Expression of DNA double-strand break repair proteins predicts the response and prognosis of colorectal cancer patients undergoing oxaliplatin-based chemotherapy

  • Authors:
    • Keisuke Ihara
    • Satoru Yamaguchi
    • Nozomi Ueno
    • Yukiko Tani
    • Yosuke Shida
    • Hideo Ogata
    • Yasushi Domeki
    • Kentaro Okamoto
    • Masanobu Nakajima
    • Kinro Sasaki
    • Takashi Tsuchioka
    • Hiroyuki Mitomi
    • Hiroyuki Kato
  • View Affiliations

  • Published online on: December 16, 2015     https://doi.org/10.3892/or.2015.4488
  • Pages: 1349-1355
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA intrastrand cross-linking agents such as oxaliplatin induce DNA double-strand breaks (DSBs) during DNA repair and replication. In the present study, we hypothesized that DNA intrastrand cross-linking agents may significantly benefit colorectal cancer patients with deficiencies in DSB repair. Seventy-eight patients with metastatic or recurrent colorectal cancer who had measurable target lesions and who underwent resection for primary colorectal cancer in our institution between April 2007 and March 2013 were included in the present study. The median age was 64.5 years, and the cohort consisted of 49 males and 29 females. The median progression-free survival (PFS) was 10.9 months. The expression of DSB repair proteins such as RAD51 and MRE11 was investigated by immunohistochemistry, and associations between RAD51 and MRE11 expression and clinicopathological factors or chemotherapeutic effect were assessed. MRE11-negative cases and RAD51-negative cases achieved significantly better tumor reduction compared with cases with positive expression. Cases with negative expression of both proteins or negative expression of either protein had significantly longer PFS than cases with positive expression for both proteins. In conclusion, DSB repair protein expression-negative colorectal cancer cases may be more highly sensitive to chemotherapy, and thus DSB repair protein expression may be a useful prognostic indicator for colorectal cancer patients.

Introduction

Remarkable progress has been made in chemotherapy for colorectal cancer during the last decade. Currently, standard first-line treatments for unresectable advanced or recurrent colorectal cancer include fluorouracil with irinotecan or oxaliplatin, alone or in combination with molecular-targeted agents, such as the monoclonal antibody against vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) (1,2). Combinations of drug therapies in unresectable advanced or recurrent colorectal cancer patients has prolonged the survival time to more than 30 months (35), but the therapeutic effects vary depending on each case. Thus, it is important to predict the chemotherapeutic effect and select patients who will benefit from cancer chemotherapy. Several studies have shown that various biomarkers predict the sensitivity to chemotherapy or chemoradiation therapy. The presence of microsatellite instability (MSI) and mutations of the KRAS gene are reliable biomarkers for sensitivity to fluorouracil and anti-EGFR monoclonal antibodies, respectively (5,6). However, there is no reliable biomarker for oxaliplatin and irinotecan.

DNA intrastrand cross-linking agents such as oxaliplatin induce DNA double-strand breaks (DSBs) during the process of DNA replication and repair (7). BRCA1, 2, the MRE11-RAD50-NBS1 (MRN) complex and RAD51 play an important role in homologous recombination during DSB repair. A previous study showed that DNA damage repair competence varies among individual breast tumors, and is closely correlated with chemosensitivity (8). The Fanconi anemia-BRCA pathway plays an important role in restoring cytotoxic damage by anticancer agents and radiation (9,10). Furthermore, previous studies have shown that BRCA-associated cancer is particularly sensitive to DNA interstrand cross-linking agents such as mitomycin C or platinum-based drugs (11,12). Differences in the expression of DNA DSB repair proteins (DDRPs) among individual colon cancer cases may also be related to the sensitivity to treatment, as well as breast cancer.

MRE11 forms the core of the MRN complex, which has essential roles in detection, signaling, protection and repair of DSBs (13,14). RAD51 is an important factor in homologous recombination as well as MRE11, and is a predictive factor for chemoradiotherapy response in a variety of human cancers. Moreover, overexpression of RAD51 confers resistance to DNA interstrand cross-linking agents such as cisplatin in non-small cell lung cancer (1517).

