Open Access

The clinical significance of SWI/SNF complex in pancreatic cancer

  • Authors:
    • Masakatsu Numata
    • Soichiro Morinaga
    • Takuo Watanabe
    • Hiroshi Tamagawa
    • Naoto Yamamoto
    • Manabu Shiozawa
    • Yoshiyasu Nakamura
    • Yoichi Kameda
    • Shinichi Okawa
    • Yasushi Rino
    • Makoto Akaike
    • Munetaka Masuda
    • Yohei Miyagi
  • View Affiliations

  • Published online on: November 30, 2012     https://doi.org/10.3892/ijo.2012.1723
  • Pages: 403-410
  • Copyright: © Numata et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chromatin remodeling factors have been the subject of great interest in oncology. However, little is known about their role in pancreatic cancer. The objective of this study was to clarify the clinical significance of the SWItch/sucrose non-fermentable (SWI/SNF) complex in patients with pancreatic cancer. A total of 68 patients with pancreatic cancer who underwent R0, 1 resection were enrolled. Cancer tissues were processed to tissue microarray, then stained immunohistochemically by using antibody of SWI/SNF components; BRM, BRG1, BAF250a, BAF180 and BAF47. The correlation of expression levels and clinicopathological outcomes were analyzed, followed by the multivariate analysis of prognostic factors for overall survival. The expression levels of the SWI/SNF components were categorized as low or high according to the median value of Histoscore. Statistical analysis revealed that BRM expression was related to tumor size, T factor, M factor, lymphatic invasion and stage BRG1 expression to histology and stage BAF180 expression to tumor size and BAF47 expression to lymphatic invasion, respectively. Multivariate Cox proportional hazard analysis showed that high BRM and low BAF180 expression levels were independent predictors of worse survival in patients with pancreatic cancer. High BRM, and low BAF180 were also independent prognostic factors for poor survival in the subgroup with adjuvant gemcitabine. These results suggest that the specific cofactors of SWI/SNF chromatin remodeling complex certainly have roles in pancreatic cancer. High BRM, and low BAF180 are useful biomarkers for poor prognosis in pancreatic cancer.

Introduction

Pancreatic cancer remains a leading cause of cancer deaths in the advanced nation (1,2). The overall 5-year survival rate is reported to be less than 5% (3). A reliable and clinically relevant prognostic biomarker which can stratify the disease is needed for developing new strategies.

It is a known fact that chromatin, highly condensed and dynamically structured, can be temporally rearranged so that specific genes can be expressed or repressed (4). Studies have shown that modification of chromatin structure is an essential step in gene regulation primarily mediated by chromatin remodeling proteins. Among these proteins, histone is known to play a dynamic role in the regulation of transcription (57). Often, transcription is also regulated by other cofactors, and the balance of chromatin remodeling activities may be crucial to ensure accurate responses to developmental or environmental cues and to prevent the transition of normal cells into cancer cells (8).

The SWItch/sucrose non-fermentable (SWI/SNF) complex is a major complex of adenosine triphosphate (ATP)-dependent chromatin remodeling factors and controls the transcriptional activity of a variety of genes involved in cellular growth and transformation by altering chromatin structure (913). SWI/SNF complex, originally identified in yeast, is composed of more than 10 characterized subunits (14,15) and human SWI/SNF complexes contain one of the two core ATPase subunits, BRM or BRG1 (13,1618). Growing genetic and molecular evidence indicates that specific subunits of the SWI/SNF complex can act as tumor suppressors (6,19). However, there is no report on the relationship between SWI/SNF components expression and the clinical significance of pancreatic cancer. In this study, we investigated the expression levels of SWI/SNF components to clarify the clinical impact of SWI/SNF complex on pancreatic cancer.

Materials and methods

Patients and samples

The surgical specimens of pancreatic cancer tissue obtained from 68 patients were evaluated. All of the patients had undergone macroscopically curative resection (R0, 1) at Kanagawa Cancer Center between July 2006 and April 2010. The clinicopathological characteristics of these patients are shown in Table I. In all cases, archival hematoxylin and eosin-stained (H&E) slides of the primary tumor were retrieved and reviewed to confirm the pathological features as well as to select suitable tissue blocks for immunohistochemical analysis. Informed consent was obtained from each patient. The Ethics Committees of the Kanagawa Cancer Center approved the protocol before initiation of the study. We declare no conflicts of interest.

Table I.

The clinicopathological characteristics of all patients.

Table I.

The clinicopathological characteristics of all patients.

Clinicopathological characteristicsNo. of patients (n=68)
Age
  <6530
  ≥6538
Sex
  Male36
  Female32
Tumor location in pancreas
  Head46
  Body/tail22
Tumor size (cm)
  <429
  ≥439
Histological type
  Well/mod32
  Poor36
T
  T1–338
  T430
N
  N017
  N151
M
  M053
  M115
Curability of surgery
  R043
  R125
Stage
  0–III53
  IV15
Adjuvant gemcitabine
  Yes42
  No26

[i] Well, well differentiated adenocarcinoma; mod, moderately differentiated adenocarcinoma; poor, poorly differentiated adenocarcinoma.

