PRKDC regulates chemosensitivity and is a potential prognostic and predictive marker of response to adjuvant chemotherapy in breast cancer patients

  • Authors:
    • Gang Sun
    • Le Yang
    • Chao Dong
    • Bin Ma
    • Meihui Shan
    • Binlin Ma
  • View Affiliations

  • Published online on: May 9, 2017     https://doi.org/10.3892/or.2017.5634
  • Pages: 3536-3542
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA-dependent kinase catalytic subunit (DNA-PKcs) is a critical component of DNA repair machinery and is found to be up- or down-regulated in different cancer types. However, its clinical significance in breast cancer remains unclear. To this end, quantitative PCR was performed to measure PRKDC expression level in 59 pairs of breast cancer tissues and the non-tumor adjacent tissues (NATs). The correlation between PRKDC expression and overall survival (OS) as well as the prognostic value of PRKDC were analyzed. In vitro and in vivo effects of PRKDC on chemosensitivity were evaluated in MCF-7 cells. We found that PRKDC expression was significantly increased in breast cancer tissue samples compared with NATs. High PRKDC expression was associated with higher tumor grade (P=0.001), positive lymph node metastasis (P=0.0357) and chemoresistance (P=0.0006). Furthermore, PRKDC expression was significantly correlated with OS in breast cancer patients with (0.0101) or without (P=0.0216) receiving chemotherapy. PRKDC was an independent prognostic factor of OS in breast cancer (P=0.022, hazard ratio=2.69, 95% confidence interval: 1.81-3.84). Moreover, downregulation of PRKDC sensitized MCF-7 cells to chemo-drugs both in vitro and in a xenografted mouse model. Collectively, our study demonstrated that PRKDC is a prognostic biomarker for chemoresistance in breast cancer patients.

Introduction

Breast cancer is the most common type of tumor among women and is the second leading cause of cancer-related mortality worldwide. It is estimated that, in 2016, approximately 250,000 new cases of breast cancer will be diagnosed and 40,890 deaths will result from breast cancer (1). Breast cancer is regarded as a systemic disease, with micrometastatic involvement at diagnosis in many patients. Although targeted therapies, including hormonal therapy in hormone receptor-positive and trastuzumab therapy in human epidermal growth factor receptor 2 (HER2)-positive patients are well established, chemotherapy remains an important mainstay in systemic treatment, especially in patients with high risk of relapse (24). Anthracycline-based chemotherapy regimens with the addition of taxanes into chemotherapy has improved survival outcome in the adjuvant setting (5). Unfortunately, long-term adverse effects and chemoresistance, pose challenges to further improving the efficacy of adjuvant breast cancer chemotherapy (6). Therefore, there is an urgent need to identify new reliable predictive biomarkers for chemosensitivity in breast cancer to improve disease management and patient survival.

PRKDC is a nuclear protein serine/threonine kinase that is activated upon association with DNA (7). It is a critical component of DNA repair machinery that plays a pivotal role in the DNA damage response (DDR), primarily double stranded break (DSB) response, and maintenance of genomic stability (8). In response to DSB formation, PRKDC is recruited to DSBs by the Ku70/80 heterodimer where it is rapidly activated after phosphorylation at multiple serine and threonine residues (9). Both aberrant expression or genetic mutations of PRKDC have been observed in various cancer types or pre-malignant cells, indicating that PRKDC may have paradoxically opposing roles in carcinogenesis, depending on the cell context or the tissue type (911). Moreover, due to its role in DDR, increased activity of PRKDC has been found to be associated with resistance to chemotherapeutic drugs and radiotherapy in glioma cells and oral squamous cell carcinoma, respectively (12,13). Therefore, it is plausible to hypothesize that inhibitors of PRKDC might be useful in sensitizing cancer cells to chemotherapy and radiotherapy (9,13).