In the present study, we hypothesized that DNA intrastrand cross-linking agents may significantly benefit colorectal cancer patients with deficiencies in DSB repair. We investigated the expression of MRE11 and RAD51 by immunohistochemistry. Associations between expression and therapeutic effect in colorectal cancer patients were also explored.

Materials and methods

Patients

Seventy-eight patients with metastatic or recurrent colorectal cancer who had measurable target lesions such as hepatic, pulmonary, lymphatic and peritoneal metastases, underwent resection for primary colon and rectal cancer at our institution between April 2007 and March 2013. All patients underwent combination chemotherapy including oxaliplatin.

Assessments of therapeutic effect

Descriptions of the therapeutic effects were evaluated using the best overall response to first-line chemotherapy using RECIST version 1.1. Changes in tumor size were expressed as the relative change in the sum of the longest diameters of the target lesions. Non-target lesions and newly occurring lesions were not considered in the measurement of tumor size changes (18). The endpoints of the long-term outcome study were progression-free survival (PFS). PFS was calculated by progression of target lesions as the only events for survival analyses.

Immunohistochemistry

Five-micrometer sections were deparaffinized with xylene and rehydrated with alcohol, and placed in 0.1 M NaOH citrase buffer (pH 7.0) for RAD51 immunostaining or 0.01 M NaOH citrate buffer (pH 6.0) for MRE11 immunostaining, and heated in an autoclave at 121°C for 15 min. Sections were then preincubated with 3% H2O2 in methanol for 30 min at room temperature to quench endogenous peroxidase activity. After blocking with normal goat serum, the sections were incubated with mouse anti-RAD51 3C10 monoclonal antibody (1:800; clone 51RAD01; Thermo Scientific, Fremont, CA, USA) and mouse anti-MRE11 12D7 monoclonal antibody (1:1600; ab214; Abcam, Cambridge, UK) for 60 min at 4°C. Thereafter, the sections were incubated with a secondary antibody (Vectastain Elite ABC kit; Vector Laboratories, Burlingame, CA, USA) for 30 min, washed with phosphate-buffered saline (PBS) and treated with peroxidase-conjugated streptavidin for 30 min. The sections were visualized by incubation with diaminobenzidine tetrahydrochloride (Liquid DAB+ Substrate Chromogen System; Dako, Carpinteria, CA, USA) and counterstained with hematoxylin.

Evaluation of immunohistochemical staining

Immunohistochemistry (IHC) scores for <10% of nuclear staining in cancer cells were negative, whereas those cases with IHC scores for >10% stained cells were deemed positive.

Statistical analysis

Categorical analysis of variables was performed using either the Chi-square or Fisher's exact test, as appropriate. Continuous data were compared with the Mann-Whitney U test. Survival curves were plotted according to the Kaplan-Meier method, and any differences were analyzed using the log-rank test. A multivariate analysis with Cox proportional hazards model was adopted to clarify the independent prognostic factors. Differences were considered to be significant if the P-value was <0.05. All statistical analyses were carried out using the R software (version 3.1.1).

Results

Patient clinicopathological characteristics

Patient clinical characteristics are detailed in Table I. The median age was 64.5 years, and the cohort consisted of 49 males and 29 females. Most patients received combination chemotherapy in addition to bevacizumab (n=70, 90%). Fifty patients received mFOLFOX6 + bevacizumab and 20 patients received CapeOX + bevacizumab. The median PFS was 10.9 months.

Table I

Clinicopathological characteristics of the patients (n=78).

Table I

Clinicopathological characteristics of the patients (n=78).

