ABSTRACT
Poly(ADP-ribose) polymerases (PARPs) are a members of family of enzymes that catalyze poly(ADP-ribosyl)ation (PARylation) and/or mono(ADP-ribosyl)ation (MARylation), two post-translational protein modifications involved in crucial cellular processes including (but not limited to) the DNA damage response (DDR). PARP1, the most abundant family member, is a nuclear protein that is activated upon sensing distinct types of DNA damage and contributes to their resolution by PARylating multiple DDR players. Recent evidence suggests that, along with DDR, activated PARP1 mediates a series of prosurvival and proapoptotic processes aimed at preserving genomic stability. Despite this potential oncosuppressive role, upregulation and/or overactivation of PARP1 or other PARP enzymes has been reported in a variety of human neoplasms. Over the last few decades, several pharmacologic inhibitors of PARP1 and PARP2 have been assessed in preclinical and clinical studies showing potent antineoplastic activity, particularly against homologous recombination (HR)-deficient ovarian and breast cancers. In this Trial Watch, we describe the impact of PARP enzymes and PARylation in cancer, discuss the mechanism of cancer cell killing by PARP1 inactivation, and summarize the results of recent clinical studies aimed at evaluating the safety and therapeutic profile of PARP inhibitors in cancer patients.
KEYWORDS: Cell death, DNA damage, drug resistance, metabolism, NAD, olaparib, synthetic lethality
Abbreviations
- ATM
ataxia telangiectasia mutated
- BER
base excision repair
- BRCA1
breast cancer 1 early onset
- BRCA2
breast cancer 2 early onset
- DDR
DNA damage response
- DSBs
double-strand breaks
- FDA
Food and Drug Administration
- GI, gastrointestinal; HER
human epidermal growth factor receptor
- HR
homologous recombination
- MAR
mono(ADP-ribose)
- NAD+
nicotinamide adenine dinucleotide
- NER
nucleotide excision repair
- NHEJ
non-homologous end joining
- ORR
overall response rate
- PAR
poly(ADP-ribose)
- PARG
poly(ADP-ribose) glycohydrolase
- PARP
poly(ADP-ribose) polymerase
- PARylation
poly(ADP-ribosyl)ation
- PFS
progression-free survival
- PI3K
phosphatidylinositol 3-kinase
- Polθ, polymerase (DNA directed) theta SSB
single-strand break
- TNBC
triple negative breast carcinoma
- VEGF
vascular endothelial growth factor
- WGR
tryptophan (W) glycin (G) and arginine (R) domain
- Zn
zinc finger domain
Introduction
Poly(ADP-ribosyl)ation (PARylation) is a post-translational protein modification that is catalyzed by members of the poly(ADP-ribose) polymerase (PARP) enzyme family (i.e., PARP1, PARP2, tankyrase, TRF1-interacting ankyrin-related ADP-ribose polymerase [TNKS] and TNKS2) and has crucial functions in multiple physiologic and pathologic processes.1,2 This modification involves the covalent addition of a linear or multibranched polymer of ADP-ribose units from the respiratory co-enzyme nicotinamide adenine dinucleotide (NAD+) to DNA or multiple acceptor proteins involved in cellular processes as diverse as DNA damage, DNA replication, DNA transcription, differentiation, gene regulation, protein degradation, and mitotic spindle maintenance.3-5 However, most members of the PARP family, which includes 17 identified enzymes to date, catalyze the covalent attachment of ADP-ribose onto acceptor proteins as a monomer (mono(ADP-ribose), MAR) instead of a polymer (PAR) (reviewed in ref. 6).6 This prompted some authors to suggest the use of ADP-ribosyltransferase (ARTD) as an official unifying name for this family.7 A complete overview of functions of the PARP family can be found in several reviews.1,2,8-16
PARylation is considered to be a key step in the DNA damage response (DDR), a complex signal transduction pathway that is triggered in response to DNA lesions and is fundamental for the preservation of genomic stability.17-21 To date, PARP1, PARP2, and PARP3 are the only family members that have been shown to contribute to DDR by promptly detecting and repairing DNA damage,7,10,15,22 with PARP1 being responsible for the resolution of up to 90% of the lesions.3,4,23 PARP1, the most abundant isoform of the PARP enzyme family, is a nuclear protein that acts as a sensor of both single-strand breaks (SSBs) and double-strand breaks (DSBs),24,25 and thereby participates in a variety of DNA damage repair pathways, including base excision repair (BER),26-31 nucleotide excision repair (NER),32,33 non-homologous end joining (NHEJ),29,34,35 and homologous recombination (HR).36-39
PARP1 contains 6 functional domains: 3 amino (N)-terminal homologous zinc finger (Zn) domains, 2 of which (Zn1 and Zn2) are responsible for DNA damage detection whereas the third (Zn3) couples DNA binding and catalytic activity; 2 central domains, the breast cancer 1, early onset (BRCA1) C-terminal (BRCT) region and the tryptophan, glycine, and arginine (WGR) module, both of which are involved in protein-protein interactions; and carboxy (C)-terminal catalytic domain, which sequentially transfers ADP-ribose subunits from the donor NAD+ to acceptor proteins.5,24,40
Soon after the appearance of DNA lesions, such as SSBs, DSBs, DNA crosslinks, and stalled replication forks, PARP1 engages at the site of damage.8,15,41 The currently accepted model postulates that the binding of PARP1 to damaged DNA via the 3 N-terminal Zn domains induces a conformational rearrangement in the WGR domain, which in turn leads to activation of the catalytic domain.8,40,42 Of note, the catalytic activity of PARP is allosterically enhanced 10- to 500-fold within 15 to 30 s of DNA binding. The activation of PARP results in massive synthesis and covalent conjugation of long linear and branched PAR chains (PAR polymers) on several proteins surrounding the DNA lesions, including nucleosomal/linker histones, topoisomerase I, and PARP1 itself.3,43-45
PARylation contributes to DNA damage repair at multiple levels. First, the increased amount of negative charge carried by PAR polymers induces extensive chromatin relaxation at the damaged site.46,47 Second, once formed, PAR polymers are recognized by a panoply of proteins (PAR-binding proteins) via one of the 4 PAR-binding domains identified so far (reviewed in ref. 6).6 This PAR-mediated recruitment contributes to chromatin remodeling and sets the stage for DNA repair.27,28,48-54 Third, the formation of PAR polymers diminishes the affinity of PARP1 and histones for DNA, providing a mechanism for removing PARP1 from damaged DNA and restoring chromatin compaction.55-57 Furthermore, polymer growth is also antagonized by 2 enzymes that hydrolyze PAR, poly(ADP-ribose) glycohydrolase (PARG) and ADP-ribosylhydrolase like 2 (ADPRHL2, best known as ARH3).58,59 Of note, O-acyl-ADP-ribose deacylase 1 (OARD1, best known as TARG1) is believed to be responsible for removal of the terminal ADP-ribose monomer.60 The concerted action of these enzymes removes PAR polymers from PARP1, restoring its ability to recognize DNA strand breaks and initiate a new round of damage signaling.
Recent evidence indicates that PARylation by this arsenal of enzymatic tools not only contributes to the repair of potentially deleterious alterations in the structure of DNA but also drives cell fate decisions in response to a variety of extrinsic and intrinsic perturbations.14 Indeed, the roles of PARP1 in the cell span from maintaining life to inducing metabolic death (termed parthanatos).61-64 Moreover, PARP hyperactivation (for instance during DDR) may deplete intracellular stores of NAD+ thus undermining NAD+-dependent processes, including (but not limited to) glucose metabolism,3,41 and/or leading to bioenergetic catastrophe and cell death.3,41,65,66
Given their contribution to DDR, PARP enzymes, and in particular PARP1, have been extensively evaluated as targets for anticancer therapy. The large wave of preclinical and clinical studies launched during the last decade has highlighted the therapeutic benefit of PARP1/2 inhibitors, especially in cancers bearing mutations in components of DNA damage repair pathways, including (but not limited to) the HR players BRCA1 and BRCA2. On the basis of these observations, the US Food and Drug Administration (FDA) approved for the first time the PARP inhibitor olaparib for patients with BRCA-mutated ovarian cancers who have had at least 3 lines of therapy (http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm427598.htm) and granted the designation of breakthrough therapy to the PARP inhibitor rucaparib for the treatment of women with BRCA-mutated advanced ovarian cancer who have received at least 2 prior lines of platinum-based chemotherapy (http://www.fda.gov/).
As part of our Trial Watch series,67,68 here we describe the contribution of PARP to cancer and discuss preclinical and clinical investigations assessing the therapeutic use of PARP inhibitors for cancer therapy.
PARP activity and cancer
The precise effect of PARP enzymes and PARylation on tumorigenesis is complex and not yet fully elucidated. Some observations, including the high rate of spontaneous genomic instability of Parp-1−/− and Parp-2−/− cells,69-72 seem to suggest an oncosuppressive role of PARP1 and PARP2. Nevertheless, Parp-1−/− and Parp-2−/− mice demonstrate, respectively, a modest incidence/long latency73,74 or the absence71,72 of spontaneous tumor development, suggesting the existence of compensatory mechanisms acting in the absence of one of these enzymes.75 In both knockout models, however, the absence of PARP1 or PARP2 rendered mice sensitive to DNA damaging agents, including alkylating compounds and ionizing radiation.72 Moreover, the simultaneous deletion of transformation related protein 53 (Tp53) in Parp1−/− and Parp2−/− mice69,71,73 or of protein kinase, DNA activated, catalytic polypeptide (Prkdc, best known as DNAPk) in Parp1−/− mice76 promoted or accelerated spontaneous tumor development. Of note, Parp1−/− mice displayed accelerated aging and an shortened life span77 as well as an altered hypoxic response,78 whereas mice with double knockout of Parp1 and Parp2,72,79 Parp1 and ataxia telangiectasia mutated (Atm),80 Parp1 and X-ray repair complementing defective repair in Chinese hamster cells 5 (Xrcc5, best known as Ku80),74 or Parp2 and Atm81 presented early embryonic lethality. Also, despite some contrasting observations,82,83 a large body of evidence suggests an increased susceptibility to mutation and/or carcinogen-induced tumorigenesis in mice carrying a homozygous deletion of the gene encoding PARP174,77,84-90 or PARP4.91 A similar effect was also found in Parg−/− mice (lacking poly(ADP-ribose) glycohydrolase) treated with diethylnitrosamine92 suggesting the importance of a balanced level of PAR for maintaining genomic stability.
Along with its role in suppressing tumorigenesis, PARP1 activity also seems to be required for cancer cell survival. Mutations of PARP1 are in fact quite rare in cancer, even though single-nucleotide polymorphisms of the gene encoding PARP1, including V762A, have been associated with an increased risk of human cancers.93-100 On the contrary, upregulation of the expression of PARP1 and/or an increased level of PAR has been found in multiple cancer cell lines as well as in samples from patients affected by tumors of different origins, including the blood,101,102 breast,103-106 cervix,107 colon,108-112 endometrium,103,113 liver,114 lung,103,115 ovary,101,103,116-118 prostate,119-121 and skin.122,123 Overexpression or overactivation of PARP enzyme has also been observed in Ewing sarcoma,124,125 glioblastoma,126,127 meningiomas of higher grade,128 and laryngeal cancer.129 Importantly, in some of these settings PARP deregulation has been linked to malignant transformation,103,105,110,112,119,121,123 tumor aggressiveness,9,116,122,128,130 and poor survival or resistance to therapy.105,116,117,130
Taken together, these findings indicate that PARP activity has a dual role in cancer, acting as a barrier against tumorigenesis (presumably by contributing to the preservation of genome stability) and promoting survival once the tumors have established.
PARP1 in cancer therapy
The idea of employing PARP1 inhibitors as antineoplastic agents stems from the early 1980s, and seminal work by Sydney Shall's group.4 Since then, multiple companies have begun to invest in the strategic development of specific PARP1 inhibitors. On theoretical grounds PARP1 may be indirectly inhibited by depleting NAD+, the substrate of PARylation.131 Nonetheless, the use of pharmacologic inhibitors of the catalytic activity of PARP1 and PARP2 (hereafter referred to as PARP inhibitors for the sake of simplicity), including olaparib (also known as AZD2281),132 rucaparib (also known as AG-014699),133 veliparib (also known as ABT-888),134 niraparib (also known as MK-4827),135 iniparib (also known as BSI-201),136-138 CEP-9722 (the pro-drug of CEP-8983),139 E7016 (also known as GPI21016),140 and INO-1001,141 has been the most widely explored approach in the context of cancer therapy.6,12,16,75,142-147
Over the last 40 years, 2 major antineoplastic strategies have been pursued, both based on the role of PARP1 in repairing DNA lesions. In the first approach, PARP inhibitors are given in combinatorial regimens to sensitize tumor cells to conventional DNA damaging therapy. A wide range of preclinical studies showing the activity of PARP inhibitors in boosting the tumor killing effects of alkylating agents, topoisomerase I poisons, and ionizing radiation, either in vitro in tumor cell lines or in vivo in human tumor xenografts, support the validity of this approach (reviewed in ref. 146, 148, 149),146,148,149) even though the off-target and toxic effects of some of these combinations are slowing their further progression into clinical use (see below). In the second approach, the abrogation of PARP enzymatic activity is used to target tumor cells with a specific and pre-existing genomic defect in the mechanism of DNA repair. The rationale of this strategy, which is based on synthetic lethality (a concept first described by Dobzhansky in 1946150 that was at the time a novelty in cancer therapy), is that cell killing occurs only if 2 conditions (e.g., the simultaneous inhibition or ablation of 2 cellular pathways) are combined whereas neither by itself is sufficient to affect viability. Evidence of the feasibility and efficacy of this synthetic lethality approach was first provided in 2005, when 2 seminal articles that were simultaneously published provided strong evidence that PARP inhibitor monotherapy efficiently depleted cancer cells with dysfunctional BRCA1 and BRCA2.151,152 In these settings, PARP inhibitors as a standalone therapeutic intervention for tumors with specific mutations in HR also displayed limited off-target toxicity and potential tumor specificity. Besides killing BRCA-mutant cancers, PARP inhibitors have also been shown to be synthetically lethal with a variety of other HR dysfunctions, thus extending the potential patient population that may benefit from these agents.23,153-163 This phenomenon, named “BRCAness,” includes deficiency in DDR players such as ATM in T-cell pro-lymphocytic leukemia, B-cell chronic lymphocytic leukemia, mantle cell lymphoma, and breast cancer;156,157,159,164,165 in checkpoint kinase 2 (CHEK2, best known as CHK2) in sarcoma, breast cancer, ovarian cancer, and brain tumors;166 in phosphatase and tensin homolog (PTEN) in glioblastoma as well as prostate, lung, and endometrial cancers;167-171 and in meiotic recombination 11 homolog A (MRE11A) in colorectal cancer and myeloid malignancies.172-174 Of relevance in the context of cancer therapy, basal-like triple negative breast cancers (TNBCs), an aggressive subtype of breast cancer frequently associated with poor prognosis,175 are believed to have a BRCAness phenotype and thus might be targeted by PARP inhibitors.176-181 Recent evidence suggests a predictive role of polymerase (DNA directed) theta (Polθ, also known as POLQ); a DNA polymerase involved in the error-prone alternative non–homologous end joining [alt-NHEJ] DNA repair) in the response of HR-deficient tumors to PARP inhibitors.182 In particular, it has been reported that HR-deficient ovarian tumors overexpress POLQ and rely on POLQ-mediated repair for their survival.182 In this study, depletion of POLQ enhanced the killing effect of PARP inhibitors in HR-deficient cells.182 This result together with evidence that PARP1 recruits POLQ to DSBs,183 highlight a role of PARP activity in alt-NHEJ. Further supporting the use of PARP inhibitors in HR-impaired tumors, inhibition of phosphatidylinositol 3-kinases (PI3K) or cyclin-dependent kinase 1 (CDK1) has been reported to sensitize BRCA-proficient cancer to PARP inactivation by affecting BRCA1/2 expression184-186 or function,187 respectively. Moreover, recent observations in BRCA1-deficient mice seem to suggest a cancer chemopreventive effect of PARP inhibitors,188 although clinical confirmation is required. Also, high PAR levels are reported to predict the response to PARP inhibitors.189-191
An intense wave of investigations is underway to elucidate the precise mechanism underlying the synthetic lethality of PARP inhibitors in HR-defective tumor cells. Two non-mutually exclusive models postulate that inhibition of the catalytic activity of PARP results in abrogation of BER and the consequent accumulation of SSBs, which, once converted to DSBs during S-phase or upon replication fork collapse, are either left not repaired because of HR impairment151,152,192 or are managed by the error-prone DSB repair pathway of NHEJ.193,194 A third model suggests that PARP inhibitors trap PARP1 and PARP2 at the DNA lesion site thereby preventing access of other DDR players and provoking obstruction of the replicative fork.195,196 In support of this latter model, recent evidence suggests that PARP inhibitors exert killing effects and synergize with the DNA-damaging agent temozolomide primarily by trapping PARP-DNA complexes rather than by inhibiting the catalytic activity of PARP.197-199
Taken together, these results indicate that inhibition of PARP activity may be an effective therapeutic strategy for the treatment of a variety of tumors bearing deficiencies in the HR pathway or displaying BRCAness properties.
Clinical investigation of PARP inhibitors
Completed clinical studies
When this Trial Watch was being redacted (March 2015), more than 40 studies had been published aimed at evaluating PARP inhibitors as standalone agents or combined with other chemotherapeutic agents in patients with cancers often harboring BRCA mutations or displaying BRCAness (Table 1), sources http://www.ncbi.nlm.nih.gov/pubmed). Of these studies, 24 involved olaparib, 9 veliparib, 2 rucaparib, 1 CEP-9722, 1 INO-1001, and 1 niraparib. In this section we will not discuss results of the studies using iniparib200-204 because this compound has recently been reported to non-selectively modify cysteine-containing proteins in tumor cells and is therefore probably not a bona fide PARP inhibitor.136,137
Table 1.
Agent | Indication(s) | Phase | Notes | Ref. |
---|---|---|---|---|
CEP-9722 | Advanced solid tumors | I | As single agent or combined with temozolomide | 264 |
Iniparib | Brain metastases | II | Combined with irinotecan and paclitaxel | 200 |
Breast cancer | II | Combined with carboplatin and gemcitabine | 203 | |
III | Combined with carboplatin and gemcitabine | 204 | ||
Non-small cell lung cancer | II | Combined with cisplatin and gemcitabine | 202 | |
Uterine cancer | II | Combined with carboplatin and paclitaxel | 201 | |
INO-1001 | Melanoma | I | Combined with temozolomide | 234 |
Niraparib | Advanced solid tumors | I | As single agent | 217 |
Olaparib | Advanced solid tumors | I | As single agent | 206 |
As single agent | 207 | |||
As single agent | 208 | |||
Combined with bevacizumab | 249 | |||
Combined with cisplatin and gemcitabine | 228 | |||
Combined with dacarbazine | 221 | |||
Combined with topotecan | 230 | |||
II | As single agent | 205 | ||
Breast cancer | I | As single agent | 210 | |
Combined with paclitaxel | 255 | |||
II | As single agent | 211 | ||
Breast or ovarian cancer | I | Combined with carboplatin | 242 | |
Combined with cediranib | 250 | |||
II | As single agent | 214 | ||
Ewing sarcoma | II | As single agent | 216 | |
Ovarian cancer | I | As single agent | 209 | |
II | As single agent | 212 | ||
As single agent | 213 | |||
As single agent | 215 | |||
As single agent | 219 | |||
Combined with carboplatin and paclitaxel | 256 | |||
Combined with cediranib | 251 | |||
Pancreatic cancer | I | Combined with gemcitabine | 329 | |
Solid tumors | I | Combined with cisplatin | 238 | |
Rucaparib | Advanced solid tumors | I | Combined with temozolomide | 265 |
Melanoma | II | Combined with temozolomide | 237 | |
Veliparib | Advanced solid tumors | I | Combined with low-dose fractionated whole abdominal radiation | 262 |
Advanced solid tumors or lymphomas | 0 | As single agent | 330 | |
I | Combined with cyclophosphamide | 261 | ||
Combined with topotecan | 231 | |||
Brain metastases | I | Combined with whole brain radiation therapy | 263 | |
CNS tumors | I | Combined with temozolomide | 233 | |
Ovarian, fallopian tube or peritoneal cancer | I | Combined with cyclophosphamide | 266 | |
II | As single agent | 218 | ||
Prostate cancer | Pilot study | Combined with temozolomide | 260 | |
Uterine cancer | I/II | Combined with filgrastim and topotecan or pegfilgrastim | 232 |
sources: http://www.ncbi.nlm.nih.gov/pubmed, searching with the following terms: “PARP AND cancer AND patient” or “PARP1 AND cancer AND patient” or “NAME OF THE COMPOUND (e.g., olaparib) AND cancer AND patient”.
