SCGB2A1 is a novel prognostic marker for colorectal cancer associated with chemoresistance and radioresistance

  • Authors:
    • Koji Munakata
    • Mamoru Uemura
    • Ichiro Takemasa
    • Miyuki Ozaki
    • Masamitsu Konno
    • Junichi Nishimura
    • Taishi Hata
    • Tsunekazu Mizushima
    • Naotsugu Haraguchi
    • Shingo Noura
    • Masakazu Ikenaga
    • Shu Okamura
    • Mutsumi Fukunaga
    • Kohei Murata
    • Hirofumi Yamamoto
    • Yuichiro Doki
    • Masaki Mori
  • View Affiliations

  • Published online on: February 28, 2014     https://doi.org/10.3892/ijo.2014.2316
  • Pages: 1521-1528
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

We recently showed that liver metastatic tissue from patients with colorectal cancer (CRC) was a useful model for identifying novel, hypoxia-inducible genes and prognostic markers. We showed that the expression of secretoglobin, family 2A, member 1 (SCGB2A1) was a potential prognostic factor for CRC. Here, we further evaluated the prognostic impact and function of SCGB2A1 in 222 patients with CRC. The impact of SCGB2A1 expression on disease-free survival (DFS) and overall survival (OS) was assessed with mRNA expression profiling. The function of SCGB2A1 was analyzed by evaluating mRNA expression profiles in cells derived from patients with CRC and by testing the effects of transfecting SCGB2A1 into different CRC-derived cell lines. We evaluated the effects of SCGB2A1 on proliferation, chemosensitivity, radiation sensitivity and sphere formation. Univariate and multivariate analyses indicated that the expression of SCGB2A1 was an independent prognostic factor for CRC (p<0.05), together with lymph node metastasis (p<0.05). Enforced expression of SCGB2A1 in CRC-derived cell lines promoted proliferation (DLD1, SW480 and LoVo cells; p<0.05), decreased chemosensitivity to 5-fluorouracil and oxaliplatin (DLD1 and SW480 cell lines; p<0.05), and significantly increased the viability of irradiated cells (DLD1, SW480 and LoVo cell lines; p<0.05). SCGB2A1 expression was also correlated to cancer stemness-related genes (Wnt, Zeb1 and Twist). Consistent with this correlation, SCGB2A1 expressing cells (SW480) showed increased sphere formation (p<0.05). These results indicated that SCGB2A1 represented a novel, prognostic factor for CRC, and that expression of SCGB2A1 correlated with chemoresistance, radioresistance and cancer cell stemness.

Introduction

Colorectal cancer (CRC) is one of the most common human malignancies worldwide. Despite recent advances in treatment with chemotherapy and radiation therapy, CRC remains a major cause of cancer death (1). Thus, there is a crucial need to explore novel cancer-related genes that may serve as diagnostic markers and molecular targets in CRC therapy.

A hypoxic microenvironment is associated with many solid tumors, including breast cancer (2), prostate cancer (3,4), brain tumor (5), malignant melanomas (6,7), lung cancer (8), liver cancer (9,10), ovarian cancer (11,12), and CRC (13). Furthermore, intratumoral hypoxia affects every major aspect of cancer biology, including cell invasion, metastasis, and determination of cell death (14).

Many molecules in the hypoxia-response pathway are good candidates for therapeutic targeting (1517). The anti-VEGF antibody, bevacizumab, is used clinically for treating several human cancers (18); this supports the notion that hypoxia-induced genes are clinically-relevant therapeutic targets. Therefore, the identification of novel hypoxia-inducible genes holds great potential for the development of additional cancer therapies.

We previously reported that liver metastatic tissue derived from patients with CRC was a useful in vivo model for identifying novel hypoxia-inducible genes and prognostic markers. These markers included the mRNA expression levels of Jumonji domain containing 1A (JMJD1A), adrenomedullin (ADM), Ephrin-A1 (EFNA1), and procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) (1922). In those experiments, we also found that the mRNA expression of secretoglobin, family 2A, member 1 (SCGB2A1) was highly induced in hypoxic regions of metastasized liver (fold-change, 1.57). Thus, we hypothesized that SCGB2A1 expression may be a novel prognostic factor in patients with CRC.