FactorsDate
Median age (years)64.5
Gender, n
 Men49
 Female29
Location, n
 Proximal20
 Distal58
Median tumor size (mm)50
Histology, n
 Well differentiated25
 Moderately differentiated44
 Poorly differentiated4
 Others5
Site of metastases, n
 Liver44
 Lung21
 Peritoneum11
 Lymph node8
 Bone1
Regimen, n
 mFOLFOX6 + bevacizumab50
 CapeOX + bevacizumab20
 mFOLFOX67
 CapeOX1
Treatment cycles, mean number10
Median overall survival (months)32.5
Median progression-free survival (months)10.9
MRE11 expression and clinical outcome

Positive nuclear staining of MRE11 was observed in 48 (61.5%) of the 78 cases (Fig. 1A). The association between MRE11 expression and clinicopathological characteristics is shown in Table II. There was no significant association between MRE11 expression and age, tumor location or tumor size. Male gender and undifferentiated type tended to be associated with MRE11 positivity. There was no significant association between MRE11 expression and CEA reduction ratio. MRE11-negative cases had significantly better relative change compared with MRE1-positive cases. Thus, MRE11-negative cases achieved better size reduction of the target lesion when compared with MRE11-positive cases. The association between MRE11 expression and prognosis is shown in Fig. 2A. MRE11-positive cases exhibited poorer PFS when compared with MRE11-negative cases, but no significant association was identified between MRE11 expression and PFS.

Table II

Relationship between DDRP expression and clinicopathological characteristics of the patients (n=90).

Table II

Relationship between DDRP expression and clinicopathological characteristics of the patients (n=90).

FactorsMRE11
P-valueRAD51
P-value
Negative (n=30)Positive (n=48)Negative (n=38)Positive (n=40)
Median age (years)6465.50.264670.3
Gender
 Male15340.06426230.3
 Female15141217
Location
 Proximal10100.29110.7
 Distal20382929
Median tumor size (mm)55500.455500.2
Median treatment cycles11.59.50.211.580.1
Histological type
 Differentiated29400.08035340.3
 Undifferentiated1836
Median serum CEA level (ng/ml)
 Before treatment6.621.050.0457.2522.350.07
 3 months later5.2510.10.0474.4512.650.003
CEA reduction ratio0.700.610.40.500.760.033
Relative change0.650.920.0290.481.01<0.001

[i] DDRP, DNA double-strand break repair protein; CEA, carcinoembryonic antigen.

RAD51 expression and clinical outcome

Positive nuclear staining of RAD51 was observed in 40 (51.2%) of the 78 cases (Fig. 1B). There was no significant association between RAD51 expression and age, gender, tumor location, tumor size or histological type (Table II). RAD51-negative cases had significantly better CEA reduction ratios and relative change. RAD51-positive cases had significantly poorer PFS when compared with RAD51-negative cases (Fig. 2B). However, for multivariate analysis, RAD51 was not an independent prognostic factor (Table III).

Table III

Univariate and multivariate analyses for progression-free survival.

Table III

Univariate and multivariate analyses for progression-free survival.

FactorUnivariate
Multivariate
Median PFSP-valueHR (95% CI)P-value
 Age (years)
 >6011.80.8
 ≤6010.6
Gender
 Male13.20.070
 Female10
Location
 Proximal9.30.0020.56 (0.31–1.00)0.051
 Distal13.5
Tumor size (mm)
 <5011.30.5
 ≥5011.3
Histology
 Undifferentiated11.80.1
 Differentiated9.7
Treatment cycles
 <106.40.0050.49 (0.30–0.80)0.005
 ≥1014.2
CEA reduction ratio
 <0.611.30.4
 ≥0.611.3
Relative change
 <0.718.4<0.0012.53 (1.39–4.62)0.002
 ≥0.79,0
MRE11
 Negative11.80.5
 Positive11.3
RAD51
 Negative13.50.0350.80 (0.35–1.83)0.6
 Positive9.7
DSB repair protein expression pattern
 Defective13.20.0151.39 (0.58–3.34)0.5
 Abundant10.1

[i] HR, hazard ratio; PFS, progression-free survival; CI, confidence interval; DSB, double-strand break.