In the present study, we evaluated the expression level of PRKDC in breast cancer patients receiving NAC and explored its potential as a prognostic biomarker as well as predictor of chemosensitivity in breast cancer.

Materials and methods

Patient and tissue samples

This study used archived material from Affiliated Cancer Hospital of Xinjiang Medical University admitted between March 2011 and December 2015, including breast cancer tissue samples from patients with stage IIIA-C disease for whom matching biopsies were available for pathological and immunohistochemical analysis. All tissues were immediately snap-frozen in liquid nitrogen and stored at −80°C until use. The patients with any other tumor were excluded from the study. A total of 159 breast cancer tissues and 59 matched non-tumor adjacent tissues (NATs) were examined in the study. None of the subjects had received any therapeutic procedures prior to this study, including surgery, chemotherapy, and radiotherapy. In addition, a total of 89 patients who received six cycles of an anthracycline based-therapy (FEC: 5-fluorouracil (5-FU) 500 mg/m2, epirubicin 75–100 mg/m2, cyclophosphamide 500 mg/m2, on day 1 of a 21-day cycle) were also included. All patients underwent surgery (mastectomy and axillary node clearance) 4 weeks after the last cycle of chemotherapy, followed by radiotherapy to the chest wall. The pathological complete response (pCR) was defined as the absence of any residual invasive carcinoma at both the primary site and in axillary lymph nodes. Patients with pCR were defined as responders whereas others were defined as non-responders. The prognosis was evaluated in all breast cancer patients in April 2015. Overall survival was defined as the time from cancer onset until death or by censoring at the last follow-up date. The study was approved by Affiliated Cancer Hospital of Xinjiang Medical University, and informed consent was obtained from all patients.

Total RNA extraction

Tissue sections were minced with scissors into small fragments (1–2 mm3) and homogenized with TRIzol™ reagent (Takara Bio, Inc., Otsu, Japan). Chloroform (200 µl; Sigma-Aldrich, Santa Clara, CA, USA) was added to the TRIzol homogenate. The preparations were then centrifuged at 12,000 × g for 15 min at 4°C, and the upper aqueous layer was transferred to a clean Eppendorf tube, containing an equal volume of isopropanol (Sigma-Aldrich). The mixed suspensions were centrifuged at 12,000 × g for a further 15 min at 4°C. The precipitations were then collected. After washing with 70% ethanol, total RNA was dissolved in RNase-free water and the quality of RNA was evaluated by gel electrophoresis. RNA concentrations were measured by optical density (260 nm, Q5000, Quawell, San Jose, CA, USA) and the preparations stored at −80°C for subsequent analysis.

RT-qPCR analysis

cDNA was reverse transcribed on the Bio-Rad S1000 Thermal Cycler (Bio-Rad Laboratories, Hercules, CA, USA) using oligo (dT) as primers. Briefly, the total RNA (1 µg) from each sample was reverse transcribed in a 20 µl reaction volume, containing 0.5 µg of oligo (dT) and 200 units M-MLV (MBI Fermentas, Vilnius, Lithuania). All samples were amplified in triplicate under the following conditions: 95°C for 2 min, 35 cycles of 95°C for 15 sec, 60°C for 30 sec and 72 C for 20 sec. qPCR reaction was performed on the Bio-Rad C1000 Real-time fluorescence thermal cycler (Bio-Rad Laboratories), using the following cycling conditions: Initiation at 95°C for 10 min; amplification for 35 cycles, with denaturation at 95°C for 30 sec; annealing at 56°C for 30 sec; and elongation at 72°C for 30 sec. A final extension at 72°C was performed for 10 min. GAPDH mRNA level was used for normalization.

Chemosensitivity assay

Cells were seeded at a density of 5×103 cells/well in 96-well microtiter plates and allowed to attach overnight. Chemo drugs were then added and cultured for an additional 72 h. Cell viability was assessed using CellTiter-Glo® assay (Promega, Madison, WI, USA). Each value was normalized to cells treated with DMSO and the IC50 values are calculated using Graphpad Prism software.

