RAD51 is a key protein in the homologous recombination (HR) pathway of DNA double-strand break repair, and HR represents a novel target for cancer therapy. Because imatinib (Gleevec) has been reported to reduce RAD51 protein levels, we tested the clonogenic survival for RT112, H1299, PANC1, and PC3 tumor cell lines of varying p53 status and normal GM05757 normal fibroblasts after exposure to single agent imatinib (0–20 μmol/L; 0–72 hours). We also combined imatinib with DNA damaging agents that are toxic to RAD51-deficient cells, including ionizing radiation, gemcitabine, and mitomycin C. We observed decreased nuclear expression and chromatin binding of RAD51 protein following imatinib treatment. Imatinib also resulted in decreased error-free HR as determined by a flow cytometry–based integrated direct repeat-green fusion protein reporter system; this correlated to reduced RAD51 expression. Clonogenic survival experiments revealed increased cell kill for imatinib-treated cells in combination with ionizing radiation, gemcitabine, and mitomycin C, due in part to mitotic catastrophe. In experiments using imatinib and gemcitabine, tumor cell lines were sensitized to a greater extent than normal fibroblasts. This preservation of the therapeutic ratio was confirmed in vivo using PC3 xenograft growth delay and intestinal crypt cell clonogenic assays. HR inhibition may be an additional mechanism of action for the chemosensitization and radiosensitization of solid tumors with imatinib with preservation of the therapeutic ratio. [Mol Cancer Ther 2009;8(1):203–13]

DNA is a major target for many cancer therapeutics including cytotoxic chemotherapy and radiotherapy. These agents can induce DNA double-strand breaks (DNA-dsb) whereby unrepaired lethal lesions lead to apoptosis, mitotic catastrophe, terminal growth arrest, and finally, increased clonogenic tumor cell kill. Human DNA-dsbs are primarily repaired by the homologous recombination (HR) pathway, which is active specifically in S- and G2-phase cells, and by the nonhomologous end-joining (NHEJ) pathway, which can be activated in all cell cycle phases (but has a preference for the G1 phase). The HR and NHEJ pathways therefore interact with each other across cell cycle transitions to effect DNA-dsb repair and prevent cellular mutation and toxicity in normal tissues (1, 2).

The expression and function of HR and NHEJ proteins may differ between normal and tumor cells, which could provide a therapeutic window for novel therapies (3, 4). Increasingly, multimodality combination treatment (e.g., chemoradiotherapy or radiotherapy with molecular-targeted agents) is being used in many bladder, lung, cervix, and other solid tumors to improve local control and survival. In this regard, correlations have been observed between radiotherapy or chemotherapy outcome and expression or function of DNA-dsb repair proteins. This suggests that an understanding of DNA-dsb repair across the malignant axis could give rise to prediction of therapy response and judicious choice of molecular-targeted agents in combination with chemotherapy and radiotherapy as tumor cell sensitizers (2, 46). Novel strategies with HR repair pathways could enhance tumor cell kill based on an increased fraction of cells in S and G2 phases of the cell cycle, altered DNA damage checkpoints (based on p53 mutation), synthetic lethality with poly(ADP-ribose) polymerase inhibition, and the presence of intratumoral hypoxia, which may modify DNA-dsb repair (2, 7, 8). HR-defective cells are more sensitive to certain agents [e.g., mitomycin C (MMC), cisplatinum, gemcitabine, ionizing radiation (IR), poly(ADP-ribose) polymerase inhibitors] compared with NHEJ-defective cells, suggesting an increased specificity for targeted therapeutics if the underlying defect in DNA-dsb repair is understood (911). Indeed, these differences in HR capacity can be assayed by determining the relative sensitivities to cross-linking and alkylating agents such as gemcitabine and MMC, which are also among the most potent radiosensitizers when combined with radiotherapy (12, 13).

A specific HR-related target for cancer therapy is the RAD51 protein. RAD51 protein expression has been linked to MMC, IR, and gemcitabine resistance (14). Recent data also suggest that RAD51 overexpression leads to a worse clinical outcome in lung cancer (15). RAD51 expression can be manipulated pharmacologically in vitro by the use of imatinib mesylate. Imatinib is an inhibitor of c-ABL, c-KIT, and platelet-derived growth factor receptor tyrosine kinases. However, c-ABL also up-regulates Rad51 gene expression and increases RAD51 protein phosphorylation at Tyr315, resulting in increased RAD51 focus formation in a complex with the c-ABL or RAD52 and ATM proteins (16, 17). Treatment of leukemia, prostate, and glioma cells with imatinib can decrease RAD51 expression and sensitize them to experimental chemotherapy and radiotherapy in vitro (16, 18). However, whether imatinib directly decreases error-free HR as a mechanism for this increased tumor cell chemosensitivity or radiosensitivity is not known. If true, this would give unique information about the judicious selection of agents to combine with imatinib and potential biomarkers relating to the use of imatinib in combination with HR-sensitizing agents.

Herein, we have studied the effects of imatinib on tumor cell lines of varying p53 status and histopathology in combination with agents that may preferentially be toxic to HR-defective cells (e.g., IR, MMC, and gemcitabine). We observed increased cell kill in tumor cells in vitro compared with fibroblasts. This was associated with reduced RAD51 expression and function. These findings were recapitulated in a prostate xenograft model treated with radiotherapy and imatinib without attendant gut toxicity. Together, these results support the targeting of HR with imatinib as a cancer therapy that preserves the therapeutic ratio.

Cell Culture Conditions

The growth characteristics and culture of the cell lines used in this study have been previously described (19, 20). GM05757 (normal diploid fibroblasts), PC3 (prostate adenocarcinoma), and PANC1 (pancreatic adenocarcinoma) were used in these experiments and were obtained from Coriell Cell Repository or American Type Culture Collection. RT112M (transitional bladder cell carcinoma) was a generous gift from Prof. Maggie Knowles. The H1299-DR-GFP human lung carcinoma cell line was a kind gift of Dr. Simon Powell (Washington University, St. Louis, MO) and expresses an integrated direct repeat-green fusion protein (DR-GFP) HR reporter system as previously described (8).