The aim of the present study was to examine the prognostic impact of SCGB2A1 and the biological significance of SCGB2A1 in CRC. We reported previously (2325) that SCGB2A1 was a useful marker in several cancers for the molecular detection of minimal residual disease in lymph nodes, but the mechanism remains unknown (2629).

Here, we found that SCGB2A1 was an independent prognostic factor, and expression of SCGB2A1 promoted both chemoresistance and radioresistance.

Cancer cell stemness is a primary underlying mechanism that contributes to resistance to chemotherapy and radiation therapy (30,31). Our data indicated that cancer stemness was enhanced by forced expression of SCGB2A1. Thus, our findings may provide clues for the development of a novel anticancer therapy.

Materials and methods

Cell lines and culture conditions

Human CRC-derived cell lines, DLD1, SW480, and LoVo, were obtained from the American Type Culture Collection. Cells were grown in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum, 100 U/ml penicillin and 100 μg/ml streptomycin at 37°C in a humidified incubator with 5% CO2.

Clinical samples and microarray analysis

For microarray analysis, we prospectively collected 1,978 primary CRC samples from consecutive patients who had curative operations in 2003 to 2006 from Osaka University Hospital and its nine associated hospitals (32). Tumor samples were consecutively collected from a total of 222 CRC patients for microarray analysis. Microarray analysis was carried out as described previously (33) using an oligonucleotide microarray covering 30,000 human probes (AceGene; DNA Chip Research, Inc. and Hitachi Software Engineering Co. Ltd.). The mean follow-up times were 42.9±28.9 months for patients with disease-free survival (DFS) and 56.7±20.4 months for all surviving patients [overall survival (OS)]. Table I shows the clinicopathological features of patients from each institute, including gender, tumor location, extent of wall invasion, lymph node metastasis, histologic grade, Dukes’ stage, and vessel invasion. In this study, no patient received preoperative chemotherapy or irradiation. After surgery, patients with Dukes’ stage C/D tumors were generally treated with 5-fluorouracil (5-FU)-based chemotherapy.

Table I.

Relationship between SCGB2A1 expression and clinicopathological factors in patients with colorectal cancer.

Table I.

Relationship between SCGB2A1 expression and clinicopathological factors in patients with colorectal cancer.

Total n=222SCGB2A1 mRNA expressionP-value

Low (n=111)High (n=111)
Age (years)66.2±10.065.8±9.766.7±10.30.61
Gender
  Male/female133/8962/4971/400.27
Tumor location
  Colon/rectum141/8177/3464/470.09
Depth of invasiona
  mp/ss26/1968/10318/930.06
Lymph node metastasis
  Present/absent109/11352/5957/540.59
Histological grade
  Well differentiated/others53/16928 /8325/860.75
Dukes’ stage
  AB/CD108/11458/5350/610.35
Vessel invasion
  Present/absent184/3889/2295/160.37

a mp, muscularis propria layer or above; ss, subserosa level or below.

Transfection of vector

DLD1, SW480 and LoVo were transfected with an SCGB2A1 expression vector with the FuGENE® 6 transfection reagent (Promega, USA), according to the manufacturer’s protocol. Control cells were transfected with the same method, but with an empty control vector.

Proliferation assay

Overexpressing SCGB2A1 cell lines and control cell lines were seeded in 96-well plates (2,000 cells/well) and grown at 37°C with 5% CO2. After 24 and 72 h, we assayed cell viability with a Cell Counting Kit-8 (Dojindo, Japan), according to themanufacturer’s instructions. After 2-h pre-incubations in the assay solution, the viable cell number in each well was determined from the absorbance at 450 nm (OD 450), measured with a microplate reader (Bio-Rad Model 680 XR, USA).

Chemosensitivity assay

We tested cell sensitivity to 5-FU and oxaliplatin (L-OHP), which are generally used in chemo-therapy for CRC. We seeded 2,000 cells/well in 96-well plates and incubated at 37°C with 5% CO2. After 24 h, the culture medium was replaced with fresh medium in the presence of 5-FU or L-OHP at predetermined IC50 concentrations. After 24 and 48 h of treatment with chemotherapy, we assayed cell viability with same method used in the proliferation assay.