Viral transductions and stable selections

For lentivirus production, 1 µg of shPRKDC (Origene) together with 1 µg of helper plasmids (0.4 µg pMD2G and 0.6 µg psPAX2) were transfected into 293FT cells with Effectene reagent (Qiagen, Valencia, CA, USA). Viral supernatants were collected 48 h after transfections and cleared through a 0.45-µm filter. Cells were infected with viral supernatants containing 4 µg/ml polybrene (Sigma, St. Louis, MO, USA) and selected with puromycin for 7 days.

Immunohistochemistry (IHC) staining

The paraffin-embedded sections were subjected to antigen retrieval by heating the slides in a microwave at 100°C for 10 min in 0.1 M citric acid buffer (pH 6.0), and then incubated with corresponding antibodies at 4°C overnight. After secondary antibody incubation at room temperature for 1 h, the slides were developed in 0.05% diaminobenzidine containing 0.01% hydrogen peroxidase. For negative controls, specific antibodies were replaced with normal goat serum by co-incubation at 4°C overnight preceding the immunohistochemical staining procedure.

Xenograft experiments

All animal experiments were approved by Institutional Animal Care and Use Committee of National Cancer Center. MCF-7 cells (3×106 cells/injection) expressing control shRNA or PRKDC shRNA-2 were subcutaneously injected into both flanks of 5-week-old female nude mice. Vehicle or 5-FU (25 mg/kg) was injected i.p. into mice daily for 12 days. Tumor volumes were measured using caliper and determined by a formula [volume = (length × width2)/2] from day 6 to day 18 post-implantation. The results were expressed as mean tumor volumes with SD.

Statistical analysis

For cell culture and mouse experiments, quantitative data are expressed as mean ± SD. Statistical significance was assessed by the Student's t-test. Differences were considered to be significant when P<0.05. For patient sample data, statistical analysis was performed using IBM SPSS Statistics version 16 (SPSS Inc., Chicago, IL, USA) and GraphPad Prism v5.0 (Graphpad Software Inc.). The Wilcoxon test was used to compare PRKDC expression in paired tumor tissue samples and NATs. The Mann-Whitney U test was used to perform statistical analysis of PRKDC mRNA level between unpaired groups. The Pearson's Chi-squared test and Fisher's exact test were used to evaluate the association between tissue PRKDC mRNA level and clinicopathological parameters. In addition, survival curves were constructed with the Kaplan-Meier method and compared using log-rank test. Cox proportional hazards regression analysis was used for univariate and multivariate analyses of prognostic values. P-value of two-sided <0.05 was considered statistically significant.

Results

PRKDC expression is upregulated in breast cancer tissues

The expression level of PRKDC was evaluated in 59 pairs of breast cancer tissues and the NATs using quantitative real-time PCR. The relative mean expression value of PRKDC mRNA in cancer tissues (4.25±2.68, normalized to GAPDH gene expression) was significantly higher than that (1.96±1.42) in the corresponding normal tissues (P<0.010). Expression fold changes above 1 was considered as upregulation of PRKDC mRNA in the cancer tissues. The results revealed that 72.9% (43/59) of breast cancer tissues expressed a higher level of PRKDC compared with the matched normal tissues. We also measured the PRKDC expression levels in tissue samples from 44 breast cancer patients who responded to NAC and 45 patients who did not. We found that the expression levels of PRKDC were significantly higher in the non-responder group than the responder group (P<0.0001) (Fig. 1B).