PARP inhibitors as monotherapeutic or maintenance agents
With one single exception, in which serious adverse effects were reported for more than 50% of individuals,205 in the vast majority of cases olaparib as a single agent was generally well tolerated in cancer patients.206-216 In these studies, the adverse effects were predominantly low grade and included mild gastrointestinal (GI) symptoms, fatigue, and anemia. Similar low-grade toxic effects were also found for niraparib monotherapy at the maximum tolerated dose of 300 mg/day,217 whereas some adverse effects, in particular GI symptoms, are reported for veliparib as a single agent.218
Preliminary evidence of antitumor activity of olaparib was reported in some phase I trials performed in patients with neoplasms, including ovarian cancer.206-209 In line with preclinical studies, olaparib monotherapy was particularly effective in cancers harboring BRCA1 or BRCA2 mutations;206,209 an overall clinical benefit rate of 46% was observed in BRCA1/2-mutated ovarian cancer, associated with platinum sensitivity.209 Moreover, in a phase I dose escalation study enrolling 100 patients, niraparib displayed antineoplastic activity in ovarian and breast cancer from BRCA1 or BRCA2 mutation carriers as well as in sporadic high-grade serous ovarian, non-small cell lung cancer, and prostate cancer217 (Table 1).
The clinical efficacy of olaparib against BRCA-deficient tumors has been confirmed in subsequent phase II interventional studies, including one trial enrolling individuals with a panel of advanced solid tumors (tumor response rate of 26.2% overall and 31.1%, 12.9%, 21.7%, and 50.0% in ovarian, breast, pancreatic, and prostate cancers, respectively).205 Along similar lines, olaparib was effective in BRCA-deficient patients with breast or ovarian cancers, with an overall response rate (ORR) of 41% and 22% in breast cancers211 and 33% and 13% in ovarian cancer213 in cohorts assigned to 400 mg and 100 mg twice daily, respectively. A similar antineoplastic effect of olaparib monotherapy was observed in 2 phase II studies performed in BRCA1- or BRCA2-mutated ovarian cancer patients.212,214 Nevertheless, in the former study no statistically significant differences in progression-free survival (PFS) were found between olaparib versus pegylated liposomal doxorubicin,212 and in the latter no objective responses were reported in subjects with breast cancer.214 Of note, in one of these trials, olaparib was also effective in subjects with non-mutated BRCA1 or BRCA2 (ORR 24%),214 whereas no significant responses or durable disease control were observed in a phase II study aimed at assessing the therapeutic potential of olaparib in individuals with metastatic Ewing sarcoma.216 Finally, in an open-label phase II clinical trial veliparib was active in women with BRCA-mutationed recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer with an ORR of 26% (20% and 35% for platinum-resistant and platinum-sensitive patients, respectively)218 (Table 1).
When given as maintenance treatment, olaparib significantly improved PFS among patients who had received 2 or more platinum-based regimens and who had a partial or complete response to their most recent platinum-based regimen (median PFS of 8.4 mo compared with 4.8 mo for placebo conditions).215 Moreover, when considering BRCA mutation status, median PFS (but not overall survival) was significantly different in the olaparib versus placebo groups (11.2 mo vs. 4.3 mo)219 (Table 1).
Driven by this evidence, the FDA recently granted accelerated approval to the PARP inhibitor olaparib for patients with advanced ovarian cancer who carry inherited mutations of BRCA and have previously received at least 3 lines of therapy (http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm427598.htm) and breakthrough therapy designation to the PARP inhibitor rucaparib as monotherapy for patients with advanced ovarian cancer who have received at least 2 lines of prior platinum-containing therapy, with BRCA-mutated tumors, inclusive of both germline and somatic BRCA mutations.
PARP inhibitors as radio- or chemosensitizing agents
In contrast to olaparib monotherapy, dose-limiting toxicities, including neutropenia and thrombocytopenia, were reported for combinations of olaparib with various therapeutic agents, including dacarbazine (an alkylating agent approved by the FDA for the treatment of patients with Hodgkin lymphoma and melanoma220);221 cisplatin and gemcitabine (a platinum derivative compound and an antimetabolite nucleoside, respectively, that are currently used for the treatment of several solid neoplasms222-227);228 and topotecan (a topoisomerase I inhibitor approved by the FDA for use alone or with other drugs to treat several type of solid neoplasms, including cervical, ovarian, and small cell lung cancer229).230 Moreover, olaparib in combination with cisplatin and gemcitabine was associated with myelosuppression, even at relatively low doses.228 In line with this observation, significant and/or dose limiting myelosuppression was observed for veliparib combined with topotecan with or without filgrastim/pegfilgrastim neutrophil support231,232 as well as for veliparib,233 INO-1001,234 or rucaparib237 administered together with temozolomide (an alkylating agent currently used to treat patients with brain tumors, including anaplastic astrocytoma and glioblastoma multiforme190,235,236). It is important to note that the use of an intermittent schedule improved the tolerability of olaparib plus cisplatin regimen,238 but not that of olaparib plus carboplatin (a platinum derivative approved by the FDA for the treatment of solid tumors, including ovarian and non-small cell lung cancer239-241) regimen.242 Contrasting data have been reported on the tolerability of olaparib and agents blocking vascular endothelial growth factor (VEGF) signaling, including bevacizumab (a VEGF-targeting recombinant humanized monoclonal antibody currently approved for the treatment of several solid tumors243-245) and cediranib (a tyrosine kinase inhibitor of vascular endothelial growth factor receptor [VEGFR-1, -2, -3] that exhibits preclinical and clinical antitumoral activity246-248).249-251 Finally, the combinations of olaparib and paclitaxel (a microtubular poison of the taxane family approved by the FDA for use alone or with other drugs for the treatment of breast, non-small cell lung, and ovarian cancer252-254),255 paclitaxel and carboplatin followed by olaparib maintenance monotherapy,256 veliparib and low-dose cyclophosphamide (an alkylating agent with immunogenic properties currently approved for the treatment of multiple neoplasms257-259),260,261 veliparib and low-dose fractionated whole abdominal radiation,262 veliparib and whole-brain radiation therapy,263 CEP-9722 and temozolomide,264 and rucaparib and temozolomide265 were all well tolerated and/or had a generally manageable and acceptable toxicity profile.
Regarding the efficacy of PARP inhibitor-based regimens, the results reported so far are contrasting (Table 1).
On the one hand, limited or no objective response was observed when (1) olaparib was administered together with dacarbazine221 or with cisplatin plus gemcitabine228 in patients with advanced solid tumors; (2) rucaparib was used together with temozolomide in individuals with melanoma (response rate of 17.4%, median time to progression 3.5 mo, median overall survival 9.9 mo);237 (3) veliparib was combined with temozolomide in children with recurrent central nervous system tumors233 or patients with metastatic castration-resistant prostate cancer;260 (4) veliparib was employed in combination with low-dose fractionated whole abdominal radiation;262 (5) veliparib was co-administered with topotecan and filgrastim/pegfilgrastim neutrophil support in women with persistent or recurrent uterine cervix cancer;232 (6) CEP-9722 was given with temozolomide to patients with advanced solid tumors;264 and (7) INO-1001 was combined with temozolomide in subjects with melanoma.234 Also, the addition of veliparib to a low-dose cyclophosphamide regimen did not improve the response rate or the median PFS in patients with pretreated primary peritoneal, fallopian tube, or high-grade serous ovarian cancers266 (Table 1).
On the other hand, promising evidence of antineoplastic activity was reported for (1) olaparib combined with cisplatin in breast or ovarian cancer patients with germline BRCA1/2 mutations (ORR of 43% and 71%, respectively);238 (2) olaparib in combination with paclitaxel for first- or second-line treatment of patients with metastatic TNBC (37% of patients with a confirmed partial response);255 (3) olaparib together with cediranib in ovarian (but not breast) cancer patients (ORR of 44%; median PFS of 17.7 and 9.0 mo for the women treated with cediranib plus olaparib and those administered with olaparib alone, respectively);250,251 (4) olaparib combined with carboplatin in BRCA1- or BRCA2-mutated breast or ovarian cancer (~50% of patients with complete or partial responses);242 (5) olaparib together with paclitaxel and carboplatin followed by olaparib maintenance monotherapy in patients with platinum-sensitive, recurrent, high-grade serous ovarian cancer (PFS 12.2 mo vs 9.6 mo for olaparib plus chemotherapy group versus chemotherapy alone group, with the greatest clinical benefit reported for patients with BRCA mutations);256 (6) veliparib in combination with cyclophosphamide in a subset of BRCA-mutated patients (7 patients with partial responses and 6 patients with disease stabilization among 35 patients enrolled in the study);261 and (7) veliparib combined with whole-brain radiation therapy (median survival time of 10 mo and 7.7 mo for the non-small cell lung cancer and the breast cancer subgroups, respectively, compared to nomogram model-predicted values of 3.5 mo and 4.9 mo)263 (Table 1). These latter results (and in particular the impressive efficacy of the combination of olaparib and cediranib in patients with BRCA-mutated and BRCA-WT ovarian tumors) support the further development of PARP-based regimens.
Ongoing clinical trials
At the time of writing this Trial Watch (March 2015), official sources listed 84 ongoing (not terminated, withdrawn, suspended, or completed) clinical trials launched after January 1 2012 with the aim of testing the safety and therapeutic potential of PARP inhibitors in cancer patients as single agents, in maintenance monotherapy, or in combination with other chemotherapeutic agents (source: http://www.clinicaltrials.gov/) (Table 2). Of these trials, 35 involve olaparib, 25 veliparib, 13 talazoparib, 5 niraparib, 3 rucaparib, 1 BGB-290, and 1 E7016. As mentioned above, clinical trials employing iniparib will not be discussed in this section.
Table 2.
Agent | Indication(s) | Phase | Status | Notes | Ref. |
---|---|---|---|---|---|
BGB-290 | Advanced solid tumors | I | Recruiting | As single agent | NCT02361723 |
E7016 | Melanoma | II | Active, not recruiting | Combined with temozolomide | NCT01605162 |
Iniparib | Advanced solid tumors | III | Active, not recruiting | Alone or combined with others chemotherapeutics, including carboplatin, doxorubicin, gemcitabine, irinotecan, paclitaxel, and topotecan | NCT01593228 |
Niraparib | Breast cancer | III | Recruiting | As single agent vs. 4 standard of care metastatic breast cancer chemotherapies (HER2-) | NCT01905592 |
Ewing sarcoma | I | Recruiting | Combined with temozolomide | NCT02044120 | |
Ovarian cancer | I/II | Not yet recruiting | Alone or combined with bevacizumab | NCT02354131 | |
II | Not yet recruiting | As single agent | NCT02354586 | ||
III | Recruiting | As single agent | NCT01847274 | ||
Olaparib | Advanced solid tumors | I | Recruiting | As single agent | NCT01894243 |
Recruiting | As single agent | NCT01894256 | |||
Recruiting | Combined with anastrozole, letrozole, or tamoxifen | NCT02093351 | |||
Recruiting | Combined with AZD5363 | NCT02338622 | |||
Active, not recruiting | As single agent | NCT01921140 | |||
Active, not recruiting | As single agent | NCT01851265 | |||
Active, not recruiting | Combined with itraconazole | NCT01900028 | |||
Active, not recruiting | Combined with rifampicin | NCT01929603 | |||
I/II | Recruiting | Combined with AZD6738 | NCT02264678 | ||
Breast cancer | I | Recruiting | Combined with RT | NCT02227082 | |
III | Recruiting | As single agent | NCT02032823 | ||
Recruiting | As single agent vs. capecitabine, eribulin or vinorelbine | NCT02000622 | |||
Breast or female reproductive system cancer | I/II | Recruiting | Combined with AZD2014 and AZD5363 | NCT02208375 | |
Breast or ovarian cancer | I | Recruiting | Combined with BKM120 | NCT01623349 | |
Ewing sarcoma | I | Recruiting | Combined with temozolomide | NCT01858168 | |
II | Active, not recruiting | As single agent | NCT01583543 | ||
Gastric cancer | III | Recruiting | Combined with paclitaxel | NCT01924533 | |
Head and neck cancer | I | Not yet recruiting | Combined with cisplatin and intensity modulated RT | NCT02308072 | |
Recruiting | Combined with cetuximab and RT | NCT01758731 | |||
Recruiting | Combined with RT | NCT02229656 | |||
Non-small cell lung cancer | I | Recruiting | Combined with cisplatin and RT | NCT01562210 | |
II | Recruiting | As single agent | NCT01788332 | ||
Esophageal cancer | I | Recruiting | Combined with RT | NCT01460888 | |
Ovarian cancer | II | Not yet recruiting | Combined with cediranib | NCT02340611 | |
III | Not yet recruiting | As single agent | NCT02392676 | ||
Recruiting | As single agent | NCT01844986 | |||
Recruiting | As single agent | NCT02282020 | |||
Active, not recruiting | As single agent | NCT01874353 | |||
Ovarian, fallopian tube or peritoneal cancer | I | Recruiting | Combined with bevacizumab, cisplatin, and paclitaxel | NCT02121990 | |
II | Not yet recruiting | Combined with cediranib | NCT02345265 | ||
Ovarian or uterine cancer | I/II | Active, not recruiting | Combined with carboplatin and paclitaxel | NCT01650376 | |
Pancreatic cancer | III | Recruiting | As single agent | NCT02184195 | |
Prostate cancer | I | Not yet recruiting | As single agent or combined with degarelix | NCT02324998 | |
II | Recruiting | As single agent | NCT01682772 | ||
Recruiting | Combined with abiraterone and prednisone/prednisolone | NCT01972217 | |||
Rucaparib | Ovarian, fallopian tube, or peritoneal cancer | II | Recruiting | As single agent | NCT01891344 |
III | Recruiting | As single agent | NCT01968213 | ||
Pancreatic cancer | II | Recruiting | As single agent | NCT02042378 | |
Talazoparib | Advanced solid tumors | I | Not yet recruiting | Combined with carboplatin and paclitaxel | NCT02317874 |
Not yet recruiting | Combined with carboplatin and paclitaxel | NCT02358200 | |||
Recruiting | Combined with temozolomide or irinotecan | NCT02049593 | |||
I/II | Recruiting | As single agent | NCT01989546 | ||
Recruiting | Combined with temozolomide | NCT02116777 | |||
II | Recruiting | As single agent | NCT02286687 | ||
Breast cancer | II | Not yet recruiting | As single agent | NCT02282345 | |
Recruiting | As single agent | NCT02034916 | |||
III | Recruiting | As single agent vs. capecitabine, eribulin, gemcitabine, or vinorelbine | NCT01945775 270 | ||
Childhood solid tumors | I | Not yet recruiting | Combined with irinotecan and temozolomide | NCT02392793 | |
Endometrial cancer | II | Not yet recruiting | As single agent | NCT02127151 | |
Ovarian cancer | II | Recruiting | As single agent | NCT02326844 | |
Ovarian, fallopian tube or peritoneal cancer | 0 | Not yet recruiting | As single agent | NCT02316834 | |
Veliparib | Advanced solid tumors | I | Active, not recruiting | As single agent | NCT02210663 |
Active, not recruiting | As single agent or combined with carboplatin and paclitaxel or with FOLFIRI | NCT02033551 | |||
Breast cancer | I | Recruiting | Combined with RT | NCT01618357 | |
Active, not recruiting | Combined with RT | NCT01477489 | |||
II | Recruiting | Combined with carboplatin, paclitaxel, and temozolomide | NCT01506609 | ||
III | Recruiting | Combined with carboplatin, cyclophosphamide, doxorubicin, and paclitaxel | NCT02032277 | ||
Recruiting | Combined with carboplatin and paclitaxel | NCT02163694 | |||
Not provided | Recruiting | Combined with lapatinib | NCT02158507 | ||
Breast cancer or ovarian cancer | I | Active, not recruiting | As single agent | NCT01853306 | |
Colorectal cancer | II | Recruiting | Combined with bevacizumab and FOLFIRI | NCT02305758 | |
Glioblastoma multiforme | II/III | Recruiting | Combined with temozolomide | NCT02152982 | |
Head and neck cancer | I/II | Recruiting | Combined with carboplatin, cisplatin, hydroxyurea, fluorouracil, paclitaxel, and RT | NCT01711541 | |
Hematologic neoplasms | I | Active, not recruiting | Combined with carboplatin and topotecan | NCT00588991 | |
Lung cancer | I | Recruiting | Combined with carboplatin and etoposide | NCT02289690 | |
I/II | Recruiting | Combined with cisplatin and etoposide | NCT01642251 | ||
II | Recruiting | Combined with temozolomide | NCT01638546 | ||
III | Recruiting | Combined with carboplatin, cisplatin, paclitaxel and pemetrexed | NCT02264990 | ||
Recruiting | Combined with carboplatin and paclitaxel | NCT02106546 | |||
Ovarian cancer | I/II | Recruiting | Combined with topotecan | NCT01690598 | |
Ovarian, fallopian tube, or peritoneal cancer | I | Recruiting | Combined with floxuridine | NCT01749397 | |
II | Active, not recruiting | As single agent | NCT01540565 | ||
Pancreatic cancer | I | Recruiting | Combined with gemcitabine and RT | NCT01908478 | |
I/II | Recruiting | Combined with fluorouracil, leucovorin, and oxaliplatin | NCT01489865 | ||
II | Recruiting | Combined with cisplatin and gemcitabine | NCT01585805 | ||
Prostate cancer | II | Recruiting | Combined with abiraterone and prednisone | NCT01576172 |
Not terminated, suspended, withdrawn, unknown or completed on the date of submission (March 20, 2015). Sources: http://www.clinicaltrials.gov, searching with the following terms: “PARP AND cancer” or “PARP1 AND cancer” or “NAME OF THE COMPOUND (e.g., olaparib) AND cancer”
The clinical profile of olaparib as a monotherapeutic agent (in some cases following conventional [mostly platinum-based] chemotherapy regimens) is being assessed (1) in 7 phase III trials performed in patients with BRCA1/2 (or other HR player)-mutated breast cancer (NCT02000622 and NCT02032823; the second is an adjuvant study), ovarian cancer (NCT01844986, NCT01874353, NCT02282020, and NCT02392676), or pancreatic cancer (NCT02184195); (2) in 3 phase II trials conducted in individuals with recurrent/metastatic Ewing sarcoma (NCT01583543), advanced non-small cell lung cancer (NCT01788332), or advanced castration-resistant prostate cancer (NCT01682772); and (3) in 5 phase I studies performed in patients with advanced solid tumors (NCT01851265, NCT01894243, NCT01894256, NCT01921140, and NCT02324998) (Table 2). The clinical study NCT01661868 (evaluating the safety and efficacy of olaparib in patients with recurrent BRCA-deficient ovarian cancer) has been withdrawn prior to enrollment due to lack of availability of the drug, whereas results of NCT01583543, which is currently listed as ongoing, but not recruiting participants, are reported in reference 216.216
In addition to olaparib, 3 other PARP inhibitors are being employed in phase III interventional studies as single agents or maintenance monotherapy: niraparib, whose efficacy is being investigated in 2 studies performed in patients with platinum-sensitive ovarian cancer (NCT01847274) or in individuals with human epidermal growth factor 2 (HER2)-negative, BRCA-mutated breast cancer (NCT01905592); rucaparib, whose therapeutic profile is being evaluated in 1 trial conducted in subjects with ovarian, fallopian tube, or peritoneal cancer who have received platinum-based chemotherapy (NCT01968213); and talazoparib, whose safety and efficacy is being compared to treatment of the physician's choice (e.g., capecitabine [an antimetabolite currently used for the treatment of human neoplasms, including breast and colorectal cancers267], the nucleoside analog gemcitabine, or eribulin or vinorelbine [2 antimitotic agents, the first of which was approved by the FDA for use alone or with cisplatin to treat non-small cell lung cancer patients268,269]) in subjects with BRCA-mutated advanced breast cancer (EMBRACA) (NCT01945775)270 (Table 2).
Among the other PARP inhibitors given as stand-alone therapeutic regimens in phase I and/or II clinical trials, (1) BGB-290 is being used in patients with advanced solid tumors (NCT02361723); (2) niraparib is being administered to individuals with ovarian cancer who have received at least 3 previous chemotherapy regimens (NCT02354586); (3) rucaparib is being used in patients with platinum-sensitive ovarian, fallopian tube, or peritoneal cancer (NCT01891344) or BRCA-mutated pancreatic cancer (NCT02042378); (4) talazoparib is being used in subjects with advanced solid tumors (NCT01989546 and NCT02286687), BRCA-mutated breast cancer (NCT02034916 and NCT02282345; the latter is a neoadjuvant study), endometrial cancer (NCT02127151), or BRCA1/2 mutation-associated ovarian cancer (NCT02326844); and (5) veliparib is being used in patients with advanced solid tumors (NCT02210663), ovarian, fallopian tube, or peritoneal cancer (NCT01540565), or solid neoplasms, including high-grade serous ovarian cancer and BRCA-mutated breast cancer (NCT01853306) (Table 2). In addition, a pilot study is evaluating the effects of talazoparib on patients with ovarian, fallopian tube, or peritoneal cancer (NCT02316834). To the best of our knowledge, the results of the clinical trial NCT02009631 (evaluating the clinical profile of veliparib in advanced solid tumors) have not yet been released. (http://www.clinicaltrials.gov).
According to official sources (http://www.clinicaltrials.gov), 52 ongoing clinical trials have been launched worldwide to evaluate the safety and tolerability of PARP inhibitors in combination with conventional radio- or chemotherapy (Table 2).
PARP inhibitors employed in Phase III interventional studies include (1) olaparib, which is being combined with paclitaxel in individuals with gastric cancer who have progressed following first-line therapy (NCT01924533), and (2) veliparib, which is being combined with carboplatin and paclitaxel or paclitaxel-based regimens in patients with breast (NCT02163694 and NCT02032277) or lung (NCT02264990 and NCT02106546) cancer. In addition, veliparib is being combined with temozolomide in a phase II/III study to treat patients with newly diagnosed glioblastoma multiforme with O-6-methylguanine-DNA methyltransferase (MGMT) promoter hypermethylation (NCT02152982) (Table 2).
