Abstract
Impaired DNA damage response pathways may create vulnerabilities of cancer cells that can be exploited therapeutically. One such selective vulnerability is the sensitivity of BRCA1- or BRCA2-defective tumors (hence defective in DNA repair by homologous recombination, HR) to inhibitors of the poly(ADP-ribose) polymerase-1 (PARP-1), an enzyme critical for repair pathways alternative to HR. While promising, treatment with PARP-1 inhibitors (PARP-1i) faces some hurdles, including (1) acquired resistance, (2) search for other sensitizing, non-BRCA1/2 cancer defects and (3) lack of biomarkers to predict response to PARP-1i. Here we addressed these issues using PARP-1i on 20 human cell lines from carcinomas of the breast, prostate, colon, pancreas and ovary. Aberrations of the Mre11-Rad50-Nbs1 (MRN) complex sensitized cancer cells to PARP-1i, while p53 status was less predictive, even in response to PARP-1i combinations with camptothecin or ionizing radiation. Furthermore, monitoring PARsylation and Rad51 foci formation as surrogate markers for PARP activity and HR, respectively, supported their candidacy for biomarkers of PARP-1i responses. As to resistance mechanisms, we confirmed the role of the multidrug resistance efflux transporters and its reversibility. More importantly, we demonstrated that shRNA lentivirus-mediated depletion of 53BP1 in human BRCA1-mutant breast cancer cells increased their resistance to PARP-1i. Given the preferential loss of 53BP1 in BRCA-defective and triple-negative breast carcinomas, our findings warrant assessment of 53BP1 among candidate predictive biomarkers of response to PARPi. Overall, this study helps characterize genetic and functional determinants of cellular responses to PARP-1i and contributes to the search for biomarkers to exploit PARP inhibitors in cancer therapy.
Keywords: 53BP1, BRCA1, DNA damage response, MRN complex, PARP-1 inhibitor, cancer treatment, p53, parsylation, predictive biomarkers, synthetic lethality or viability
Introduction
Aberrations in the DNA damage response (DDR) machinery are common in cancer and represent potential targets for therapeutic intervention.1,2 This is because normal cells possess the full spectrum of DNA damage checkpoints and repair pathways, while in cancer cells only some of these mechanisms are often intact, and targeting such remaining operational DDR pathways may selectively kill cancer cells.2-4 PARP-1 activity is important in sensing and signaling DNA damage that arises both endogenously, for example through generation of oxidative DNA lesions and DNA single-strand breaks (SSBs), or exogenously, such as due to radiation exposure or treatment with cytotoxic chemotherapy.5,6 Continuous exposure of cycling cells to PARP-1 inhibitors results in excessive formation of SSBs which, when encountered by replication forks, may cause replication fork collapse and formation of DNA double-strand breaks (DSBs).7 DNA breaks arising during replication are preferentially repaired by HR, an accurate mechanism that maintains genomic integrity.8 When HR is defective due to mutations or silencing of BRCA1 or BRCA2, cells are extremely sensitive to inhibitors of PARP-dependent alternative repair pathway(s).9,10 Based on this synthetic lethality principle, PARP-1 inhibitors are under clinical evaluation as a promising strategy of tumor-selective mono-therapy for tumors bearing BRCA1/2 mutations.2,11
Apart from its direct role in SSB repair, PARP-1 is involved in modulation of DSB repair pathways by physical association as well as PARsylation of various repair proteins.12,13 DSBs are identified by phosphorylation of the core histone variant H2AX (forming γH2AX) that occurs independently of PARP-1 or PAR.13 On the other hand, the rapid relaxation of chromatin around DSBs can be attributed to local PARsylation mediated by PARP-1, which associates with γH2AX.5 Furthermore, PARP-1 forms a complex with Mre11 and is required for rapid DNA breakage-induced subcellular relocalization of the MRN complex, a critical sensor of DSBs.14 However, accumulation and activation of PARP-1 at DSBs enhances, but is not absolutely required for, the DSB signaling and repair processes such as HR and the less precise non-homologous end joining (NHEJ).15
Inspired by motivation to further develop the treatment strategy with PARP inhibitors, additional DDR-related defects that sensitize cells to PARP-1i have been identified, such as in DNA damage sensors and signaling kinases, nucleotide excision repair or Aurora A kinase. 16-18 These results suggest that the therapeutic potential of PARP inhibitors might extend beyond tumors with defective BRCA1/2 and HR and warrant further investigation. Despite the enthusiasm evoked by the promising studies performed so far, treatment with PARP inhibitors also faces the difficulties and challenges broadly analogous to those encountered by other innovative cancer treatments. First, examples of resistance mechanisms to PARP inhibitors are emerging, and these must be better understood to be overcome.19 Second, as much of the data about the additional, non-BRCA1/2 determinants of sensitivity to PARP-1i are based on studies with a limited number of cancer cell lines,16,20 such candidate DDR defects need to be validated on additional cancer models of different tissue origin and within the context of various genetic backgrounds. Third, there is an urgent need to identify and validate potential biomarkers to predict responses of individual tumors to PARP inhibitors. In this study, we attempted to address some aspects of these challenging issues by analyzing responses of a panel of human cell lines from carcinomas of the breast, prostate, colon, pancreas and ovary, and genetic derivatives of selected models, to the PARP-1 inhibitors KU 58948 and its close derivative olaparib, the latter already under investigation in clinical trials.11
Results
Deficiency in the MRN complex and sensitivity to PARP-1 inhibition
To examine whether PARP-1 inhibition is selectively lethal to various cellular models deficient in components of the DSB-sensing and -processing complex of Mre11, Rad50 and Nbs1, we tested sensitivity of a panel of human cancer cell lines with differential status of this important tumor suppressor complex.3,21 Given that only a subset of carcinoma-derived cell lines can perform robustly in a clonogenic assay, we first established a more universally applicable, shorter-term viability assay to determine sensitivity to PARP-1i. We chose as a positive control the pancreatic cancer cell line CAPAN-1 (harboring the naturally occurring BRCA2 617delT frameshift mutation accompanied by the loss of the second allele)22 and, in parallel, examined an isogenic pair of human SV40-immortalized fibroblasts either deficient in Nbs1, NBS-1LBI or complemented with wt-Nbs1, NBS-1LBI+Nbs1. The CAPAN-1 and NBS-1LBI cell lines have been reported to show profound sensitivity to PARP-1i due to their defect in BRCA2 and Nbs1, respectively.16,23 After 4 d of exponential growth (see Methods for details), viable cells were counted and the viability expressed as a cell number normalized to untreated control (Fig. 1). These initial experiments confirmed differences in responses of Nbs1-deficient vs. Nbs1-proficient human fibroblasts and pronounced sensitivity of the BRCA2-deficient CAPAN-1 cells to PARP-1i treatment, at the same time providing support for our assay as an informative approach to monitor the impact of PARP-1 inhibition on cellular viability.
Figure 1. Human Nbs1-deficient fibroblasts NBS-1LBI and pancreatic cancer cell line CAPAN-1 are highly sensitive to KU 58948. (A) Cell survival curve of CAPAN-1 cells upon PARP-1i treatment. After 4 d (see Methods), cell numbers were counted and expressed as percentage of untreated control cells. Results are means ± s.d. (B) Western blot analysis of Mre11-Rad50-Nbs1 complex proteins shows deficiency of Nbs1 in CAPAN-1 cells; γ-tubulin was used as a loading control. (C) Proliferation curves of Nbs1-deficient NBS-1LBI and isogenic Nbs1-wt complemented NBS-1LBI+Nbs1 human fibroblasts in response to PARP-1i. Results are means ± s.d. (D) Western blot analysis of MRN complex proteins showing selective Nbs1 protein defect in NBS-1LBI and re-expression of Nbs1-wt protein NBS-1LBI+Nbs1; γ-tubulin served as a loading control.