Association between tissue PRKDC expression level and clinicopathological factors or chemotherapy response

The correlation between PRKDC expression level and clinicopathological factors or chemotherapy response was assessed using χ2 test. As shown in Table I, a total of 62 of 102 (60.7%) patients who were Grade II or III had significantly higher expression level of PRKDC than Grade I patients (P=0.001). We detected high PRKDC level in patients who were lymph node metastasis positive (P=0.0357). High PRKDC expression level was observed in 34% and 71.1% of patients who were responders or non-responders to neoadjuvant chemotherapy (NAC), respectively (P=0.0006). However, no significant correlation was observed between PRKDC expression levels and age (P=0.1541), tumor size (P=0.8736), tumor stage (P=0.3691), ER status (P=0.1354), PR status (P=0.191) or Her-2 status (P=0.4189).

Table I.

Correlation between tissue PRKDC expression level and clinicopathological factors or chemosensitivity.

Table I.

Correlation between tissue PRKDC expression level and clinicopathological factors or chemosensitivity.

CharacteristicsNo. of patientsPRKDC low expressionPRKDC high expressionP-value
Age (years) 0.1541
  ≤501175760
  >50  422616
Tumor size (cm) 0.8736
  ≤2  833944
  >2  763442
Tumor stage 0.3691
  I, II1186256
  III  412516
Tumor grade   0.0010a
  I  573819
  II, III1024062
Lymph node metastasis   0.0357a
  Negative  654025
  Positive  944153
ER status 0.1354
  Negative  562333
  Positive1035647
PR status 0.1910
  Negative  745024
  Positive  854837
Her-2 status 0.4189
  Negative  964848
  Positive  632736
Clinical response to NAC   0.0006a
  Responder  442915
  Non-responder  451332

a P<0.05, statistical significance. ER, estrogen receptor; PR, progesterone receptor.

High expression of PRKDC predicts poor prognosis in breast cancer patients with or without receiving NAC

Univariate and multivariate survival analyses were performed to examine the effects of clinicopathological factors and PRKDC expression on prognosis in patients receiving NAC. As shown in Table II, lymph node metastasis (P=0.044), tumor grade (P=0.038), PRKDC expression (P=0.011) and response to NAC (P=0.008) were significantly associated with overall survival (OS). Tumor grade (P=0.037, HR=2.16, 95% confidence interval: 1.35–3.04), PRKDC expression (P=0.022, HR=2.69, 95% confidence interval: 1.81–3.84) and response to NAC (P=0.014, HR=3.96, 95% confidence interval: 2.61–5.47) were independent predictors of OS in these patients (Table II).

Table II.

Univariate and multivariate analysis of factors associated with overall survival for patients with breast cancer (stage IIIA-C) who received NAC prior to surgery.

Table II.

Univariate and multivariate analysis of factors associated with overall survival for patients with breast cancer (stage IIIA-C) who received NAC prior to surgery.

Multivariate

VariablesUnivariate P-valueHR95% CIP-value
Age0.446 NA
Tumor size0.645 NA
Tumor stage0.297 NA
Lymph node metastasis   0.044a 0.079
Tumor grade   0.038a2.161.35–3.040.037a
ER status0.058 NA
PR status0.112 NA
Her-2 status0.265 NA
PRKDC expression
  Positive vs. negative   0.011a2.691.81–3.840.022a
Response to NAC
  Non-responder vs. responder   0.008a3.962.61–5.470.014a

a P<0.05, statistical significance. HR, hazard ratio; CI, confidence interval.

A survival analysis of OS was performed in patients with or without NAC treatment to determine whether PRKDC expression level can predict prognosis. The estimated Kaplan-Meier OS curves showed that high expression of PRKDC was significantly correlated with poor OS in patients without receiving NAC (P=0.0216) (Fig. 2A). In patients receiving NAC treatment, the OS was significantly worse in patients with high levels of PRKDC (P=0.0101) with a median survival of 30 months (Fig. 2B). These results indicate that high PRKDC expression was associated with poor OS in patients with or without NAC treatment and was an independent prognostic factor.