The vast majority of phase II or I studies assessing the therapeutic profile of PARP inhibitors in combinatorial regimens involves olaparib or veliparib (Table 2).
Olaparib is being administered together with (1) anastrozole or letrozole (2 aromatase inhibitors currently used for the treatment of breast cancer in postmenopausal women271-273) or tamoxifen (a nonsteroidal selective estrogen receptor modulator approved by the FDA for the treatment and prevention of breast cancer273,274) in patients with advanced solid tumors (NCT02093351); (2) itraconazole (an FDA-approved antifungal compound with antineoplastic activity275-277) in individuals with advanced solid neoplasms (NCT01900028); (3) rifampicin (a cytochrome P450, family 3, subfamily A, polypeptide 4, CYP3A4, inducer278) in subjects with solid tumors (NCT01929603); (4) radiotherapy or radiotherapy-based regimens in individuals with breast cancer (NCT02227082), in subjects with head and neck cancer (NCT02229656, NCT01758731 and NCT02308072), in patients with non-small cell lung cancer (NCT01562210), or in individuals with esophageal cancer (NCT01460888); (5) AZD2014 or AZD5363 (inhibitors of mammalian target of rapamycin complex 1/2 [mTORC1/2] or AKT, respectively, with antitumor activity279-282) in subjects with breast cancer or female reproductive system cancer (NCT02208375) or advanced solid tumors (NCT02338622); (6) BKM120 (a PI3K inhibitor283 whose therapeutic potential is being evaluated in clinical studies) in breast or ovarian cancer patients (NCT01623349); (7) bevacizumab, cisplatin, and paclitaxel in individuals with ovarian, fallopian tube, or peritoneal cancer (NCT02121990); (8) carboplatin and paclitaxel in subjects with ovarian or uterine cancer (NCT01650376); (9) temozolomide in patients with Ewing sarcoma (NCT01858168); (10) abiraterone (an FDA-approved androgen biosynthesis inhibitor284) and prednisone/prednisolone (2 FDA-approved glucocorticosteroids285,286) in individuals with prostate cancer (NCT01972217); (11) degarelix (a gonadotropin-releasing hormone antagonistic approved by the FDA for the treatment of advanced prostate cancers287,288) in patients with prostate cancer prior to radical prostatectomy (NCT02324998); (12) cediranib in patients with ovarian, fallopian tube, peritoneal (NCT02345265) or ovarian (NCT02340611) cancer; and (13) AZD6738 (an ATR serine/threonine kinase inhibitor whose activity is being evaluated in some clinical trials http://www.clinicaltrials.gov) in subjects with advanced solid malignancies (NCT02264678) (Table 2).
Veliparib is being (1) combined with carboplatin and paclitaxel-based regimens in patients with head and neck cancer (NCT01711541); (2) used in combination with carboplatin and paclitaxel or either FOLFIRI (irinotecan, leucovorin, 5-flourouracil, and irinotecan) in advanced solid tumors (NCT02033551) or temozolomide in breast cancer (NCT01506609); (3) administered together with radiotherapy in individuals with breast cancer (NCT01477489 and NCT01618357); (4) given in combination with radiotherapy and gemcitabine in pancreatic cancer patients (NCT01908478); (5) combined with temozolomide in patients with lung cancer (NCT01638546); (6) used together with floxuridine (an antimetabolite that is metabolized to fluorouracil and other metabolites with antineoplastic activity289) in females with ovarian, fallopian tube, or peritoneal cancer (NCT01749397); (7) used in combination with topotecan and/or carboplatin in individuals with ovarian cancer (NCT01690598) and hematologic neoplasms (NCT00588991); (8) given together with 5-fluorouracil (an antimetabolite fluoropyrimidine commonly used for the adjuvant and palliative treatment of several neoplasms290,291), leucovorin (an active metabolite of folic acid approved by the FDA in combination with fluorouracil as palliative treatment in patients with advanced colorectal cancer292,293), and oxaliplatin (an FDA-approved cisplatin derivative294,295) in pancreatic cancer patients (NCT01489865); (9) combined with bevacizumab and FOLFIRI in subjects with colorectal cancer (NCT02305758); (10) used in combination with platinum-based agents and either etoposide (an FDA-approved inhibitor of topoisomerase II currently used for the treatment of patients affected by a variety of solid malignancies296,297) or gemcitabine in individuals with lung (NCT01642251 and NCT02289690) or pancreatic cancer (NCT01585805), respectively; and (11) administered together with abiraterone and prednisone in patients with prostate cancer (NCT01576172) (Table 2). In addition, an open-label pilot study is evaluating the effectiveness and safety of veliparib together with lapatinib (a tyrosine kinase inhibitor currently used in combination with capecitabine or letrozole to treat advanced or metastasized breast cancer298-301) in patients with metastatic, TNBCs (NCT02158507). To the best of our knowledge, the results of clinical trials NCT01589419 (evaluating the safety and efficacy of veliparib in combination with radiotherapy and capecitabine in colorectal cancer), NCT01657799 (determining the activity, safety, and tolerability of veliparib combined with whole-brain radiation therapy for brain metastases from non-small cell lung cancer), and NCT01560104 and NCT01617928 (both assessing the clinical profile of veliparib in combination with carboplatin and paclitaxel in advanced solid tumors) have not yet been released (http://www.clinicaltrials.gov). The recruitment status of the clinical trial NCT01495351 is unknown, while NCT01818063 has suspended participant recruitment due to lack of funding.
Regarding the other PARP inhibitors, niraparib and E7016 are being combined with temozolomide in subjects with Ewing sarcoma (NCT02044120) or melanoma (NCT01605162), respectively. Niraparib is also being administered alone or combined with bevacizumab in patients with ovarian cancer (NCT02354131). Finally, talazoparib is being used together with (1) carboplatin and paclitaxel in individuals with advanced solid tumors (NCT02317874) or in subjects with advanced BRCA-mutated solid tumors or metastatic TNBCs (NCT02358200); and (2) temozolomide and/or irinotecan in individuals with advanced solid tumors (NCT02049593 and NCT02116777) or in patients with childhood solid tumors (NCT02392793) (Table 2).
Concluding remarks
A large number of preclinical and clinical studies have demonstrated the safety and therapeutic benefits of PARP inhibitors alone or in combination with other cytotoxic drugs (mostly DNA damaging agents), in particular against breast and ovarian cancers harboring mutations in components of DNA repair pathways.
Nonetheless, the role of PARP1 in the preservation of genomic stability, the non-selective inactivation of PARP family members other than PARP1 ascribed to PARP inhibitors, the toxicity and limited efficacy of some (but not all) combinatorial regimens, and the development of resistance to synthetic lethal strategies based on PARP inhibitors may cast doubt on the use of these agents for cancer therapy. It is important to note that the mechanisms of resistance to PARP inhibitors acquired by HR-deficient tumors include novel deletions or secondary mutations that restore the open reading frame (ORF), sequence and/or function of BRCA2;302-304 somatic mutations of tumor protein p53 binding protein 1 (TP53BP1 best known as 53BP1) or loss of MAD2 mitotic arrest deficient-like 2 (MAD2L2, best known as REV7), which are believed to restore HR proficiency;194,305-307 and upregulation of ATP-binding cassette, sub-family B (MDR/TAP), member 1 (ABCB1, also known as multidrug resistance protein 1, MDR1, or P-glycoprotein, P-GP), which increases drug efflux.308 Moreover, besides being involved in DDR, PARP enzymes regulate many functions relevant for cancer, including DNA transcription,309 energy metabolism,11 metabolic checkpoints (i.e., molecular mechanisms triggered in response to metabolic fluctuations),310 cell death,61-64 mitosis,311-313 ploidy status,314 cell migration,315-317 angiogenesis,318-320 and metastasis.321 To add further layers of complexity, PARP enzymes have been linked to signaling pathways, including extracellular signal-regulated kinase (ERK), nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-kB), and wingless-type (Wnt)/β-catenin pathways.322-325 PARP activity is also involved in cancer stem cell (CSC) maintenance,326 whereas MARylation has been shown to regulate processes relevant to cancer, including the unfolded protein response (UPR).6,327 Finally, recent evidence suggests a cytoprotective role of PARP1 and PAR in response to intrinsic or extrinsic perturbations.6,14,190,328
The recent approval of olaparib by the FDA as a maintenance therapy for patients with platinum-sensitive high-grade serous ovarian cancer and promising clinical evidence showing the efficacy of PARP inhibitors in the adjuvant setting75 are both paving the way for the advancement of PARP1 inhibitors in clinics. Nevertheless, the results of large phase III trials are awaited in order to uncover the true potential of these agents for cancer therapy. Moreover, a better understanding of the biology of the PARP enzyme family and the identification of potential predictive biomarkers for PARP inhibitor-based therapies are urgently needed in order to increase the specificity and efficacy of these agents and expand the patient population that might benefit from these targeting strategies.
Disclosure of potential conflicts of interest
No potential conflicts of interest were disclosed.
Acknowledgments
We thank Tania Merlino for technical assistance.
Funding
The authors are supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC: MFAG 2013 #14641 and Triennial Fellowship “Antonietta Latronico," 2014), Ministero Italiano della Salute (RF_GR-2011–02351355), the Programma per i Giovani Ricercatori “Rita Levi Montalcini” 2011 and Ligue Nationale contre le Cancer.
References
- 1.Gibson BA, Kraus WL. New insights into the molecular and cellular functions of poly(ADP-ribose) and PARPs. Nat Rev Mol Cell Biol 2012; 13:411-24; PMID:22713970; http://dx.doi.org/ 10.1038/nrm3376 [DOI] [PubMed] [Google Scholar]
- 2.Schreiber V, Dantzer F, Ame JC, de Murcia G. Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol 2006; 7:517-28; PMID:16829982; http://dx.doi.org/ 10.1038/nrm1963 [DOI] [PubMed] [Google Scholar]
- 3.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; PMID:10455009; http://dx.doi.org/ 10.1042/0264-6021:3420249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Durkacz BW, Omidiji O, Gray DA, Shall S. (ADP-ribose)n participates in DNA excision repair. Nature 1980; 283:593-6; PMID:6243744; http://dx.doi.org/ 10.1038/283593a0 [DOI] [PubMed] [Google Scholar]
- 5.Burkle A. Poly(ADP-ribose). The most elaborate metabolite of NAD+. FEBS J 2005; 272:4576-89; PMID:16156780; http://dx.doi.org/ 10.1111/j.1742-4658.2005.04864.x [DOI] [PubMed] [Google Scholar]
- 6.Vyas S, Chang P. New PARP targets for cancer therapy. Nat Rev Cancer 2014; 14:502-9; PMID:24898058; http://dx.doi.org/ 10.1038/nrc3748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hottiger MO, Hassa PO, Luscher B, Schuler H, Koch-Nolte F. Toward a unified nomenclature for mammalian ADP-ribosyltransferases. Trends Biochem Sci 2010; 35:208-19; PMID:20106667; http://dx.doi.org/ 10.1016/j.tibs.2009.12.003 [DOI] [PubMed] [Google Scholar]
- 8.Li M, Yu X. The role of poly(ADP-ribosyl)ation in DNA damage response and cancer chemotherapy. Oncogene 2015; 34(26):3349-56; PMID:25220415; http://dx.doi.org/ 10.1038/onc.2014.295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Masutani M, Fujimori H. Poly(ADP-ribosyl)ation in carcinogenesis. Mol Aspects Med 2013; 34:1202-16; PMID:23714734; http://dx.doi.org/ 10.1016/j.mam.2013.05.003 [DOI] [PubMed] [Google Scholar]
- 10.Burkle A, Virag L. Poly(ADP-ribose): PARadigms and PARadoxes. Mol Aspects Med 2013; 34:1046-65; PMID:23290998; http://dx.doi.org/ 10.1016/j.mam.2012.12.010 [DOI] [PubMed] [Google Scholar]
- 11.Bai P, Canto C. The role of PARP-1 and PARP-2 enzymes in metabolic regulation and disease. Cell Metab 2012; 16:290-5; PMID:22921416; http://dx.doi.org/ 10.1016/j.cmet.2012.06.016 [DOI] [PubMed] [Google Scholar]
- 12.Davar D, Beumer JH, Hamieh L, Tawbi H. Role of PARP inhibitors in cancer biology and therapy. Curr Med Chem 2012; 19:3907-21; PMID:22788767; http://dx.doi.org/ 10.2174/092986712802002464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Leung AK. Poly(ADP-ribose): an organizer of cellular architecture. J Cell Biol 2014; 205:613-9; PMID:24914234; http://dx.doi.org/ 10.1083/jcb.201402114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Luo X, Kraus WL. On PAR with PARP: cellular stress signaling through poly(ADP-ribose) and PARP-1. Genes Dev 2012; 26:417-32; PMID:22391446; http://dx.doi.org/ 10.1101/gad.183509.111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ame JC, Spenlehauer C, de Murcia G. The PARP superfamily. Bioessays 2004; 26:882-93; PMID:15273990; http://dx.doi.org/ 10.1002/bies.20085 [DOI] [PubMed] [Google Scholar]
- 16.Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. PARP inhibition: PARP1 and beyond. Nat Rev Cancer 2010; 10:293-301; PMID:20200537; http://dx.doi.org/ 10.1038/nrc2812 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ciccia A, Elledge SJ. The DNA damage response: making it safe to play with knives. Mol Cell 2010; 40:179-204; PMID:20965415; http://dx.doi.org/ 10.1016/j.molcel.2010.09.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144:646-74; PMID:21376230; http://dx.doi.org/ 10.1016/j.cell.2011.02.013 [DOI] [PubMed] [Google Scholar]
- 19.Giglia-Mari G, Zotter A, Vermeulen W. DNA damage response. Cold Spring Harb Perspect Biol 2011; 3:a000745; PMID:20980439; http://dx.doi.org/ 10.1101/cshperspect.a000745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature 2009; 461:1071-8; PMID:19847258; http://dx.doi.org/ 10.1038/nature08467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Harper JW, Elledge SJ. The DNA damage response: ten years after. Mol Cell 2007; 28:739-45; PMID:18082599; http://dx.doi.org/ 10.1016/j.molcel.2007.11.015 [DOI] [PubMed] [Google Scholar]
- 22.Beck C, Robert I, Reina-San-Martin B, Schreiber V, Dantzer F. Poly(ADP-ribose) polymerases in double-strand break repair: focus on PARP1, PARP2 and PARP3. Exp Cell Res 2014; 329:18-25; PMID:25017100; http://dx.doi.org/ 10.1016/j.yexcr.2014.07.003 [DOI] [PubMed] [Google Scholar]
- 23.Papeo G, Forte B, Orsini P, Perrera C, Posteri H, Scolaro A, Montagnoli A. Poly(ADP-ribose) polymerase inhibition in cancer therapy: are we close to maturity? Expert Opin Ther Pat 2009; 19:1377-400; PMID:19743897; http://dx.doi.org/ 10.1517/13543770903215883 [DOI] [PubMed] [Google Scholar]
- 24.Ali AA, Timinszky G, Arribas-Bosacoma R, Kozlowski M, Hassa PO, Hassler M, Ladurner AG, Pearl LH, Oliver AW. The zinc-finger domains of PARP1 cooperate to recognize DNA strand breaks. Nat Struct Mol Biol 2012; 19:685-92; PMID:22683995; http://dx.doi.org/ 10.1038/nsmb.2335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Okano S, Lan L, Caldecott KW, Mori T, Yasui A. Spatial and temporal cellular responses to single-strand breaks in human cells. Mol Cell Biol 2003; 23:3974-81; PMID:12748298; http://dx.doi.org/ 10.1128/MCB.23.11.3974-3981.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.De Vos M, Schreiber V, Dantzer F. The diverse roles and clinical relevance of PARPs in DNA damage repair: current state of the art. Biochem Pharmacol 2012; 84:137-46; PMID:22469522; http://dx.doi.org/ 10.1016/j.bcp.2012.03.018 [DOI] [PubMed] [Google Scholar]
- 27.El-Khamisy SF, Masutani M, Suzuki H, Caldecott KW. A requirement for PARP-1 for the assembly or stability of XRCC1 nuclear foci at sites of oxidative DNA damage. Nucleic Acids Res 2003; 31:5526-33; PMID:14500814; http://dx.doi.org/ 10.1093/nar/gkg761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Masson M, Niedergang C, Schreiber V, Muller S, Menissier-de Murcia J, de Murcia G. XRCC1 is specifically associated with poly(ADP-ribose) polymerase and negatively regulates its activity following DNA damage. Mol Cell Biol 1998; 18:3563-71; PMID:9584196 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Li M, Lu LY, Yang CY, Wang S, Yu X. The FHA and BRCT domains recognize ADP-ribosylation during DNA damage response. Genes Dev 2013; 27:1752-68; PMID:23964092; http://dx.doi.org/ 10.1101/gad.226357.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Pleschke JM, Kleczkowska HE, Strohm M, Althaus FR. Poly(ADP-ribose) binds to specific domains in DNA damage checkpoint proteins. J Biol Chem 2000; 275:40974-80; PMID:11016934; http://dx.doi.org/ 10.1074/jbc.M006520200 [DOI] [PubMed] [Google Scholar]
- 31.Fisher AE, Hochegger H, Takeda S, Caldecott KW. Poly(ADP-ribose) polymerase 1 accelerates single-strand break repair in concert with poly(ADP-ribose) glycohydrolase. Mol Cell Biol 2007; 27:5597-605; PMID:17548475; http://dx.doi.org/ 10.1128/MCB.02248-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Robu M, Shah RG, Petitclerc N, Brind'Amour J, Kandan-Kulangara F, Shah GM. Role of poly(ADP-ribose) polymerase-1 in the removal of UV-induced DNA lesions by nucleotide excision repair. Proc Natl Acad Sci U S A 2013; 110:1658-63; PMID:23319653; http://dx.doi.org/ 10.1073/pnas.1209507110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Pines A, Vrouwe MG, Marteijn JA, Typas D, Luijsterburg MS, Cansoy M, Hensbergen P, Deelder A, de Groot A, Matsumoto S, et al.. PARP1 promotes nucleotide excision repair through DDB2 stabilization and recruitment of ALC1. J Cell Biol 2012; 199:235-49; PMID:23045548; http://dx.doi.org/ 10.1083/jcb.201112132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Audebert M, Salles B, Calsou P. Effect of double-strand break DNA sequence on the PARP-1 NHEJ pathway. Biochem Biophys Res Commun 2008; 369:982-8; PMID:18054777; http://dx.doi.org/ 10.1016/j.bbrc.2007.11.132 [DOI] [PubMed] [Google Scholar]