DSB repair protein expression pattern

In the DSB repair pathway, the role of MRE11 and RAD51 are sequential. When one of either is defect, it may be impossible to recovery from DSBs. Therefore, we defined two groups. The 'defective pattern' is described as the state when expression of both MRE11 and RAD51 expression is negative or expression of either one of these protein is negative. The 'abundant pattern' is described as the state when expression of both proteins is positive. In addition, we investigated the association between the two groups and therapeutic effect. The association between expression patterns and clinical characteristics or chemotherapeutic effect is shown in Table IV. None of the examined clinicopathological characteristics correlated with the expression pattern. For the therapeutic effects, there was no significant difference between expression pattern and CEA reduction ratio. The defective pattern had significantly better relative change compared with the abundant pattern. As shown in Fig. 2C, the median PFS for the defective pattern and abundant pattern was 13.2 and 10.1 months, respectively, and there was a significant difference between the defective pattern and abundant pattern for PFS (Fig. 2C). Nonetheless, for the multivariate analysis, the expression pattern or RAD51 expression alone were not independent prognostic factors (Table III).

Table IV

Relationship between the DSB protein expression pattern and clinicopathological characteristics.

Table IV

Relationship between the DSB protein expression pattern and clinicopathological characteristics.

FactorsDefective (n=47)Abundant (n=31)P-value
Median age (years)64680.3
Gender, n
 Male30190.8
 Female1712
Location, n
 Right1370.8
 Left3424
Median tumor size (mm)55500.6
Median treatment cycles1180.1
Histological type, n
 Differentiated4425 0.08a
 Undifferentiated36
Median serum CEA level (ng/ml)
 Before treatment7240.009
 3 months later4.712.80.001
CEA reduction ratio0.550.680.1
Relative change0.51.02<0.001

[i] DSB, DNA double-strand break.

Distal colon cancer patients benefit more from these ex vivo tests

MRE11 mutations occur in 83.7% of MMR-defective primary colorectal cancers. MSI is displayed in ~15% of colorectal cancer cases. We reviewed all subjects in the present study with the exception of one case of tumor localization to a site proximal to the splenic flexure due to the association of high-frequency MSI with this tumor site. In these cases, MRE11-negative cases exhibited longer PFS than the positive cases (P=0.077) (Fig. 2D). Moreover, by multivariate analysis, DSB repair protein expression pattern was an independent prognostic factor (P=0.036) (Table V).

Table V

Univariate and multivariate analyses for PFS in the distal colon.

Table V

Univariate and multivariate analyses for PFS in the distal colon.

FactorUnivariate
Multivariate
MedianP-valueHR (95% CI)P-value
Treatment cycles
 <108.80.0090.46 (0.27–0.78)0.004
 ≥1014.5
CEA reduction ratio
 <0.613.50.2
 ≥0.612.6
Relative change
 <0.718.9<0.0012.23 (1.17–4.24)0.014
 ≥0.79.7
MRE11
 Negative18.90.077
 Positive11.3
RAD51
 Negative15.60.0360.44 (0.15–1.37)0.2
 Positive10.1
DSB repair protein expression pattern
 Defective16.90.0013.62 (1.09–11.99)0.036
 Abundant10.1

[i] DSB, double-strand break.

Discussion

The recent development of chemotherapies such as FOLFOX and FOLFIRI along with several molecular-targeting agents has markedly improved the survival of unresectable colorectal cancer patients. Previous studies have shown that the median survival time was prolonged to 11–26 months in unresectable advanced or recurrent colorectal cancer (19,20). In the present study, we calculated the curative effect of chemotherapy by determining the correlations between DSB repair protein expression pattern and chemotherapeutic effect; those patients exhibiting better relative changes in tumor size had longer survival times. According to a previous study, the chemotherapeutic effect by first-line treatment may be a prognostic factor. Therefore, it is necessary to detect biomarkers that predict the effect of first-line treatment to obtain further chemotherapeutic effects.

Oxaliplatin is a DNA intrastrand cross-linking agent and is frequently used to treat colorectal cancer that has spread. The cytotoxic reaction of oxaliplatin is dependent on DSBs. In DSBs defective cases, the chemotherapeutic effect of oxaliplatin may be greater. MRE11 and RAD51 are important components of homologous recombination, which functions in the repair of DSB. In the present study, we examined the correlation between the expression of these proteins and the chemotherapeutic effect in colorectal cancer patients. RAD51 protein forms a helical nucleoprotein filament to promote DNA strand exchange and stimulate DNA-pairing activity, the basic steps of homologous recombination (21,22). In the present study, patients negative for MRE11 or RAD51 expression obtained better size reduction of target lesions. We showed that RAD51-negative cases achieved longer survival times than positive cases. Several previous studies demonstrated that RAD51 expression is correlated with resistance to chemotherapy and survival in various types of cancer such as lung, breast and esophageal cancer (2325).