The p53 gene status of each cell strain was determined by direct DNA sequencing of exons 2 to 11 using automated DNA sequencing (DNA Sequenator, Becton-Coulter) as previously described (21). The GM05757 strain and RT112 cell line express two wild-type p53 alleles and functional wild-type p53 protein, whereas the PC3, H1299, and PANC1 cell lines express truncated p53 alleles leading to a lack of expression of the wild-type p53 protein (i.e., null p53). All cells were incubated in 5% CO2 at 37°C. RT112 cells were grown in RPMI 1640 supplemented with 10% FCS and 1% l-glutamine; H1299 in RPMI 1640, 10% FCS, and 20 mmol/L HEPES; PC3 in F12K with 10% FCS; PANC1 in Dulbecco's and 10% FCS; and NDF-GM05757 in α-MEM with 15% FCS. All media contained broad-spectrum antibiotics (100 mg/L penicillin V and 100 mg/L streptomycin). All cell lines were negative for Mycoplasma contamination.

Western Blotting of Whole Cell or Nuclear Lysates

Whole cell lysates from asynchronously growing and synchronized G0-G1 cell cultures were prepared for SDS-PAGE/Western blot analyses (20). Briefly, nonirradiated and irradiated cultures were lysed on ice in lysis buffer [150 mmol/L NaCl, 0.1% SDS, 1% sodium deoxycholate, 1% Triton X-100, 10 mmol/L Tris-HCl (pH 7.4), plus Complete Protease Inhibitor (Roche)] and 20 to 50 μg of protein were equally loaded for SDS-PAGE. Gels were electroblotted onto nitrocellulose membranes (S&S) using semi-dry transfer (Helixx). Membranes were placed in blocking solution (TBST/10% low fat milk) before incubation with the primary antibody of interest. The anti–c-ABL monoclonal antihuman antibody was obtained from Cell Signaling Technology; the anti-Ku70 and anti-RAD51 antibodies from Santa Cruz Biotechnology, Inc.; and the anti-actin antibody from Sigma-Aldrich, Inc. Membranes were then washed with TBST, incubated with the appropriate horseradish peroxidase–conjugated secondary antibody (Santa Cruz Biotechnology and Oncogene Research Products), washed again in TBST, and processed for chemiluminescence detection using the Renaissance chemiluminescence detection kit (NEN). Membranes were then exposed to enhanced chemiluminescence film (Amersham).

For selected experiments, we isolated insoluble nuclear biochemical fractions (chromatin fractions) using a modified Dignam method (22). Briefly, cells were lysed and incubated for 5 min on ice in cytoplasmic buffer [25 mmol/L KCl, 5 mmol/L MgCl2, 10 mmol/L Tris-HCl (pH 8.0), 0.5% NP40, 1 mmol/L DTT, 1× protease inhibitors (Complete EDTA-free, Roche), 1× phosphatase inhibitors (Cocktail Set II, Calbiochem)]. Lysates were centrifuged at 3,000 rpm for 5 min and the supernatant (cytoplasmic fraction) was separated. The nuclear pellet was rinsed thrice with cytoplasmic buffer, then resuspended in nuclear buffer [10 mmol/L Tris-HCl (pH 8.0), 500 mmol/L NaCl, 1.5 mmol/L MgCl2, 0.1% NP40, 5 mmol/L EDTA, 1× protease inhibitors (Complete EDTA-free, Roche), 1× phosphatase inhibitors (Cocktail Set II, Calbiochem)]. The nuclei were lysed by vigorous pipetting, incubated on ice for 15 min, and then centrifuged at 3,000 rpm for 5 min. The supernatant was separated and the remaining pellet was rinsed and resuspended in nuclear buffer before sonication. The insoluble nuclear fraction was analyzed by Western blotting. The expression of actin, GRB2, or p84 protein was used as loading controls for total, cytoplasmic, or nuclear fractions, respectively.

Cell Cycle Analysis and Apoptosis

Cellular apoptosis was scored as previously described (21) using a nuclear morphology end point. Both adherent and floating cells were collected from cell cultures following treatment with imatinib and gemcitabine or radiation and stained with 10 μmol/L Hoechst 33342 dye in 4% formalin-PBS for 30 min. Stained cells were visualized under a fluorescent microscope to look for distinct apoptotic bodies and nuclear fragmentation.

Cell cycle distributions pretreatment and posttreatment were quantitated by flow cytometry. Treated cells were collected, washed, and fixed in 70% ethanol. The cells were washed with magnesium- and calcium-free PBS before staining with 10 μg/mL propidium iodide and addition of 10 μg/mL DNase-free RNase. Flow cytometry was done using a FACSCalibur flow cytometer (BD Biosciences), and cell cycle distribution profiles were analyzed using CellQuest software (BD Biosciences).

To evaluate error-free HR repair following imatinib treatment, we used H1299 cells containing a DR-GFP construct as previously described (8). In this system, transient expression of an I-SceI endonuclease generates a DNA-dsb at integrated GFP-deletion gene sequences in which error-free repair leads to a full-length GFP product detectable by flow cytometry. H1299 cells were therefore transfected with pGFP (transfection efficiency control) or pCMV3xnlsI-SceI (functional endonuclease) or phCMV-1 I-SceI (negative control) and in the presence or absence of imatinib. GFP signal was quantitated at 3 d posttransfection on a FACSCalibur flow cytometer (BD Biosciences). For each experiment, 50,000 cells were scored per treatment group and the frequency of recombination events was calculated from the number of GFP-positive cells divided by the number of cells analyzed following correction for transfection efficiency as previously described (8).

Rad51-Validated Stealth RNAi DuoPak and the negative oligo (Invitrogen) were used at a concentration of 0.10 nmol/L for a 24-h transfection to obtain ∼50% down-regulation of RAD51 in the cells. Transfection of siRNA or negative oligo was achieved using Lipofectamine 2000 reagent (Invitrogen) as previously described (8).

Clonogenic Cell Survival In vitro

Cells were irradiated with doses between 0 and 10 Gy under aerobic conditions, at room temperature, using a 137Cs unit (Nordion Gammacell) at a dose rate of 1.03 Gy/min, as previously described (23). In other experiments, cells were treated with gemcitabine (Lilly; 0–100 nmol/L), MMC (0–750 nmol/L), or imatinib (Novartis; 0–50 μmol/L). Imatinib was reconstituted in acidified PBS (pH 5.3) to a stock concentration of 10 mmol/L and stored at −20°C. Gemcitabine and MMC were obtained at stock concentrations of 0.127 mol/L and 1.5 μmol/L and stored at room temperature and −4°C, respectively. Asynchronously growing cultures were washed with PBS, trypsinized, and plated at appropriate density into six-well tissue culture dishes before irradiation or treatment with chemotherapy. Dishes were treated for 16 to 18 h after plating to ensure a cell multiplicity of <1.1 (23). At 10 to 14 d posttreatment, dishes were stained with methylene blue-ethanol, and surviving colonies of >50 cells were scored under a dissection microscope. The surviving fraction for a given treatment dose was calculated as the relative plating efficiency of treated versus untreated (control) cultures. The ICD50 was defined by the concentration of drug that results in 50% clonogenic cell survival. For each cell line, three to six independent experiments were done.