- 35.Wang M, Wu W, Rosidi B, Zhang L, Wang H, Iliakis G. PARP-1 and Ku compete for repair of DNA double strand breaks by distinct NHEJ pathways. Nucleic Acids Res 2006; 34:6170-82; PMID:17088286; http://dx.doi.org/ 10.1093/nar/gkl840 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bryant HE, Petermann E, Schultz N, Jemth AS, Loseva O, Issaeva N, Johansson F, Fernandez S, McGlynn P, Helleday T. PARP is activated at stalled forks to mediate Mre11-dependent replication restart and recombination. EMBO J 2009; 28:2601-15; PMID:19629035; http://dx.doi.org/ 10.1038/emboj.2009.206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sugimura K, Takebayashi S, Taguchi H, Takeda S, Okumura K. PARP-1 ensures regulation of replication fork progression by homologous recombination on damaged DNA. J Cell Biol 2008; 183:1203-12; PMID:19103807; http://dx.doi.org/ 10.1083/jcb.200806068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Li M, Yu X. Function of BRCA1 in the DNA damage response is mediated by ADP-ribosylation. Cancer Cell 2013; 23:693-704; PMID:23680151; http://dx.doi.org/ 10.1016/j.ccr.2013.03.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hu Y, Petit SA, Ficarro SB, Toomire KJ, Xie A, Lim E, Cao SA, Park E, Eck MJ, Scully R, et al.. PARP1-driven poly-ADP-ribosylation regulates BRCA1 function in homologous recombination-mediated DNA repair. Cancer Discov 2014; 4:1430-47; PMID:25252691; http://dx.doi.org/ 10.1158/2159-8290.CD-13-0891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Langelier MF, Planck JL, Roy S, Pascal JM. Structural basis for DNA damage-dependent poly(ADP-ribosyl)ation by human PARP-1. Science 2012; 336:728-32; PMID:22582261; http://dx.doi.org/ 10.1126/science.1216338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Alvarez-Gonzalez R, Pacheco-Rodriguez G, Mendoza-Alvarez H. Enzymology of ADP-ribose polymer synthesis. Mol Cell Biochem 1994; 138:33-7; PMID:7898472; http://dx.doi.org/ 10.1007/BF00928440 [DOI] [PubMed] [Google Scholar]
- 42.Langelier MF, Pascal JM. PARP-1 mechanism for coupling DNA damage detection to poly(ADP-ribose) synthesis. Curr Opin Struct Biol 2013; 23:134-43; PMID:23333033; http://dx.doi.org/ 10.1016/j.sbi.2013.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kim MY, Zhang T, Kraus WL. Poly(ADP-ribosyl)ation by PARP-1: ‘PAR-laying’ NAD+ into a nuclear signal. Genes Dev 2005; 19:1951-67; PMID:16140981; http://dx.doi.org/ 10.1101/gad.1331805 [DOI] [PubMed] [Google Scholar]
- 44.Haince JF, McDonald D, Rodrigue A, Dery U, Masson JY, Hendzel MJ, Poirier GG. PARP1-dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites. J Biol Chem 2008; 283:1197-208; PMID:18025084; http://dx.doi.org/ 10.1074/jbc.M706734200 [DOI] [PubMed] [Google Scholar]
- 45.Hassa PO, Hottiger MO. The diverse biological roles of mammalian PARPS, a small but powerful family of poly-ADP-ribose polymerases. Front Biosci 2008; 13:3046-82; PMID:17981777; http://dx.doi.org/ 10.2741/2909 [DOI] [PubMed] [Google Scholar]
- 46.Martinez-Zamudio R, Ha HC. Histone ADP-ribosylation facilitates gene transcription by directly remodeling nucleosomes. Mol Cell Biol 2012; 32:2490-502; PMID:22547677; http://dx.doi.org/ 10.1128/MCB.06667-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Messner S, Hottiger MO. Histone ADP-ribosylation in DNA repair, replication and transcription. Trends Cell Biol 2011; 21:534-42; PMID:21741840; http://dx.doi.org/ 10.1016/j.tcb.2011.06.001 [DOI] [PubMed] [Google Scholar]
- 48.Kulkarni A, Oza J, Yao M, Sohail H, Ginjala V, Tomas-Loba A, Horejsi Z, Tan AR, Boulton SJ, Ganesan S. Tripartite Motif-containing 33 (TRIM33) protein functions in the poly(ADP-ribose) polymerase (PARP)-dependent DNA damage response through interaction with Amplified in Liver Cancer 1 (ALC1) protein. J Biol Chem 2013; 288:32357-69; PMID:23926104; http://dx.doi.org/ 10.1074/jbc.M113.459164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kraus WL. New functions for an ancient domain. Nat Struct Mol Biol 2009; 16:904-7; PMID:19739287; http://dx.doi.org/ 10.1038/nsmb0909-904 [DOI] [PubMed] [Google Scholar]
- 50.Timinszky G, Till S, Hassa PO, Hothorn M, Kustatscher G, Nijmeijer B, Colombelli J, Altmeyer M, Stelzer EH, Scheffzek K, et al.. A macrodomain-containing histone rearranges chromatin upon sensing PARP1 activation. Nat Struct Mol Biol 2009; 16:923-9; PMID:19680243; http://dx.doi.org/ 10.1038/nsmb.1664 [DOI] [PubMed] [Google Scholar]
- 51.Gottschalk AJ, Timinszky G, Kong SE, Jin J, Cai Y, Swanson SK, Washburn MP, Florens L, Ladurner AG, Conaway JW, et al.. Poly(ADP-ribosyl)ation directs recruitment and activation of an ATP-dependent chromatin remodeler. Proc Natl Acad Sci U S A 2009; 106:13770-4; PMID:19666485; http://dx.doi.org/ 10.1073/pnas.0906920106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ahel D, Horejsi Z, Wiechens N, Polo SE, Garcia-Wilson E, Ahel I, Flynn H, Skehel M, West SC, Jackson SP, et al.. Poly(ADP-ribose)-dependent regulation of DNA repair by the chromatin remodeling enzyme ALC1. Science 2009; 325:1240-3; PMID:19661379; http://dx.doi.org/ 10.1126/science.1177321 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gagne JP, Isabelle M, Lo KS, Bourassa S, Hendzel MJ, Dawson VL, Dawson TM, Poirier GG. Proteome-wide identification of poly(ADP-ribose) binding proteins and poly(ADP-ribose)-associated protein complexes. Nucleic Acids Res 2008; 36:6959-76; PMID:18981049; http://dx.doi.org/ 10.1093/nar/gkn771 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Malanga M, Althaus FR. The role of poly(ADP-ribose) in the DNA damage signaling network. Biochem Cell Biol 2005; 83:354-64; PMID:15959561; http://dx.doi.org/ 10.1139/o05-038 [DOI] [PubMed] [Google Scholar]
- 55.Tulin A, Spradling A. Chromatin loosening by poly(ADP)-ribose polymerase (PARP) at Drosophila puff loci. Science 2003; 299:560-2; PMID:12543974; http://dx.doi.org/ 10.1126/science.1078764 [DOI] [PubMed] [Google Scholar]
- 56.Poirier GG, de Murcia G, Jongstra-Bilen J, Niedergang C, Mandel P. Poly(ADP-ribosyl)ation of polynucleosomes causes relaxation of chromatin structure. Proc Natl Acad Sci U S A 1982; 79:3423-7; PMID:6808510; http://dx.doi.org/ 10.1073/pnas.79.11.3423 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ogata N, Ueda K, Kagamiyama H, Hayaishi O. ADP-ribosylation of histone H1. Identification of glutamic acid residues 2, 14, and the COOH-terminal lysine residue as modification sites. J Biol Chem 1980; 255:7616-20; PMID:6772638 [PubMed] [Google Scholar]
- 58.Oka S, Kato J, Moss J. Identification and characterization of a mammalian 39-kDa poly(ADP-ribose) glycohydrolase. J Biol Chem 2006; 281:705-13; PMID:16278211; http://dx.doi.org/ 10.1074/jbc.M510290200 [DOI] [PubMed] [Google Scholar]
- 59.Meyer-Ficca ML, Meyer RG, Coyle DL, Jacobson EL, Jacobson MK. Human poly(ADP-ribose) glycohydrolase is expressed in alternative splice variants yielding isoforms that localize to different cell compartments. Exp Cell Res 2004; 297:521-32; PMID:15212953; http://dx.doi.org/ 10.1016/j.yexcr.2004.03.050 [DOI] [PubMed] [Google Scholar]
- 60.Sharifi R, Morra R, Appel CD, Tallis M, Chioza B, Jankevicius G, Simpson MA, Matic I, Ozkan E, Golia B, et al.. Deficiency of terminal ADP-ribose protein glycohydrolase TARG1/C6orf130 in neurodegenerative disease. EMBO J 2013; 32:1225-37; PMID:23481255; http://dx.doi.org/ 10.1038/emboj.2013.51 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.David KK, Andrabi SA, Dawson TM, Dawson VL. Parthanatos, a messenger of death. Front Biosci (Landmark Ed) 2009; 14:1116-28; PMID:19273119; http://dx.doi.org/ 10.2741/3297 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Aredia F, Scovassi AI. Poly(ADP-ribose): a signaling molecule in different paradigms of cell death. Biochem Pharmacol 2014; 92:157-63; PMID:24976506; http://dx.doi.org/ 10.1016/j.bcp.2014.06.021 [DOI] [PubMed] [Google Scholar]
- 63.Wang Y, Kim NS, Haince JF, Kang HC, David KK, Andrabi SA, Poirier GG, Dawson VL, Dawson TM. Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos). Sci Signal 2011; 4:ra20; PMID:21467298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang Y, Kim NS, Li X, Greer PA, Koehler RC, Dawson VL, Dawson TM. Calpain activation is not required for AIF translocation in PARP-1-dependent cell death (parthanatos). J Neurochem 2009; 110:687-96; PMID:19457082; http://dx.doi.org/ 10.1111/j.1471-4159.2009.06167.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Berger NA, Sims JL, Catino DM, Berger SJ. Poly(ADP-ribose) polymerase mediates the suicide response to massive DNA damage: studies in normal and DNA-repair defective cells. Princess Takamatsu Symp 1983; 13:219-26; PMID:6317637 [PubMed] [Google Scholar]
- 66.Andrabi SA, Kang HC, Haince JF, Lee YI, Zhang J, Chi Z, West AB, Koehler RC, Poirier GG, Dawson TM, et al.. Iduna protects the brain from glutamate excitotoxicity and stroke by interfering with poly(ADP-ribose) polymer-induced cell death. Nat Med 2011; 17:692-9; PMID:21602803; http://dx.doi.org/ 10.1038/nm.2387 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Manic G, Obrist F, Kroemer G, Vitale I, Galluzzi L. Chloroquine and hydroxychloroquine for cancer therapy. Mol Cell Oncol 2014; 1:e29911; http://dx.doi.org/ 10.4161/mco.29911 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Obrist F, Manic G, Kroemer G, Vitale I, Galluzzi L. Trial Watch: Proteasomal inhibitors for anticancer therapy. Mol Cell Oncol 2014; 2:e974463; http://dx.doi.org/ 10.4161/23723556.2014.974463 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tong WM, Hande MP, Lansdorp PM, Wang ZQ. DNA strand break-sensing molecule poly(ADP-Ribose) polymerase cooperates with p53 in telomere function, chromosome stability, and tumor suppression. Mol Cell Biol 2001; 21:4046-54; PMID:11359911; http://dx.doi.org/ 10.1128/MCB.21.12.4046-4054.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Trucco C, Oliver FJ, de Murcia G, Menissier-de Murcia J. DNA repair defect in poly(ADP-ribose) polymerase-deficient cell lines. Nucleic Acids Res 1998; 26:2644-9; PMID:9592149; http://dx.doi.org/ 10.1093/nar/26.11.2644 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Nicolas L, Martinez C, Baro C, Rodriguez M, Baroja-Mazo A, Sole F, Flores JM, Ampurdanes C, Dantzer F, Martin-Caballero J, et al.. Loss of poly(ADP-ribose) polymerase-2 leads to rapid development of spontaneous T-cell lymphomas in p53-deficient mice. Oncogene 2010; 29:2877-83; PMID:20154718; http://dx.doi.org/ 10.1038/onc.2010.11 [DOI] [PubMed] [Google Scholar]
- 72.Menissier de Murcia J, Ricoul M, Tartier L, Niedergang C, Huber A, Dantzer F, Schreiber V, Ame JC, Dierich A, LeMeur M, et al.. Functional interaction between PARP-1 and PARP-2 in chromosome stability and embryonic development in mouse. EMBO J 2003; 22:2255-63; PMID:12727891; http://dx.doi.org/ 10.1093/emboj/cdg206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Tong WM, Yang YG, Cao WH, Galendo D, Frappart L, Shen Y, Wang ZQ. Poly(ADP-ribose) polymerase-1 plays a role in suppressing mammary tumourigenesis in mice. Oncogene 2007; 26:3857-67; PMID:17160013; http://dx.doi.org/ 10.1038/sj.onc.1210156 [DOI] [PubMed] [Google Scholar]
- 74.Tong WM, Cortes U, Hande MP, Ohgaki H, Cavalli LR, Lansdorp PM, Haddad BR, Wang ZQ. Synergistic role of Ku80 and poly(ADP-ribose) polymerase in suppressing chromosomal aberrations and liver cancer formation. Cancer Res 2002; 62:6990-6; PMID:12460917 [PubMed] [Google Scholar]
- 75.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; PMID:25286972; http://dx.doi.org/ 10.1038/nrclinonc.2014.163 [DOI] [PubMed] [Google Scholar]
- 76.Morrison C, Smith GC, Stingl L, Jackson SP, Wagner EF, Wang ZQ. Genetic interaction between PARP and DNA-PK in V(D)J recombination and tumorigenesis. Nat Genet 1997; 17:479-82; PMID:9398855; http://dx.doi.org/ 10.1038/ng1297-479 [DOI] [PubMed] [Google Scholar]
- 77.Piskunova TS, Yurova MN, Ovsyannikov AI, Semenchenko AV, Zabezhinski MA, Popovich IG, Wang ZQ, Anisimov VN. Deficiency in Poly(ADP-ribose) Polymerase-1 (PARP-1) Accelerates Aging and Spontaneous Carcinogenesis in Mice. Curr Gerontol Geriatr Res 2008; PMID:19415146; http://dx.doi.org/ 10.1155/2008/754190 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gonzalez-Flores A, Aguilar-Quesada R, Siles E, Pozo S, Rodriguez-Lara MI, Lopez-Jimenez L, Lopez-Rodriguez M, Peralta-Leal A, Villar D, Martin-Oliva D, et al.. Interaction between PARP-1 and HIF-2alpha in the hypoxic response. Oncogene 2014; 33:891-8; PMID:23455322; http://dx.doi.org/ 10.1038/onc.2013.9 [DOI] [PubMed] [Google Scholar]
- 79.Boehler C, Gauthier L, Yelamos J, Noll A, Schreiber V, Dantzer F. Phenotypic characterization of Parp-1 and Parp-2 deficient mice and cells. Methods Mol Biol 2011; 780:313-36; PMID:21870269 [DOI] [PubMed] [Google Scholar]
- 80.Menisser-de Murcia J, Mark M, Wendling O, Wynshaw-Boris A, de Murcia G. Early embryonic lethality in PARP-1 Atm double-mutant mice suggests a functional synergy in cell proliferation during development. Mol Cell Biol 2001; 21:1828-32; PMID:11238919; http://dx.doi.org/ 10.1128/MCB.21.5.1828-1832.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Huber A, Bai P, de Murcia JM, de Murcia G. PARP-1, PARP-2 and ATM in the DNA damage response: functional synergy in mouse development. DNA Repair (Amst) 2004; 3:1103-8; PMID:15279798; http://dx.doi.org/ 10.1016/j.dnarep.2004.06.002 [DOI] [PubMed] [Google Scholar]
- 82.Ogawa K, Masutani M, Kato K, Tang M, Kamada N, Suzuki H, Nakagama H, Sugimura T, Shirai T. Parp-1 deficiency does not enhance liver carcinogenesis induced by 2-amino-3-methylimidazo; [4,5-f]quinoline in mice. Cancer Lett 2006; 236:32-8; PMID:15955622; http://dx.doi.org/ 10.1016/j.canlet.2005.04.030 [DOI] [PubMed] [Google Scholar]
- 83.Gunji A, Uemura A, Tsutsumi M, Nozaki T, Kusuoka O, Omura K, Suzuki H, Nakagama H, Sugimura T, Masutani M. Parp-1 deficiency does not increase the frequency of tumors in the oral cavity and esophagus of ICR/129Sv mice by 4-nitroquinoline 1-oxide, a carcinogen producing bulky adducts. Cancer Lett 2006; 241:87-92; PMID:16338061; http://dx.doi.org/ 10.1016/j.canlet.2005.10.003 [DOI] [PubMed] [Google Scholar]
- 84.Shibata A, Kamada N, Masumura K, Nohmi T, Kobayashi S, Teraoka H, Nakagama H, Sugimura T, Suzuki H, Masutani M. Parp-1 deficiency causes an increase of deletion mutations and insertions/rearrangements in vivo after treatment with an alkylating agent. Oncogene 2005; 24:1328-37; PMID:15608683; http://dx.doi.org/ 10.1038/sj.onc.1208289 [DOI] [PubMed] [Google Scholar]
- 85.Shibata A, Maeda D, Ogino H, Tsutsumi M, Nohmi T, Nakagama H, Sugimura T, Teraoka H, Masutani M. Role of Parp-1 in suppressing spontaneous deletion mutation in the liver and brain of mice at adolescence and advanced age. Mutat Res 2009; 664:20-7; PMID:19428377; http://dx.doi.org/ 10.1016/j.mrfmmm.2009.02.001 [DOI] [PubMed] [Google Scholar]
- 86.Simbulan-Rosenthal CM, Haddad BR, Rosenthal DS, Weaver Z, Coleman A, Luo R, Young HM, Wang ZQ, Ried T, Smulson ME. Chromosomal aberrations in PARP(−/−) mice: genome stabilization in immortalized cells by reintroduction of poly(ADP-ribose) polymerase cDNA. Proc Natl Acad Sci U S A 1999; 96:13191-6; PMID:10557296; http://dx.doi.org/ 10.1073/pnas.96.23.13191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Piskunova TS, Zabezhinskii MA, Popovich IG, Tyndyk ML, Iurova MN, Anisimov VN. ; [Diethylnitrosamine-induced carcinogenesis in PARP-1(−/−) and PARP-1(+/+) mice]. Vopr Onkol 2009; 55:608-11; PMID:20020658 [PubMed] [Google Scholar]
- 88.Tsutsumi M, Masutani M, Nozaki T, Kusuoka O, Tsujiuchi T, Nakagama H, Suzuki H, Konishi Y, Sugimura T. Increased susceptibility of poly(ADP-ribose) polymerase-1 knockout mice to nitrosamine carcinogenicity. Carcinogenesis 2001; 22:1-3; PMID:11159733; http://dx.doi.org/ 10.1093/carcin/22.1.1 [DOI] [PubMed] [Google Scholar]
- 89.Nozaki T, Fujihara H, Watanabe M, Tsutsumi M, Nakamoto K, Kusuoka O, Kamada N, Suzuki H, Nakagama H, Sugimura T, et al.. Parp-1 deficiency implicated in colon and liver tumorigenesis induced by azoxymethane. Cancer Sci 2003; 94:497-500; PMID:12824873; http://dx.doi.org/ 10.1111/j.1349-7006.2003.tb01472.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Tanori M, Mancuso M, Pasquali E, Leonardi S, Rebessi S, Di Majo V, Guilly MN, Giangaspero F, Covelli V, Pazzaglia S, et al.. PARP-1 cooperates with Ptc1 to suppress medulloblastoma and basal cell carcinoma. Carcinogenesis 2008; 29:1911-9; PMID:18660545; http://dx.doi.org/ 10.1093/carcin/bgn174 [DOI] [PubMed] [Google Scholar]
- 91.Raval-Fernandes S, Kickhoefer VA, Kitchen C, Rome LH. Increased susceptibility of vault poly(ADP-ribose) polymerase-deficient mice to carcinogen-induced tumorigenesis. Cancer Res 2005; 65:8846-52; PMID:16204055; http://dx.doi.org/ 10.1158/0008-5472.CAN-05-0770 [DOI] [PubMed] [Google Scholar]
- 92.Min W, Cortes U, Herceg Z, Tong WM, Wang ZQ. Deletion of the nuclear isoform of poly(ADP-ribose) glycohydrolase (PARG) reveals its function in DNA repair, genomic stability and tumorigenesis. Carcinogenesis 2010; 31:2058-65; PMID:20926829; http://dx.doi.org/ 10.1093/carcin/bgq205 [DOI] [PubMed] [Google Scholar]
- 93.Masutani M, Nakagama H, Sugimura T. Poly(ADP-ribosyl)ation in relation to cancer and autoimmune disease. Cell Mol Life Sci 2005; 62:769-83; PMID:15868402; http://dx.doi.org/ 10.1007/s00018-004-4509-x [DOI] [PubMed] [Google Scholar]
- 94.Lockett KL, Hall MC, Xu J, Zheng SL, Berwick M, Chuang SC, Clark PE, Cramer SD, Lohman K, Hu JJ. The ADPRT V762A genetic variant contributes to prostate cancer susceptibility and deficient enzyme function. Cancer Res 2004; 64:6344-8; PMID:15342424; http://dx.doi.org/ 10.1158/0008-5472.CAN-04-0338 [DOI] [PubMed] [Google Scholar]
- 95.Hao B, Wang H, Zhou K, Li Y, Chen X, Zhou G, Zhu Y, Miao X, Tan W, Wei Q, et al.. Identification of genetic variants in base excision repair pathway and their associations with risk of esophageal squamous cell carcinoma. Cancer Res 2004; 64:4378-84; PMID:15205355; http://dx.doi.org/ 10.1158/0008-5472.CAN-04-0372 [DOI] [PubMed] [Google Scholar]
- 96.Zhang X, Miao X, Liang G, Hao B, Wang Y, Tan W, Li Y, Guo Y, He F, Wei Q, et al.. Polymorphisms in DNA base excision repair genes ADPRT and XRCC1 and risk of lung cancer. Cancer Res 2005; 65:722-6; PMID:15705867 [PubMed] [Google Scholar]