MRE11 is the core component of the MRN complex, the primary sensor of DSBs (12,13). In the present study, MRE11 expression was not an independent prognostic factor. The combined evaluation of MRE11, which acts as a sensor, and RAD51, which functions in repair, may lead to a better indication of the chemotherapeutic effect (Fig. 3). In fact, the relative change and PFS were significantly different between the defective pattern and abundant pattern, the expression pattern of DSB repair proteins.

We investigated the reason for the difference between MRE11 and RAD51 despite it also being a DSB repair protein, and postulated that some factors that may affect MRE11 alone intervened in the result. MRE11 mutations occur in 83.7% of MMR-defective primary colorectal cancers (26,27). Microsatellite instability (MSI) is displayed by ~15% of colorectal cancer cases, and high levels of MSI may be a predictive marker for lack of efficacy of fluorouracil-based therapy (28,29). This raises the possibility that some MRE11-negative patients have poor prognosis as a consequence of their MSI status. We reviewed the subjects of our study with the exception of one case of tumor localization to a site proximal to the splenic flexure due to the association of high-frequency MSI with this tumor site (6). In this instance, MRE11 expression tended to be related to PFS, and DSB repair protein expression pattern is a factor that independently predicts PFS. In cases of tumor development in the distal colon, DSB repair protein expression may predict prognosis.

In the present study, we only examined cases with a targeted lesion to clarify associations with chemotherapeutic effect. Therefore, it is necessary to confirm expression in other cases. Moreover, we only investigated cases receiving oxaliplatin-based chemotherapy regimen as first-line treatment since there were few cases undergoing other regimens as first-line treatment. In future studies, we will investigate the association between the expression of DSB repair proteins and the chemotherapeutic effect for other regimens, and compare these findings with the results of the present study.

In conclusion, cases with a defective pattern of DSB repair protein expression may possess higher sensitivity to chemotherapy for colorectal cancer. The expression pattern of DSB repair proteins may be a useful prognostic indicator for colorectal cancer patients.

Acknowledgments

The authors would like to thank H. Ozeki, Y. Ohashi, and M. Asano for their secretarial assistance and N. Suzuki, a member of the Research Support Center Division of Clinical Science, for her technical assistance. The present study was supported in part by JSPS KAKENHI grant no. 25462065.

References

1 

Tournigand C, André T, Achille E, Lledo G, Flesh M, Mery-Mignard D, Quinaux E, Couteau C, Buyse M, Ganem G, et al: FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: A randomized GERCOR study. J Clin Oncol. 22:229–237. 2004. View Article : Google Scholar

2 

Yamaguchi S, Ogata H, Katsumata D, Nakajima M, Fujii T, Tsutsumi S, Asao T, Sasaki K, Kuwano H and Kato H: Early serum carcinoembryonic antigen reduction predicts tumor shrinkage and overall survival in colorectal cancer patients with distant metastasis, after primary surgery followed by mFOLFOX6 plus bevacizumab treatment. Hepatogastroenterology. In press.