Radiation sensitivity assay

To measure radiation sensitivity, each cell line was seeded at 2,000 cells/well into 96-well plates and incubated at 37°C with 5% CO2. After 24 h, a 137Cs Gamma Cell 40 Exactor (MDS Nordion, Canada) was used to irradiate DLD1 and SW480 cells at 8 Gy and LoVo cells at 6 Gy. After 24 and 72 h of treatment with radiation therapy, we assayed cell viability with same method used in the proliferation assay.

Sphere formation assay

The sphere formation assay was performed essentially as described previously (34). In brief, single cells were plated in 96-well ultralow attachment plates (Corning Inc., USA) at a density of 100 cells/well and grown in tumorspheric culture medium (Dulbecco’s modified Eagle’s medium, DMEM/F-12), supplemented with 20 ng/ml human platelet growth factor (Sigma-Aldrich, USA), 20 ng/ml epidermal growth factor (Invitrogen, USA) and 1% antibiotic-antimycotic solution (Invitrogen) at 37°C in a humidified atmosphere of 95% air and 5% CO2. We counted the number of spheres ≥100 μm in all wells and evaluated differences in the average number/well.

Statistical analysis

Statistical analysis was performed with the StatView 5.0 program (Abacus Concepts, Inc., USA). The Kaplan-Meier method was used to examine DFS and OS, and the log-rank test was used to determine statistical significance. A Cox proportional hazard model was used to assess the risk ratio with simultaneous contributions from several covariates. Statistical analysis was performed with the Student’s t-test or Fisher’s exact test for categorical data and with the Mann-Whitney U test for non-parametric data. Correlation significance was assessed with Pearson’s correlation coefficient-test. Values of p<0.05 denoted a statistically significant difference.

Results

Relationship between SCGB2A1 expression and clinicopathological factors

Patients were divided into two groups (high or low) according to whether the SCGB1A1 mRNA expression level was above or below the median SCGB2A1 expression value. The relationships between SCGB2A1 mRNA expression and clinicopathological factors were examined (Table I). There were no significant differences between groups in age, gender, tumor location, depth of tumor invasion, lymph node metastasis, histological grade, Dukes’ stage or vessel invasion.

Impact of SCGB2A1 mRNA expression on disease-free survival

The DFS curves were stratified according to SCGB2A1 expression levels. The DFS was significantly longer in the low SCGB2A1 expression group compared to that in the high SCGB2A1 expression group (p<0.0001; Fig. 1A).

In a univariate analysis, the level of SCGB2A1 mRNA expression and various other clinicopathological parameters were evaluated for their impact on DFS (Table II). The DFS was significantly associated with the depth of invasion (p=0.0082), lymph node metastasis (p<0.0001), histological grade (p=0.0193), vessel invasion (p=0.0001) and the expression of SCGB2A1 (p<0.0001).

Table II.

Analysis of associations between disease-free survival and clinicopathological factors, including SCGB2A1 mRNA expression.

Table II.

Analysis of associations between disease-free survival and clinicopathological factors, including SCGB2A1 mRNA expression.

VariableUnivariate analysisMultivariate analysis


P-valueRelative risk95 % CIP-value
Tumor location (colon/rectum)0.0655
Depth of invasiona (mp/ss)0.00820.3270.118–0.9090.0321
Lymph node metastasis (+/−)<0.00011.5921.025–2.4730.0386
Histological grade (well differentiated/others)0.01931.2760.711–2.2930.4141
Dukes’ stage (AB/CD)<0.0001
Vessel invasion (yes/no)0.00013.2071.121–9.1770.0298
SCGB2A1 (high/low)<0.00012.7841.775–4.369<0.0001

a mp, muscularis propria layer or above; ss, subserosa level or below.

A multivariate Cox regression analysis demonstrated that the mRNA expression of SCGB2A1 was a significant prognostic factor for DFS (p<0.0001; Table II). Among the other covariates, the depth of invasion, lymph node metastasis and vessel invasion were significant prognostic factors (p=0.0321, 0.0386 and 0.0298, respectively; Table II).

Impact of SCGB2A1 expression on overall survival

The OS curves were stratified according to SCGB2A1 expression levels. The OS was significantly longer in the low SCGB2A1 expression group compared to that in the high SCGB2A1 expression group (p=0.0092; Fig. 1B).

In a univariate analysis, mRNA expression of SCGB2A1 and various clinicopathological parameters were evaluated for their impact on OS. The OS was significantly associated with lymph node metastasis (p<0.0001), vessel invasion (p=0.0201) and the expression of SCGB2A1 (p=0.0092) (Table III).