- 97.Ye F, Cheng Q, Hu Y, Zhang J, Chen H. PARP-1 Val762Ala polymorphism is associated with risk of cervical carcinoma. PLoS One 2012; 7:e37446; PMID:22624032; http://dx.doi.org/ 10.1371/journal.pone.0037446 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Roszak A, Lianeri M, Sowinska A, Jagodzinski PP. Involvement of PARP-1 Val762Ala polymorphism in the onset of cervical cancer in caucasian women. Mol Diagn Ther 2013; 17:239-45; PMID:23633189; http://dx.doi.org/ 10.1007/s40291-013-0036-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Yu H, Ma H, Yin M, Wei Q. Association between PARP-1 V762A polymorphism and cancer susceptibility: a meta-analysis. Genet Epidemiol 2012; 36:56-65; PMID:22127734; http://dx.doi.org/ 10.1002/gepi.20663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Qin Q, Lu J, Zhu H, Xu L, Cheng H, Zhan L, Yang X, Zhang C, Sun X. PARP-1 Val762Ala polymorphism and risk of cancer: a meta-analysis based on 39 case-control studies. PLoS One 2014; 9:e98022; PMID:24853559; http://dx.doi.org/ 10.1371/journal.pone.0098022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Singh N. Enhanced poly ADP-ribosylation in human leukemia lymphocytes and ovarian cancers. Cancer Lett 1991; 58:131-5; PMID:1904797; http://dx.doi.org/ 10.1016/0304-3835(91)90035-G [DOI] [PubMed] [Google Scholar]
- 102.Pournazari P, Padmore RF, Kosari F, Scalia P, Shahbani-Rad MT, Shariff S, Demetrick DJ, Bosch M, Mansoor A. B-lymphoblastic leukemia/lymphoma: overexpression of nuclear DNA repair protein PARP-1 correlates with antiapoptotic protein Bcl-2 and complex chromosomal abnormalities. Hum Pathol 2014; 45:1582-7; PMID:24856976; http://dx.doi.org/ 10.1016/j.humpath.2013.11.024 [DOI] [PubMed] [Google Scholar]
- 103.Ossovskaya V, Koo IC, Kaldjian EP, Alvares C, Sherman BM. Upregulation of Poly (ADP-Ribose) Polymerase-1 (PARP1) in triple-negative breast cancer and other primary human tumor types. Genes Cancer 2010; 1:812-21; PMID:21779467; http://dx.doi.org/ 10.1177/1947601910383418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Bieche I, Pennaneach V, Driouch K, Vacher S, Zaremba T, Susini A, Lidereau R, Hall J. Variations in the mRNA expression of poly(ADP-ribose) polymerases, poly(ADP-ribose) glycohydrolase and ADP-ribosylhydrolase 3 in breast tumors and impact on clinical outcome. Int J Cancer 2013; 133:2791-800; PMID:23736962 [DOI] [PubMed] [Google Scholar]
- 105.Rojo F, Garcia-Parra J, Zazo S, Tusquets I, Ferrer-Lozano J, Menendez S, Eroles P, Chamizo C, Servitja S, Ramirez-Merino N, et al.. Nuclear PARP-1 protein overexpression is associated with poor overall survival in early breast cancer. Ann Oncol 2012; 23:1156-64; PMID:21908496; http://dx.doi.org/ 10.1093/annonc/mdr361 [DOI] [PubMed] [Google Scholar]
- 106.Domagala P, Huzarski T, Lubinski J, Gugala K, Domagala W. PARP-1 expression in breast cancer including BRCA1-associated, triple negative and basal-like tumors: possible implications for PARP-1 inhibitor therapy. Breast Cancer Res Treat 2011; 127:861-9; PMID:21409392; http://dx.doi.org/ 10.1007/s10549-011-1441-2 [DOI] [PubMed] [Google Scholar]
- 107.Hassumi-Fukasawa MK, Miranda-Camargo FA, Zanetti BR, Galano DF, Ribeiro-Silva A, Soares EG. Expression of BAG-1 and PARP-1 in precursor lesions and invasive cervical cancer associated with human papillomavirus (HPV). Pathol Oncol Res 2012; 18:929-37; PMID:22454210; http://dx.doi.org/ 10.1007/s12253-012-9523-y [DOI] [PubMed] [Google Scholar]
- 108.Tang Y, Wang YL, Yang L, Xu JX, Xiong W, Xiao M, Li M. Inhibition of arginine ADP-ribosyltransferase 1 reduces the expression of poly(ADP-ribose) polymerase-1 in colon carcinoma. Int J Mol Med 2013; 32:130-6; PMID:23652727 [DOI] [PubMed] [Google Scholar]
- 109.Yalcintepe L, Turker-Sener L, Sener A, Yetkin G, Tiryaki D, Bermek E. Changes in NAD/ADP-ribose metabolism in rectal cancer. Braz J Med Biol Res 2005; 38:361-5; PMID:15761615; http://dx.doi.org/ 10.1590/S0100-879X2005000300006 [DOI] [PubMed] [Google Scholar]
- 110.Nosho K, Yamamoto H, Mikami M, Taniguchi H, Takahashi T, Adachi Y, Imamura A, Imai K, Shinomura Y. Overexpression of poly(ADP-ribose) polymerase-1 (PARP-1) in the early stage of colorectal carcinogenesis. Eur J Cancer 2006; 42:2374-81; PMID:16809031; http://dx.doi.org/ 10.1016/j.ejca.2006.01.061 [DOI] [PubMed] [Google Scholar]
- 111.Idogawa M, Masutani M, Shitashige M, Honda K, Tokino T, Shinomura Y, Imai K, Hirohashi S, Yamada T. Ku70 and poly(ADP-ribose) polymerase-1 competitively regulate β-catenin and T-cell factor-4-mediated gene transactivation: possible linkage of DNA damage recognition and Wnt signaling. Cancer Res 2007; 67:911-8; PMID:17283121; http://dx.doi.org/ 10.1158/0008-5472.CAN-06-2360 [DOI] [PubMed] [Google Scholar]
- 112.Idogawa M, Yamada T, Honda K, Sato S, Imai K, Hirohashi S. Poly(ADP-ribose) polymerase-1 is a component of the oncogenic T-cell factor-4/β-catenin complex. Gastroenterology 2005; 128:1919-36; PMID:15940627; http://dx.doi.org/ 10.1053/j.gastro.2005.03.007 [DOI] [PubMed] [Google Scholar]
- 113.Postawski K, Monist M, Keith G. PARP-1 activity in normal and cancerous human endometrium and its relationship with quantity of abasic sites (AP). Ginekol Pol 2011; 82:16-21; PMID:21469519 [PubMed] [Google Scholar]
- 114.Nomura F, Yaguchi M, Togawa A, Miyazaki M, Isobe K, Miyake M, Noda M, Nakai T. Enhancement of poly-adenosine diphosphate-ribosylation in human hepatocellular carcinoma. J Gastroenterol Hepatol 2000; 15:529-35; PMID:10847440; http://dx.doi.org/ 10.1046/j.1440-1746.2000.02193.x [DOI] [PubMed] [Google Scholar]
- 115.Byers LA, Wang J, Nilsson MB, Fujimoto J, Saintigny P, Yordy J, Giri U, Peyton M, Fan YH, Diao L, et al.. Proteomic profiling identifies dysregulated pathways in small cell lung cancer and novel therapeutic targets including PARP1. Cancer Discov 2012; 2:798-811; PMID:22961666; http://dx.doi.org/ 10.1158/2159-8290.CD-12-0112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Brustmann H. Poly(adenosine diphosphate-ribose) polymerase expression in serous ovarian carcinoma: correlation with p53, MIB-1, and outcome. Int J Gynecol Pathol 2007; 26:147-53; PMID:17413981; http://dx.doi.org/ 10.1097/pgp.0b013e3180555999 [DOI] [PubMed] [Google Scholar]
- 117.Gan A, Green AR, Nolan CC, Martin S, Deen S. Poly(adenosine diphosphate-ribose) polymerase expression in BRCA-proficient ovarian high-grade serous carcinoma; association with patient survival. Hum Pathol 2013; 44:1638-47; PMID:23574784; http://dx.doi.org/ 10.1016/j.humpath.2013.01.015 [DOI] [PubMed] [Google Scholar]
- 118.Bi FF, Li D, Yang Q. Promoter hypomethylation, especially around the E26 transformation-specific motif, and increased expression of poly (ADP-ribose) polymerase 1 in BRCA-mutated serous ovarian cancer. BMC Cancer 2013; 13:90; PMID:23442605; http://dx.doi.org/ 10.1186/1471-2407-13-90 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wu W, Zhu H, Liang Y, Kong Z, Duan X, Li S, Zhao Z, Yang D, Zeng G. 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; PMID:24436031; http://dx.doi.org/ 10.1007/s11255-014-0642-0 [DOI] [PubMed] [Google Scholar]
- 120.Salemi M, Galia A, Fraggetta F, La Corte C, Pepe P, La Vignera S, Improta G, Bosco P, Calogero AE. Poly (ADP-ribose) polymerase 1 protein expression in normal and neoplastic prostatic tissue. Eur J Histochem 2013; 57:e13; PMID:23807292; http://dx.doi.org/ 10.4081/ejh.2013.e13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.McNealy T, Frey M, Trojan L, Knoll T, Alken P, Michel MS. Intrinsic presence of poly (ADP-ribose) is significantly increased in malignant prostate compared to benign prostate cell lines. Anticancer Res 2003; 23:1473-8; PMID:12820412 [PubMed] [Google Scholar]
- 122.Staibano S, Pepe S, Lo Muzio L, Somma P, Mascolo M, Argenziano G, Scalvenzi M, Salvatore G, Fabbrocini G, Molea G, et al.. Poly(adenosine diphosphate-ribose) polymerase 1 expression in malignant melanomas from photoexposed areas of the head and neck region. Hum Pathol 2005; 36:724-31; PMID:16084940; http://dx.doi.org/ 10.1016/j.humpath.2005.04.017 [DOI] [PubMed] [Google Scholar]
- 123.Tentori L, Muzi A, Dorio AS, Bultrini S, Mazzon E, Lacal PM, Shah GM, Zhang J, Navarra P, Nocentini G, et al.. Stable depletion of poly (ADP-ribose) polymerase-1 reduces in vivo melanoma growth and increases chemosensitivity. Eur J Cancer 2008; 44:1302-14; PMID:18440222; http://dx.doi.org/ 10.1016/j.ejca.2008.03.019 [DOI] [PubMed] [Google Scholar]
- 124.Newman RE, Soldatenkov VA, Dritschilo A, Notario V. Poly(ADP-ribose) polymerase turnover alterations do not contribute to PARP overexpression in Ewing's sarcoma cells. Oncol Rep 2002; 9:529-32; PMID:11956622 [PubMed] [Google Scholar]
- 125.Prasad SC, Thraves PJ, Bhatia KG, Smulson ME, Dritschilo A. Enhanced poly(adenosine diphosphate ribose) polymerase activity and gene expression in Ewing's sarcoma cells. Cancer Res 1990; 50:38-43; PMID:2104538 [PubMed] [Google Scholar]
- 126.Galia A, Calogero AE, Condorelli R, Fraggetta F, La Corte A, Ridolfo F, Bosco P, Castiglione R, Salemi M. PARP-1 protein expression in glioblastoma multiforme. Eur J Histochem 2012; 56:e9; PMID:22472897; http://dx.doi.org/ 10.4081/ejh.2012.e9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wharton SB, McNelis U, Bell HS, Whittle IR. Expression of poly(ADP-ribose) polymerase and distribution of poly(ADP-ribosyl)ation in glioblastoma and in a glioma multicellular tumour spheroid model. Neuropathol Appl Neurobiol 2000; 26:528-35; PMID:11123719; http://dx.doi.org/ 10.1046/j.0305-1846.2000.00288.x [DOI] [PubMed] [Google Scholar]
- 128.Csonka T, Murnyak B, Szepesi R, Kurucz A, Klekner A, Hortobagyi T. Poly(ADP-ribose) polymerase-1 (PARP1) and p53 labelling index correlates with tumour grade in meningiomas. Folia Neuropathol 2014; 52:111-20; PMID:25118896; http://dx.doi.org/ 10.5114/fn.2014.43782 [DOI] [PubMed] [Google Scholar]
- 129.Rajaee-Behbahani N, Schmezer P, Ramroth H, Burkle A, Bartsch H, Dietz A, Becher H. Reduced poly(ADP-ribosyl)ation in lymphocytes of laryngeal cancer patients: results of a case-control study. Int J Cancer 2002; 98:780-4; PMID:11920651; http://dx.doi.org/ 10.1002/ijc.10234 [DOI] [PubMed] [Google Scholar]
- 130.von Minckwitz G, Muller BM, Loibl S, Budczies J, Hanusch C, Darb-Esfahani S, Hilfrich J, Weiss E, Huober J, Blohmer JU, et al.. Cytoplasmic poly(adenosine diphosphate-ribose) polymerase expression is predictive and prognostic in patients with breast cancer treated with neoadjuvant chemotherapy. J Clin Oncol 2011; 29:2150-7; PMID:21519019; http://dx.doi.org/ 10.1200/JCO.2010.31.9079 [DOI] [PubMed] [Google Scholar]
- 131.Nobori T, Yamanaka H, Carson DA. Poly(ADP-ribose) polymerase inhibits DNA synthesis initiation in the absence of NAD. Biochem Biophys Res Commun 1989; 163:1113-8; PMID:2506850; http://dx.doi.org/ 10.1016/0006-291X(89)92336-X [DOI] [PubMed] [Google Scholar]
- 132.Menear KA, Adcock C, Boulter R, Cockcroft XL, Copsey L, Cranston A, Dillon KJ, Drzewiecki J, Garman S, Gomez S, et al. Four-; [3-(4-cyclopropanecarbonylpiperazine-1-carbonyl)-4-fluorobenzyl]-2H-phthalazin- 1-one: a novel bioavailable inhibitor of poly(ADP-ribose) polymerase-1. J Med Chem 2008; 51:6581-91; PMID:18800822; http://dx.doi.org/ 10.1021/jm8001263 [DOI] [PubMed] [Google Scholar]
- 133.Thomas HD, Calabrese CR, Batey MA, Canan S, Hostomsky Z, Kyle S, Maegley KA, Newell DR, Skalitzky D, Wang LZ, et al.. Preclinical selection of a novel poly(ADP-ribose) polymerase inhibitor for clinical trial. Mol Cancer Ther 2007; 6:945-56; PMID:17363489; http://dx.doi.org/ 10.1158/1535-7163.MCT-06-0552 [DOI] [PubMed] [Google Scholar]
- 134.Donawho CK, Luo Y, Luo Y, Penning TD, Bauch JL, Bouska JJ, Bontcheva-Diaz VD, Cox BF, DeWeese TL, Dillehay LE, et al.. ABT-888, an orally active poly(ADP-ribose) polymerase inhibitor that potentiates DNA-damaging agents in preclinical tumor models. Clin Cancer Res 2007; 13:2728-37; PMID:17473206; http://dx.doi.org/ 10.1158/1078-0432.CCR-06-3039 [DOI] [PubMed] [Google Scholar]
- 135.Jones P, Altamura S, Boueres J, Ferrigno F, Fonsi M, Giomini C, Lamartina S, Monteagudo E, Ontoria JM, Orsale MV, et al.. Discovery of 2-{4-; [(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): a novel oral poly(ADP-ribose)polymerase (PARP) inhibitor efficacious in BRCA-1 and -2 mutant tumors. J Med Chem 2009; 52:7170-85; PMID:19873981; http://dx.doi.org/ 10.1021/jm901188v [DOI] [PubMed] [Google Scholar]
- 136.Chuang HC, Kapuriya N, Kulp SK, Chen CS, Shapiro CL. Differential anti-proliferative activities of poly(ADP-ribose) polymerase (PARP) inhibitors in triple-negative breast cancer cells. Breast Cancer Res Treat 2012; 134:649-59; PMID:22678161; http://dx.doi.org/ 10.1007/s10549-012-2106-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Patel AG, De Lorenzo SB, Flatten KS, Poirier GG, Kaufmann SH. Failure of iniparib to inhibit poly(ADP-Ribose) polymerase in vitro. Clin Cancer Res 2012; 18:1655-62; PMID:22291137; http://dx.doi.org/ 10.1158/1078-0432.CCR-11-2890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.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; PMID:7669074; http://dx.doi.org/ 10.1016/0006-2952(95)00189-7 [DOI] [PubMed] [Google Scholar]
- 139.Miknyoczki S, Chang H, Grobelny J, Pritchard S, Worrell C, McGann N, Ator M, Husten J, Deibold J, Hudkins R, et al.. The selective poly(ADP-ribose) polymerase-1(2) inhibitor, CEP-8983, increases the sensitivity of chemoresistant tumor cells to temozolomide and irinotecan but does not potentiate myelotoxicity. Mol Cancer Ther 2007; 6:2290-302; PMID:17699724; http://dx.doi.org/ 10.1158/1535-7163.MCT-07-0062 [DOI] [PubMed] [Google Scholar]
- 140.Russo AL, Kwon HC, Burgan WE, Carter D, Beam K, Weizheng X, Zhang J, Slusher BS, Chakravarti A, Tofilon PJ, et al.. In vitro and in vivo radiosensitization of glioblastoma cells by the poly (ADP-ribose) polymerase inhibitor E7016. Clin Cancer Res 2009; 15:607-12; PMID:19147766; http://dx.doi.org/ 10.1158/1078-0432.CCR-08-2079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Brock WA, Milas L, Bergh S, Lo R, Szabo C, Mason KA. Radiosensitization of human and rodent cell lines by INO-1001, a novel inhibitor of poly(ADP-ribose) polymerase. Cancer Lett 2004; 205:155-60; PMID:15036647; http://dx.doi.org/ 10.1016/j.canlet.2003.10.029 [DOI] [PubMed] [Google Scholar]
- 142.Michels J, Vitale I, Saparbaev M, Castedo M, Kroemer G. Predictive biomarkers for cancer therapy with PARP inhibitors. Oncogene 2014; 33:3894-907; PMID:24037533; http://dx.doi.org/ 10.1038/onc.2013.352 [DOI] [PubMed] [Google Scholar]
- 143.Michels J, Obrist F, Castedo M, Vitale I, Kroemer G. PARP and other prospective targets for poisoning cancer cell metabolism. Biochem Pharmacol 2014; 92:164-71; PMID:25199458; http://dx.doi.org/ 10.1016/j.bcp.2014.08.026 [DOI] [PubMed] [Google Scholar]
- 144.Curtin NJ, Szabo C. Therapeutic applications of PARP inhibitors: anticancer therapy and beyond. Mol Aspects Med 2013; 34:1217-56; PMID:23370117; http://dx.doi.org/ 10.1016/j.mam.2013.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Wahlberg E, Karlberg T, Kouznetsova E, Markova N, Macchiarulo A, Thorsell AG, Pol E, Frostell A, Ekblad T, Oncu D, et al.. Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitors. Nat Biotechnol 2012; 30:283-8; PMID:22343925; http://dx.doi.org/ 10.1038/nbt.2121 [DOI] [PubMed] [Google Scholar]
- 146.Kummar S, Chen A, Parchment RE, Kinders RJ, Ji J, Tomaszewski JE, Doroshow JH. Advances in using PARP inhibitors to treat cancer. BMC Med 2012; 10:25; PMID:22401667; http://dx.doi.org/ 10.1186/1741-7015-10-25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Leung M, Rosen D, Fields S, Cesano A, Budman DR. Poly(ADP-ribose) polymerase-1 inhibition: preclinical and clinical development of synthetic lethality. Mol Med 2011; 17:854-62; PMID:21424107; http://dx.doi.org/ 10.2119/molmed.2010.00240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zaremba T, Curtin NJ. PARP inhibitor development for systemic cancer targeting. Anticancer Agents Med Chem 2007; 7:515-23; PMID:17896912; http://dx.doi.org/ 10.2174/187152007781668715 [DOI] [PubMed] [Google Scholar]
- 149.Ratnam K, Low JA. Current development of clinical inhibitors of poly(ADP-ribose) polymerase in oncology. Clin Cancer Res 2007; 13:1383-8; PMID:17332279; http://dx.doi.org/ 10.1158/1078-0432.CCR-06-2260 [DOI] [PubMed] [Google Scholar]
- 150.Dobzhansky T. Genetics of natural populations. Xiii. recombination and variability in populations of drosophila pseudoobscura. Genetics 1946; 31:269-90; PMID:17247197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, Santarosa M, Dillon KJ, Hickson I, Knights C, et al.. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005; 434:917-21; PMID:15829967; http://dx.doi.org/ 10.1038/nature03445 [DOI] [PubMed] [Google Scholar]
- 152.Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, Kyle S, Meuth M, Curtin NJ, Helleday T. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005; 434:913-7; PMID:15829966; http://dx.doi.org/ 10.1038/nature03443 [DOI] [PubMed] [Google Scholar]
- 153.Lee JM, Ledermann JA, Kohn EC. PARP Inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann Oncol 2014; 25:32-40; PMID:24225019; http://dx.doi.org/ 10.1093/annonc/mdt384 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Turner N, Tutt A, Ashworth A. Hallmarks of ‘BRCAness’ in sporadic cancers. Nat Rev Cancer 2004; 4:814-9; PMID:15510162; http://dx.doi.org/ 10.1038/nrc1457 [DOI] [PubMed] [Google Scholar]
- 155.McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, Giavara S, O'Connor MJ, Tutt AN, Zdzienicka MZ, 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; PMID:16912188; http://dx.doi.org/ 10.1158/0008-5472.CAN-06-0140 [DOI] [PubMed] [Google Scholar]
- 156.Williamson CT, Muzik H, Turhan AG, Zamo A, O'Connor MJ, Bebb DG, Lees-Miller SP. ATM deficiency sensitizes mantle cell lymphoma cells to poly(ADP-ribose) polymerase-1 inhibitors. Mol Cancer Ther 2010; 9:347-57; PMID:20124459; http://dx.doi.org/ 10.1158/1535-7163.MCT-09-0872 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Williamson CT, Kubota E, Hamill JD, Klimowicz A, Ye R, Muzik H, Dean M, Tu L, Gilley D, Magliocco AM, et al.. Enhanced cytotoxicity of PARP inhibition in mantle cell lymphoma harbouring mutations in both ATM and p53. EMBO Mol Med 2012; 4:515-27; PMID:22416035; http://dx.doi.org/ 10.1002/emmm.201200229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Huehls AM, Wagner JM, Huntoon CJ, Karnitz LM. Identification of DNA repair pathways that affect the survival of ovarian cancer cells treated with a poly(ADP-ribose) polymerase inhibitor in a novel drug combination. Mol Pharmacol 2012; 82:767-76; PMID:22833573; http://dx.doi.org/ 10.1124/mol.112.080614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Weston VJ, Oldreive CE, Skowronska A, Oscier DG, Pratt G, Dyer MJ, Smith G, Powell JE, Rudzki Z, Kearns P, et al.. The PARP inhibitor olaparib induces significant killing of ATM-deficient lymphoid tumor cells in vitro and in vivo. Blood 2010; 116:4578-87; PMID:20739657; http://dx.doi.org/ 10.1182/blood-2010-01-265769 [DOI] [PubMed] [Google Scholar]