3 

Grothey A, Sargent D, Goldberg RM and Schmoll HJ: Survival of patients with advanced colorectal cancer improves with the availability of fluorouracil-leucovorin, irinotecan, and oxaliplatin in the course of treatment. J Clin Oncol. 22:1209–1214. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Saltz LB, Clarke S, Díaz-Rubio E, Scheithauer W, Figer A, Wong R, Koski S, Lichinitser M, Yang TS, Rivera F, et al: Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: A randomized phase III study. J Clin Oncol. 26:2013–2019. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Van Cutsem E, Köhne CH, Hitre E, Zaluski J, Chang Chien CR, Makhson A, D'Haens G, Pintér T, Lim R, Bodoky G, et al: Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 360:1408–1417. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM, Hamilton SR, Laurent-Puig P, Gryfe R, Shepherd LE, et al: Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med. 349:247–257. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Chiu SJ, Lee YJ, Hsu TS and Chen WS: Oxaliplatin-induced gamma-H2AX activation via both p53-dependent and -independent pathways but is not associated with cell cycle arrest in human colorectal cancer cells. Chem Biol Interact. 182:173–182. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Asakawa H, Koizumi H, Koike A, Takahashi M, Wu W, Iwase H, Fukuda M and Ohta T: Prediction of breast cancer sensitivity to neoadjuvant chemotherapy based on status of DNA damage repair proteins. Breast Cancer Res. 12:R172010. View Article : Google Scholar : PubMed/NCBI

9 

Turner N, Tutt A and Ashworth A: Hallmarks of 'BRCAness' in sporadic cancers. Nat Rev Cancer. 4:814–819. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Chu G: Double strand break repair. J Biol Chem. 272:24097–24100. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Gallagher DJ, Konner JA, Bell-McGuinn KM, Bhatia J, Sabbatini P, Aghajanian CA, Offit K, Barakat RR, Spriggs DR and Kauff ND: Survival in epithelial ovarian cancer: A multivariate analysis incorporating BRCA mutation status and platinum sensitivity. Ann Oncol. 22:1127–1132. 2011. View Article : Google Scholar

12 

Moynahan ME, Cui TY and Jasin M: Homology-directed DNA repair, mitomycin-c resistance, and chromosome stability is restored with correction of a Brca1 mutation. Cancer Res. 61:4842–4850. 2001.PubMed/NCBI

13 

Stracker TH and Petrini JH: The MRE11 complex: Starting from the ends. Nat Rev Mol Cell Biol. 12:90–103. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Williams RS, Williams JS and Tainer JA: Mre11-Rad50-Nbs1 is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochem Cell Biol. 85:509–520. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Baumann P and West SC: Role of the human RAD51 protein in homologous recombination and double-stranded-break repair. Trends Biochem Sci. 23:247–251. 1998. View Article : Google Scholar : PubMed/NCBI

16 

Slupianek A, Schmutte C, Tombline G, Nieborowska-Skorska M, Hoser G, Nowicki MO, Pierce AJ, Fishel R and Skorski T: BCR/ABL regulates mammalian RecA homologs, resulting in drug resistance. Mol Cell. 8:795–806. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Takenaka T, Yoshino I, Kouso H, Ohba T, Yohena T, Osoegawa A, Shoji F and Maehara Y: Combined evaluation of Rad51 and ERCC1 expressions for sensitivity to platinum agents in non-small cell lung cancer. Int J Cancer. 121:895–900. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Piessevaux H, Buyse M, Schlichting M, Van Cutsem E, Bokemeyer C, Heeger S and Tejpar S: Use of early tumor shrinkage to predict long-term outcome in metastatic colorectal cancer treated with cetuximab. J Clin Oncol. 31:3764–3775. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Yoshida M, Goto M, Kii T, Nishitani H, Kawabe S, Kuwakado S, Asaishi K, Miyamoto T and Higuchi K: Retrospective study as first-line chemotherapy combined anti-VEGF antibody with fluoropyrimidine for frail patients with unresectable or metastatic colorectal cancer. Digestion. 87:59–64. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Maeda K, Shibutani M, Otani H, Nagahara H, Sugano K, Ikeya T, Kubo N, Amano R, Kimura K, Muguruma K, et al: Low nutritional prognostic index correlates with poor survival in patients with stage IV colorectal cancer following palliative resection of the primary tumor. World J Surg. 38:1217–1222. 2014. View Article : Google Scholar

21 

Thompson LH and Schild D: The contribution of homologous recombination in preserving genome integrity in mammalian cells. Biochimie. 81:87–105. 1999. View Article : Google Scholar : PubMed/NCBI