Table III.

Analysis of associations between overall survival and clinicopathological factors, including SCGB2A1 mRNA expression.

Table III.

Analysis of associations between overall survival and clinicopathological factors, including SCGB2A1 mRNA expression.

VariableUnivariate analysisMultivariate analysis


P-valueRelative risk95 % CIP-value
Tumor location (colon/rectum)0.8824
Depth of invasiona (mp/ss)0.2763
Lymph node metastasis (+/−)<0.00014.0651.911–8.6430.0003
Histological grade (well differentiated/others)0.0632
Dukes’ stage (AB/CD)<0.0001
Vessel invasion (yes/no)0.02012.1920.506–9.4930.2939
SCGB2A1 (high/low)0.00921.9881.067–3.7040.0305

a mp, muscularis propria layer or above; ss, subserosa level or below.

A multivariate Cox regression analysis demonstrated that the mRNA expression of SCGB2A1 was a significant prognostic factor for OS (p=0.0305; Table III). Among the other covariates, only lymph node metastasis was a significant prognostic factor (p=0.0003; Table III).

Proliferation assay

To explore SCGB2A1 gene function, we first transfected a plasmid that encoded SCGB2A1 or the empty control vector into CRC-derived cell lines, DLD1, SW480 and LoVo. Our results showed that upregulation of SCGB2A1 elicited significant cell proliferation compared to cells transfected with the empty control vector at 72 h (p<0.05) (Fig. 2).

Chemoresistance and radioresistance conferred by SCGB2A1 in CRC cells

CRC-derived cells (DLD1 and SW480) transfected with SCGB2A1 exhibited much greater resistance to the anticancer drugs, 5-FU and L-OHP, than cells transfected with empty control vector at 48 h (p<0.05) (Fig. 3). Likewise, upregulation of SCGB2A1 in DLD1, SW480 and LoVo cells conferred stronger radioresistance compared to cells transfected with empty control vector at 72 h (p<0.05) (Fig. 4).

Correlation of SCGB2A1 with cancer stemness-related genes and sphere formation

For elucidation of the chemoresistance and radioresistance mechanism, we analyzed the microarray data to identify genes that were correlated with SCGB2A1 expression. We found that SCGB2A1 was correlated with the expression of wingless and INT-1 (Wnt; Fig. 5A), zinc finger E-box binding homeobox 1 (Zeb1; Fig. 5B) and Twist (Fig. 5C).

Recently, the development of cancer stem cells (CSCs) was proposed to be one of the major mechanisms underlying treatment resistance. One study demonstrated a relationship between colon CSCs and Wnt activity (35). Another study showed that Zeb1 induction was associated with the epithelialmesenchymal transition, and it was also related to CSCs (36).

We conducted a sphere formation assay to evaluate whether SCGB2A1 overexpressing cells acquired cancer cell stemness. The appearance of spheres, which form after several weeks, is considered indicative of the ability to self-renew. This phenomenon would be consistent with development of a CSC phenotype (34). After 3 weeks in culture, sphere formation was observed in the SW480 cell line, but not the other cell lines (Fig. 6A). Also, we found that upregulation of SCGB2A1 could generate many more spheres compared to baseline SCGB2A1 levels in cells transfected with the empty control vector (p<0.0001) (Fig. 6B).

Discussion

The SCGB2A1 (mammaglobin B) gene encodes a small secreted protein of the uteroglobin superfamily. This super-family includes nine human secretoglobins that are localized on chromosome 11q12.2 (37). SCGB2A1 was first isolated by Becker et al in 1998 (38).

SCGB2A1 is considered a candidate marker for the molecular detection of several minimal cancers in lymph nodes (2329) and for the diagnosis of occult tumor cells in effusions from patients with various malignancies, including gynecological cancers (39). However, the biological function of SCGB2A1 has not yet been clarified in CRC.

The present report was the first to show that SCGB2A1 could be an important prognostic factor for patients with CRC. We showed that enhanced expression of SCGB2A1 in CRC cells might confer the property of treatment resistance.