- 160.Gilardini Montani MS, Prodosmo A, Stagni V, Merli D, Monteonofrio L, Gatti V, Gentileschi MP, Barila D, Soddu S. ATM-depletion in breast cancer cells confers sensitivity to PARP inhibition. J Exp Clin Cancer Res 2013; 32:95; PMID:24252502; http://dx.doi.org/ 10.1186/1756-9966-32-95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Huntoon CJ, Flatten KS, Wahner Hendrickson AE, Huehls AM, Sutor SL, Kaufmann SH, Karnitz LM. ATR inhibition broadly sensitizes ovarian cancer cells to chemotherapy independent of BRCA status. Cancer Res 2013; 73:3683-91; PMID:23548269; http://dx.doi.org/ 10.1158/0008-5472.CAN-13-0110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Iorns E, Lord CJ, Turner N, Ashworth A. Utilizing RNA interference to enhance cancer drug discovery. Nat Rev Drug Discov 2007; 6:556-68; PMID:17599085; http://dx.doi.org/ 10.1038/nrd2355 [DOI] [PubMed] [Google Scholar]
- 163.Taniguchi T, Tischkowitz M, Ameziane N, Hodgson SV, Mathew CG, Joenje H, Mok SC, D'Andrea AD. Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med 2003; 9:568-74; PMID:12692539; http://dx.doi.org/ 10.1038/nm852 [DOI] [PubMed] [Google Scholar]
- 164.Renwick A, Thompson D, Seal S, Kelly P, Chagtai T, Ahmed M, North B, Jayatilake H, Barfoot R, Spanova K, et al.. ATM mutations that cause ataxia-telangiectasia are breast cancer susceptibility alleles. Nat Genet 2006; 38:873-5; PMID:16832357; http://dx.doi.org/ 10.1038/ng1837 [DOI] [PubMed] [Google Scholar]
- 165.Boultwood J. Ataxia telangiectasia gene mutations in leukaemia and lymphoma. J Clin Pathol 2001; 54:512-6; PMID:11429421; http://dx.doi.org/ 10.1136/jcp.54.7.512 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Nevanlinna H, Bartek J. The CHEK2 gene and inherited breast cancer susceptibility. Oncogene 2006; 25:5912-9; PMID:16998506; http://dx.doi.org/ 10.1038/sj.onc.1209877 [DOI] [PubMed] [Google Scholar]
- 167.Mendes-Pereira AM, Martin SA, Brough R, McCarthy A, Taylor JR, Kim JS, Waldman T, Lord CJ, Ashworth A. Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO Mol Med 2009; 1:315-22; PMID:20049735; http://dx.doi.org/ 10.1002/emmm.200900041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Dedes KJ, Wetterskog D, Mendes-Pereira AM, Natrajan R, Lambros MB, Geyer FC, Vatcheva R, Savage K, Mackay A, Lord CJ, et al.. PTEN deficiency in endometrioid endometrial adenocarcinomas predicts sensitivity to PARP inhibitors. Sci Transl Med 2010; 2:53ra75; PMID:20944090; http://dx.doi.org/ 10.1126/scitranslmed.3001538 [DOI] [PubMed] [Google Scholar]
- 169.McEllin B, Camacho CV, Mukherjee B, Hahm B, Tomimatsu N, Bachoo RM, Burma S. PTEN loss compromises homologous recombination repair in astrocytes: implications for glioblastoma therapy with temozolomide or poly(ADP-ribose) polymerase inhibitors. Cancer Res 2010; 70:5457-64; PMID:20530668; http://dx.doi.org/ 10.1158/0008-5472.CAN-09-4295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Minami D, Takigawa N, Takeda H, Takata M, Ochi N, Ichihara E, Hisamoto A, Hotta K, Tanimoto M, Kiura K. Synergistic effect of olaparib with combination of cisplatin on PTEN-deficient lung cancer cells. Mol Cancer Res 2013; 11:140-8; PMID:23239809; http://dx.doi.org/ 10.1158/1541-7786.MCR-12-0401 [DOI] [PubMed] [Google Scholar]
- 171.Chatterjee P, Choudhary GS, Sharma A, Singh K, Heston WD, Ciezki J, Klein EA, Almasan A. PARP inhibition sensitizes to low dose-rate radiation TMPRSS2-ERG fusion gene-expressing and PTEN-deficient prostate cancer cells. PLoS One 2013; 8:e60408; PMID:23565244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Vilar E, Bartnik CM, Stenzel SL, Raskin L, Ahn J, Moreno V, Mukherjee B, Iniesta MD, Morgan MA, Rennert G, et al.. MRE11 deficiency increases sensitivity to poly(ADP-ribose) polymerase inhibition in microsatellite unstable colorectal cancers. Cancer Res 2011; 71:2632-42; PMID:21300766; http://dx.doi.org/ 10.1158/0008-5472.CAN-10-1120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.McPherson LA, Shen Y, Ford JM. Poly (ADP-ribose) polymerase inhibitor LT-626: Sensitivity correlates with MRE11 mutations and synergizes with platinums and irinotecan in colorectal cancer cells. Cancer Lett 2014; 343:217-23; PMID:24215868; http://dx.doi.org/ 10.1016/j.canlet.2013.10.034 [DOI] [PubMed] [Google Scholar]
- 174.Gaymes TJ, Mohamedali AM, Patterson M, Matto N, Smith A, Kulasekararaj A, Chelliah R, Curtin N, Farzaneh F, Shall S, et al.. Microsatellite instability induced mutations in DNA repair genes CtIP and MRE11 confer hypersensitivity to poly (ADP-ribose) polymerase inhibitors in myeloid malignancies. Haematologica 2013; 98:1397-406; PMID:23349304; http://dx.doi.org/ 10.3324/haematol.2012.079251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Metzger-Filho O, Tutt A, de Azambuja E, Saini KS, Viale G, Loi S, Bradbury I, Bliss JM, Azim HA Jr., Ellis P, et al.. Dissecting the heterogeneity of triple-negative breast cancer. J Clin Oncol 2012; 30:1879-87; PMID:22454417; http://dx.doi.org/ 10.1200/JCO.2011.38.2010 [DOI] [PubMed] [Google Scholar]
- 176.Jacot W, Thezenas S, Senal R, Viglianti C, Laberenne AC, Lopez-Crapez E, Bibeau F, Bleuse JP, Romieu G, Lamy PJ. BRCA1 promoter hypermethylation, 53BP1 protein expression and PARP-1 activity as biomarkers of DNA repair deficit in breast cancer. BMC Cancer 2013; 13:523; PMID:24191908; http://dx.doi.org/ 10.1186/1471-2407-13-523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Lips EH, Mulder L, Oonk A, van der Kolk LE, Hogervorst FB, Imholz AL, Wesseling J, Rodenhuis S, Nederlof PM. Triple-negative breast cancer: BRCAness and concordance of clinical features with BRCA1-mutation carriers. Br J Cancer 2013; 108:2172-7; PMID:23558900; http://dx.doi.org/ 10.1038/bjc.2013.144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Hartman AR, Kaldate RR, Sailer LM, Painter L, Grier CE, Endsley RR, Griffin M, Hamilton SA, Frye CA, Silberman MA, et al.. Prevalence of BRCA mutations in an unselected population of triple-negative breast cancer. Cancer 2012; 118:2787-95; PMID:22614657; http://dx.doi.org/ 10.1002/cncr.26576 [DOI] [PubMed] [Google Scholar]
- 179.Gonzalez-Angulo AM, Timms KM, Liu S, Chen H, Litton JK, Potter J, Lanchbury JS, Stemke-Hale K, Hennessy BT, Arun BK, et al.. Incidence and outcome of BRCA mutations in unselected patients with triple receptor-negative breast cancer. Clin Cancer Res 2011; 17:1082-9; PMID:21233401; http://dx.doi.org/ 10.1158/1078-0432.CCR-10-2560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Anders CK, Winer EP, Ford JM, Dent R, Silver DP, Sledge GW, Carey LA. Poly(ADP-Ribose) polymerase inhibition: “targeted” therapy for triple-negative breast cancer. Clin Cancer Res 2010; 16:4702-10; PMID:20858840; http://dx.doi.org/ 10.1158/1078-0432.CCR-10-0939 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Alli E, Sharma VB, Sunderesakumar P, Ford JM. Defective repair of oxidative dna damage in triple-negative breast cancer confers sensitivity to inhibition of poly(ADP-ribose) polymerase. Cancer Res 2009; 69:3589-96; PMID:19351835; http://dx.doi.org/ 10.1158/0008-5472.CAN-08-4016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Ceccaldi R, Liu JC, Amunugama R, Hajdu I, Primack B, Petalcorin MI, O'Connor KW, Konstantinopoulos PA, Elledge SJ, Boulton SJ, et al.. Homologous-recombination-deficient tumours are dependent on Poltheta-mediated repair. Nature 2015; 518:258-62; PMID:25642963; http://dx.doi.org/ 10.1038/nature14184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Mateos-Gomez PA, Gong F, Nair N, Miller KM, Lazzerini-Denchi E, Sfeir A. Mammalian polymerase theta promotes alternative NHEJ and suppresses recombination. Nature 2015; 518:254-7; PMID:25642960; http://dx.doi.org/ 10.1038/nature14157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Ibrahim YH, Garcia-Garcia C, Serra V, He L, Torres-Lockhart K, Prat A, Anton P, Cozar P, Guzman M, Grueso J, et al.. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov 2012; 2:1036-47; PMID:22915752; http://dx.doi.org/ 10.1158/2159-8290.CD-11-0348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Juvekar A, Burga LN, Hu H, Lunsford EP, Ibrahim YH, Balmana J, Rajendran A, Papa A, Spencer K, Lyssiotis CA, et al.. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov 2012; 2:1048-63; PMID:22915751; http://dx.doi.org/ 10.1158/2159-8290.CD-11-0336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.De P, Sun Y, Carlson JH, Friedman LS, Leyland-Jones BR, Dey N. Doubling down on the PI3K-AKT-mTOR pathway enhances the antitumor efficacy of PARP inhibitor in triple negative breast cancer model beyond BRCA-ness. Neoplasia 2014; 16:43-72; PMID:24563619; http://dx.doi.org/ 10.1593/neo.131694 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Johnson N, Li YC, Walton ZE, Cheng KA, Li D, Rodig SJ, Moreau LA, Unitt C, Bronson RT, Thomas HD, et al.. Compromised CDK1 activity sensitizes BRCA-proficient cancers to PARP inhibition. Nat Med 2011; 17:875-82; PMID:21706030; http://dx.doi.org/ 10.1038/nm.2377 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.To C, Kim EH, Royce DB, Williams CR, Collins RM, Risingsong R, Sporn MB, Liby KT. The PARP inhibitors, veliparib and olaparib, are effective chemopreventive agents for delaying mammary tumor development in BRCA1-deficient mice. Cancer Prev Res (Phila) 2014; 7:698-707; PMID:24817481; http://dx.doi.org/ 10.1158/1940-6207.CAPR-14-0047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Gottipati P, Vischioni B, Schultz N, Solomons J, Bryant HE, Djureinovic T, Issaeva N, Sleeth K, Sharma RA, Helleday T. Poly(ADP-ribose) polymerase is hyperactivated in homologous recombination-defective cells. Cancer Res 2010; 70:5389-98; PMID:20551068; http://dx.doi.org/ 10.1158/0008-5472.CAN-09-4716 [DOI] [PubMed] [Google Scholar]
- 190.Michels J, Vitale I, Galluzzi L, Adam J, Olaussen KA, Kepp O, Senovilla L, Talhaoui I, Guegan J, Enot DP, et al.. Cisplatin resistance associated with PARP hyperactivation. Cancer Res 2013; 73:2271-80; PMID:23554447; http://dx.doi.org/ 10.1158/0008-5472.CAN-12-3000 [DOI] [PubMed] [Google Scholar]
- 191.Nowsheen S, Bonner JA, Yang ES. The poly(ADP-Ribose) polymerase inhibitor ABT-888 reduces radiation-induced nuclear EGFR and augments head and neck tumor response to radiotherapy. Radiother Oncol 2011; 99:331-8; PMID:21719137; http://dx.doi.org/ 10.1016/j.radonc.2011.05.084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Shall S, de Murcia G. Poly(ADP-ribose) polymerase-1: what have we learned from the deficient mouse model? Mutat Res 2000; 460:1-15; PMID:10856830; http://dx.doi.org/ 10.1016/S0921-8777(00)00016-1 [DOI] [PubMed] [Google Scholar]
- 193.Patel AG, Sarkaria JN, Kaufmann SH. Nonhomologous end joining drives poly(ADP-ribose) polymerase (PARP) inhibitor lethality in homologous recombination-deficient cells. Proc Natl Acad Sci U S A 2011; 108:3406-11; PMID:21300883; http://dx.doi.org/ 10.1073/pnas.1013715108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Bouwman P, Aly A, Escandell JM, Pieterse M, Bartkova J, van der Gulden H, Hiddingh S, Thanasoula M, Kulkarni A, Yang Q, et al.. 53BP1 loss rescues BRCA1 deficiency and is associated with triple-negative and BRCA-mutated breast cancers. Nat Struct Mol Biol 2010; 17:688-95; PMID:20453858; http://dx.doi.org/ 10.1038/nsmb.1831 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Murai J, Huang SY, Das BB, Renaud A, Zhang Y, Doroshow JH, Ji J, Takeda S, Pommier Y. Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors. Cancer Res 2012; 72:5588-99; PMID:23118055; http://dx.doi.org/ 10.1158/0008-5472.CAN-12-2753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Helleday T. The underlying mechanism for the PARP and BRCA synthetic lethality: clearing up the misunderstandings. Mol Oncol 2011; 5:387-93; PMID:21821475; http://dx.doi.org/ 10.1016/j.molonc.2011.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Shen Y, Aoyagi-Scharber M, Wang B. Trapping Poly(ADP-Ribose) Polymerase. J Pharmacol Exp Ther 2015; 353:446-57; PMID:25758918; http://dx.doi.org/ 10.1124/jpet.114.222448 [DOI] [PubMed] [Google Scholar]
- 198.Murai J, Zhang Y, Morris J, Ji J, Takeda S, Doroshow JH, Pommier Y. Rationale for poly(ADP-ribose) polymerase (PARP) inhibitors in combination therapy with camptothecins or temozolomide based on PARP trapping vs. catalytic inhibition. J Pharmacol Exp Ther 2014; 349:408-16; PMID:24650937; http://dx.doi.org/ 10.1124/jpet.113.210146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Murai J, Huang SY, Renaud A, Zhang Y, Ji J, Takeda S, Morris J, Teicher B, Doroshow JH, Pommier Y. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol Cancer Ther 2014; 13:433-43; PMID:24356813; http://dx.doi.org/ 10.1158/1535-7163.MCT-13-0803 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Anders C, Deal AM, Abramson V, Liu MC, Storniolo AM, Carpenter JT, Puhalla S, Nanda R, Melhem-Bertrandt A, Lin NU, et al.. TBCRC 018: phase II study of iniparib in combination with irinotecan to treat progressive triple negative breast cancer brain metastases. Breast Cancer Res Treat 2014; 146:557-66; PMID:25001612; http://dx.doi.org/ 10.1007/s10549-014-3039-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Aghajanian C, Sill MW, Secord AA, Powell MA, Steinhoff M. Iniparib plus paclitaxel and carboplatin as initial treatment of advanced or recurrent uterine carcinosarcoma: a Gynecologic Oncology Group Study. Gynecol Oncol 2012; 126:424-7; PMID:22634397; http://dx.doi.org/ 10.1016/j.ygyno.2012.05.024 [DOI] [PubMed] [Google Scholar]
- 202.Novello S, Besse B, Felip E, Barlesi F, Mazieres J, Zalcman G, von Pawel J, Reck M, Cappuzzo F, Ferry D, et al.. A phase II randomized study evaluating the addition of iniparib to gemcitabine plus cisplatin as first-line therapy for metastatic non-small-cell lung cancer. Ann Oncol 2014; 25:2156-62; PMID:25139550; http://dx.doi.org/ 10.1093/annonc/mdu384 [DOI] [PubMed] [Google Scholar]
- 203.O'Shaughnessy J, Osborne C, Pippen JE, Yoffe M, Patt D, Rocha C, Koo IC, Sherman BM, Bradley C. Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N Engl J Med 2011; 364:205-14; PMID:21208101; http://dx.doi.org/ 10.1056/NEJMoa1011418 [DOI] [PubMed] [Google Scholar]
- 204.O'Shaughnessy J, Schwartzberg L, Danso MA, Miller KD, Rugo HS, Neubauer M, Robert N, Hellerstedt B, Saleh M, Richards P, et al.. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J Clin Oncol 2014; 32:3840-7; PMID:25349301; http://dx.doi.org/ 10.1200/JCO.2014.55.2984 [DOI] [PubMed] [Google Scholar]
- 205.Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, Friedlander M, Balmana J, Mitchell G, Fried G, Stemmer SM, Hubert A, et al.. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 2015; 33:244-50; PMID:25366685; http://dx.doi.org/ 10.1200/JCO.2014.56.2728 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O'Connor MJ, et al.. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 2009; 361:123-34; PMID:19553641; http://dx.doi.org/ 10.1056/NEJMoa0900212 [DOI] [PubMed] [Google Scholar]
- 207.Yamamoto N, Nokihara H, Yamada Y, Goto Y, Tanioka M, Shibata T, Yamada K, Asahina H, Kawata T, Shi X, et al.. A Phase I, dose-finding and pharmacokinetic study of olaparib (AZD2281) in Japanese patients with advanced solid tumors. Cancer Sci 2012; 103:504-9; PMID:22145984; http://dx.doi.org/ 10.1111/j.1349-7006.2011.02179.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Del Conte G, Sessa C, von Moos R, Vigano L, Digena T, Locatelli A, Gallerani E, Fasolo A, Tessari A, Cathomas R, et al.. Phase I study of olaparib in combination with liposomal doxorubicin in patients with advanced solid tumours. Br J Cancer 2014; 111:651-9; PMID:25025963; http://dx.doi.org/ 10.1038/bjc.2014.345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Fong PC, Yap TA, Boss DS, Carden CP, Mergui-Roelvink M, Gourley C, De Greve J, Lubinski J, Shanley S, Messiou C, et al.. Poly(ADP)-ribose polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum-free interval. J Clin Oncol 2010; 28:2512-9; PMID:20406929; http://dx.doi.org/ 10.1200/JCO.2009.26.9589 [DOI] [PubMed] [Google Scholar]
- 210.Bundred N, Gardovskis J, Jaskiewicz J, Eglitis J, Paramonov V, McCormack P, Swaisland H, Cavallin M, Parry T, Carmichael J, et al.. Evaluation of the pharmacodynamics and pharmacokinetics of the PARP inhibitor olaparib: a phase I multicentre trial in patients scheduled for elective breast cancer surgery. Invest New Drugs 2013; 31:949-58; PMID:23315029; http://dx.doi.org/ 10.1007/s10637-012-9922-7 [DOI] [PubMed] [Google Scholar]
- 211.Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, Friedlander M, Arun B, Loman N, Schmutzler RK, 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; PMID:20609467; http://dx.doi.org/ 10.1016/S0140-6736(10)60892-6 [DOI] [PubMed] [Google Scholar]
- 212.Kaye SB, Lubinski J, Matulonis U, Ang JE, Gourley C, Karlan BY, Amnon A, Bell-McGuinn KM, Chen LM, Friedlander M, et al.. Phase II, open-label, randomized, multicenter study comparing the efficacy and safety of olaparib, a poly (ADP-ribose) polymerase inhibitor, and pegylated liposomal doxorubicin in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer. J Clin Oncol 2012; 30:372-9; PMID:22203755; http://dx.doi.org/ 10.1200/JCO.2011.36.9215 [DOI] [PubMed] [Google Scholar]
- 213.Audeh MW, Carmichael J, Penson RT, Friedlander M, Powell B, Bell-McGuinn KM, Scott C, Weitzel JN, Oaknin A, Loman N, 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; PMID:20609468; http://dx.doi.org/ 10.1016/S0140-6736(10)60893-8 [DOI] [PubMed] [Google Scholar]
- 214.Gelmon KA, Tischkowitz M, Mackay H, Swenerton K, Robidoux A, Tonkin K, Hirte H, Huntsman D, Clemons M, Gilks B, et al.. Olaparib in patients with recurrent high-grade serous or poorly differentiated ovarian carcinoma or triple-negative breast cancer: a phase 2, multicentre, open-label, non-randomised study. Lancet Oncol 2011; 12:852-61; PMID:21862407; http://dx.doi.org/ 10.1016/S1470-2045(11)70214-5 [DOI] [PubMed] [Google Scholar]