Immunofluorescent Microscopy

Cells were fixed and permeabilized as previously described (24). Cells were stained with mouse monoclonal γ-H2AX antibody (Upstate) at 1:500 dilution or RAD51 antibody (Cell Signaling) at 1:200 dilution and counterstained with Alexa 488–conjugated antimouse secondary at 1:500 dilution (Molecular Bioprobes). DNA staining was accomplished with 4′,6-diamidino-2-phenylindole before microscopy. Cells were examined using either an Axiovert wide-field microscope under control of Northern Eclipse software or a Zeiss two-photon confocal microscope under control of LSM software as previously described (19).

In vivo Studies

To derive PC3 prostate xenografts for in vivo growth delay studies of combined treatments with imatinib and radiotherapy (IR), male BALB/c nude mice were injected s.c. in the flank with 1.5 × 106 cells. Drug and radiation treatments were started when xenografts reached a volume of about 150 to 200 mm3. Mice were randomly divided into four groups of 9 to 10 mice adjusted by initial tumor size and treated with (a) PBS alone, (b) imatinib 50 mg/kg i.p. daily for 8 d, (c) 5 × 4 Gy plus 8 d PBS i.p., and (d) 5 × 4 Gy plus 8 d of imatinib (IR starting after 3 d of pretreatment with imatinib). Animals were weighed and flank tumors measured twice a week with calipers; measurements were converted into tumor volume. Growth delay was calculated as the time difference (in days) between treatment and control groups to grow to 250 mm3. In a separate experiment, selected tumors were treated similarly and removed 4 h following a single dose of 4 Gy and prepared for Western blotting and paraffin-fixed sections for immunohistochemical staining for RAD51 expression (25).

Gut toxicity was determined using intestinal clonogenic survival in vivo using a modified protocol (26). Briefly, doses of whole-body irradiation at 0 to 18 Gy were administered to BALB/c mice following 50 mg/kg/d × 5 d of imatinib or PBS i.p. dosing. Three days later, the small intestine was removed, washed, and fixed in formalin; gut cross sections were stained with H&E before scoring surviving crypts in each treatment group. Two independent experiments were completed. Crypt radiation surviving fraction in vivo was calculated as the ratio of surviving crypts in the presence and absence of imatinib. All animal experiments were carried out in accordance with the Ontario Cancer Institute/Princess Margaret Hospital Animal Care Committee–approved protocol.

Statistical Analyses

Unless otherwise stated, quantitative experiments were conducted with a minimum of three independent experiments and data expressed as mean ± SE. Significant differences between experimental groups were compared using one-sided or paired t tests.

Imatinib Inhibits Nuclear RAD51 Expression in Tumor Cell Lines

We initially investigated whether RAD51 protein expression could be decreased by imatinib (Fig. 1; Supplementary Fig. S1).3

3

Supplementary material for this article is available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

A concentration of 10 or 20 μmol/L imatinib (reflecting an inhibition of 50% survival; ICD50) significantly decreased RAD51 protein expression in a time-dependent manner in all cell lines by 40% to 50% (P < 0.05; Fig. 1A). RAD51 expression at 72 hours was markedly lower than at earlier time points independent of p53 status or histologic cell type. In contrast, there was no effect on Ku70 with similar treatment (Fig. 1A). Although RAD51 is differentially expressed in the S and G2 phases of the cell cycle (reviewed in ref. 4), the imatinib-mediated decrease in RAD51 was not be explained by altered S-phase fraction because cell cycle distributions remained unchanged following imatinib treatment (Supplementary Fig. S1C).3

Figure 1.

Imatinib inhibition of nuclear RAD51 expression in RT112 and other tumor cell lines. A,top, Western blots showing decreased RAD51 protein, but not Ku70 protein, in RT112 and PC3 cells following treatment with ICD50 concentration of imatinib (20 μmol/L for RT112 and 10 μmol/L for PC3) for up to 72 h. Actin is shown as a loading control. Bottom, Western blots showing decreased RAD51 and Ku70 protein at 72 h in four tumor cell lines were treated with ICD50 concentrations of imatinib (RT112, 20 μmol/L; H1299, 10 μmol/L; PC3, 10 μmol/L; PANC1, 5 μmol/L). Columns, mean band intensity based on three independent experiments; bars, SE. *, P < 0.05, significant decreases in RAD51 expression in cells treated with imatinib. Ku70 expression was not significantly altered. B, immunoprecipitation (IP)-Western blots for RAD51 or c-Abl in RT112 and H1299 cell lysates following imatinib treatment. Imatinib inhibited cAbl-RAD51 complex formation post-IR. RT112 and H1299 cells were pretreated with imatinib for 48 h before lysing at 1 h following irradiation (20 Gy). C, relative expression of RAD51 in insoluble nuclear (chromatin) extracts of RT112, PC-3, and H1299 cells treated with ICD50 concentrations of imatinib (RT112, 20 μmol/L; H1299, 10 μmol/L; PC3, 10 μmol/L) before and after 10-Gy irradiation. Shown also are actin loading controls. Columns, mean for three independent experiments; bars, SE. *, P < 0.05, significant decreases in RAD51 expression in cells treated with radiation plus imatinib experiments versus radiation alone. D, microscopic analyses of RT112 cells treated with 20 μmol/L imatinib and stained for γ-H2AX or RAD51. Note increased RAD51 protein sequestered in cytoplasm following imatinib treatment.

Figure 1.

Imatinib inhibition of nuclear RAD51 expression in RT112 and other tumor cell lines. A,top, Western blots showing decreased RAD51 protein, but not Ku70 protein, in RT112 and PC3 cells following treatment with ICD50 concentration of imatinib (20 μmol/L for RT112 and 10 μmol/L for PC3) for up to 72 h. Actin is shown as a loading control. Bottom, Western blots showing decreased RAD51 and Ku70 protein at 72 h in four tumor cell lines were treated with ICD50 concentrations of imatinib (RT112, 20 μmol/L; H1299, 10 μmol/L; PC3, 10 μmol/L; PANC1, 5 μmol/L). Columns, mean band intensity based on three independent experiments; bars, SE. *, P < 0.05, significant decreases in RAD51 expression in cells treated with imatinib. Ku70 expression was not significantly altered. B, immunoprecipitation (IP)-Western blots for RAD51 or c-Abl in RT112 and H1299 cell lysates following imatinib treatment. Imatinib inhibited cAbl-RAD51 complex formation post-IR. RT112 and H1299 cells were pretreated with imatinib for 48 h before lysing at 1 h following irradiation (20 Gy). C, relative expression of RAD51 in insoluble nuclear (chromatin) extracts of RT112, PC-3, and H1299 cells treated with ICD50 concentrations of imatinib (RT112, 20 μmol/L; H1299, 10 μmol/L; PC3, 10 μmol/L) before and after 10-Gy irradiation. Shown also are actin loading controls. Columns, mean for three independent experiments; bars, SE. *, P < 0.05, significant decreases in RAD51 expression in cells treated with radiation plus imatinib experiments versus radiation alone. D, microscopic analyses of RT112 cells treated with 20 μmol/L imatinib and stained for γ-H2AX or RAD51. Note increased RAD51 protein sequestered in cytoplasm following imatinib treatment.

