluorodeoxyuridine but not 5-fluorouracil in ovarian cancer cells
.
Cancer Res
2011
;
71
:
4944
54
.
42.
Bauer
PI
,
Mendeleyeva
J
,
Kirsten
E
,
Comstock
JA
,
Hakam
A
,
Buki
KG
, et al
Anti-cancer action of 4-iodo-3-nitrobenzamide in combination with buthionine sulfoximine: inactivation of poly(ADP-ribose) polymerase and tumor glycolysis and the appearance of a poly(ADP-ribose) polymerase protease
.
Biochem Pharmacol
2002
;
63
:
455
62
.
43.
Reed
E
. 
Cisplatin, carboplatin and oxaliplatin
.
In
:
Chabner
BA
,
Longo
DL
,
editors
.
Cancer chemotherapy & biotherapy: principles and practice
. 4th ed.
Philadelphia, PA
:
Lippincott Williams and Wilkins
; 
2006
. p.
332
43
.
44.
O'Shaughnessy
J
,
Schwartzberg
LS
,
Danso
MA
,
Rugo
HS
,
Miller
K
,
Yardley
DA
, et al
A randomized phase III study of iniparib (BSI-201) in combination with gemcitabine/carboplatin (G/C) in metastatic triple-negative breast cancer (TNBC)
.
J Clin Oncol
29
: 
2011
(
suppl; abstr 1007
).

Supplementary data