We previously established a method for finding clinically important, hypoxia-inducible genes from samples of liver that had metastasized from CRC (1922). In a chronically hypoxic environment, cancer cells undergo genetic and adaptive changes that allow them to become more clinically aggressive, and they develop resistance to irradiation and chemotherapy (15,16,40). An efficient therapeutic strategy for combating those cell types is essential for overcoming cancer. However, it is difficult to identify important hypoxia-inducible genes that are related to clinical cancer biology in vitro, because cancer cells typically exist in chronically hypoxic conditions in vivo. Therefore, cancer cells develop complex interactions that affect several pathways.

The most important finding in this study was the reciprocal relationship between SCGB2A1 expression and treatment resistance. We hypothesized that treatment resistance was caused by the development of cancer stemness, because our data showed that the expression of SCGB2A1 was correlated with the expression of Wnt, Zeb1 and Twist. The sphere formation assay is commonly used to detect CSCs in vitro. As we expected, SCGB2A1-expressing cells showed more abundant sphere formation than control cells. This cancer-stemness property may partly explain why SCGB2A1 expression was associated with chemoresistance and radioresistance in CRC. These findings suggested that SCGB2A1-expressing cells have enhanced malignancy potential. Our data also suggested that SCGB2A1 may represent a novel therapeutic target.

In conclusion, we showed that SCGB2A1 represents a novel prognostic factor for CRC. SCGB2A1 correlated with chemoresistance, radioresistance, and cancer cell stemness.

Acknowledgements

This study was supported by a Grant-in-Aid for Cancer Research from the Ministry of Education, Science, Sports and Culture Technology, Japan (grant 21390360).

References

1. 

Jemal A, Siegel R, Xu J and Ward E: Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. 2010. View Article : Google Scholar

2. 

Vaupel P, Briest S and Hockel M: Hypoxia in breast cancer: pathogenesis, characterization and biological/therapeutic implications. Wien Med Wochenschr. 152:334–342. 2002. View Article : Google Scholar : PubMed/NCBI

3. 

Sooriakumaran P and Kaba R: Angiogenesis and the tumour hypoxia response in prostate cancer: a review. Int J Surg. 3:61–67. 2005. View Article : Google Scholar : PubMed/NCBI

4. 

Higgins LH, Withers HG, Garbens A, et al: Hypoxia and the metabolic phenotype of prostate cancer cells. Biochim Biophys Acta. 1787:1433–1443. 2009. View Article : Google Scholar : PubMed/NCBI

5. 

Jensen RL: Brain tumor hypoxia: tumorigenesis, angiogenesis, imaging, pseudoprogression, and as a therapeutic target. J Neurooncol. 92:317–335. 2009. View Article : Google Scholar : PubMed/NCBI

6. 

O’Connell MP, Marchbank K, Webster MR, et al: Hypoxia induces phenotypic plasticity and therapy resistance in melanoma via the tyrosine kinase receptors ROR1 and ROR2. Cancer Discov. Oct 8–2013.(Epub ahead of print). View Article : Google Scholar : 2013.

7. 

Zeng W, Yang D, Long T, et al: CD147 promotes melanoma progression through hypoxia-induced MMP2 activation. Curr Mol Med. Oct 3–2013.(Epub ahead of print).

8. 

Lee GW, Go SI, Cho YJ, et al: Hypoxia-inducible factor-1alpha and excision repair cross-complementing 1 in patients with small cell lung cancer who received front-line platinum-based chemotherapy: a retrospective study. J Thorac Oncol. 7:528–534. 2012. View Article : Google Scholar

9. 

Zhang L, Huang G, Li X, et al: Hypoxia induces epithelial-mesenchymal transition via activation of SNAI1 by hypoxia-inducible factor -1alpha in hepatocellular carcinoma. BMC Cancer. 13:1082013. View Article : Google Scholar : PubMed/NCBI

10. 

Liu Y, Zhang JB, Qin Y, et al: PROX1 promotes hepatocellular carcinoma metastasis by way of up-regulating hypoxia-inducible factor 1alpha expression and protein stability. Hepatology. 58:692–705. 2013. View Article : Google Scholar

11. 

Selvendiran K, Bratasz A, Kuppusamy ML, Tazi MF, Rivera BK and Kuppusamy P: Hypoxia induces chemoresistance in ovarian cancer cells by activation of signal transducer and activator of transcription 3. Int J Cancer. 125:2198–2204. 2009. View Article : Google Scholar : PubMed/NCBI

12. 