Close modal

The coimmunoprecipitation of c-ABL/RAD51 complexes following radiation was also decreased by imatinib treatment (Fig. 1B). Given the effect of imatinib on RAD51 levels, we investigated the effect of imatinib on the pool of chromatin-bound RAD51 by fractionating imatinib-treated cells into a nuclear-insoluble protein pool (Fig. 1C; Supplementary Fig. S1A and B)3 and by observing RAD51 subcellular localization using immunofluorescent microscopy (Fig. 1D). We observed that imatinib significantly decreased RAD51 protein in the chromatin-bound fraction in two of the three irradiated tumor cell lines (P < 0.05) with a strong trend to decrease in the remaining cell line (Fig. 1C; Supplementary Fig. S1A and B).3 Microscopy experiments showed increased nuclear extrusion of RAD51 in imatinib-treated cells (Fig. 1D). We conclude that treatment with ICD50 concentrations of imatinib leads to decreased RAD51 nuclear expression in tumor cell lines in a cell cycle–, p53-, and histology-independent manner.

Imatinib Decreases Error-Free Homologous Recombination

To determine whether the decrease in nuclear RAD51 protein expression led to altered error-free HR, we used a specialized H1299 lung cancer cell model that contains an integrated DR-GFP HR reporter system (8). The reporter construct contains a GFP gene, which is interrupted by a rare restriction site (I-SceI), and a downstream GFP fragment. Cells were transfected with a plasmid encoding for either a negative control, GFP (transfection efficiency control), or the endonuclease to generate a specific DNA-dsb. In this assay, functional GFP can only be restored if the DNA-dsb is repaired in an error-free manner using the downstream GFP fragment as a template for HR (see Fig. 2A; refs. 8, 27). We tested the HR frequency in H1299 cells following treatment with imatinib (see Fig. 2A and B). The results in Fig. 2B showed that RAD51 protein was significantly reduced to 40% to 70% following treatment with 10 or 20 μmol/L of imatinib (P < 0.05). In a parallel assay, 10 and 20 μmol/L of imatinib decreased the frequency of recombination to 62% and 77%, respectively (P < 0.05), supporting a concordance between RAD51 expression and HR following imatinib treatment.

Figure 2.

Imatinib decreases error-free HR. A, to evaluate error-free HR repair of DNA-dsbs, H1299 cells containing a DR-GFP construct were transfected with pGFP (transfection efficiency control), pCMV3xnlsI-SceI (functional endonuclease), or phCMV-1 I-SceI (negative control) and placed under 21% or 0.2% O2 conditions. Transient expression of I-SceI endonuclease generates a DNA-dsb at the integrated GFP gene sequences and stimulates HR. GFP signal was assayed at 3 d posttransfection on a FACSCalibur flow cytometer (BD Biosciences). For each experiment, 50,000 cells were scored per treatment group and the frequency of recombination events was calculated from the number of GFP-positive cells divided by the number of cells analyzed following correction for transfection efficiency. Shown on the left is the schema for production of GFP+ cells, and on the right are representative dot plots from flow cytometry with a reduction of HR events in imatinib-treated cells. B, data from DR-GFP flow analysis showing reduction in HR frequency (white columns) and RAD51 expression based on Western blotting (gray columns) following imatinib pretreatment at 10 or 20 μmol/L. Columns, mean for three independent experiments; bars, SE. Also shown is Western blot illustrating reduction in RAD51 protein levels. *, P < 0.05, significant decreases in RAD51 expression in imatinib-treated cells. C, siRNA to Rad51 leads to decreased recombination (based on DR-GFP assay) and clonogenic survival following IR (2 Gy) or MMC (0.5 μg/mL) treatment. The decrease in RAD51 levels attained with siRNA is similar to that achieved with imatinib treatment (∼50% reduction) as in B.

Figure 2.

Imatinib decreases error-free HR. A, to evaluate error-free HR repair of DNA-dsbs, H1299 cells containing a DR-GFP construct were transfected with pGFP (transfection efficiency control), pCMV3xnlsI-SceI (functional endonuclease), or phCMV-1 I-SceI (negative control) and placed under 21% or 0.2% O2 conditions. Transient expression of I-SceI endonuclease generates a DNA-dsb at the integrated GFP gene sequences and stimulates HR. GFP signal was assayed at 3 d posttransfection on a FACSCalibur flow cytometer (BD Biosciences). For each experiment, 50,000 cells were scored per treatment group and the frequency of recombination events was calculated from the number of GFP-positive cells divided by the number of cells analyzed following correction for transfection efficiency. Shown on the left is the schema for production of GFP+ cells, and on the right are representative dot plots from flow cytometry with a reduction of HR events in imatinib-treated cells. B, data from DR-GFP flow analysis showing reduction in HR frequency (white columns) and RAD51 expression based on Western blotting (gray columns) following imatinib pretreatment at 10 or 20 μmol/L. Columns, mean for three independent experiments; bars, SE. Also shown is Western blot illustrating reduction in RAD51 protein levels. *, P < 0.05, significant decreases in RAD51 expression in imatinib-treated cells. C, siRNA to Rad51 leads to decreased recombination (based on DR-GFP assay) and clonogenic survival following IR (2 Gy) or MMC (0.5 μg/mL) treatment. The decrease in RAD51 levels attained with siRNA is similar to that achieved with imatinib treatment (∼50% reduction) as in B.

Close modal

We compared this effect of imatinib on RAD51 expression and HR with the effect of RAD51 knockdown in the same cell line using siRNA treatment. RAD51 knockdown led to a 40% to 50% decrease in RAD51 protein expression (similar to that achieved by imatinib treatment; compare Fig. 2B and C), reduced HR by 30% to 40%, and increased sensitivity to IR and MMC. We conclude that imatinib and RAD51-siRNA are both capable of inhibiting HR in the H1299/DR-GFP cellular model.