22 

Sung P and Robberson DL: DNA strand exchange mediated by a RAD51-ssDNA nucleoprotein filament with polarity opposite to that of RecA. Cell. 82:453–461. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Nakanoko T, Saeki H, Morita M, Nakashima Y, Ando K, Oki E, Ohga T, Kakeji Y, Toh Y and Maehara Y: Rad51 expression is a useful predictive factor for the efficacy of neoadjuvant chemoradiotherapy in squamous cell carcinoma of the esophagus. Ann Surg Oncol. 21:597–604. 2014. View Article : Google Scholar :

24 

Tsai MS, Kuo YH, Chiu YF, Su YC and Lin YW: Down-regulation of Rad51 expression overcomes drug resistance to gemcitabine in human non-small-cell lung cancer cells. J Pharmacol Exp Ther. 335:830–840. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Hannay JA, Liu J, Zhu QS, Bolshakov SV, Li L, Pisters PW, Lazar AJ, Yu D, Pollock RE and Lev D: Rad51 overexpression contributes to chemoresistance in human soft tissue sarcoma cells: A role for p53/activator protein 2 transcriptional regulation. Mol Cancer Ther. 6:1650–1660. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Giannini G, Rinaldi C, Ristori E, Ambrosini MI, Cerignoli F, Viel A, Bidoli E, Berni S, D'Amati G, Scambia G, et al: Mutations of an intronic repeat induce impaired MRE11 expression in primary human cancer with microsatellite instability. Oncogene. 23:2640–2647. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Vilar E, Bartnik CM, Stenzel SL, Raskin L, Ahn J, Moreno V, Mukherjee B, Iniesta MD, Morgan MA, Rennert G, et al: MRE11 deficiency increases sensitivity to poly(ADP-ribose) polymerase inhibition in microsatellite unstable colorectal cancers. Cancer Res. 71:2632–2642. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Sargent DJ, Marsoni S, Monges G, Thibodeau SN, Labianca R, Hamilton SR, French AJ, Kabat B, Foster NR, Torri V, et al: Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. J Clin Oncol. 28:3219–3226. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Yamaguchi S, Ogata H, Katsumata D, Nakajima M, Fujii T, Tsutsumi S, Asao T, Sasaki K, Kuwano H and Kato H: MUTYH- associated colorectal cancer and adenomatous polyposis. Surg Today. 44:593–600. 2014. View Article : Google Scholar

Related Articles

Journal Cover

March-2016
Volume 35 Issue 3

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
Ihara K, Yamaguchi S, Ueno N, Tani Y, Shida Y, Ogata H, Domeki Y, Okamoto K, Nakajima M, Sasaki K, Sasaki K, et al: Expression of DNA double-strand break repair proteins predicts the response and prognosis of colorectal cancer patients undergoing oxaliplatin-based chemotherapy. Oncol Rep 35: 1349-1355, 2016
APA
Ihara, K., Yamaguchi, S., Ueno, N., Tani, Y., Shida, Y., Ogata, H. ... Kato, H. (2016). Expression of DNA double-strand break repair proteins predicts the response and prognosis of colorectal cancer patients undergoing oxaliplatin-based chemotherapy. Oncology Reports, 35, 1349-1355. https://doi.org/10.3892/or.2015.4488
MLA
Ihara, K., Yamaguchi, S., Ueno, N., Tani, Y., Shida, Y., Ogata, H., Domeki, Y., Okamoto, K., Nakajima, M., Sasaki, K., Tsuchioka, T., Mitomi, H., Kato, H."Expression of DNA double-strand break repair proteins predicts the response and prognosis of colorectal cancer patients undergoing oxaliplatin-based chemotherapy". Oncology Reports 35.3 (2016): 1349-1355.
Chicago
Ihara, K., Yamaguchi, S., Ueno, N., Tani, Y., Shida, Y., Ogata, H., Domeki, Y., Okamoto, K., Nakajima, M., Sasaki, K., Tsuchioka, T., Mitomi, H., Kato, H."Expression of DNA double-strand break repair proteins predicts the response and prognosis of colorectal cancer patients undergoing oxaliplatin-based chemotherapy". Oncology Reports 35, no. 3 (2016): 1349-1355. https://doi.org/10.3892/or.2015.4488