Deficiency in the MRN complex sensitizes breast cancer cells to PARP-1i
Exposure of BRCA1- or BRCA2-depleted cells to PARP-1i reportedly leads to cell cycle arrest, predominantly in G2 phase, followed by an increase of apoptotic cell death (Farmer et al., 2005). To test whether exposure of Cal51 breast cancer cells to PARP-1i induces cell death, rather than cell cycle arrest only, we performed a well- established XTT-based cell proliferation assay coupled with lactate dehydrogenase (LDH) activity measurement. LDH is an intracellular enzyme, which, when released into the media by dying/dead cells, catalyzes the conversion of lactate to pyruvate while reducing NAD+ to NADH/H+. In the second step of this assay, NADH/H+ is used to reduce tetrazolium salt into colorimetrically detectable formazan. As shown in Figure 2A, reduced cell proliferation caused by PARP-1i treatment is accompanied by increased cell death as indicated by the higher activity of LDH.
Figure 2. Deficiency in the MRN complex sensitizes breast cancer cells to PARP-1i. (A) Proliferation activity of Cal51-wt cells measured by XTT test after 4 d treatment with KU 58948 and complemented with the cell death assessment performed by LDH activity measurement from the same cells. Results represent means ± s.d. (B) Survival curves of Cal51 MRN-wt, Nbs1 and Mre11 knockdown cells upon 2-d treatment with KU 58948 followed by 2 d growth in DMEM. (C) Western blot analysis of Cal51 breast cancer cell line expressing wt-MRN complex and stable shRNA mediated knockdown of Nbs1 and Mre11. Detection of SMC1 was used as a loading control.
To further validate the hypothesis that MRN deficiency increases sensitivity to PARP-1i, we compared the Cal51 cells that harbor a functional MRN complex (wt-MRN) with stable Cal51-derived transfectants expressing shRNA against either Mre11 or Nbs1 to knockdown the respective MRN-complex proteins (Fig. 2B and C). The 48-h treatment with PARPi was sufficient to abolish PARsylation spontaneously occurring in these cells (Fig. S1), and the 4 d viability assay showed enhanced toxicity of PARPi toward Cal51 cells upon partial knockdown of Mre11 and Nbs1 proteins, respectively, compared with the parental wt-MRN cells (Fig. 2B). These results extend the previous analyses to defects of different MRN complex components and suggest that even partial depletion of Mre11 or NBS1 translates into enhanced sensitivity of human breast cancer cells to PARP-1 inhibition.
Efflux transporters confer resistance of Mre11-deficient colon cancer cells to single-agent treatment with PARP-1i
Next, we focused on human colon cancer, a tumor type with known MRN defects. Among the five colorectal cancer cell lines we examined for expression of proteins of the MRN complex, the HT29 cell line lacked the Nbs1 protein and showed an aberrantly reduced level of Mre11 complex (Fig. 3B). The cell line HCT116, from which p53-proficient and p53-deleted variants are available, harbors a mixture of wild-type Mre11 and a splice variant that leads to a partial deletion of sequence in the N-terminal nuclease domain. This mutant allele of Mre11 preserves some functions of the wild-type protein and suppresses other functions of the MRN complex in a dominant-negative manner.24,25 Our immunoblotting analysis of total lysates from HCT116 cells revealed detectable but significantly reduced levels of components of the MRN complex (Fig. 3B), suggesting destabilization of the MRN complex by the Mre11 mutation in this cell line. Exposure of colon cancer cells to PARP-1i revealed mild sensitivity of the p53-proficient version of HCT116 cells, while the p53-deleted HCT116 cells were more resistant, similar to the MRN-proficient colon cancer cell line SW620 (Fig. 3A). Ectopic expression of Mre11 in HCT116 cells resulted in increased levels of endogenous Nbs1 and Rad50 proteins (Fig. 3D), and such reconstitution led to a shift in the PARP-1i response curve (Fig. 3C).
Figure 3. MRN defect does not cause marked sensitivity of colon cancer cells to PARP-1 inhibitor in a short-term assay. (A) Proliferation curves of MRN-proficient (SW620) colon cancer cell line or deficient in Nbs1 (HT29) and Mre11 proteins (HCT116 p53-wt, HCT116 p53-deleted), respectively. NBS-1LBI was used as a sensitive cell line to compare the response of colon cancer cells. Cells were exposed to a range of concentrations of PARP-1 inhibitor for 2 d, split and grown for additional 2 d in DMEM only. Proliferation activity was determined by cell counting and is expressed as a percentage of control cells. Results represent means ± s.d. (B) MRN complex protein expression is normal in osteosarcoma U2OS cells and SW620, SW480 and HCT15 colon cancer cell lines while HCT116 colon cancer cells show low levels of endogenous wild type Mre11 and expresses a shorter dominant-negative splice variant of Mre11 (Wen et al., 2008) with a strong impact on total abundance of the MRN complex. HT29 cells show deficiencies of Nbs1 and Mre11 proteins. (C) Survival curves of MRN deficient and Mre11-wt reconstituted HCT116 (p53-wt) cells upon the standard treatment with PARP-1 inhibitor. Results represent means ± s.d. (D) MRN protein expression in wild-type SW620 cells and HCT116 (p53-wt) cells with/ without Mre11-wt transfection. The same membrane was also blotted for SMC1 as a protein loading control.
We speculated that the inefficient response of MRN-deficient colon cancer cells to PARP-1i could be related to multidrug resistance, a frequent feature of colorectal cancer, as P-glycoprotein (P-gp) overexpression was associated with PARP-1i resistance.26-28 To address this possibility, we treated the P-gp efflux pump expressing HCT116 cells with PARP-1i and co-treated them or not with the P-gp inhibitor Verapamil. Monitoring the intracellular levels of PARP-1i directly by mass spectrometry showed (Fig. S3) that administration of a non-toxic concentration of Verapamil more than doubled the intracellular concentration of PARP-1i, and this correlated with sensitization of the HCT116 cells in our 4-d proliferation test (Fig. 4). We conclude that one mechanism of resistance to PARP-1i treatment in human MRN-deficient colorectal cancer is the enhanced P-gp-mediated drug efflux, and that this type of resistance can potentially be overcome, at least in part, by suitable inhibitors of the multidrug resistance pumps.
Figure 4. Combination of KU 58948 and P-gp inhibitor Verapamil decreases survival of MRN-deficient colon cancer cells. HCT116 cells were treated with KU 58948 in concentrations 10−8 – 10−4 M or pre-treated for 1 h with 5 μg/ ml Verapamil prior to addition of KU 58948. The cells were grown for 4 d and subsequently the cell viability was assessed using the XTT test.
p53 status and response of MRN-deficient cancer cells to PARP-1i
Mutation or loss of p53 can confer resistance of diverse types of cancer cells to multiple classes of drugs.29 Impaired apoptotic and/or checkpoint mechanisms in p53-mutant tumors allow such cancer cells to proliferate even under exposure to genotoxic stress. BRCA2-depleted cells, however, are sensitive to PARP-1i regardless of p53 status.9 Our findings, on the other hand, suggested there might be differential responses of HCT116 colon cancer cells with wild-type vs. deleted p53 to PARP-1i treatment (Fig. 3A). Intrigued by this observation, we further investigated the possibility that p53 aberrations could contribute to the observed resistance of the MRN-deficient colon cancer cells to PARP-1i. First, we noted the p53 status in the 20 cell lines of our panel, tested for sensitivity to PARP-1i (Table S1). To further examine the potential effect of p53 status on response to PARP-1i using another MRN-deficient cancer cell type, we silenced the wt-p53 in the breast cancer cell line Cal51, in which the MRN complex was disabled by a knockdown of Nbs1 through stable expression of shRNA (Nbs1 kd) (Fig. 5A). Consistent with the data obtained for colon cancer cells in the 4-d XTT assay, we observed an increased resistance to PARP-1i in the p53-depleted Cal51-(Nbs1 kd) cells, compared with their Cal51-(Nbs1 kd)/wt-p53 counterparts (Fig. 5B). However, in contrast to the differences observed in the short-term proliferation assay, p53 status did not appear to have an impact on sensitivity of MRN-deficient cells to PARP-1i assessed by the longer-term colony formation assay, at least in the HCT116 colon cancer model (Fig. 5C).