Imatinib Increases the Chemosensitivity and Radiosensitivity of Cancer Cell Lines in Part by Mitotic Catastrophe

Given the decreased RAD51 expression and function in selected tumor cell lines, we determined whether the ability of imatinib to render tumor cells deficient in RAD51 expression led to increased tumor cell increased sensitivity to IR, MMC, or gemcitabine, agents that may selectively be toxic to DNA-dsb repair–deficient cells (2, 4, 7, 23). We observed that imatinib also reduced RAD51 expression following an initial IR or gemcitabine treatment in vitro (see Fig. 3A and B). Tumor cells treated with imatinib and IR, MMC, or gemcitabine had significantly decreased clonogenic survival (P < 0.05) compared to cells treated with IR or chemotherapy alone (Fig. 3B and C). The effect of imatinib as a chemosensitizer was more pronounced in tumor cells compared with normal diploid fibroblasts (P < 0.05; Fig. 3D).

Figure 3.

Imatinib increases the chemosensitivity and radiosensitivity of RT112 and other cancer cell lines. A,top, imatinib decreases RAD51 expression in the presence and absence of 2-Gy IR in RT112 cells (top). Cells were preincubated for 24 h with 20 μmol/L ICD50 imatinib and then irradiated under 2-Gy IR. Cells were harvested at 24 h post-IR for Western blotting. Actin is shown as a loading control. Bottom, imatinib decreases RAD51 expression in the presence and absence of gemcitabine in RT112 cells. Cells were pretreated for 24 h with imatinib 20 mmol/L followed by gemcitabine 6.5 nmol/L for 3 h. Actin is shown as a loading control. B, clonogenic survival of RT112 and PC3 cells under IR with (solid line) or without (dashed line) imatinib exposure for 24 h (ICD50 concentrations). Points, mean survival for three independent experiments; bars, SE. *, P < 0.05, significant differences in survival with imatinib-radiation treatment compared with radiation alone. DMF, dose modifying factor (based on relative dose to achieve 10% survival). C, clonogenic survival in RT112 and PC3 cells following treatment with imatinib (IMT; ICD50 concentrations) and 0.5 μg/mL MMC (1 h). Columns, mean of three to seven independent experiments; bars, SE. *, P < 0.05, significant differences in survival with imatinib-drug treatment versus drug alone. D, clonogenic survival of normal fibroblasts (GM05757 cells) versus tumor cell lines following treatment with gemcitabine (GEM) and imatinib (alone or in combination). For each cell line or strain, cells were plated and treated with 10 μmol/L imatinib and 6.5 nmol/L gemcitabine to allow for direct comparison between normal and malignant cells. Columns, mean survival values of three independent experiments; bars, SE. *, P < 0.05, significant differences in survival with combined treatment for tumor cell lines compared with survival in fibroblasts.

Figure 3.

Imatinib increases the chemosensitivity and radiosensitivity of RT112 and other cancer cell lines. A,top, imatinib decreases RAD51 expression in the presence and absence of 2-Gy IR in RT112 cells (top). Cells were preincubated for 24 h with 20 μmol/L ICD50 imatinib and then irradiated under 2-Gy IR. Cells were harvested at 24 h post-IR for Western blotting. Actin is shown as a loading control. Bottom, imatinib decreases RAD51 expression in the presence and absence of gemcitabine in RT112 cells. Cells were pretreated for 24 h with imatinib 20 mmol/L followed by gemcitabine 6.5 nmol/L for 3 h. Actin is shown as a loading control. B, clonogenic survival of RT112 and PC3 cells under IR with (solid line) or without (dashed line) imatinib exposure for 24 h (ICD50 concentrations). Points, mean survival for three independent experiments; bars, SE. *, P < 0.05, significant differences in survival with imatinib-radiation treatment compared with radiation alone. DMF, dose modifying factor (based on relative dose to achieve 10% survival). C, clonogenic survival in RT112 and PC3 cells following treatment with imatinib (IMT; ICD50 concentrations) and 0.5 μg/mL MMC (1 h). Columns, mean of three to seven independent experiments; bars, SE. *, P < 0.05, significant differences in survival with imatinib-drug treatment versus drug alone. D, clonogenic survival of normal fibroblasts (GM05757 cells) versus tumor cell lines following treatment with gemcitabine (GEM) and imatinib (alone or in combination). For each cell line or strain, cells were plated and treated with 10 μmol/L imatinib and 6.5 nmol/L gemcitabine to allow for direct comparison between normal and malignant cells. Columns, mean survival values of three independent experiments; bars, SE. *, P < 0.05, significant differences in survival with combined treatment for tumor cell lines compared with survival in fibroblasts.

Close modal

Given the results of the clonogenic assays, we characterized the expression of the DNA-dsb biomarker γ-H2AX in imatinib-treated cells. Using an apoptotic assay, we found no evidence that imatinib altered the apoptotic response in the cell lines treated with IR or chemotherapy. For example, the AI for RT112 cells with radiation was 8% after 10 Gy at 72 hours and 6% after treatment with gemcitabine at 72 hours (data not shown). However, treatment with the combination of gemcitabine or IR and imatinib resulted in increased residual γ-H2AX signal and mitotic catastrophe (see Fig. 4). We conclude that imatinib can sensitize tumor cells to DNA-dsb–inducing agents and induce mitotic catastrophe.

Figure 4.

Imatinib leads to mitotic catastrophe in RT112 cells treated with either gemcitabine or IR. A, microscopic images of γ-H2AX activation in RT112 cells treated with a combination of imatinib, gemcitabine, or IR (alone or in combination: 20 μmol/L imatinib and 6.5 nmol/L gemcitabine). Imatinib activated γ-H2AX in the absence of exogenous DNA damage. Shown also is 4′,6-diamidino-2-phenylindole-DNA staining to outline nuclei. B, microscopic images of γ-H2AX activation in RT112 cells treated with a combination of imatinib (20 μmol/L) and IR at 24 h posttreatment. In contrast to resolved γ-H2AX signal in irradiated RT112 cells, the addition of imatinib to irradiation led to fragmented mitotic nuclei and increased residual activation of γ-H2AX. There was little evidence for apoptosis.

Figure 4.