Downregulation of PRKDC sensitizes breast cancer cells to chemo-drugs in vitro and in vivo

To examine the regulatory role of PRKDC in chemoresistance in breast cancer cells, we generated stable PRKDC knockdown MCF-7 cell lines using two independent shRNAs targeting PRKDC. The knockdown efficiency was confirmed by qPCR (Fig. 3) and MCF-7 cells expressing shPRKDC-2 which exhibited better knockdown was used in the following experiments. Cytotoxicity of 4 commonly used chemo-drugs was evaluated in these cells by performing dose response analysis. As shown in Fig. 4, stable knockdown of PRKDC sensitized MCF-7 cells to all the drugs. In addition, downregulation of PRKDC significantly reduced tumor growth rate as well as tumor weight in MCF-7 xenografted mice receiving 5-FU whereas injection of 5-FU alone had negligible effects on tumor growth or tumor weight (Fig. 5A and B). Immunohistochemical staining showed that 5-FU treatment led to more induction of cleaved caspase-3 in PRKDC knockdown xenografts compared with control xenografts (Fig. 5C). Collectively, the results showed that PRKDC directly regulates chemosensitivity of breast cancer cells both in vitro and in vivo by enhancing drug-induced apoptotic cell death.

Discussion

PRKDC, a serine/threonine-protein kinase, is a member of the phosphatidylinositol 3-kinase-related kinase (PIKK) family and is abundantly expressed in almost all mammalian cells (14). It forms the DNAPK complex with DNA-binding Ku70/80 heterodimer which serves as a key regulator of the non-homologous end-joining (NHEJ) pathway (15). Additionally, PRKDC plays an important role in maintaining proper cell cycle progression by coordinating mitosis, microtubule dynamics and chromosomal segregation and has been shown to prevent mitotic catastrophe after ionization (9,16,17). Interestingly, both up- and down-regulation of PRKDC have been observed in various cancer types. For example, loss of PRKDC expression was found in gastric cancer, and was associated with advanced tumor stage, lymphatic invasion, lymph node metastasis and poor patient survival (18). Moreover, loss of PRKDC expression was shown to be predictive markers for poor prognosis of patients with gall bladder malignancies (19). On the other hand, PRKDC has been found to be upregulated associated with advanced clinical stages and poor prognosis in numerous tumor types (2022). An elevated expression of PRKDC has been observed in esophageal cancer tissues compared with adjacent normal mucosae (21). High tumor/normal expression ratio of PRKDC is correlated with increased risk of death in non-small cell lung cancer (22). An increased expression of PRKDC was observed in high-grade lymphoma lymph node samples by staining compared with those from low-grade lymphoma patients (23). However, the expression of PRKDC and its association with clinicopathological factors has not been investigated in breast cancer patients. In our study, we observed significantly higher levels of PRKDC in tumor tissues compared with NATs in 72.9% of the patients analyzed (Fig. 1A) and the mean expression level difference was significant (P<0.01). In addition, PRKDC expression levels were significantly correlated with tumor grade, lymph node metastasis, and response to NAC (Table I). Moreover, the breast cancer patient with elevated levels of PRKDC showed significantly worse OS than those with low PRKDC expression (Fig. 2A). These results suggest that PRKDC may function as an oncogene in breast cancer tumorigenesis. The potential mechanism by which PRKDC regulates metastasis is through mediation of transcriptional network or through regulation of secreted proteins involved in migration and invasion (24,25).

Due to its role in mediating DDR, aberrant expression/activation of PRKDC has been associated with chemo- or radio-resistance (26,27). A previous study showed that PRKDC regulated AKT activation and inhibited apoptosis in ovarian cancer cells with acquired platinum resistance (28,29). In oral squamous cell carcinoma, a significant upregulation of PRKDC proteins was detected after radiotherapy in the residual cancer cells and this upregulation was associated with radioresistance (13). On the contrary, inhibition of PRKDC sensitized cancer cells to radiotherapy and chemotherapy (27,30,31). For example, inhibition of PRKDC activity by a small molecule inhibitor, NU7441, sensitized breast cancer cells to ionizing radiation and doxorubicin (30). High expression of PRKDC was detected in glioma patients who were resistant to cisplatin-based chemotherapy and was shown to be a predictor for response to radiation therapy in esophageal cancer and early breast cancer (12,32,33).