Liang D, Ma Y, Liu J, et al: The hypoxic microenvironment upgrades stem-like properties of ovarian cancer cells. BMC Cancer. 12:2012012. View Article : Google Scholar : PubMed/NCBI

13. 

Chang LH, Chen CH, Huang DY, Pai HC, Pan SL and Teng CM: Thrombin induces expression of twist and cell motility via the hypoxia-inducible factor-1alpha translational pathway in colorectal cancer cells. J Cell Physiol. 226:1060–1068. 2011. View Article : Google Scholar

14. 

Semenza GL: Hypoxia and cancer. Cancer Metastasis Rev. 26:223–224. 2007. View Article : Google Scholar

15. 

Harris AL: Hypoxia - a key regulatory factor in tumour growth. Nat Rev Cancer. 2:38–47. 2002. View Article : Google Scholar : PubMed/NCBI

16. 

Kizaka-Kondoh S, Inoue M, Harada H and Hiraoka M: Tumor hypoxia: a target for selective cancer therapy. Cancer Sci. 94:1021–1028. 2003. View Article : Google Scholar : PubMed/NCBI

17. 

Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 3:721–732. 2003. View Article : Google Scholar

18. 

Hurwitz H, Fehrenbacher L, Novotny W, et al: Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 350:2335–2342. 2004. View Article : Google Scholar : PubMed/NCBI

19. 

Uemura M, Yamamoto H, Takemasa I, et al: Jumonji domain containing 1A is a novel prognostic marker for colorectal cancer: in vivo identification from hypoxic tumor cells. Clin Cancer Res. 16:4636–4646. 2010. View Article : Google Scholar : PubMed/NCBI

20. 

Uemura M, Yamamoto H, Takemasa I, et al: Hypoxia-inducible adrenomedullin in colorectal cancer. Anticancer Res. 31:507–514. 2011.PubMed/NCBI

21. 

Yamamoto H, Tei M, Uemura M, et al: Ephrin-A1 mRNA is associated with poor prognosis of colorectal cancer. Int J Oncol. 42:549–555. 2012.

22. 

Noda T, Yamamoto H, Takemasa I, et al: PLOD2 induced under hypoxia is a novel prognostic factor for hepatocellular carcinoma after curative resection. Liver Int. 32:110–118. 2012. View Article : Google Scholar : PubMed/NCBI

23. 

Aihara T, Fujiwara Y, Ooka M, Sakita I, Tamaki Y and Monden M: Mammaglobin B as a novel marker for detection of breast cancer micrometastases in axillary lymph nodes by reverse transcription-polymerase chain reaction. Breast Cancer Res Treat. 58:137–140. 1999. View Article : Google Scholar

24. 

Okami J, Dohno K, Sakon M, et al: Genetic detection for micro-metastasis in lymph node of biliary tract carcinoma. Clin Cancer Res. 6:2326–2332. 2000.PubMed/NCBI

25. 

Aihara T, Fujiwara Y, Miyake Y, et al: Mammaglobin B gene as a novel marker for lymph node micrometastasis in patients with abdominal cancers. Cancer Lett. 150:79–84. 2000. View Article : Google Scholar : PubMed/NCBI

26. 

Ouellette RJ, Richard D and Maicas E: RT-PCR for mamma-globin genes, MGB1 and MGB2, identifies breast cancer micrometastases in sentinel lymph nodes. Am J Clin Pathol. 121:637–643. 2004. View Article : Google Scholar : PubMed/NCBI

27. 

Tassi RA, Bignotti E, Falchetti M, et al: Mammaglobin B expression in human endometrial cancer. Int J Gynecol Cancer. 18:1090–1096. 2008. View Article : Google Scholar : PubMed/NCBI

28. 

Tassi RA, Calza S, Ravaggi A, et al: Mammaglobin B is an independent prognostic marker in epithelial ovarian cancer and its expression is associated with reduced risk of disease recurrence. BMC Cancer. 9:2532009. View Article : Google Scholar : PubMed/NCBI

29. 

Bellone S, Tassi R, Betti M, et al: Mammaglobin B (SCGB2A1) is a novel tumour antigen highly differentially expressed in all major histological types of ovarian cancer: implications for ovarian cancer immunotherapy. Br J Cancer. 109:462–471. 2013. View Article : Google Scholar

30. 

Mohrin M, Bourke E, Alexander D, et al: Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem Cell. 7:174–185. 2010. View Article : Google Scholar : PubMed/NCBI

31. 