Imatinib leads to mitotic catastrophe in RT112 cells treated with either gemcitabine or IR. A, microscopic images of γ-H2AX activation in RT112 cells treated with a combination of imatinib, gemcitabine, or IR (alone or in combination: 20 μmol/L imatinib and 6.5 nmol/L gemcitabine). Imatinib activated γ-H2AX in the absence of exogenous DNA damage. Shown also is 4′,6-diamidino-2-phenylindole-DNA staining to outline nuclei. B, microscopic images of γ-H2AX activation in RT112 cells treated with a combination of imatinib (20 μmol/L) and IR at 24 h posttreatment. In contrast to resolved γ-H2AX signal in irradiated RT112 cells, the addition of imatinib to irradiation led to fragmented mitotic nuclei and increased residual activation of γ-H2AX. There was little evidence for apoptosis.

Close modal

Combined Imatinib and Experimental Radiotherapy Leads to Sensitization in a PC-3 Prostate Cancer Xenograft Model

To support the in vitro imatinib-IR sensitization data in PC-3 cells, we used a PC3 prostate xenograft model to test whether we could observe radiosensitization in vivo following a combination of imatinib and experimental fractionated radiotherapy (Fig. 5). We also tested whether the combination of radiation plus imatinib would be toxic in an associated normal tissue pelvic irradiation model based on intestinal crypt cell survival.

Figure 5.

Imatinib increases radiotherapy-induced tumor growth delay without increasing intestinal toxicity in a PC-3 prostate cancer xenograft. A, intestinal crypt cell survival in BALB/c mice following 0 to 18 Gy in vivo in the presence or absence of 50 mg/kg/d for 5 d. Surviving crypts were scored 3 d after irradiation based on three mice per treatment group. Points, mean survival; bars, SD. Inset, H&E-stained section of intestinal crypts before and after 14 Gy. B, Western blot of RAD51 expression from lysates prepared from sample PC3 xenografts treated with PBS alone, imatinib (50 mg/kg/d × 4 d), 4 Gy, or 4 Gy plus imatinib at 4 h following treatment. Actin is shown as a loading control. C, immunohistochemical staining for RAD51 in sample PC3 xenografts treated with PBS alone, imatinib (50 mg/kg/d × 4 d), 4 Gy, or 4 Gy plus imatinib. D,top, plot of growth delay of PC3 xenografts treated with PBS alone, imatinib (50 mg/kg/d × 8 d) alone, 4 Gy × 5 plus PBS, or 4 Gy × 5 plus imatinib. Irradiated mice were either pretreated with 3 d PBS (sham) or 3 d imatinib (50 mg/kg/d i.p.) before irradiation and received daily PBS or imatinib i.p. dosing during 4-Gy daily radiation fractions for a total treatment period of 8 d similar to drug alone. For clarity purposes, shown is the median animal per group. The calculated mean growth delay based on 9 to 10 mice per group was 100 ± 7 d for the combined treatment group, compared with 74 ± 14 d for the radiotherapy alone group (P = 0.003). There was no difference in growth delay between the imatinib alone and control groups. Bottom, plot of median body weight over time for the treatment groups in D. There were not significant differences in body weight among the treatment groups.

Figure 5.

Imatinib increases radiotherapy-induced tumor growth delay without increasing intestinal toxicity in a PC-3 prostate cancer xenograft. A, intestinal crypt cell survival in BALB/c mice following 0 to 18 Gy in vivo in the presence or absence of 50 mg/kg/d for 5 d. Surviving crypts were scored 3 d after irradiation based on three mice per treatment group. Points, mean survival; bars, SD. Inset, H&E-stained section of intestinal crypts before and after 14 Gy. B, Western blot of RAD51 expression from lysates prepared from sample PC3 xenografts treated with PBS alone, imatinib (50 mg/kg/d × 4 d), 4 Gy, or 4 Gy plus imatinib at 4 h following treatment. Actin is shown as a loading control. C, immunohistochemical staining for RAD51 in sample PC3 xenografts treated with PBS alone, imatinib (50 mg/kg/d × 4 d), 4 Gy, or 4 Gy plus imatinib. D,top, plot of growth delay of PC3 xenografts treated with PBS alone, imatinib (50 mg/kg/d × 8 d) alone, 4 Gy × 5 plus PBS, or 4 Gy × 5 plus imatinib. Irradiated mice were either pretreated with 3 d PBS (sham) or 3 d imatinib (50 mg/kg/d i.p.) before irradiation and received daily PBS or imatinib i.p. dosing during 4-Gy daily radiation fractions for a total treatment period of 8 d similar to drug alone. For clarity purposes, shown is the median animal per group. The calculated mean growth delay based on 9 to 10 mice per group was 100 ± 7 d for the combined treatment group, compared with 74 ± 14 d for the radiotherapy alone group (P = 0.003). There was no difference in growth delay between the imatinib alone and control groups. Bottom, plot of median body weight over time for the treatment groups in D. There were not significant differences in body weight among the treatment groups.

Close modal

Figure 5A shows that imatinib at 50 mg/kg/d for 5 days did not lead to increased radiation gut toxicity. Studies using the PC-3 xenografts showed decreased RAD51 protein expression in vivo based on Western blotting or immunohistochemical staining of RAD51 protein of PC3 cells within irradiated xenografts (Fig. 5B and C). The combination of imatinib and fractionated radiotherapy (five fractions of 4 Gy) led to an increased growth delay (100 ± 7 days) compared with radiotherapy alone (74 ± 14 days; P = 0.003; Fig. 5D). There was no significant growth delay in the imatinib-alone treatment group. Over the duration of the growth delay experiment, we did not observe significant differences in body weight among the treatment groups (Fig. 5D).

Imatinib has been used in the clinic for a number of years in the setting of chronic myelogenous leukemia and rare gastric sarcomas and is considered to be safe and well tolerated (24). Previous studies have shown that imatinib can reduce the levels of cellular RAD51, a pivotal protein involved in DNA-dsb repair, leading to enhanced radiosensitization (28, 29). However, whether this was secondary to functional decreases in HR was not studied. The latter is important to choose the most effective therapies to combine with imatinib and lead to the development of additional biomarkers relating to HR. Our data suggest that imatinib can decrease functional HR leading to increased chemosensitivity and radiosensitivity in vitro and in vivo without undue toxicity toward normal cells and tissues; we believe that this effect is, in part, optimizing the therapeutic ratio for these combined treatments.