- 215.Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, Scott C, Meier W, Shapira-Frommer R, Safra T, et al.. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med 2012; 366:1382-92; PMID:22452356; http://dx.doi.org/ 10.1056/NEJMoa1105535 [DOI] [PubMed] [Google Scholar]
- 216.Choy E, Butrynski JE, Harmon DC, Morgan JA, George S, Wagner AJ, D'Adamo D, Cote GM, Flamand Y, Benes CH, et al.. Phase II study of olaparib in patients with refractory Ewing sarcoma following failure of standard chemotherapy. BMC Cancer 2014; 14:813; PMID:25374341; http://dx.doi.org/ 10.1186/1471-2407-14-813 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Sandhu SK, Schelman WR, Wilding G, Moreno V, Baird RD, Miranda S, Hylands L, Riisnaes R, Forster M, Omlin A, et al.. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: a phase 1 dose-escalation trial. Lancet Oncol 2013; 14:882-92; PMID:23810788; http://dx.doi.org/ 10.1016/S1470-2045(13)70240-7 [DOI] [PubMed] [Google Scholar]
- 218.Coleman RL, Sill MW, Bell-McGuinn K, Aghajanian C, Gray HJ, Tewari KS, Rubin SC, Rutherford TJ, Chan JK, Chen A, et al.. A phase II evaluation of the potent, highly selective PARP inhibitor veliparib in the treatment of persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer in patients who carry a germline BRCA1 or BRCA2 mutation – An NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol 2015; 137:386-91; PMID:25818403; doi: 10.1016/j.ygyno.2015.03.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Ledermann J, Harter P, Gourley C, Friedlander M, Vergote I, Rustin G, Scott CL, Meier W, Shapira-Frommer R, Safra T, et al.. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 2014; 15:852-61; PMID:24882434; http://dx.doi.org/ 10.1016/S1470-2045(14)70228-1 [DOI] [PubMed] [Google Scholar]
- 220.Eggermont AM, Kirkwood JM. Re-evaluating the role of dacarbazine in metastatic melanoma: what have we learned in 30 years? Eur J Cancer 2004; 40:1825-36; PMID:15288283; http://dx.doi.org/ 10.1016/j.ejca.2004.04.030 [DOI] [PubMed] [Google Scholar]
- 221.Khan OA, Gore M, Lorigan P, Stone J, Greystoke A, Burke W, Carmichael J, Watson AJ, McGown G, Thorncroft M, et al.. A phase I study of the safety and tolerability of olaparib (AZD2281, KU0059436) and dacarbazine in patients with advanced solid tumours. Br J Cancer 2011; 104:750-5; PMID:21326243; http://dx.doi.org/ 10.1038/bjc.2011.8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Galluzzi L, Vitale I, Senovilla L, Olaussen KA, Pinna G, Eisenberg T, Goubar A, Martins I, Michels J, Kratassiouk G, et al.. Prognostic impact of vitamin B6 metabolism in lung cancer. Cell Rep 2012; 2:257-69; PMID:22854025; http://dx.doi.org/ 10.1016/j.celrep.2012.06.017 [DOI] [PubMed] [Google Scholar]
- 223.Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M, Kroemer G. Molecular mechanisms of cisplatin resistance. Oncogene 2012; 31:1869-83; PMID:21892204; http://dx.doi.org/ 10.1038/onc.2011.384 [DOI] [PubMed] [Google Scholar]
- 224.Tajeddine N, Galluzzi L, Kepp O, Hangen E, Morselli E, Senovilla L, Araujo N, Pinna G, Larochette N, Zamzami N, et al.. Hierarchical involvement of Bak, VDAC1 and Bax in cisplatin-induced cell death. Oncogene 2008; 27:4221-32; PMID:18362892; http://dx.doi.org/ 10.1038/onc.2008.63 [DOI] [PubMed] [Google Scholar]
- 225.Cirone M, Garufi A, Di Renzo L, Granato M, Faggioni A, D'Orazi G. Zinc supplementation is required for the cytotoxic and immunogenic effects of chemotherapy in chemoresistant p53-functionally deficient cells. Oncoimmunology 2013; 2:e26198; PMID:24228232; http://dx.doi.org/ 10.4161/onci.26198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Pauwels B, Korst AE, Lardon F, Vermorken JB. Combined modality therapy of gemcitabine and radiation. Oncologist 2005; 10:34-51; PMID:15632251; http://dx.doi.org/ 10.1634/theoncologist.10-1-34 [DOI] [PubMed] [Google Scholar]
- 227.Heinemann V. Role of gemcitabine in the treatment of advanced and metastatic breast cancer. Oncology 2003; 64:191-206; PMID:12697958; http://dx.doi.org/ 10.1159/000069315 [DOI] [PubMed] [Google Scholar]
- 228.Rajan A, Carter CA, Kelly RJ, Gutierrez M, Kummar S, Szabo E, Yancey MA, Ji J, Mannargudi B, Woo S, et al.. A phase I combination study of olaparib with cisplatin and gemcitabine in adults with solid tumors. Clin Cancer Res 2012; 18:2344-51; PMID:22371451; http://dx.doi.org/ 10.1158/1078-0432.CCR-11-2425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Pirker R, Berzinec P, Brincat S, Kasan P, Ostoros G, Pesek M, Plate S, Purkalne G, Rooneem R, Skrickova J, et al.. Therapy of small cell lung cancer with emphasis on oral topotecan. Lung Cancer 2010; 70:7-13; PMID:20576312; http://dx.doi.org/ 10.1016/j.lungcan.2010.05.020 [DOI] [PubMed] [Google Scholar]
- 230.Samol J, Ranson M, Scott E, Macpherson E, Carmichael J, Thomas A, Cassidy J. Safety and tolerability of the poly(ADP-ribose) polymerase (PARP) inhibitor, olaparib (AZD2281) in combination with topotecan for the treatment of patients with advanced solid tumors: a phase I study. Invest New Drugs 2012; 30:1493-500; PMID:21590367; http://dx.doi.org/ 10.1007/s10637-011-9682-9 [DOI] [PubMed] [Google Scholar]
- 231.Kummar S, Chen A, Ji J, Zhang Y, Reid JM, Ames M, Jia L, Weil M, Speranza G, Murgo AJ, et al.. Phase I study of PARP inhibitor ABT-888 in combination with topotecan in adults with refractory solid tumors and lymphomas. Cancer Res 2011; 71:5626-34; PMID:21795476; http://dx.doi.org/ 10.1158/0008-5472.CAN-11-1227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Kunos C, Deng W, Dawson D, Lea JS, Zanotti KM, Gray HJ, Bender DP, Guaglianone PP, Carter JS, Moore KN. A phase I-II evaluation of veliparib (NSC #737664), topotecan, and filgrastim or pegfilgrastim in the treatment of persistent or recurrent carcinoma of the uterine cervix: an NRG oncology/gynecologic oncology group study. Int J Gynecol Cancer 2015; 25:484-92; PMID:25594147; http://dx.doi.org/ 10.1097/IGC.0000000000000380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Su JM, Thompson P, Adesina A, Li XN, Kilburn L, Onar-Thomas A, Kocak M, Chyla B, McKeegan E, Warren KE, et al.. A phase I trial of veliparib (ABT-888) and temozolomide in children with recurrent CNS tumors: a pediatric brain tumor consortium report. Neuro Oncol 2014; 16:1661-8; PMID:24908656; http://dx.doi.org/ 10.1093/neuonc/nou103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Bedikian AY, Papadopoulos NE, Kim KB, Hwu WJ, Homsi J, Glass MR, Cain S, Rudewicz P, Vernillet L, Hwu P. A phase IB trial of intravenous INO-1001 plus oral temozolomide in subjects with unresectable stage-III or IV melanoma. Cancer Invest 2009; 27:756-63; PMID:19440934; http://dx.doi.org/ 10.1080/07357900802709159 [DOI] [PubMed] [Google Scholar]
- 235.Margison GP, Santibanez Koref MF, Povey AC. Mechanisms of carcinogenicity/chemotherapy by O6-methylguanine. Mutagenesis 2002; 17:483-7; PMID:12435845; http://dx.doi.org/ 10.1093/mutage/17.6.483 [DOI] [PubMed] [Google Scholar]
- 236.Danson SJ, Middleton MR. Temozolomide: a novel oral alkylating agent. Expert Rev Anticancer Ther 2001; 1:13-9; PMID:12113120; http://dx.doi.org/ 10.1586/14737140.1.1.13 [DOI] [PubMed] [Google Scholar]
- 237.Plummer R, Lorigan P, Steven N, Scott L, Middleton MR, Wilson RH, Mulligan E, Curtin N, Wang D, Dewji R, et al.. A phase II study of the potent PARP inhibitor, Rucaparib (PF-01367338, AG014699), with temozolomide in patients with metastatic melanoma demonstrating evidence of chemopotentiation. Cancer Chemother Pharmacol 2013; 71:1191-9; PMID:23423489; http://dx.doi.org/ 10.1007/s00280-013-2113-1 [DOI] [PubMed] [Google Scholar]
- 238.Balmana J, Tung NM, Isakoff SJ, Grana B, Ryan PD, Saura C, Lowe ES, Frewer P, Winer E, Baselga J, et al.. Phase I trial of olaparib in combination with cisplatin for the treatment of patients with advanced breast, ovarian and other solid tumors. Ann Oncol 2014; 25:1656-63; PMID:24827126; http://dx.doi.org/ 10.1093/annonc/mdu187 [DOI] [PubMed] [Google Scholar]
- 239.Vonderheide RH, Burg JM, Mick R, Trosko JA, Li D, Shaik MN, Tolcher AW, Hamid O. Phase I study of the CD40 agonist antibody CP-870,893 combined with carboplatin and paclitaxel in patients with advanced solid tumors. Oncoimmunology 2013; 2:e23033; PMID:23483678; http://dx.doi.org/ 10.4161/onci.23033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Weiss RB, Christian MC. New cisplatin analogues in development. A review. Drugs 1993; 46:360-77; PMID:7693428; http://dx.doi.org/ 10.2165/00003495-199346030-00003 [DOI] [PubMed] [Google Scholar]
- 241.Harrap KR. Preclinical studies identifying carboplatin as a viable cisplatin alternative. Cancer Treat Rev 1985; 12:21-33; PMID:3910219; http://dx.doi.org/ 10.1016/0305-7372(85)90015-5 [DOI] [PubMed] [Google Scholar]
- 242.Lee JM, Hays JL, Annunziata CM, Noonan AM, Minasian L, Zujewski JA, Yu M, Gordon N, Ji J, Sissung TM, et al.. Phase I/Ib study of olaparib and carboplatin in BRCA1 or BRCA2 mutation-associated breast or ovarian cancer with biomarker analyses. J Natl Cancer Inst 2014; 106:dju089; PMID:24842883 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.White D, Kassim A, Bhaskar B, Yi J, Wamstad K, Paton VE. Results from AMBER, a randomized phase 2 study of bevacizumab and bortezomib versus bortezomib in relapsed or refractory multiple myeloma. Cancer 2013; 119:339-47; PMID:22811009; http://dx.doi.org/ 10.1002/cncr.27745 [DOI] [PubMed] [Google Scholar]
- 244.Mansfield AS, Nevala WK, Lieser EA, Leontovich AA, Markovic SN. The immunomodulatory effects of bevacizumab on systemic immunity in patients with metastatic melanoma. Oncoimmunology 2013; 2:e24436; PMID:23762809; http://dx.doi.org/ 10.4161/onci.24436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Foy KC, Miller MJ, Moldovan N, Carson Iii WE, Kaumaya PT. Combined vaccination with HER-2 peptide followed by therapy with VEGF peptide mimics exerts effective anti-tumor and anti-angiogenic effects in vitro and in vivo. Oncoimmunology 2012; 1:1048-60; PMID:23170253; http://dx.doi.org/ 10.4161/onci.20708 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Goss G, Shepherd FA, Laurie S, Gauthier I, Leighl N, Chen E, Feld R, Powers J, Seymour L. A phase I and pharmacokinetic study of daily oral cediranib, an inhibitor of vascular endothelial growth factor tyrosine kinases, in combination with cisplatin and gemcitabine in patients with advanced non-small cell lung cancer: a study of the national cancer institute of canada clinical trials group. Eur J Cancer 2009; 45:782-8; PMID:19091548; http://dx.doi.org/ 10.1016/j.ejca.2008.10.022 [DOI] [PubMed] [Google Scholar]
- 247.Gomez-Rivera F, Santillan-Gomez AA, Younes MN, Kim S, Fooshee D, Zhao M, Jasser SA, Myers JN. The tyrosine kinase inhibitor, AZD2171, inhibits vascular endothelial growth factor receptor signaling and growth of anaplastic thyroid cancer in an orthotopic nude mouse model. Clin Cancer Res 2007; 13:4519-27; PMID:17671138; http://dx.doi.org/ 10.1158/1078-0432.CCR-06-2636 [DOI] [PubMed] [Google Scholar]
- 248.Wedge SR, Kendrew J, Hennequin LF, Valentine PJ, Barry ST, Brave SR, Smith NR, James NH, Dukes M, Curwen JO, et al.. AZD2171: a highly potent, orally bioavailable, vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor for the treatment of cancer. Cancer Res 2005; 65:4389-400; PMID:15899831; http://dx.doi.org/ 10.1158/0008-5472.CAN-04-4409 [DOI] [PubMed] [Google Scholar]
- 249.Dean E, Middleton MR, Pwint T, Swaisland H, Carmichael J, Goodege-Kunwar P, Ranson M. Phase I study to assess the safety and tolerability of olaparib in combination with bevacizumab in patients with advanced solid tumours. Br J Cancer 2012; 106:468-74; PMID:22223088; http://dx.doi.org/ 10.1038/bjc.2011.555 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Liu JF, Tolaney SM, Birrer M, Fleming GF, Buss MK, Dahlberg SE, Lee H, Whalen C, Tyburski K, Winer E, et al.. A Phase 1 trial of the poly(ADP-ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti-angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple-negative breast cancer. Eur J Cancer 2013; 49:2972-8; PMID:23810467; http://dx.doi.org/ 10.1016/j.ejca.2013.05.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Liu JF, Barry WT, Birrer M, Lee JM, Buckanovich RJ, Fleming GF, Rimel B, Buss MK, Nattam S, Hurteau J, et al.. Combination cediranib and olaparib versus olaparib alone for women with recurrent platinum-sensitive ovarian cancer: a randomised phase 2 study. Lancet Oncol 2014; 15:1207-14; PMID:25218906; http://dx.doi.org/ 10.1016/S1470-2045(14)70391-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Hoffmann J, Vitale I, Buchmann B, Galluzzi L, Schwede W, Senovilla L, Skuballa W, Vivet S, Lichtner RB, Vicencio JM, et al.. Improved cellular pharmacokinetics and pharmacodynamics underlie the wide anticancer activity of sagopilone. Cancer Res 2008; 68:5301-8; PMID:18593931; http://dx.doi.org/ 10.1158/0008-5472.CAN-08-0237 [DOI] [PubMed] [Google Scholar]
- 253.Domenech E, Malumbres M. Mitosis-targeting therapies: a troubleshooting guide. Curr Opin Pharmacol 2013; 13:519-28; PMID:23583638; http://dx.doi.org/ 10.1016/j.coph.2013.03.011 [DOI] [PubMed] [Google Scholar]
- 254.Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer 2004; 4:253-65; PMID:15057285; http://dx.doi.org/ 10.1038/nrc1317 [DOI] [PubMed] [Google Scholar]
- 255.Dent RA, Lindeman GJ, Clemons M, Wildiers H, Chan A, McCarthy NJ, Singer CF, Lowe ES, Watkins CL, Carmichael J. Phase I trial of the oral PARP inhibitor olaparib in combination with paclitaxel for first- or second-line treatment of patients with metastatic triple-negative breast cancer. Breast Cancer Res 2013; 15:R88; PMID:24063698; http://dx.doi.org/ 10.1186/bcr3484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Oza AM, Cibula D, Benzaquen AO, Poole C, Mathijssen RH, Sonke GS, Colombo N, Spacek J, Vuylsteke P, Hirte H, et al.. Olaparib combined with chemotherapy for recurrent platinum-sensitive ovarian cancer: a randomised phase 2 trial. Lancet Oncol 2015; 16:87-97; PMID:25481791; http://dx.doi.org/ 10.1016/S1470-2045(14)71135-0 [DOI] [PubMed] [Google Scholar]
- 257.Ziccheddu G, Proietti E, Moschella F. The Janus face of cyclophosphamide: a sterile inflammatory response that potentiates cancer immunotherapy. Oncoimmunology 2013; 2:e25789; PMID:24244905; http://dx.doi.org/ 10.4161/onci.25789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Walter S, Weinschenk T, Reinhardt C, Singh-Jasuja H. Single-dose cyclophosphamide synergizes with immune responses to the renal cell cancer vaccine IMA901. Oncoimmunology 2013; 2:e22246; PMID:23482454; http://dx.doi.org/ 10.4161/onci.22246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Chen X, Wakefield LM, Oppenheim JJ. Synergistic antitumor effects of a TGFbeta inhibitor and cyclophosphamide. Oncoimmunology 2014; 3:e28247; PMID:25050195; http://dx.doi.org/ 10.4161/onci.28247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Hussain M, Carducci MA, Slovin S, Cetnar J, Qian J, McKeegan EM, Refici-Buhr M, Chyla B, Shepherd SP, Giranda VL, et al.. Targeting DNA repair with combination veliparib (ABT-888) and temozolomide in patients with metastatic castration-resistant prostate cancer. Invest New Drugs 2014; 32:904-12; PMID:24764124; http://dx.doi.org/ 10.1007/s10637-014-0099-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Kummar S, Ji J, Morgan R, Lenz HJ, Puhalla SL, Belani CP, Gandara DR, Allen D, Kiesel B, Beumer JH, et al.. A phase I study of veliparib in combination with metronomic cyclophosphamide in adults with refractory solid tumors and lymphomas. Clin Cancer Res 2012; 18:1726-34; PMID:22307137; http://dx.doi.org/ 10.1158/1078-0432.CCR-11-2821 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Reiss KA, Herman JM, Zahurak M, Brade A, Dawson LA, Scardina A, Joffe C, Petito E, Hacker-Prietz A, Kinders RJ, et al.. A phase I study of veliparib (ABT-888) in combination with low-dose fractionated whole abdominal radiation therapy in patients with advanced solid malignancies and peritoneal carcinomatosis. Clin Cancer Res 2015; 21:68-76; PMID:25355929; http://dx.doi.org/ 10.1158/1078-0432.CCR-14-1552 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Mehta MP, Wang D, Wang F, Kleinberg L, Brade A, Robins HI, Turaka A, Leahy T, Medina D, Xiong H, et al.. Veliparib in combination with whole brain radiation therapy in patients with brain metastases: results of a phase 1 study. J Neurooncol 2015; 122:409-17; PMID:25682091; http://dx.doi.org/ 10.1007/s11060-015-1733-1 [DOI] [PubMed] [Google Scholar]
- 264.Plummer R, Stephens P, Aissat-Daudigny L, Cambois A, Moachon G, Brown PD, Campone M. Phase 1 dose-escalation study of the PARP inhibitor CEP-9722 as monotherapy or in combination with temozolomide in patients with solid tumors. Cancer Chemother Pharmacol 2014; 74:257-65; PMID:24880570; http://dx.doi.org/ 10.1007/s00280-014-2486-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Plummer R, Jones C, Middleton M, Wilson R, Evans J, Olsen A, Curtin N, Boddy A, McHugh P, Newell D, et al.. Phase I study of the poly(ADP-ribose) polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced solid tumors. Clin Cancer Res 2008; 14:7917-23; PMID:19047122; http://dx.doi.org/ 10.1158/1078-0432.CCR-08-1223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Kummar S, Oza AM, Fleming GF, Sullivan DM, Gandara DR, Naughton MJ, Villalona-Calero MA, Morgan RJ Jr., Szabo PM, Youn A, et al.. Randomized trial of oral cyclophosphamide and veliparib in high-grade serous ovarian, primary peritoneal, or fallopian tube cancers, or BRCA-mutant ovarian cancer. Clin Cancer Res 2015; 21:1574-82; PMID:25589624; http://dx.doi.org/ 10.1158/1078-0432.CCR-14-2565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Walko CM, Lindley C. Capecitabine: a review. Clin Ther 2005; 27:23-44; PMID:15763604; http://dx.doi.org/ 10.1016/j.clinthera.2005.01.005 [DOI] [PubMed] [Google Scholar]