Our study suggested an association between PRKDC expression and chemosensitivity in breast cancer. We found that the expression of PRKDC was elevated in tissue samples from patients who do not respond to NAC compared to those who respond (Fig. 1B). There was a significant correlation between PRKDC expression level and chemosensitivity in these patients (Table I). In addition, high PRKDC expression level predicted poor survival in patients receiving NAC treatment (Fig. 2B). Moreover, multivariate analysis demonstrated that PRKDC was an independent prognostic factor for OS in breast cancer patients receiving NAC (Table II). Importantly, knockdown of PRKDC sensitized MCF-7 breast cancer cells to chemo-drugs in vitro and in vivo with increased induction of apoptosis (Figs. 4 and 5), demonstrating the direct involvement of PRKDC in regulating chemosensitivity in breast cancer cells. These results also highlight the potential of PRKDC as a drug target for chemosensitization or as an anti-cancer therapeutic strategy.

In summary, our study showed that PRKDC regulates chemosensitivity in breast cancer cells and its expression was significantly associated with chemoresistance in patients receiving NAC. PRKDC may serve as prognostic biomarker for poor survival and a predictor for NAC response in breast cancer patients. Further studies are warranted to understand the precise mechanism underlying the role of PRKDCs in regulation of chemosensitivity which may lend support to the development of new therapeutic strategies to overcome chemoresistance in breast cancer.

Acknowledgements

The present study was funded by Natural Science Foundation of China (NSFC grant no. 81360391) and Youth Science and Technology Innovative Talent Training Project of Xinjiang (grant no. 2014721043).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Cuzick J, Sestak I, Baum M, Buzdar A, Howell A, Dowsett M and Forbes JF: ATAC/LATTE investigators: Effect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 10-year analysis of the ATAC trial. Lancet Oncol. 11:1135–1141. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Perez EA, Romond EH, Suman VJ, Jeong JH, Davidson NE, Geyer CE Jr, Martino S, Mamounas EP, Kaufman PA and Wolmark N: Four-year follow-up of trastuzumab plus adjuvant chemotherapy for operable human epidermal growth factor receptor 2-positive breast cancer: Joint analysis of data from NCCTG N9831 and NSABP B-31. J Clin Oncol. 29:3366–3373. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Slamon D, Eiermann W, Robert N, Pienkowski T, Martin M, Press M, Mackey J, Glaspy J, Chan A, Pawlicki M, et al: Breast Cancer International Research Group: Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med. 365:1273–1283. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Palmieri C and Jones A: The 2011 EBCTCG polychemotherapy overview. Lancet. 379:390–392. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Gonzalez-Angulo AM, Morales-Vasquez F and Hortobagyi GN: Overview of resistance to systemic therapy in patients with breast cancer. Adv Exp Med Biol. 608:1–22. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Jackson SP: DNA-dependent protein kinase. Int J Biochem Cell Biol. 29:935–938. 1997. View Article : Google Scholar : PubMed/NCBI

8 

Goodwin JF and Knudsen KE: Beyond DNA repair: DNA-PK function in cancer. Cancer Discov. 4:1126–1139. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Hsu FM, Zhang S and Chen BP: Role of DNA-dependent protein kinase catalytic subunit in cancer development and treatment. Transl Cancer Res. 1:22–34. 2012.PubMed/NCBI