Imatinib can inhibit c-ABL, platelet-derived growth factor receptor, mitogen-activated protein kinase, and c-KIT signaling pathways to decrease interstitial fluid pressure and angiogenesis in vivo, but there are also reports of altered DNA repair (3038). Additionally, c-ABL has also been shown to be involved in the transcription and posttranslational modification of RAD51 (17). Because the platelet-derived growth factor-mitogen-activated protein kinase pathway is not always intact in malignant cell lines, our results suggest that our observed increases in cytotoxicity in vitro and in vivo following combined treatment with imatinib and DNA damaging agents may be due, in part, to inhibition of DNA repair (30, 31). Indeed, initial Western blotting for phosphorylated platelet-derived growth factor receptor and mitogen-activated protein kinase among the tumor cell lines in this study following treatment with imatinib was variable from cell line to cell line (data not shown). In contrast, RAD51 was uniformly reduced in all cell lines after 72 hours. This was not due to differential mRNA expression of the Rad51 gene (data not shown) and may therefore be secondary to imatinib effects on RAD51 translation or posttranslational modification. A further mechanism for reduced HR could stem from our observation that RAD51 expression was specifically inhibited within the cell nucleus rather than the cytoplasm. Recent reports confirm that intracellular trafficking of the RAD51 protein and functional HR can be modified by insulin-like growth factor type I receptor or hepatocyte growth factor (MET) signaling pathways (39, 40). Future research using defined RAD51 or signaling isogenic systems will be required to test whether these mechanisms are operational in imatinib-treated cells.

In the presence of decreased RAD51, IR or chemotherapy combined with imatinib was shown to significantly decrease tumor cell survival. Ku70 levels were not affected by imatinib in this study. This suggests that a differential effect between HR and NHEJ could be exploited in radiotherapy protocols because these pathways may be differentially used in malignant cells and normal cells, respectively (reviewed in ref. 4). Indeed, the survival of the normal fibroblasts was higher following imatinib treatment in comparison with tumor cells, and we speculate that this is due to the increased number of cells in the G1 phase in the GM05757 strain (∼85%), which is preferential to NHEJ-directed DNA repair. The increased cell kill in tumor cells would be secondary to higher S and G2 fractions in which HR inhibition would be maximal (4).

Previous studies have shown enhanced radiosensitization in glioblastoma cell lines and xenografts (29, 34, 35). Podtcheko et al. (36) have shown that imatinib enhanced radiosensitivity in anaplastic thyroid cells both in vitro and in vivo. They concluded that not only inhibition of proliferative cell growth but also terminal growth arrest and senescence were important (36). Senescence refers to irreversible arrest and loss of clonogenicity. Mitotic cell death occurs in cells with abnormal cell cycling and defective apoptosis with irradiation or chemotherapy (37). A recent study by Rink et al. (32) has implicated NBS1, a key protein in the MRE11-NBS1-RAD50 complex involved in DNA-dsb signaling and cell cycle control, in leukemic cell killing following imatinib treatment (33). In this study, the treatment resistance of BCR/ABL-positive leukemic cells was reversed by imatinib through inhibition of NBS1 and cell cycle arrest, leading to decreased capacity for DNA repair following DNA damage. Whether our combined treatment results can be explained by effects on the NBS1 pathway will require specific study, but they are consistent with our observation of increased mitotic catastrophe following imatinib treatment in combination with chemotherapy or radiotherapy.

Recently, Bertino et al. (38) investigated the combination of gemcitabine or premetrexed with imatinib in mesothelioma cells and found the combinations to be synergistic. Gemcitabine is used in the treatment of lung, pancreas, and bladder cancer. Interestingly, in this study, these cell lines showed a greater effect with combination gemcitabine and imatinib treatment when compared with the prostatic cell line. Although further study is required with multiple cell lines within each histopathologic category in vitro and in vivo, we cautiously suggest that the combination may be optimal in patients with lung, pancreas, or bladder cancer. In the United Kingdom, a phase II trial of concurrent gemcitabine with radiotherapy in the treatment of bladder cancer (radiation therapy weekly with gemcitabine)4

has just closed and results are awaited with interest. If the gemcitabine and radiotherapy combination is shown to be more efficacious than radiotherapy alone in a phase III trial, then future studies may look at the outcome of trimodality treatment using radiation, chemotherapy, and imatinib, with possible further improvements in clinical outcome and reduced toxicity compared with current treatments. Finally, given the increasing data that RAD51 overexpression is associated with malignancy and can lead to chemoresistance and radioresistance (41), HR-related biomarkers such as RAD51 nuclear expression could potentially be used as biomarkers of efficacy in trials testing HR-targeted therapies (42).

The authors received grant support from AstraZeneca-KuDOS, Xceed, and SuperGen for preclinical research. No other potential conflicts of interest were disclosed.

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

Note: A. Choudhury and H. Zhao contributed equally to this work.

We thank Carla Coackley for her technical expertise.