Figure 5. Effect of p53 status on sensitivity of MRN-deficient colon and breast cancer cell lines. (A) Wild-type p53 protein was transiently silenced by RNA interference in Cal51 (Nbs1 knockdown) cell line as shown by western blotting. SMC1 protein was used to verify equal protein loading. (B) Proliferation curves represent sensitivity of Cal51 (Nbs1 knockdown) cell line possessing either wild-type or silenced p53 protein to PARP-1 inhibitor treatment. (C) Evaluation of sensitivity of MRN-deficient HCT116 p53 wt and p53-null and MRN-proficient HCT15 colon cancer cells to PARP-1 inhibitor treatment in 12-d colony-forming assay.
PARP-1i can sensitize cancer cells to Camptothecin or ionizing radiation regardless of the p53 status
Apart from the BRCA1 and BRCA2 defects that strongly sensitize cells to PARP-1i, the other genetic defects that show synthetic sickness effects when combined with PARPi have an overall less pronounced impact. Consequently, tumors with such non-BRCA1/2 aberrations are less suitable for a single-agent treatment with PARPi. Rather, in such clinical scenarios, it could potentially be advantageous to apply PARP-1i in combination treatments to sensitize tumors to standard-of-care chemo- or radiotherapy.
To explore this possibility in a model system, we first examined the impact of PARP-1i combined with the genotoxic drug camptothecin (CPT, a topoisomerase I inhibitor commonly used in clinical oncology) in our model of MRN-deficient colon cancer cells HCT116, either with wt-p53 or deleted for p53, the latter being more resistant to PARP-1i treatment alone (see Fig. 3A). First, we noted that the p53 status affected the response of this cell line to CPT alone, in that the p53-deleted HCT116 cells were clearly more resistant compared with their p53-wild-type counterparts (Fig. 6A). Notably, concomitant treatment with PARP-1i (at a moderate dose that alone showed virtually no effect on proliferation of the p53-deleted HCT116 cells in this assay, Fig. 3A) enhanced the anti-proliferative effects of CPT in either variant of the HCT116 cells, suggesting that PARP-1i sensitizes colorectal carcinoma cells to chemotherapy independently of p53 status (Fig. 6A).
Figure 6. Combined treatment with KU 58948 and camptothecin or irradiation effectively reduces cell survival. (A) Proliferation curves of HCT116 p53-wt and HCT116 p53- deficient cells treated with various concentrations of CPT alone or combined and 1 μM KU 58948. After 2-d exposure to the drugs, cells were split and grown for additional 2 d in DMEM only. Proliferation activity was determined by cell counting and is expressed as a percentage of untreated control cells. Results are means ± s.d. (B) Evaluation of sensitivity of prostate cancer cell lines PC3 and DU145 to KU 58948 alone or in combination with irradiation in a 12-d colony-forming assay. Results are means ± s.d.
Second, we explored a conceptually analogous scenario, testing the impact of added PARP-1i to treatment of two p53-mutant human prostate carcinoma cell lines by ionizing radiation (IR), a modality commonly used to treat prostate cancer. As can be seen from the results of clonogenic assays showed in Figure 6B, both PC3 and DU145 prostate cancer cell lines responded similarly to clinically relevant doses of 2 Gy and 4 Gy of IR alone. Interestingly, addition of a moderate dose of the PARP-1i potentiated the impact of IR on the PC3 cell line, while the response of DU145 cells to IR remained unaffected regardless of PARP-1i treatment (Fig. 6B).
Together, these results suggest that PARP-1i can sensitize subsets of diverse types of common human cancers to CPT and/or ionizing radiation, and that the added PARP-1i in such combined treatment may help eliminate even p53-mutant tumors that are otherwise often more resistant to standard-of-care non-surgical therapies.
Spontaneous PARsylation and Rad51 foci formation as candidate biomarkers of response to PARP-1i
Next, we thought to explore the potential of two functional aspects of the DDR machinery as candidate predictive biomarkers relevant for PARP-1i treatment, namely endogenous PARsylation and focus formation by the HR protein Rad51.
First, as the primary function of PARP-1i is to block the enzymatic activity of PARP-1 and thereby prevent or reduce PAR formation, we argued that cells which do not spontaneously produce detectable PAR without exogenous stimuli are unlikely to be successfully targeted with PARP-1i as a single agent. To this end, we first investigated whether there are any differences in spontaneous PARsylation among the cell lines tested for sensitivity to PARP-1i. Western blot analysis of whole-cell lysates derived from the cells exponentially grown under standard conditions revealed notable variation in endogenous PAR levels among the cell lines of our panel (see examples in Fig. 7B). When comparing the ability of the cells to produce PAR, with response to PARP-1i treatment, we noted a trend for cells with undetectable endogenous PAR to show greater resistance to PARP-1i, as compared with more robust responses of those cancer cell lines producing PAR at detectable levels (Fig. 7A and B). These findings support our working hypothesis of PAR as a potentially useful predictive biomarker, a notion that is further considered in the discussion.
Figure 7. Correlation between spontaneous PARsylation, Rad51 foci formation and sensitivity to PARP-1 inhibitor. (A) Sensitivity of different osteosarcoma, pancreatic, prostate and ovarian cancer cell lines to PARP-1 inhibitor in a short-term (4 d) proliferation assay. (B) Western blot results showing expression of spontaneous PAR in tested cell lines. (C) Rad51 foci induction after 24 h treatment with 1 μM KU 58948 (data represent the average of two independent experiments).
Second, the inability of Rad51 protein to form subnuclear foci in response to DNA damage is regarded as an indication of a functional defect in the HR repair pathway, and therefore a potential surrogate marker that could be useful in predicting responses to PARP-1i. To assess this notion in our panel of human cancer cell lines, we examined the extent of spontaneous Rad51 foci and those formed in response to PARP-1i treatment using immunofluorescence with a well-validated antibody to Rad51.30 The assay was also validated by the fact that neither spontaneous nor PARP-1i-induced Rad51 foci were observed in SUM149 and CAPAN1 cancer cell lines defective in BRCA1 and BRCA2, respectively, and used here as controls expected to be deficient in this assay (Fig. 7C). In contrast to the BRCA1/BRCA2-defective cell lines, however, we could detect sizeable fractions of cells with spontaneous Rad51 foci,\ and an increased formation of such foci after a 24-exposure to PARP-1i, in all other cancer cell lines of our panel examined in this assay (Fig. 7C; Fig. S2 and Table S2). As the cell lines capable of forming Rad51 foci included also several MRN-deficient cell types and other models that showed enhanced sensitivity to PARP-1i (Table S1), our data raise some concerns about the general applicability of this assay in predicting responses to PARP-1i.
Loss of 53BP1 and resistance of BRCA1-deficient breast cancer cells to PARP-1i
Recent evidence suggests that there is a biological selection and enhanced viability and growth among the BRCA-defective human tumors and mouse models, for those cells with aberrantly reduced or lost 53BP1, an important DDR mediator protein that channels DNA DSB lesions preferentially for repair by NHEJ, at the expense of HR.31,32 As the loss of 53BP1 allows at least partial rescue of the recombination repair, including the ability to form Rad51 foci, these results suggested that absence of 53BP1 could highlight a new class of resistance mechanism to PARP-1i in the otherwise exquisitely sensitive BRCA1-defective cancer cells. To test this possibility in our experimental models, we first examined the total levels of 53BP1 protein in whole-cell lysates by immunoblotting. Despite some differences in protein abundance, 53BP1 was expressed in all cancer cell lines of our panel, including the two BRCA1-defective breast cancer cell lines SUM149 and MDA-MB-436 (Fig. 8B and data not shown). Therefore, we created lentivirus vectors and knocked-down 53BP1 in the MDA- MB-436 and control Cal51 cells, by two different shRNAs (Fig. 8B and data not shown). While the knockdown of 53BP1 had no impact on response of the Cal51 cells to PARP-1i, reduction of 53BP1 resulted in a partial, yet statistically significant (p < 0.01 for both concentrations), reduction in sensitivity to PARP-1i in the BRCA1-deficient MDA-MB-436 cells (Fig. 8A). Considering the robust but nevertheless incomplete loss of 53BP1 in these knockdown experiments (Fig. 8B and C), it is possible that a complete lack of 53BP1 would cause an even more pronounced degree of resistance to PARP-1i. In an attempt to relate these results to clinical settings, we evaluated the proportion of complete vs. partial loss of 53BP1 by sensitive immunohistochemical analysis in our previously published cohort (n = 81) of human breast carcinomas.33 Notably, while complete or near-complete lack of 53BP1 protein was very rare, the vast majority of cases among the 25% of tumors with aberrantly reduced 53BP1 featured heterogeneous expression of the protein within the tumor cell areas (Fig. 8D), reminiscent of the patterns observed in our MDA-436 cells after shRNA-mediated knockdown (Figs. 8B and C). Overall, these results indicate that our model for aberrant 53BP1 reduction in breast cancer cells mimicks the patterns seen in clinical specimens, and that such reduced levels of 53BP1 can result in enhanced resistance to treatment with PARP-1i, particularly in HR-deficient tumors, such as those with BRCA1 mutation.