Adikrisna R, Tanaka S, Muramatsu S, et al: Identification of pancreatic cancer stem cells and selective toxicity of chemotherapeutic agents. Gastroenterology. 143:234–245. 2012. View Article : Google Scholar : PubMed/NCBI

32. 

Miyake M, Takemasa I, Matoba R, et al: Heterogeneity of colorectal cancers and extraction of discriminator gene signatures for personalized prediction of prognosis. Int J Oncol. 39:781–789. 2011.PubMed/NCBI

33. 

Takeno A, Takemasa I, Doki Y, et al: Integrative approach for differentially overexpressed genes in gastric cancer by combining large-scale gene expression profiling and network analysis. Br J Cancer. 99:1307–1315. 2008. View Article : Google Scholar

34. 

Takaishi S, Okumura T, Tu S, et al: Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cells. 27:1006–1020. 2009. View Article : Google Scholar : PubMed/NCBI

35. 

Vermeulen L, De Sousa EMF, van der Heijden M, et al: Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 12:468–476. 2010. View Article : Google Scholar : PubMed/NCBI

36. 

Singh A and Settleman J: EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 29:4741–4751. 2010. View Article : Google Scholar : PubMed/NCBI

37. 

Ni J, Kalff-Suske M, Gentz R, Schageman J, Beato M and Klug J: All human genes of the uteroglobin family are localized on chromosome 11q12.2 and form a dense cluster. Ann NY Acad Sci. 923:25–42. 2000. View Article : Google Scholar : PubMed/NCBI

38. 

Becker RM, Darrow C, Zimonjic DB, Popescu NC, Watson MA and Fleming TP: Identification of mammaglobin B, a novel member of the uteroglobin gene family. Genomics. 54:70–78. 1998. View Article : Google Scholar : PubMed/NCBI

39. 

Fiegl M, Haun M, Massoner A, et al: Combination of cytology, fluorescence in situ hybridization for aneuploidy, and reverse-transcriptase polymerase chain reaction for human mammaglobin/mammaglobin B expression improves diagnosis of malignant effusions. J Clin Oncol. 22:474–483. 2004. View Article : Google Scholar

40. 

Brennan DJ, Jirstrom K, Kronblad A, et al: CA IX is an independent prognostic marker in premenopausal breast cancer patients with one to three positive lymph nodes and a putative marker of radiation resistance. Clin Cancer Res. 12:6421–6431. 2006. View Article : Google Scholar

Related Articles

Journal Cover

May-2014
Volume 44 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Munakata K, Uemura M, Takemasa I, Ozaki M, Konno M, Nishimura J, Hata T, Mizushima T, Haraguchi N, Noura S, Noura S, et al: SCGB2A1 is a novel prognostic marker for colorectal cancer associated with chemoresistance and radioresistance. Int J Oncol 44: 1521-1528, 2014
APA
Munakata, K., Uemura, M., Takemasa, I., Ozaki, M., Konno, M., Nishimura, J. ... Mori, M. (2014). SCGB2A1 is a novel prognostic marker for colorectal cancer associated with chemoresistance and radioresistance. International Journal of Oncology, 44, 1521-1528. https://doi.org/10.3892/ijo.2014.2316
MLA
Munakata, K., Uemura, M., Takemasa, I., Ozaki, M., Konno, M., Nishimura, J., Hata, T., Mizushima, T., Haraguchi, N., Noura, S., Ikenaga, M., Okamura, S., Fukunaga, M., Murata, K., Yamamoto, H., Doki, Y., Mori, M."SCGB2A1 is a novel prognostic marker for colorectal cancer associated with chemoresistance and radioresistance". International Journal of Oncology 44.5 (2014): 1521-1528.
Chicago
Munakata, K., Uemura, M., Takemasa, I., Ozaki, M., Konno, M., Nishimura, J., Hata, T., Mizushima, T., Haraguchi, N., Noura, S., Ikenaga, M., Okamura, S., Fukunaga, M., Murata, K., Yamamoto, H., Doki, Y., Mori, M."SCGB2A1 is a novel prognostic marker for colorectal cancer associated with chemoresistance and radioresistance". International Journal of Oncology 44, no. 5 (2014): 1521-1528. https://doi.org/10.3892/ijo.2014.2316