10 

Zhang L, Komurov K, Wright WE and Shay JW: Identification of novel driver tumor suppressors through functional interrogation of putative passenger mutations in colorectal cancer. Int J Cancer. 132:732–737. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Zhang L, Kim S, Jia G, Buhmeida A, Dallol A, Wright WE, Fornace AJ, Al-Qahtani M and Shay JW: Exome sequencing of normal and isogenic transformed human colonic epithelial cells (HCECs) reveals novel genes potentially involved in the early stages of colorectal tumorigenesis. BMC Genomics. 16 Suppl 1:S82015. View Article : Google Scholar : PubMed/NCBI

12 

Mukherjee B, McEllin B, Camacho CV, Tomimatsu N, Sirasanagandala S, Nannepaga S, Hatanpaa KJ, Mickey B, Madden C, Maher E, et al: EGFRvIII and DNA double-strand break repair: A molecular mechanism for radioresistance in glioblastoma. Cancer Res. 69:4252–4259. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Shintani S, Mihara M, Li C, Nakahara Y, Hino S, Nakashiro K and Hamakawa H: Up-regulation of DNA-dependent protein kinase correlates with radiation resistance in oral squamous cell carcinoma. Cancer Sci. 94:894–900. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Hartley KO, Gell D, Smith GC, Zhang H, Divecha N, Connelly MA, Admon A, Lees-Miller SP, Anderson CW and Jackson SP: DNA-dependent protein kinase catalytic subunit: A relative of phosphatidylinositol 3-kinase and the ataxia telangiectasia gene product. Cell. 82:849–856. 1995. View Article : Google Scholar : PubMed/NCBI

15 

Davis AJ, Chen BP and Chen DJ: DNA-PK: A dynamic enzyme in a versatile DSB repair pathway. DNA Repair (Amst). 17:21–29. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Lee KJ, Lin YF, Chou HY, Yajima H, Fattah KR, Lee SC and Chen BP: Involvement of DNA-dependent protein kinase in normal cell cycle progression through mitosis. J Biol Chem. 286:12796–12802. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Shang ZF, Huang B, Xu QZ, Zhang SM, Fan R, Liu XD, Wang Y and Zhou PK: Inactivation of DNA-dependent protein kinase leads to spindle disruption and mitotic catastrophe with attenuated checkpoint protein 2 Phosphorylation in response to DNA damage. Cancer Res. 70:3657–3666. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Lee HS, Yang HK, Kim WH and Choe G: Loss of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) expression in gastric cancers. Cancer Res Treat. 37:98–102. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Ren F, Yang ZL, Tan X, Liu D, Zou Q, Yuan Y, Li J, Liang L, Zeng G and Chen S: DNA-PKcs and Ku70 are predictive markers for poor prognosis of patients with gall bladder malignancies. Appl Immunohistochem Mol Morphol. 22:741–747. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Hosoi Y, Watanabe T, Nakagawa K, Matsumoto Y, Enomoto A, Morita A, Nagawa H and Suzuki N: Up-regulation of DNA-dependent protein kinase activity and Sp1 in colorectal cancer. Int J Oncol. 25:461–468. 2004.PubMed/NCBI

21 

Tonotsuka N, Hosoi Y, Miyazaki S, Miyata G, Sugawara K, Mori T, Ouchi N, Satomi S, Matsumoto Y, Nakagawa K, et al: Heterogeneous expression of DNA-dependent protein kinase in esophageal cancer and normal epithelium. Int J Mol Med. 18:441–447. 2006.PubMed/NCBI