- 268.Grossi F, Aita M, Defferrari C, Rosetti F, Brianti A, Fasola G, Vinante O, Pronzato P, Pappagallo G. Impact of third-generation drugs on the activity of first-line chemotherapy in advanced non-small cell lung cancer: a meta-analytical approach. Oncologist 2009; 14:497-510; PMID:19423674; http://dx.doi.org/ 10.1634/theoncologist.2008-0260 [DOI] [PubMed] [Google Scholar]
- 269.Himes RH. Interactions of the catharanthus (Vinca) alkaloids with tubulin and microtubules. Pharmacol Ther 1991; 51:257-67; PMID:1784631; http://dx.doi.org/ 10.1016/0163-7258(91)90081-V [DOI] [PubMed] [Google Scholar]
- 270.Roche H, Blum J, Eiermann W, Im YH, Martin M, Mina L, Rugo H, Visco F, Zhang C, Lokker N, et al.. P1.01A phase 3 study of the oral PARP inhibitor talazoparib (BMN 673) in BRCA mutation subjects with advanced breast cancer (EMBRACA). Ann Oncol 2015; 26 2:ii16 [Google Scholar]
- 271.Goss PE, Ingle JN, Pater JL, Martino S, Robert NJ, Muss HB, Piccart MJ, Castiglione M, Shepherd LE, Pritchard KI, et al.. Late extended adjuvant treatment with letrozole improves outcome in women with early-stage breast cancer who complete 5 years of tamoxifen. J Clin Oncol 2008; 26:1948-55; PMID:18332475; http://dx.doi.org/ 10.1200/JCO.2007.11.6798 [DOI] [PubMed] [Google Scholar]
- 272.Forbes JF, Cuzick J, Buzdar A, Howell A, Tobias JS, Baum M. Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 100-month analysis of the ATAC trial. Lancet Oncol 2008; 9:45-53; PMID:18083636; http://dx.doi.org/ 10.1016/S1470-2045(07)70385-6 [DOI] [PubMed] [Google Scholar]
- 273.Howell A, Cuzick J, Baum M, Buzdar A, Dowsett M, Forbes JF, Hoctin-Boes G, Houghton J, Locker GY, Tobias JS. Results of the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial after completion of 5 years' adjuvant treatment for breast cancer. Lancet 2005; 365:60-2; PMID:15639680; http://dx.doi.org/ 10.1016/S0140-6736(05)74803-0 [DOI] [PubMed] [Google Scholar]
- 274.Davies C, Godwin J, Gray R, Clarke M, Cutter D, Darby S, McGale P, Pan HC, Taylor C, Wang YC, et al.. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet 2011; 378:771-84; PMID:21802721; http://dx.doi.org/ 10.1016/S0140-6736(11)60993-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Kim DJ, Kim J, Spaunhurst K, Montoya J, Khodosh R, Chandra K, Fu T, Gilliam A, Molgo M, Beachy PA, et al.. Open-label, exploratory phase II trial of oral itraconazole for the treatment of basal cell carcinoma. J Clin Oncol 2014; 32:745-51; PMID:24493717; http://dx.doi.org/ 10.1200/JCO.2013.49.9525 [DOI] [PubMed] [Google Scholar]
- 276.Rudin CM, Brahmer JR, Juergens RA, Hann CL, Ettinger DS, Sebree R, Smith R, Aftab BT, Huang P, Liu JO. Phase 2 study of pemetrexed and itraconazole as second-line therapy for metastatic nonsquamous non-small-cell lung cancer. J Thorac Oncol 2013; 8:619-23; PMID:23546045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Kim J, Tang JY, Gong R, Kim J, Lee JJ, Clemons KV, Chong CR, Chang KS, Fereshteh M, Gardner D, et al.. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell 2010; 17:388-99; PMID:20385363; http://dx.doi.org/ 10.1016/j.ccr.2010.02.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Niemi M, Backman JT, Fromm MF, Neuvonen PJ, Kivisto KT. Pharmacokinetic interactions with rifampicin : clinical relevance. Clin Pharmacokinet 2003; 42:819-50; PMID:12882588; http://dx.doi.org/ 10.2165/00003088-200342090-00003 [DOI] [PubMed] [Google Scholar]
- 279.Liao H, Huang Y, Guo B, Liang B, Liu X, Ou H, Jiang C, Li X, Yang D. Dramatic antitumor effects of the dual mTORC1 and mTORC2 inhibitor AZD2014 in hepatocellular carcinoma. Am J Cancer Res 2015; 5:125-39; PMID:25628925 [PMC free article] [PubMed] [Google Scholar]
- 280.Addie M, Ballard P, Buttar D, Crafter C, Currie G, Davies BR, Debreczeni J, Dry H, Dudley P, Greenwood R, et al.. Discovery of 4-amino-N-; [(1S)-1-(4-chlorophenyl)-3-hydroxypropyl]-1-(7H-pyrrolo; [2,3-d]pyrimidin -4-yl)piperidine-4-carboxamide (AZD5363), an orally bioavailable, potent inhibitor of Akt kinases. J Med Chem 2013; 56:2059-73; PMID:23394218; http://dx.doi.org/ 10.1021/jm301762v [DOI] [PubMed] [Google Scholar]
- 281.Pike KG, Malagu K, Hummersone MG, Menear KA, Duggan HM, Gomez S, Martin NM, Ruston L, Pass SL, Pass M. Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: the discovery of AZD8055 and AZD2014. Bioorg Med Chem Lett 2013; 23:1212-6; PMID:23375793; http://dx.doi.org/ 10.1016/j.bmcl.2013.01.019 [DOI] [PubMed] [Google Scholar]
- 282.Davies BR, Greenwood H, Dudley P, Crafter C, Yu DH, Zhang J, Li J, Gao B, Ji Q, Maynard J, et al.. Preclinical pharmacology of AZD5363, an inhibitor of AKT: pharmacodynamics, antitumor activity, and correlation of monotherapy activity with genetic background. Mol Cancer Ther 2012; 11:873-87; PMID:22294718; http://dx.doi.org/ 10.1158/1535-7163.MCT-11-0824-T [DOI] [PubMed] [Google Scholar]
- 283.Burger MT, Pecchi S, Wagman A, Ni ZJ, Knapp M, Hendrickson T, Atallah G, Pfister K, Zhang Y, Bartulis S, et al.. Identification of NVP-BKM120 as a potent, selective, orally bioavailable class I PI3 kinase inhibitor for treating cancer. ACS Med Chem Lett 2011; 2:774-9; PMID:24900266; http://dx.doi.org/ 10.1021/ml200156t [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Reid AH, Attard G, Barrie E, de Bono JS. CYP17 inhibition as a hormonal strategy for prostate cancer. Nat Clin Pract Urol 2008; 5:610-20; PMID:18985049; http://dx.doi.org/ 10.1038/ncpuro1237 [DOI] [PubMed] [Google Scholar]
- 285.Raab MS, Podar K, Breitkreutz I, Richardson PG, Anderson KC. Multiple myeloma. Lancet 2009; 374:324-39; PMID:19541364; http://dx.doi.org/ 10.1016/S0140-6736(09)60221-X [DOI] [PubMed] [Google Scholar]
- 286.Ranney HM, Gellhorn A. The effect of massive prednisone and prednisolone therapy on acute leukemia and malignant lymphomas. Am J Med 1957; 22:405-13; PMID:13402792; http://dx.doi.org/ 10.1016/0002-9343(57)90096-7 [DOI] [PubMed] [Google Scholar]
- 287.Carter NJ, Keam SJ. Degarelix: a review of its use in patients with prostate cancer. Drugs 2014; 74:699-712; PMID:24756432; http://dx.doi.org/ 10.1007/s40265-014-0211-y [DOI] [PubMed] [Google Scholar]
- 288.Steinberg M. Degarelix: a gonadotropin-releasing hormone antagonist for the management of prostate cancer. Clin Ther 2009; 31 2:2312-31; PMID:20110043; http://dx.doi.org/ 10.1016/j.clinthera.2009.11.009 [DOI] [PubMed] [Google Scholar]
- 289.Power DG, Kemeny NE. The role of floxuridine in metastatic liver disease. Mol Cancer Ther 2009; 8:1015-25; PMID:19383854; http://dx.doi.org/ 10.1158/1535-7163.MCT-08-0709 [DOI] [PubMed] [Google Scholar]
- 290.Twelves C, Wong A, Nowacki MP, Abt M, Burris H 3rd, Carrato A, Cassidy J, Cervantes A, Fagerberg J, Georgoulias V, et al.. Capecitabine as adjuvant treatment for stage III colon cancer. N Engl J Med 2005; 352:2696-704; PMID:15987918; http://dx.doi.org/ 10.1056/NEJMoa043116 [DOI] [PubMed] [Google Scholar]
- 291.Meyerhardt JA, Mayer RJ. Systemic therapy for colorectal cancer. N Engl J Med 2005; 352:476-87; PMID:15689586; http://dx.doi.org/ 10.1056/NEJMra040958 [DOI] [PubMed] [Google Scholar]
- 292.Cersosimo RJ. Management of advanced colorectal cancer, Part 2. Am J Health Syst Pharm 2013; 70:491-506; PMID:23456402; http://dx.doi.org/ 10.2146/ajhp110532b [DOI] [PubMed] [Google Scholar]
- 293.Cersosimo RJ. Management of advanced colorectal cancer, Part 1. Am J Health Syst Pharm 2013; 70:395-406; PMID:23413162; http://dx.doi.org/ 10.2146/ajhp110532 [DOI] [PubMed] [Google Scholar]
- 294.Culy CR, Clemett D, Wiseman LR. Oxaliplatin. a review of its pharmacological properties and clinical efficacy in metastatic colorectal cancer and its potential in other malignancies. Drugs 2000; 60:895-924; PMID:11085200; http://dx.doi.org/ 10.2165/00003495-200060040-00005 [DOI] [PubMed] [Google Scholar]
- 295.Raymond E, Faivre S, Chaney S, Woynarowski J, Cvitkovic E. Cellular and molecular pharmacology of oxaliplatin. Mol Cancer Ther 2002; 1:227-35; PMID:12467217 [PubMed] [Google Scholar]
- 296.Bruserud O, Reikvam H, Kittang AO, Ahmed AB, Tvedt TH, Sjo M, Hatfield KJ. High-dose etoposide in allogeneic stem cell transplantation. Cancer Chemother Pharmacol 2012; 70:765-82; PMID:23053272; http://dx.doi.org/ 10.1007/s00280-012-1990-z [DOI] [PubMed] [Google Scholar]
- 297.Montecucco A, Biamonti G. Cellular response to etoposide treatment. Cancer Lett 2007; 252:9-18; PMID:17166655; http://dx.doi.org/ 10.1016/j.canlet.2006.11.005 [DOI] [PubMed] [Google Scholar]
- 298.Josephs DH, Fisher DS, Spicer J, Flanagan RJ. Clinical pharmacokinetics of tyrosine kinase inhibitors: implications for therapeutic drug monitoring. Ther Drug Monit 2013; 35:562-87; PMID:24052062 [DOI] [PubMed] [Google Scholar]
- 299.Johnston S, Pippen J Jr., Pivot X, Lichinitser M, Sadeghi S, Dieras V, Gomez HL, Romieu G, Manikhas A, Kennedy MJ, et al.. Lapatinib combined with letrozole versus letrozole and placebo as first-line therapy for postmenopausal hormone receptor-positive metastatic breast cancer. J Clin Oncol 2009; 27:5538-46; PMID:19786658; http://dx.doi.org/ 10.1200/JCO.2009.23.3734 [DOI] [PubMed] [Google Scholar]
- 300.Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, et al.. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med 2006; 355:2733-43; PMID:17192538; http://dx.doi.org/ 10.1056/NEJMoa064320 [DOI] [PubMed] [Google Scholar]
- 301.Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, Untch M, Rusnak DW, Spehar G, Mullin RJ, et al.. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res 2006; 66:1630-9; PMID:16452222; http://dx.doi.org/ 10.1158/0008-5472.CAN-05-1182 [DOI] [PubMed] [Google Scholar]
- 302.Montoni A, Robu M, Pouliot E, Shah GM. Resistance to PARP-Inhibitors in Cancer Therapy. Front Pharmacol 2013; 4:18; PMID:23450678; http://dx.doi.org/ 10.3389/fphar.2013.00018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Barber LJ, Sandhu S, Chen L, Campbell J, Kozarewa I, Fenwick K, Assiotis I, Rodrigues DN, Reis Filho JS, Moreno V, et al.. Secondary mutations in BRCA2 associated with clinical resistance to a PARP inhibitor. J Pathol 2013; 229:422-9; PMID:23165508; http://dx.doi.org/ 10.1002/path.4140 [DOI] [PubMed] [Google Scholar]
- 304.Edwards SL, Brough R, Lord CJ, Natrajan R, Vatcheva R, Levine DA, Boyd J, Reis-Filho JS, Ashworth A. Resistance to therapy caused by intragenic deletion in BRCA2. Nature 2008; 451:1111-5; PMID:18264088; http://dx.doi.org/ 10.1038/nature06548 [DOI] [PubMed] [Google Scholar]
- 305.Jaspers JE, Kersbergen A, Boon U, Sol W, van Deemter L, Zander SA, Drost R, Wientjens E, Ji J, Aly A, et al.. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov 2013; 3:68-81; PMID:23103855; http://dx.doi.org/ 10.1158/2159-8290.CD-12-0049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Bunting SF, Callen E, Wong N, Chen HT, Polato F, Gunn A, Bothmer A, Feldhahn N, Fernandez-Capetillo O, Cao L, et al.. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell 2010; 141:243-54; PMID:20362325; http://dx.doi.org/ 10.1016/j.cell.2010.03.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Xu G, Chapman JR, Brandsma I, Yuan J, Mistrik M, Bouwman P, Bartkova J, Gogola E, Warmerdam D, Barazas M, et al.. REV7 counteracts DNA double-strand break resection and affects PARP inhibition. Nature 2015; PMID:25799992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.Rottenberg S, Jaspers JE, Kersbergen A, van der Burg E, Nygren AO, Zander SA, Derksen PW, de Bruin M, Zevenhoven J, Lau A, et al.. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. Proc Natl Acad Sci U S A 2008; 105:17079-84; PMID:18971340; http://dx.doi.org/ 10.1073/pnas.0806092105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.Leung AK, Vyas S, Rood JE, Bhutkar A, Sharp PA, Chang P. Poly(ADP-ribose) regulates stress responses and microRNA activity in the cytoplasm. Mol Cell 2011; 42:489-99; PMID:21596313; http://dx.doi.org/ 10.1016/j.molcel.2011.04.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Green DR, Galluzzi L, Kroemer G. Cell biology. Metabolic control of cell death. Science 2014; 345:1250256; PMID:25237106; http://dx.doi.org/ 10.1126/science.1250256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Kanai M, Tong WM, Sugihara E, Wang ZQ, Fukasawa K, Miwa M. Involvement of poly(ADP-Ribose) polymerase 1 and poly(ADP-Ribosyl)ation in regulation of centrosome function. Mol Cell Biol 2003; 23:2451-62; PMID:12640128; http://dx.doi.org/ 10.1128/MCB.23.7.2451-2462.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Kanai M, Tong WM, Wang ZQ, Miwa M. Haploinsufficiency of poly(ADP-ribose) polymerase-1-mediated poly(ADP-ribosyl)ation for centrosome duplication. Biochem Biophys Res Commun 2007; 359:426-30; PMID:17553458; http://dx.doi.org/ 10.1016/j.bbrc.2007.05.108 [DOI] [PubMed] [Google Scholar]
- 313.Simbulan-Rosenthal CM, Ly DH, Rosenthal DS, Konopka G, Luo R, Wang ZQ, Schultz PG, Smulson ME. Misregulation of gene expression in primary fibroblasts lacking poly(ADP-ribose) polymerase. Proc Natl Acad Sci U S A 2000; 97:11274-9; PMID:11016956; http://dx.doi.org/ 10.1073/pnas.200285797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Halappanavar SS, Shah GM. Defective control of mitotic and post-mitotic checkpoints in poly(ADP-ribose) polymerase-1(−/−) fibroblasts after mitotic spindle disruption. Cell Cycle 2004; 3:335-42; PMID:14726664; http://dx.doi.org/ 10.4161/cc.3.3.670 [DOI] [PubMed] [Google Scholar]
- 315.Inbar D, Cohen-Armon M, Neumann D. Erythropoietin-driven signalling and cell migration mediated by polyADP-ribosylation. Br J Cancer 2012; 107:1317-26; PMID:22955851; http://dx.doi.org/ 10.1038/bjc.2012.395 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Caldini R, Fanti E, Magnelli L, Barletta E, Tanganelli E, Zampieri M, Chevanne M. Low doses of 3-aminobenzamide, a poly(ADP-ribose) polymerase inhibitor, stimulate angiogenesis by regulating expression of urokinase type plasminogen activator and matrix metalloprotease 2. Vasc Cell 2011; 3:12; PMID:21595892; http://dx.doi.org/ 10.1186/2045-824X-3-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Lacal PM, Tentori L, Muzi A, Ruffini F, Dorio AS, Xu W, Arcelli D, Zhang J, Graziani G. Pharmacological inhibition of poly(ADP-ribose) polymerase activity down-regulates the expression of syndecan-4 and Id-1 in endothelial cells. Int J Oncol 2009; 34:861-72; PMID:19212692 [DOI] [PubMed] [Google Scholar]
- 318.Tentori L, Lacal PM, Muzi A, Dorio AS, Leonetti C, Scarsella M, Ruffini F, Xu W, Min W, Stoppacciaro A, et al.. Poly(ADP-ribose) polymerase (PARP) inhibition or PARP-1 gene deletion reduces angiogenesis. Eur J Cancer 2007; 43:2124-33; PMID:17714938; http://dx.doi.org/ 10.1016/j.ejca.2007.07.010 [DOI] [PubMed] [Google Scholar]
- 319.Pyriochou A, Olah G, Deitch EA, Szabo C, Papapetropoulos A. Inhibition of angiogenesis by the poly(ADP-ribose) polymerase inhibitor PJ-34. Int J Mol Med 2008; 22:113-8; PMID:18575783 [PubMed] [Google Scholar]
- 320.Rajesh M, Mukhopadhyay P, Godlewski G, Batkai S, Hasko G, Liaudet L, Pacher P. Poly(ADP-ribose)polymerase inhibition decreases angiogenesis. Biochem Biophys Res Commun 2006; 350:1056-62; PMID:17046715; http://dx.doi.org/ 10.1016/j.bbrc.2006.09.160 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Li Q, Li M, Wang YL, Fauzee NJ, Yang Y, Pan J, Yang L, Lazar A. RNA interference of PARG could inhibit the metastatic potency of colon carcinoma cells via PI3-kinase/Akt pathway. Cell Physiol Biochem 2012; 29:361-72; PMID:22508044; http://dx.doi.org/ 10.1159/000338491 [DOI] [PubMed] [Google Scholar]
- 322.Riffell JL, Lord CJ, Ashworth A. Tankyrase-targeted therapeutics: expanding opportunities in the PARP family. Nat Rev Drug Discov 2012; 11:923-36; PMID:23197039; http://dx.doi.org/ 10.1038/nrd3868 [DOI] [PubMed] [Google Scholar]
- 323.Stilmann M, Hinz M, Arslan SC, Zimmer A, Schreiber V, Scheidereit C. A nuclear poly(ADP-ribose)-dependent signalosome confers DNA damage-induced IkappaB kinase activation. Mol Cell 2009; 36:365-78; PMID:19917246; http://dx.doi.org/ 10.1016/j.molcel.2009.09.032 [DOI] [PubMed] [Google Scholar]
- 324.Cohen-Armon M, Visochek L, Rozensal D, Kalal A, Geistrikh I, Klein R, Bendetz-Nezer S, Yao Z, Seger R. DNA-independent PARP-1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation. Mol Cell 2007; 25:297-308; PMID:17244536; http://dx.doi.org/ 10.1016/j.molcel.2006.12.012 [DOI] [PubMed] [Google Scholar]
- 325.Kauppinen TM, Chan WY, Suh SW, Wiggins AK, Huang EJ, Swanson RA. Direct phosphorylation and regulation of poly(ADP-ribose) polymerase-1 by extracellular signal-regulated kinases 1/2. Proc Natl Acad Sci U S A 2006; 103:7136-41; PMID:16627622; http://dx.doi.org/ 10.1073/pnas.0508606103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326.Shimo T, Kurebayashi J, Kanomata N, Yamashita T, Kozuka Y, Moriya T, Sonoo H. Antitumor and anticancer stem cell activity of a poly ADP-ribose polymerase inhibitor olaparib in breast cancer cells. Breast Cancer 2014; 21:75-85; PMID:22454224; http://dx.doi.org/ 10.1007/s12282-012-0356-z [DOI] [PubMed] [Google Scholar]
- 327.Jwa M, Chang P. PARP16 is a tail-anchored endoplasmic reticulum protein required for the PERK- and IRE1alpha-mediated unfolded protein response. Nat Cell Biol 2012; 14:1223-30; PMID:23103912; http://dx.doi.org/ 10.1038/ncb2593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.El-Hamoly T, Hegedus C, Lakatos P, Kovacs K, Bai P, El-Ghazaly MA, El-Denshary ES, Szabo E, Virag L. Activation of poly(ADP-ribose) polymerase-1 delays wound healing by regulating keratinocyte migration and production of inflammatory mediators. Mol Med 2014; 20:363-71; PMID:25014793; http://dx.doi.org/ 10.2119/molmed.2014.00130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329.Bendell J, O'Reilly EM, Middleton MR, Chau I, Hochster H, Fielding A, Burke W, Burris H 3rd. Phase I study of olaparib plus gemcitabine in patients with advanced solid tumours and comparison with gemcitabine alone in patients with locally advanced/metastatic pancreatic cancer. Ann Oncol 2015; 26:804-11; PMID:25573533; http://dx.doi.org/ 10.1093/annonc/mdu581 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Kummar S, Kinders R, Gutierrez ME, Rubinstein L, Parchment RE, Phillips LR, Ji J, Monks A, Low JA, Chen A, et al.. Phase 0 clinical trial of the poly (ADP-ribose) polymerase inhibitor ABT-888 in patients with advanced malignancies. J Clin Oncol 2009; 27:2705-11; PMID:19364967; http://dx.doi.org/ 10.1200/JCO.2008.19.7681 [DOI] [PMC free article] [PubMed] [Google Scholar]