Figure 8. Downregulation of 53BP1 protein leads to increased resistance to PARP-1 inhibitors of human BRCA1-deficient cancer cells MDA-MB-436. (A) Cell survival curve of the MDA-MB-436 and Cal51 cell line upon 53BP1 downregulation and PARP-1 inhibitor treatment. MDA-MB-436 cell line is BRCA1 deficient, whereas Cal 51 cell line was used as control cell line expressing BRCA1 protein. Cells were seeded in triplicates and exposed to 0.1 and 1.0 µM olaparib (or vehiculum) for a period of time corresponding to four cell cycles. Surviving fraction (SF) was determined by cell counting and compared with untreated cells. Results are means ± s.d. (B) Western blot analysis of BRCA1 and 53BP1proteins after stable shRNA mediated downregulation. SMC1 was used as a protein loading control. (C) Heterogeneity of shRNA mediated 53BP1 downregulation in MDA-MB-436 (lower panel; control is shown in the upper panel). (D) Heteregeneity of 53BP1 staining in different breast cancer patients. Empty arrows show positivity in stromal cells, bold arrows indicate different staining intensity of cancer cells (40× magnification in the upper panels and 100× in the lower ones).
Discussion
Recent discoveries based on the concepts of synthetic lethality/sickness and synthetic viability have provided novel mechanistic insights into the complex network of cellular signaling and effector pathways, including the DDR machinery, and opened new avenues for targeted treatments in oncology.1,2,34 The promising treatment strategy of BRCA1/2-deficient tumors with PARP inhibitors illustrates this fruitful trend in cancer research.18 The initial clinical trials with PARP-1i established their relative safety in terms of low toxicity as a single-agent therapy even upon long-term administration, as well as showing some striking examples of positive impact in the clinic.11 On the other hand, several potential molecular mechanisms of resistance to PARP-1i have been reported,19 and suitable predictive biomarkers are also lacking so far. Indeed, what could be the key to an official approval and successful introduction of this emerging treatment into the clinic is to identify those subsets of cancer patients who might most benefit from therapy with PARP-1i, while excluding the patients predicted to be resistant to such treatment. Our present study aimed to identify and/or validate genetic and functional features of human cancer cells that may enhance or decrease sensitivity to PARP-1i, and thereby contribute to the search for potential biomarkers to guide this targeted treatment strategy in the future. We believe our results can assist this global effort in three aspects, each discussed below.
First, we exploited the possibility of extending the subsets of tumors potentially suitable for treatment with PARP-1i to non-BRCA1/2 defects, focusing on the MRN complex deficiency that has been suggested as potentially synthetically lethal when combined with PARP inhibition.18 Indeed, we validated and extended this notion using breast cancer cell line Cal51 experimentally depleted of either Nbs1 or Mre11 proteins by shRNA. In these experiments, exposure to PARP-1i efficiently blocked the endogenous PARsylation in Cal51 cells whose sensitivity to PARP-1i was enhanced by creating MRN deficiency. Given that aberrant expression of the MRN complex was identified in significant fractions of both sporadic and familial breast carcinomas, and MRN defects occur more commonly among the so-called ER/PR/ERBB2 (triple)-negative carcinomas,25,35 our findings suggest that PARP-1i might prove useful in future treatment of this presently most difficult-to-treat subset of breast cancer.
Another genetic determinant we assessed in the present study was the status of the p53 tumor suppressor, the loss or mutations of which have been associated with resistance to a range of chemotherapeutics.29 Here, we found that in the isogenic background of the Mre11-deficient colon cancer cell line HCT116, selective deletion of p53 resulted in an increase in resistance to PARP-1i using the short-term assay. Assessment of the HCT116 model is relevant, in that aberrant Mre11 is known to occur relatively frequently among colorectal tumors with mismatch repair defects.36 This observation from colorectal cancer cells was further supported by the increased survival of Nbs1-deficient breast cancer cells Cal51 in which wild-type p53 was deleted. Therefore, it appears that the effect of p53 on cell survival after PARP inhibition is not specific to colon cancer cells, but can be observed in other types of common human tumors. The p53-deficient cells are often less prone to undergo apoptosis, which may also be true for the DNA damage overload scenario under conditions of PARP inhibition, when enhanced amounts of unrepaired DNA lesions would be more likely to induce apoptosis or cell senescence if p53 was functional. However, the p53-dependent impact of PARP-1i was not recapitulated when we used the long-term clonogenic assay in the same model of MRN-deficient colorectal cancer. Relevant to this point, it has been reported that breast cancer cells depleted for BRCA2 are highly sensitive to PARP-1i regardless of the p53 status.9 We assume that during multiple rounds of the cell cycle, DNA damage accumulates due to insufficient repair, ultimately leading to cell death probably via mitotic catastrophe. Both assays used in our study are well-established methods for evaluating the effect of anticancer drugs in vitro. Short-term cell culture assays determine the cell viability through the evaluation of metabolic activity and give fast and reproducible results with significant clinical relevance. Clonogenic assays, on the other hand, are used to evaluate the drug effect on proliferation potential of actively growing tumor cells and provide as well significant predictive value for further clinical translation.37-39 Faced with the discrepancy of the data obtained with the two assays, the issue of whether p53 status impacts the responses to PARP-1i in a biologically significant manner remains to be resolved.
Another aspect of our study related to p53 status and tumors that do not harbor BRCA1/2 abnormalities is the potential combined treatment with PARP-1i and either chemotherapy or ionizing radiation. Here, we explored this issue using camptothecin and ionizing radiation in colorectal and prostate cancer cell lines, respectively. The choice of CPT and IR was motivated by the fact that the DNA damage caused by CPT and IR requires PARP-1 for efficient signaling and repair,40,41 and therefore PARP-1i may potentiate the cytotoxicity of such genotoxic treatments through a direct increase of unrepaired DNA strand-breakage. These standard-of-care modalities are also clinically relevant, and our present data, together with Nguyen et al.,42 support the notion that a moderate dose of PARP-1i can sensitize colon and prostate cancer cells to such treatments, apparently even in carcinomas with mutant p53. To what extent these, and analogous combinations,43,44 are tolerated in the clinic remains to be established.
The second major aspect of our present work that deserves to be discussed is the consideration of endogenous PARsylation and Rad51 focus formation as surrogate markers of PARP activity and functional HR, respectively. The rationale for monitoring PARsylation is obvious, namely that ongoing activity of the target enzyme(s) is a prerequisite for the inhibitor to show meaningful biological effects. As many cancer cells show spontaneous activation of DDR due to replication stress evoked by the oncogenic process and potential repair deficiencies,45-47 the degree of spontaneous PARsylation can (and does) vary broadly among various cancer cell lines and clinical tumor specimens, and therefore it seems feasible to categorize a given type of tumor into subsets based on the extent of this potential biomarker. Indeed, our analyses suggest a correlation between the level of spontaneous PARsylation and the biological response to PARP-1i among the cell lines of our panel. What is encouraging is that assessment of PARsylation by western blotting of cell and tissue lysates appears to be feasible (our present study and refs. 11 and 48). On the other hand, it would be much more convenient to establish and optimize a simple immunohistochemical assay to monitor PARsylation directly in tumor biopsies, a highly challenging yet very worthwhile goal for future studies.