22 

Xing J, Wu X, Vaporciyan AA, Spitz MR and Gu J: Prognostic significance of ataxia-telangiectasia mutated, DNA-dependent protein kinase catalytic subunit, and Ku heterodimeric regulatory complex 86-kD subunit expression in patients with nonsmall cell lung cancer. Cancer. 112:2756–2764. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Holgersson A, Erdal H, Nilsson A, Lewensohn R and Kanter L: Expression of DNA-PKcs and Ku86, but not Ku70, differs between lymphoid malignancies. Exp Mol Pathol. 77:1–6. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Kotula E, Berthault N, Agrario C, Lienafa MC, Simon A, Dingli F, Loew D, Sibut V, Saule S and Dutreix M: DNA-PKcs plays role in cancer metastasis through regulation of secreted proteins involved in migration and invasion. Cell Cycle. 14:1961–1972. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Goodwin JF, Kothari V, Drake JM, Zhao S, Dylgjeri E, Dean JL, Schiewer MJ, McNair C, Jones JK, Aytes A, et al: DNA-PKcs-mediated transcriptional regulation drives prostate cancer progression and metastasis. Cancer Cell. 28:97–113. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Burdine LJ, Burdine MS, Moreland L, Fogel B, Orr LM, James J, Turnage RH and Tackett AJ: Proteomic identification of DNA-PK involvement within the RET signaling pathway. PLoS One. 10:e01279432015. View Article : Google Scholar : PubMed/NCBI

27 

Elliott SL, Crawford C, Mulligan E, Summerfield G, Newton P, Wallis J, Mainou-Fowler T, Evans P, Bedwell C, Durkacz BW, et al: Mitoxantrone in combination with an inhibitor of DNA-dependent protein kinase: A potential therapy for high risk B-cell chronic lymphocytic leukaemia. Br J Haematol. 152:61–71. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Stronach EA, Chen M, Maginn EN, Agarwal R, Mills GB, Wasan H and Gabra H: DNA-PK mediates AKT activation and apoptosis inhibition in clinically acquired platinum resistance. Neoplasia. 13:1069–1080. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Ciszewski WM, Tavecchio M, Dastych J and Curtin NJ: DNA-PK inhibition by NU7441 sensitizes breast cancer cells to ionizing radiation and doxorubicin. Breast Cancer Res Treat. 143:47–55. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Munck JM, Batey MA, Zhao Y, Jenkins H, Richardson CJ, Cano C, Tavecchio M, Barbeau J, Bardos J, Cornell L, et al: Chemosensitization of cancer cells by KU-0060648, a dual inhibitor of DNA-PK and PI-3K. Mol Cancer Ther. 11:1789–1798. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Noguchi T, Shibata T, Fumoto S, Uchida Y, Mueller W and Takeno S: DNA-PKcs expression in esophageal cancer as a predictor for chemoradiation therapeutic sensitivity. Ann Surg Oncol. 9:1017–1022. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Söderlund Leifler K, Queseth S, Fornander T and Askmalm MS: Low expression of Ku70/80, but high expression of DNA-PKcs, predict good response to radiotherapy in early breast cancer. Int J Oncol. 37:1547–1554. 2010.PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 37 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun G, Yang L, Dong C, Ma B, Shan M and Ma B: PRKDC regulates chemosensitivity and is a potential prognostic and predictive marker of response to adjuvant chemotherapy in breast cancer patients. Oncol Rep 37: 3536-3542, 2017
APA
Sun, G., Yang, L., Dong, C., Ma, B., Shan, M., & Ma, B. (2017). PRKDC regulates chemosensitivity and is a potential prognostic and predictive marker of response to adjuvant chemotherapy in breast cancer patients. Oncology Reports, 37, 3536-3542. https://doi.org/10.3892/or.2017.5634
MLA
Sun, G., Yang, L., Dong, C., Ma, B., Shan, M., Ma, B."PRKDC regulates chemosensitivity and is a potential prognostic and predictive marker of response to adjuvant chemotherapy in breast cancer patients". Oncology Reports 37.6 (2017): 3536-3542.
Chicago
Sun, G., Yang, L., Dong, C., Ma, B., Shan, M., Ma, B."PRKDC regulates chemosensitivity and is a potential prognostic and predictive marker of response to adjuvant chemotherapy in breast cancer patients". Oncology Reports 37, no. 6 (2017): 3536-3542. https://doi.org/10.3892/or.2017.5634