1
Allen C, Halbrook J, Nickoloff JA. Interactive competition between homologous recombination and non-homologous end joining.
Mol Cancer Res
2003
;
1
:
913
–20.
2
Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA. DNA repair pathways as targets for cancer therapy.
Nat Rev Cancer
2008
;
8
:
193
–204.
3
Fan R, Kumaravel TS, Jalali F, et al. Defective DNA strand break repair after DNA damage in prostate cancer cells: implications for genetic instability and prostate cancer progression.
Cancer Res
2004
;
64
:
8526
– 33.
4
Bristow RG, Ozcelik H, Jalali F, Chan N, Vesprini D. Homologous recombination and prostate cancer: a model for novel DNA repair targets and therapies.
Radiother Oncol
2007
;
83
:
220
–30.
5
Harima Y, Sawada S, Miyazaki Y, et al. Expression of Ku80 in cervical cancer correlates with response to radiotherapy and survival.
Am J Clin Oncol
2003
;
26
:
e80
–85.
6
Wilson CR, Davidson SE, Margison GP, et al. Expression of Ku70 correlates with survival in carcinoma of the cervix.
Br J Cancer
2000
;
83
:
1702
– 6.
7
Choudhury A, Cuddihy A, Bristow RG. Radiation and new molecular agents part I: targeting ATM-ATR checkpoints, DNA repair, and the proteasome.
Semin Radiat Oncol
2006
;
16
:
51
–8.
8
Chan N, Koritzinsky M, Zhao H, et al. Chronic hypoxia decreases synthesis of homologous recombination proteins to offset chemoresistance and radioresistance.
Cancer Res
2008
;
68
:
605
–14.
9
Ashworth A. A synthetic lethal therapeutic approach: poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair.
J Clin Oncol
2008
;
26
:
3785
–90.
10
Bryant HE, Schultz N, Thomas HD, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase.
Nature
2005
;
434
:
913
–7.
11
Helleday T, Lo J, van Gent DC, Engelward BP. DNA double-strand break repair: from mechanistic understanding to cancer treatment.
DNA Repair (Amst)
2007
;
6
:
923
–35.
12
Wang YY, Maher VM, Liskay RM, McCormick JJ. Carcinogens can induce homologous recombination between duplicated chromosomal sequences in mouse L cells.
Mol Cell Biol
1988
;
8
:
196
–202.
13
Wachters FM, van Putten JW, Maring JG, et al. Selective targeting of homologous DNA recombination repair by gemcitabine.
Int J Radiat Oncol Biol Phys
2003
;
57
:
553
–62.
14
Slupianek A, Hoser G, Majsterek I, et al. Fusion tyrosine kinases induce drug resistance by stimulation of homology-dependent recombination repair, prolongation of G(2)/M phase, and protection from apoptosis.
Mol Cell Biol
2002
;
22
:
4189
–201.
15
Qiao GB, Wu YL, Yang XN, et al. High-level expression of Rad51 is an independent prognostic marker of survival in non-small-cell lung cancer patients.
Br J Cancer
2005
;
93
:
137
–43.
16
Chen G, Yuan SS, Liu W, et al. Radiation-induced assembly of Rad51 and Rad52 recombination complex requires ATM and c-Abl.
J Biol Chem
1999
;
274
:
12748
–52.
17
Slupianek A, Schmutte C, Tombline G, et al. BCR/ABL regulates mammalian RecA homologs, resulting in drug resistance.
Mol Cell
2001
;
8
:
795
–806.
18
Kubler HR, van Randenborgh H, Treiber U, et al. In vitro cytotoxic effects of imatinib in combination with anticancer drugs in human prostate cancer cell lines.
Prostate
2005
;
63
:
385
–94.
19
Al Rashid ST, Dellaire G, Cuddihy A, et al. Evidence for the direct binding of phosphorylated p53 to sites of DNA breaks in vivo.
Cancer Res
2005
;
65
:
10810
–21.
20
Meng AX, Jalali F, Cuddihy A, et al. Hypoxia down-regulates DNA double strand break repair gene expression in prostate cancer cells.
Radiother Oncol
2005
;
76
:
168
–76.
21
Bromfield GP, Meng A, Warde P, Bristow RG. Cell death in irradiated prostate epithelial cells: role of apoptotic and clonogenic cell kill.
Prostate Cancer Prostatic Dis
2003
;
6
:
73
–85.
22
Dignam JD, Lebovitz RM, Roeder RG. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei.
Nucleic Acids Res
1983
;
11
:
1475
–89.
23
Bristow RG, Hu Q, Jang A, et al. Radioresistant MTp53-expressing rat embryo cell transformants exhibit increased DNA-dsb rejoining during exposure to ionizing radiation.
Oncogene
1998
;
16
:
1789
–802.
24
Hensley ML, Ford JM. Imatinib treatment: specific issues related to safety, fertility, and pregnancy.
Semin Hematol
2003
;
40
:
21
–5.
25
Liu SK, Coackley C, Krause M, et al. A novel poly(ADP-ribose) polymerase inhibitor, ABT-888, radiosensitizes malignant human cell lines under hypoxia.
Radiother Oncol
2008
;
88
:
258
–68.
26
Roberts SA, Hendry JH, Potten CS. Intestinal crypt clonogens: a new interpretation of radiation survival curve shape and clonogenic cell number.
Cell Prolif
2003
;
36
:
215
–31.
27
Richardson C, Stark JM, Ommundsen M, Jasin M. Rad51 overexpression promotes alternative double-strand break repair pathways and genome instability.
Oncogene
2004
;
23
:
546
–53.
28
Aloyz R, Grzywacz K, Xu ZY, et al. Imatinib sensitizes CLL lymphocytes to chlorambucil.
Leukemia
2004
;
18
:
409
–14.
29
Russell JS, Brady K, Burgan WE, et al. Gleevec-mediated inhibition of Rad51 expression and enhancement of tumor cell radiosensitivity.
Cancer Res
2003
;
63
:
7377
–83.
30
Platanias LC. Map kinase signaling pathways and hematologic malignancies.
Blood
2003
;
101
:
4667
–79.
31
Mawrin C, Sasse T, Kirches E, et al. Different activation of mitogen-activated protein kinase and Akt signaling is associated with aggressive phenotype of human meningiomas.
Clin Cancer Res
2005
;
11
:
4074
–82.
32
Rink L, Slupianek A, Stoklosa T, et al. Enhanced phosphorylation of Nbs1, a member of DNA repair/checkpoint complex Mre11-50-Nbs1, can be targeted to increase the efficacy of imatinib mesylate against BCR/ABL-positive leukemia cells.
Blood
2007
;
110
:
651
–60.
33
D'Amours D, Jackson SP. The Mre11 complex: at the crossroads of DNA repair and checkpoint signalling.
Nat Rev Mol Cell Biol
2002
;
3
:
317
–27.
34
Geng L, Shinohara ET, Kim D, et al. STI571 (Gleevec) improves tumor growth delay and survival in irradiated mouse models of glioblastoma.
Int J Radiat Oncol Biol Phys
2006
;
64
:
263
–71.
35
Oertel S, Krempien R, Lindel K, et al. Human glioblastoma and carcinoma xenograft tumors treated by combined radiation and imatinib (Gleevec).
Strahlenther Onkol
2006
;
182
:
400
–7.
36
Podtcheko A, Ohtsuru A, Namba H, et al. Inhibition of ABL tyrosine kinase potentiates radiation-induced terminal growth arrest in anaplastic thyroid cancer cells.
Radiat Res
2006
;
165
:
35
–42.
37
Blank M, Shiloh Y. Programs for cell death: apoptosis is only one way to go.
Cell Cycle
2007
;
6
:
686
–95.
38
Bertino P, Porta C, Barbone D, et al. Preliminary data suggestive of a novel translational approach to mesothelioma treatment: imatinib mesylate with gemcitabine or pemetrexed.
Thorax
2007
;
62
:
690
–5.
39
Torjanek J, Del Valle L, Nowicki M, et al. Role of insulin-like growth factor I/insulin receptor substrate 1 axis in Rad51 trafficking and DNA repair by homologous recombination.
Mol Cell Biol
2003
;
23
:
7510
–24.
40
Ganapathipillai SS, Medova M, Aebersold DM, et al. Coupling of mutated Met variants to DNA repair via Abl and Rad51.
Cancer Res
2008
;
68
:
5769
–77.
41
Klein HL. The consequences of Rad51 overexpression for normal and tumor cells.
DNA Repair (Amst)
2008
;
7
:
686
–93.
42
Liu S, Olive P, Bristow RG. Biomarkers for DNA DSB inhibitors and radiotherapy trials.
Cancer Met Rev
2008
;
27
:
445
–58.

Supplementary data