The value of Rad51 focus formation as a candidate biomarker of ongoing HR repair has already been documented by several recent studies, including assessment by immunofluorescence directly on human tumor biopsies.30,49 The challenge to avoid false-negative results here is to concomitantly assess the cell cycle position of each cell on the tissue section, as HR only operates in S and G2 phases of the cell cycle. Another limitation of this assay is the fact that not all defects of the HR repair machinery may be detected by this assay, and therefore the preserved capacity to form Rad51 foci may not reliably identify tumors unsuitable for PARP-1i treatment. This notion is illustrated in our present study by the observation that, in our models deficient in the MRN complex and sensitive to PARP-1i, the ability to form Rad51 was apparently not dramatically affected. Another example of potentially false results would be cancer-associated defects at some step(s) of the HR pathway that operate(s) downstream of Rad51 foci formation. Again, the preserved ability to form Rad51 foci could provide false information, suggesting to clinicians that such patients are unsuitable for treatment by PARP-1i.
The third and final aspect of our present work, which provides novel and useful information, is the notion of cell resistance mechanisms in response to PARP-1i. While the role of multidrug efflux pumps, including P-gp, in resistance to PARP-1i has already been reported,27,28 our results extend these findings to human colon cancer models and also directly support the reversibility of such resistance by manipulating the activity of P-gp, while directly monitoring the intracellular level of the PARP-1i through mass spectrometry and documenting the correlation of intracellular PARP-1i levels with the biological impact on cancer cell viability.
More important and novel, however, are our present results documenting the acquired higher resistance to PARP-1i by aberrant reduction of 53BP1 in cancer cells with defective BRCA-1. This part of the present study was inspired by our recent collaborative work that revealed preferential loss of 53BP1 in human familial breast carcinomas with BRCA1/2 defects and sporadic triple-negative cancers that are also known to exhibit defective HR.31 Also, the fact that BRCA-1-defective mouse and human cells re-gain the ability to resect DNA ends flanking DSB lesions and form Rad51 foci when 53BP1 is experimentally depleted,31,32 suggested to us that such a scenario might also occur as adaptation and increased survival of relevant types of human tumors under treatment with PARP-1i. Here, we provide the first experimental evidence, based on a clinically relevant model of human BRCA1-defective breast cancer cells, to support loss of 53BP1 as a source of acquired resistance to PARP-1i treatment. Apart from resistance to PARP-1i or other genotoxic treatments, the BRCA1-defective cells may benefit from loss of 53BP1 by gaining fitness in terms of enhanced chromosomal stability and more efficient proliferation.31,32 While the degree of resistance to PARP-1i gained upon shRNA-mediated knockdown of 53BP1 was partial, it would be predicted to be more pronounced, if the depletion of 53BP1 were more complete. Notably, the variable reduced level of 53BP1 seen among cells after the shRNA-mediated knockdown was highly reminiscent of the heterogenous patterns seen in clinical specimens of human breast carcinomas with aberrant overall reduction of 53BP1. Collectively, these results point to reduction or loss of 53BP1 as a plausible source of cancer cells better adapted to the endogenous genetic instability and genotoxic therapies, such as treatment with PARP-1i, particularly in HR-defective tumors facing highly unstable genomic landscapes, under selection pressure for more efficient DNA repair in order to survive.
Taken together, our present results support the notion that the therapeutic potential of PARP-1 inhibitors may broaden from BRCA1/2-deficient tumors to those bearing defects in the MRN complex, likely as part of a combination treatment with standard-of-care chemotherapeutics or IR. Furthermore, we highlight the potential value of endogenous PARsylation and assessment of 53BP1 expression patterns as potential new predictive markers of cellular responses to PARP-1i, at least in a single -agent therapy approach. Considering the current limitations of these assays, it is unlikely that a universal and absolutely reliable biomarker will be discovered and validated in the near future. It is therefore possible that a combination of several biomarkers, perhaps including BRCA1/2 and MRN status, PARsylation, Rad51 focus formation, and monitoring 53BP1 can find predictive applications in guiding PARP-1i treatment in the years to come.
Material and Methods
Cell culture
Human fibroblasts BJ and NBS-1LBI, NBS-1LBI+Nbs1; colon cancer cell lines HCT116, SW480, SW620, HCT15, HT29; osteosarcoma cell U2OS; breast cancer cell lines CAL51 (CAL51-wt, CAL51-Mre11 kd, CAL51-Nbs1 kd), MDA-MB-436, SUM149; prostate cancer cell lines PC3, DU-145; ovarian cancer cell line OVCAR3; and pancreatic cancer cell line CAPAN1 were cultured as described.50 Lymphoblastoma cell lines K256, SR were maintained in RPMI 1640 medium supplemented with 10% (v/v) FBS and 100 U Penicillin and 100 μl/ml Strepromycin. All cell lines were maintained at 37°C in a humidified atmosphere at 5% CO2. Reagents for cell cultivation were obtained from Gibco Invitrogen.
Stable transfections
Colon cancer cell line HCT116 p53wt was transfected with pcDNA 3.1 Mre11-GFP vector (encoding the full-length wild-type human Mre11 fused with the green fluorescent protein) together with pBABEpuro (5:1 ration) using FuGene 6 (Roche) reagent according to manufacturer’s protocol. Stable clones expressing the transgene were selected in standard DMEM with added puromycine (1 μg/ml). Breast cancer cell lines CAL-51 and MDA MB 436 were transduced with lentiviral particles containing non-targeting shRNA (NT shRNA) or 53BP1 shRNA (Sigma Aldrich). Stable cell lines were selected (5 μg/ml puromycin treatment) and maintained with 0.2 μg/ml puromycin. Breast cancer cell line CAL51-wt and CAL51 shRNA knockdowns of Mre11 and Nbs1 (CAL51-Mre11, CAL51-Nbs1) were kindly provided by KuDOS Pharmaceuticals Ltd. (now part of AstraZeneca).
The cell lines Nbs1 wt reconstituted Nbs1-Tert and NBS-1LBI cell lines were prepared and described by Horejsi et al.51
RNA interference
Silencing of p53 expression was induced by respective ON-TARGET plus SMARTpool siRNA, the control siRNA sequence used was GGGAGGACAAGACGUUCUAdTdT (Dharmacon). Introduction of siRNA into the cells was performed by an electroporator using Nucleofector kit V (program P-20) (Amaxa/Lonza), according to manufacturer’s instructions. Efficacy of gene silencing was validated by western blot analysis.
Western blot analysis
Western blotting was performed as previously described.52 Briefly, dishes with cells were washed with ice-cold PBS, and subsequently lysed in EB lysis buffer (50 mM Tris pH7.5, 150 mM NaCl, 0.5% Igepal 360, 1.0 mM EDTA, 1 mM NaF, 1.0 mM PMSF, 10 mM β-glycerolphosphate, 1 μg/ml aprotitnin, 1 μg/ml leupeptin) on the dishes, scrapped off and lysed on ice for 20 min. After centrifugation (10 min/16,000 g/5°C) supernatant was used. Protein concentration in each supernatant was determined spectrophotometrically (wavelength 595 nm) with Bradford reagent (Bio-Rad) or Pierce BCA Protein Assay Kit (Thermo Scientific). Nuclear extracts from CAL51 and MDA MB 436 cell lines to assess BRCA1 protein level were isolated using NE-PER Nuclear and Cytoplasmic Extraction Kit, according to manufacturer’s protocols (Thermo Scientific).
Protein loading buffer (4×, 200 mM Tris-HCL, pH 7.8, 400 mM DTT, 8% SDS, 0.4% bromphenol blue, and 40% glycerol) was added to each sample and heated at 95°C for 3 min. Proteins were separated on 7.5% SDS-PAGE and blotted onto nitrocellulose membrane (Advantec). Large proteins (53BP1 and BRCA1) were separated on Novex 4% Tris-Glycine gels (Invitrogen) and transferred to nitrocellulose membrane by using I-blot system and reagents, according to manufacturer’s protocols (Invitrogen). After blocking with 5% (w/v) milk in PBS containing 0.1% Tween-20, membranes were probed with indicated primary antibodies: Nbs1 (1D7, GeneTex, Inc.), Rad50 (13B3, GeneTex, Inc.), Mre11 (NB100–142, Novus Biologicals), SMC1 (ab 9262, Abcam), goat γ-tubulin (C-20, Santa Cruz Biotech., Inc.), PARP-1 (H-250, Santa Cruz Biotech., Inc.), PAR (LP96–10, BD PharMingen, 53BP1 (Santa Cruz Biotech., Inc.), α-tubulin (Sigma Aldrich), BRCA1 (Santa Cruz Biotech., Inc.) overnight at 4°C. Finally, the membranes were rinsed with PBS + 0.1% Tween-20 for 30 min and incubated with appropriate horseradish peroxidase conjugated secondary antibodies (Pierce) for 1 h at room temperature (RT). Followed by washing in PBS + 0.1% Tween-20 for 30 min, protein bands were detected using enhanced chemiluminiscence kit, ECL solution (Amersham Biosciences).
Cell proliferation assays and cell death assessment
Exponentially growing cells were seeded in 6-well culture plates at appropriate densities (20 × 104 cancer cells, 15 × 104 fibroblasts) and attached overnight. PARP-1 inhibitors KU 58948 or AZD2281/olaparib (KuDOS Pharmaceuticals Ltd, now part of AstraZeneca) and CPT (Sigma Aldrich) were added to the medium at indicated concentrations. In case of combined treatment, CPT in various concentrations (1.23 nM, 3.7 nM, 11.11 nM) plus 1 μM PARP-1 inhibitor was added to the medium. Cells were grown for 2 d then split to 50% (to keep the optimal cell density) and grown for additional 2 d with medium only. After 4 d of growth, the cells were counted on cell counter (BD Biosciences, CellQuest Pro Software), and cell number was calculated as a percentage of mock-treated cells. All experiments were performed in triplicates and repeated at least twice. After transfection with siRNA against p53, cells were seeded into 6-well plates, attached ON and treated with PARP-1 inhibitor as described above. Stable Cal51- and MDA-436-derived cell lines expressing NT shRNA or 53BP1 shRNA were seeded in 24-well culture plates (1 × 104 cells/well) in triplicate and exposed to 0.1 and 1.0 μM PARPi (or vehiculum) for a period of time corresponding to four cell cycles. Surviving fraction (SF) was determined by cell counting and compared with control (vehiculum-treated) cells. Results are means from four independent experiments ± s.d. To assess p value, two tailed Student’s t-test was used (p < 0.05).
Proliferation activity of PARP-1 inhibitor-treated CAL51 cells and their derivatives, coupled with cell death evaluation, was assessed by 96-well plate based XTT [2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxyanilide inner salt] colorimetric assay and lactate dehydrogenase (LDH) activity test, respectively, according to manufacturer’s protocols (Roche). Briefly, cells were seeded at the concentration of 5,000 cells per well and attached overnight. The following day, PARP-1 inhibitor KU 58948 was added to the cells at concentrations of 10−4 – 10−8M. After 4 d growth with the drug, medium was removed to the new 96-well plate to evaluate the LDH activity released to the medium by the death cells. Remaining cells were washed twice with PBS, and absorbance was measured 4 h after addition of XTT reagents at 480 and 690 nm using VersaMax spectrophotometer.
The XTT assay was used to evaluate proliferation activity of HCT116 cells after treatment with KU 58948 alone and in combination with Pg-p inhibitor. Briefly, cells were treated with KU 58948 in concentrations 10−8 – 10−4 M or pre-treated for 1 h with 5 μg/ ml Verapamil prior to addition of KU 58948. The cells were grown for 4 d, and subsequently the cell viability was assessed using the XTT test according to manufacturer’s instructions.
Clonogenic survival assay and irradiation
Cells were seeded in triplicates into 6-well plates (500 cells per well) and left to stabilize for 24 h. After that, the cells were incubated with 0.1 μM KU 58948 for 24 h and then irradiated. Treated cultures were incubated for additional 10 d in drug-free medium. Finally, the cultures were fixed, stained with crystal violet and colonies containing more than 50 cells were counted. Irradiation sessions were performed at room temperature using Linear accelerator Siemens primus (6MV).
Intracellular detection of KU 58948
HCT116 cells were incubated with 1μM PARP-1 inhibitor KU 58948 and in combination with 5 μg/ ml Verapamil for 1 min and 24 h, respectively. The cell pellet was resuspended in 200 μl of methanol, and an analyte (KU 58948) was extracted from the supernatant using solid-phase extraction microcolumn with a C18 phase (SPE C18). The eluates were resuspended in 30 μl of 10% methanol, vortexed and sonicated for 5 min. One-third of the solution was injected to UPLC-ESI-QqQ system (Acquity UPLC and XEVO MS, Waters). The quantitation of the analyte in the cells was performed using an external calibration with KU 58958 as calibrant.
Immunofluorescence analysis
Immunofluorescence analysis for performed as previously described.53 For in situ analysis of p53, cells were grown on glass coverslips, rinsed briefly in cold PBS and fixed in a 1:1 mixture of ice cold methanol:acetone at RT for 10 min. After drying at RT, cells on coverslips were stained with primary antibody against p53 (DO-01, prepared in house) for 1 h/RT. As secondary antibodies goat anti-mouse coupled to Alexa Flour 488 nm (Invitrogen) were used. After staining procedure, 1 h/ RT (light shielded), the coverslips were dried by ethanol series and mounted on standard microscopic glass using Vecta Shield mounting medium with DAPI (Vector Laboratories). Examination was done using fluorescent microscope (Zeiss Axioplan II). Images were acquired through a PLAN-Neofluar 40×/1.3 oil immersion objective and photographed by digital camera (Cool Snap).
For Rad51 analysis, cells were seeded into 96-well plate (Costar, black, flat bottom) and attached overnight. Cells were treated with PARP-1 inhibitor KU 58948 1 μM for 24 h and subsequently fixed in 4% formaldehyde for 15 min RT. Fixation was followed by cell permeabilization with 0.5% Triton X-100 (in PBS) for 4 min RT and washing 3× in PBS. Cells were then blocked with 3% BSA in PBS for 1 h RT. Primary antibody mix Rad51 (GeneTex Clone 14B4) and cyclinA (Santa Cruz Biotech., Inc.) was added on the cells ON at 4°C. Plate was washed 3× with TBS + 0.1% Tween, and the secondary antibody mix Alexa Fluor 488 and 546 was applied for 1 h RT. Cells were washed again 3× with TBS + 0.1% Tween and incubated with Hoechst (Invitrogen) for 5 min RT. Finally, plate was washed 1× in PBS and cells were covered with 75 μL PBS. Images were acquired on ArrayScan VTI HCS Reader (Thermo Scientific) 20x objective and Rad51 foci in cyclin A-positive cells analyzed by Cellomics software.
For 53BP1 analysis, attached cells in 96-well plate (Costar) were irradiated with 2 Gy and after 30 min fixed as described for Rad51/cyclinA. Cells were incubated with antibody against 53BP1 (Santa Cruz Biotech., Inc.) ON at 4°C. After the washing step, secondary antibody Alexa Fluor 488 was applied for 1 h RT which was followed by 5 min incubation with Hoechst (Invitrogen). The plate was washed 1× in PBS and cells were covered with 75 μL PBS. Representative images were acquired on ArrayScan VTI HCS Reader (Thermo Scientific) 20× objective.
Immunohistochemistry on paraffin sections
For immunohistochemical analysis of archival formalin-fixed, paraffin-embedded human breast carcinomas, the tissue sections were deparaffinized and processed for sensitive immunoperoxidase staining with the primary mouse monoclonal antibody to 53BP1 (kindly donated by Thanos Halazonetis, University of Geneva). The primary antibody was incubated overnight, followed by detection using the Vectastain Elite kit (Vector Laboratories) and nickel sulfate enhancement without nuclear counterstaining, as described previously,25,33 followed by evaluation of the staining patterns by an experienced oncopathologist.
Supplementary Material
Acknowledgments
This work was supported by the Danish Council for Independent Research, the Danish Cancer Society, the Danish National Research Foundation, the Lundbeck Foundation (R-93-A8990), the Novo Nordisk Foundation, the Czech Ministry of Health (NS10282–3/2009), and the European Commission (projects Infla-Care, Biomedreg: CZ.1.05/2.1.00/01.0030, DDResponse, and FP7-PEOPLE-2011-IEF, SYNvia).
Glossary
Abbreviations:
- 53BP1
p53 binding protein 1
- ATM
ataxia telangiectasia mutated
- BRCA1
breast cancer type 1 susceptibility protein
- BRCA2
breast cancer type 2 susceptibility protein
- DDR
DNA damage response
- DSB
DNA double-strand break
- γH2AX
histone variant H2AX phosphorylated on Ser139
- IR
ionizing radiation
- MRN
Mre11-Rad50-Nbs1 complex
- PARP-1
poly(ADP-ribose) polymerase-1
- P-gp
P-glycoprotein
- shRNA
short hairpin RNA
- SSB
single-strand break
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Supplemental Material
Supplemental material may be found here: www.landesbioscience.com/journals/cc/article/22026
Footnotes
Previously published online: www.landesbioscience.com/journals/cc/article/22026
References
- 1.O’Connor MJ, Martin NM, Smith GC. Targeted cancer therapies based on the inhibition of DNA strand break repair. Oncogene. 2007;26:7816–24. doi: 10.1038/sj.onc.1210879. [DOI] [PubMed] [Google Scholar]
- 2.Lord CJ, Ashworth A. The DNA damage response and cancer therapy. Nature. 2012;481:287–94. doi: 10.1038/nature10760. [DOI] [PubMed] [Google Scholar]
- 3.Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature. 2009;461:1071–8. doi: 10.1038/nature08467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Blagosklonny MV. NCI’s provocative questions on cancer: some answers to ignite discussion. Oncotarget. 2011;2:1352–67. doi: 10.18632/oncotarget.432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.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. doi: 10.1038/nrm1963. [DOI] [PubMed] [Google Scholar]
- 6.Plummer ER. Inhibition of poly(ADP-ribose) polymerase in cancer. Curr Opin Pharmacol. 2006;6:364–8. doi: 10.1016/j.coph.2006.02.004. [DOI] [PubMed] [Google Scholar]
- 7.Helleday T. The underlying mechanism for the PARP and BRCA synthetic lethality: clearing up the misunderstandings. Mol Oncol. 2011;5:387–93. doi: 10.1016/j.molonc.2011.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hoeijmakers JH. Genome maintenance mechanisms for preventing cancer. Nature. 2001;411:366–74. doi: 10.1038/35077232. [DOI] [PubMed] [Google Scholar]
- 9.Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434:913–7. doi: 10.1038/nature03443. [DOI] [PubMed] [Google Scholar]
- 10.Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434:917–21. doi: 10.1038/nature03445. [DOI] [PubMed] [Google Scholar]
- 11.Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361:123–34. doi: 10.1056/NEJMoa0900212. [DOI] [PubMed] [Google Scholar]
- 12.Gagné JP, Isabelle M, Lo KS, Bourassa S, Hendzel MJ, Dawson VL, et al. Proteome-wide identification of poly(ADP-ribose) binding proteins and poly(ADP-ribose)-associated protein complexes. Nucleic Acids Res. 2008;36:6959–76. doi: 10.1093/nar/gkn771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lukas J, Lukas C, Bartek J. More than just a focus: The chromatin response to DNA damage and its role in genome integrity maintenance. Nat Cell Biol. 2011;13:1161–9. doi: 10.1038/ncb2344. [DOI] [PubMed] [Google Scholar]
- 14.Williams RS, Williams JS, Tainer JA. Mre11-Rad50-Nbs1 is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochem Cell Biol. 2007;85:509–20. doi: 10.1139/O07-069. [DOI] [PubMed] [Google Scholar]
- 15.Haince JF, McDonald D, Rodrigue A, Déry U, Masson JY, Hendzel MJ, et al. PARP1-dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites. J Biol Chem. 2008;283:1197–208. doi: 10.1074/jbc.M706734200. [DOI] [PubMed] [Google Scholar]
- 16.McCabe N, Turner NC, Lord CJ, Kluzek K, Bialkowska A, Swift S, et al. Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res. 2006;66:8109–15. doi: 10.1158/0008-5472.CAN-06-0140. [DOI] [PubMed] [Google Scholar]
- 17.Höglund A, Strömvall K, Li Y, Forshell LP, Nilsson JA. Chk2 deficiency in Myc overexpressing lymphoma cells elicits a synergistic lethal response in combination with PARP inhibition. Cell Cycle. 2011;10:3598–607. doi: 10.4161/cc.10.20.17887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Dedes KJ, Wilkerson PM, Wetterskog D, Weigelt B, Ashworth A, Reis-Filho JS. Synthetic lethality of PARP inhibition in cancers lacking BRCA1 and BRCA2 mutations. Cell Cycle. 2011;10:1192–9. doi: 10.4161/cc.10.8.15273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Michalak EM, Jonkers J. Studying therapy response and resistance in mouse models for BRCA1-deficient breast cancer. J Mammary Gland Biol Neoplasia. 2011;16:41–50. doi: 10.1007/s10911-011-9199-z. [DOI] [PubMed] [Google Scholar]
- 20.Lord CJ, McDonald S, Swift S, Turner NC, Ashworth A. A high-throughput RNA interference screen for DNA repair determinants of PARP inhibitor sensitivity. DNA Repair (Amst) 2008;7:2010–9. doi: 10.1016/j.dnarep.2008.08.014. [DOI] [PubMed] [Google Scholar]
- 21.Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature. 2004;432:316–23. doi: 10.1038/nature03097. [DOI] [PubMed] [Google Scholar]
- 22.Gallmeier E, Kern SE. Absence of specific cell killing of the BRCA2-deficient human cancer cell line CAPAN1 by poly(ADP-ribose) polymerase inhibition. Cancer Biol Ther. 2005;4:703–6. doi: 10.4161/cbt.4.7.1909. [DOI] [PubMed] [Google Scholar]
- 23.McCabe N, Lord CJ, Tutt AN, Martin NM, Smith GC, Ashworth A. BRCA2-deficient CAPAN-1 cells are extremely sensitive to the inhibition of Poly (ADP-Ribose) polymerase: an issue of potency. Cancer Biol Ther. 2005;4:934–6. doi: 10.4161/cbt.4.9.2141. [DOI] [PubMed] [Google Scholar]
- 24.Wen Q, Scorah J, Phear G, Rodgers G, Rodgers S, Meuth M. A mutant allele of MRE11 found in mismatch repair-deficient tumor cells suppresses the cellular response to DNA replication fork stress in a dominant-negative manner. Mol Biol Cell. 2008;19:1693–705. doi: 10.1091/mbc.E07-09-0975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bartkova J, Tommiska J, Oplustilova L, Aaltonen K, Tamminen A, Heikkinen T, et al. Aberrations of the MRE11-RAD50-NBS1 DNA damage sensor complex in human breast cancer: MRE11 as a candidate familial cancer-predisposing gene. Mol Oncol. 2008;2:296–316. doi: 10.1016/j.molonc.2008.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yagi K, Kunitomo K, Ii K, Tashiro S. Relationship of P-glycoprotein and p53 protein to chemosensitivity in colorectal cancer. Int J Clin Oncol. 1997;2:81–91. doi: 10.1007/BF02488878. [DOI] [Google Scholar]
- 27.Rottenberg S, Jaspers JE, Kersbergen A, van der Burg E, Nygren AO, Zander SA, 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 USA. 2008;105:17079–84. doi: 10.1073/pnas.0806092105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hay T, Matthews JR, Pietzka L, Lau A, Cranston A, Nygren AO, et al. Poly(ADP-ribose) polymerase-1 inhibitor treatment regresses autochthonous Brca2/p53-mutant mammary tumors in vivo and delays tumor relapse in combination with carboplatin. Cancer Res. 2009;69:3850–5. doi: 10.1158/0008-5472.CAN-08-2388. [DOI] [PubMed] [Google Scholar]
- 29.O’Connor PM, Jackman J, Bae I, Myers TG, Fan S, Mutoh M, et al. Characterization of the p53 tumor suppressor pathway in cell lines of the National Cancer Institute anticancer drug screen and correlations with the growth-inhibitory potency of 123 anticancer agents. Cancer Res. 1997;57:4285–300. [PubMed] [Google Scholar]
- 30.Graeser M, McCarthy A, Lord CJ, Savage K, Hills M, Salter J, et al. A marker of homologous recombination predicts pathologic complete response to neoadjuvant chemotherapy in primary breast cancer. Clin Cancer Res. 2010;16:6159–68. doi: 10.1158/1078-0432.CCR-10-1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bouwman P, Aly A, Escandell JM, Pieterse M, Bartkova J, van der Gulden H, 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. doi: 10.1038/nsmb.1831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Bunting SF, Callén E, Wong N, Chen HT, Polato F, Gunn A, et al. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell. 2010;141:243–54. doi: 10.1016/j.cell.2010.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bartkova J, Horejsí Z, Sehested M, Nesland JM, Rajpert-De Meyts E, Skakkebaek NE, et al. DNA damage response mediators MDC1 and 53BP1: constitutive activation and aberrant loss in breast and lung cancer, but not in testicular germ cell tumours. Oncogene. 2007;26:7414–22. doi: 10.1038/sj.onc.1210553. [DOI] [PubMed] [Google Scholar]
- 34.Bauzon F, Zhu L. Racing to block tumorigenesis after pRb loss: an innocuous point mutation wins with synthetic lethality. Cell Cycle. 2010;9:2118–23. doi: 10.4161/cc.9.11.11726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hsu HM, Wang HC, Chen ST, Hsu GC, Shen CY, Yu JC. Breast cancer risk is associated with the genes encoding the DNA double-strand break repair Mre11/Rad50/Nbs1 complex. Cancer Epidemiol Biomarkers Prev. 2007;16:2024–32. doi: 10.1158/1055-9965.EPI-07-0116. [DOI] [PubMed] [Google Scholar]
- 36.Giannini G, Ristori E, Cerignoli F, Rinaldi C, Zani M, Viel A, et al. Human MRE11 is inactivated in mismatch repair-deficient cancers. EMBO Rep. 2002;3:248–54. doi: 10.1093/embo-reports/kvf044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zoli W, Ricotti L, Tesei A, Barzanti F, Amadori D. In vitro preclinical models for a rational design of chemotherapy combinations in human tumors. Crit Rev Oncol Hematol. 2001;37:69–82. doi: 10.1016/S1040-8428(00)00110-4. [DOI] [PubMed] [Google Scholar]
- 38.Testa U, Pasquini L, Petrucci E. In vitro assays of tumor chemosensitivity and chemoresistance. Drugs Future. 2004;29:1035–42. doi: 10.1358/dof.2004.029.10.863394. [DOI] [Google Scholar]
- 39.Hatok J, Babusikova E, Matakova T, Mistuna D, Dobrota D, Racay P. In vitro assays for the evaluation of drug resistance in tumor cells. Clin Exp Med. 2009;9:1–7. doi: 10.1007/s10238-008-0011-3. [DOI] [PubMed] [Google Scholar]
- 40.Bowman KJ, Newell DR, Calvert AH, Curtin NJ. Differential effects of the poly (ADP-ribose) polymerase (PARP) inhibitor NU1025 on topoisomerase I and II inhibitor cytotoxicity in L1210 cells in vitro. Br J Cancer. 2001;84:106–12. doi: 10.1054/bjoc.2000.1555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Javle M, Curtin NJ. The role of PARP in DNA repair and its therapeutic exploitation. Br J Cancer. 2011;105:1114–22. doi: 10.1038/bjc.2011.382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nguyen D, Zajac-Kaye M, Rubinstein L, Voeller D, Tomaszewski JE, Kummar S, et al. Poly(ADP-ribose) polymerase inhibition enhances p53-dependent and -independent DNA damage responses induced by DNA damaging agent. Cell Cycle. 2011;10:4074–82. doi: 10.4161/cc.10.23.18170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hawtin RE, Stockett DE, Wong OK, Lundin C, Helleday T, Fox JA. Homologous recombination repair is essential for repair of vosaroxin-induced DNA double-strand breaks. Oncotarget. 2010;1:606–19. doi: 10.18632/oncotarget.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.van Vuurden DG, Hulleman E, Meijer OL, Wedekind LE, Kool M, Witt H, et al. PARP inhibition sensitizes childhood high grade glioma, medulloblastoma and ependymoma to radiation. Oncotarget. 2011;2:984–96. doi: 10.18632/oncotarget.362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bartkova J, Horejsí Z, Koed K, Krämer A, Tort F, Zieger K, et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature. 2005;434:864–70. doi: 10.1038/nature03482. [DOI] [PubMed] [Google Scholar]
- 46.Guirouilh-Barbat JK, Wilhelm T, Lopez BS. AKT1/BRCA1 in the control of homologous recombination and genetic stability: the missing link between hereditary and sporadic breast cancers. Oncotarget. 2010;1:691–9. doi: 10.18632/oncotarget.203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kosar M, Bartkova J, Hubackova S, Hodny Z, Lukas J, Bartek J. Senescence-associated heterochromatin foci are dispensable for cellular senescence, occur in a cell type- and insult-dependent manner and follow expression of p16(ink4a) Cell Cycle. 2011;10:457–68. doi: 10.4161/cc.10.3.14707. [DOI] [PubMed] [Google Scholar]
- 48.Gottipati P, Vischioni B, Schultz N, Solomons J, Bryant HE, Djureinovic T, et al. Poly(ADP-ribose) polymerase is hyperactivated in homologous recombination-defective cells. Cancer Res. 2010;70:5389–98. doi: 10.1158/0008-5472.CAN-09-4716. [DOI] [PubMed] [Google Scholar]
- 49.Mukhopadhyay A, Elattar A, Cerbinskaite A, Wilkinson SJ, Drew Y, Kyle S, et al. Development of a functional assay for homologous recombination status in primary cultures of epithelial ovarian tumor and correlation with sensitivity to poly(ADP-ribose) polymerase inhibitors. Clin Cancer Res. 2010;16:2344–51. doi: 10.1158/1078-0432.CCR-09-2758. [DOI] [PubMed] [Google Scholar]
- 50.Aagaard L, Lukas J, Bartkova J, Kjerulff A-A, Strauss M, Bartek J. Aberrations of p16Ink4 and retinoblastoma tumour-suppressor genes occur in distinct sub-sets of human cancer cell lines. Int J Cancer. 1995;61:115–20. doi: 10.1002/ijc.2910610120. [DOI] [PubMed] [Google Scholar]
- 51.Horejsí Z, Falck J, Bakkenist CJ, Kastan MB, Lukas J, Bartek J. Distinct functional domains of Nbs1 modulate the timing and magnitude of ATM activation after low doses of ionizing radiation. Oncogene. 2004;23:3122–7. doi: 10.1038/sj.onc.1207447. [DOI] [PubMed] [Google Scholar]
- 52.Hubackova S, Novakova Z, Krejcikova K, Kosar M, Dobrovolna J, Duskova P, et al. Regulation of the PML tumor suppressor in drug-induced senescence of human normal and cancer cells by JAK/STAT-mediated signaling. Cell Cycle. 2010;9:3085–99. doi: 10.4161/cc.9.15.12521. [DOI] [PubMed] [Google Scholar]
- 53.Moudry P, Lukas C, Macurek L, Hanzlikova H, Hodny Z, Lukas J, et al. Ubiquitin-activating enzyme UBA1 is required for cellular response to DNA damage. Cell Cycle. 2012;11:1573–82. doi: 10.4161/cc.19978. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.