Tissue microarrays and immunohistochemistry

Microarrays consisting of cores, each measuring 2 mm in diameter, were prepared from formalin-fixed paraffin-embedded tissue blocks of surgically removed primary tumors. Each tissue core of the primary tumor was sampled.

Immunohistochemical staining was performed using commercially available polyclonal rabbit, or mouse antibodies raised against BRM (Abcam Inc., Cambridge, MA), BRG1 (Santa Cruz Biotechnology Inc., Santa Cruz, CA), BAF250a (Santa Cruz Biotechnology Inc.), BAF180 (Sigma-Aldrich Inc., St. Louis, MO), BAF47 (Santa Cruz Biotechnology Inc.). Tissue microarray blocks were sectioned at a thickness of 4 μm and mounted on pre-coated glass slides. The sections were de-paraffinized through a graded series of xylene and rehydrated through a graded series of alcohol to distilled water. Endogenous peroxidase was quenched with 3% hydrogen peroxide in methanol at room temperature. The sections were placed in a 95°C solution of 0.01 M sodium citrate buffer (pH 6.0) for 40 min for antigen retrieval. Normal goat serum (5%) was then applied for 15 min to block any non-specific protein binding sites. Primary polyclonal antibodies were applied for 1 h at room temperature at the following dilutions: anti-BRM at 1:250, anti-BRG1 at 1:200, anti-BAF250a at 1:100, anti-BAF180 at 1:90 and BAF47 at 1:300. Immunoreactive proteins were detected using the Simple Stain MAX-PO (Multi).

All sections were counterstained with Mayer’s hematoxylin, and negative controls were included in each staining sequence. The intensity and global level of staining were scored semi-quantitatively for each tissue microarray by an investigator blinded to all of the clinicopathological variables. The global level of staining refers to the percentage of tumor cells that stained positively for an antibody within each tissue microarray at ×200 magnification using a light microscope.

Scoring of immunohistochemical reactivity

Immunohistochemical scoring was completed using the modified Histoscore (H-score) (20), which involves a semiquantitative assessment of both the intensity of staining (graded as: 0, non-staining; 1, weak; 2, median; or 3, strong using adjacent normal mucosa as the median) and the percentage of positive cells (Fig. 1). The range of possible scores was from 0 to 300. Expression level of each component was categorized as low or high according to the median value of H-score.

Statistical analysis

The relationships between the expression level and the clinicopathological factors were evaluated with the χ2 test. The postoperative survival rate from the day of primary tumor resection was analyzed using the Kaplan-Meier method and any differences in the survival rates were assessed with the log-rank test. A Cox proportional-hazard model was used for the multivariate analyses. Differences were considered significant when P<0.05. The statistical analysis was performed using the PASW Statistics 18 (SPSS, Inc., Chicago, IL).

Results

Relation of SWI/SNF component expression to clinicopathological features

The distribution of H-score is showed in Fig. 2. Expression level of the SWI/SNF components was categorized as low or high according to the median value of the H-score. Relations between the expression levels of each component and clinicopathological features were then examined. Factors implicating significant relations were tumor size, T factor, M factor, lymphatic invasion, and stage in BRM, histology and stage in BRG1, tumor size in BAF180, lymphatic invasion in BAF47, respectively (Table II).

Table II.

Relation of SWI/SNF component expression to clinicopathological factors.

Table II.

Relation of SWI/SNF component expression to clinicopathological factors.

BRM
BRG1
BAF250a
BAF180
BAF47
FactorsLowHighp-valueLowHighp-valueLowHighp-valueLowHighp-valueLowHighp-value
Age (years)
  <65/≥6515/1915/191.00018/1912/220.14313/2117/170.32919/1511/230.05113/2117/170.329
Gender
  Male/female16/1816/181.00016/1816/181.00013/2119/150.14515/1917/170.62717/1715/190.627
Tumor size
  <4/≥4 cm19/1510/240.02712/2217/170.22014/2015/190.80610/2419/150.02715/1914/200.806
Histology
  Well, mod/poor18/1614/200.33111/2321/130.01514/2018/160.33113/2119/150.14515/1917/170.627
T
  T1-3/425/913/210.00323/1115/190.05117/1721/130.32919/1519/151.00020/1418/160.625
N
  N0/N19/258/260.77910/247/270.40110/247/270.4018/269/250.7799/258/260.779
M
  M0/M130/423/110.04127/726/80.77024/1029/50.11425/928/60.38028/625/90.380
Vessel invasion
  No/yes12/228/260.28711/239/250.5957/2713/210.11010/2410/241.0008/2612/220.287
Lymphatic invasion
  No/yes15/196/280.01813/218/260.1899/2512/220.4319/2512/220.43115/196/280.018
Stage
  0–III/IV18/165/290.00117/176/280.00510/2413/210.44211/2312/220.79814/209/250.200
Curability
  R0/R125/918/160.07823/1120/140.45120/1423/110.45121/1322/120.80120/1423/110.451

[i] Well, well differentiated adenocarcinoma; mod, moderately differentiated adenocarcinoma; poor, poorly differentiated adenocarcinoma; inv, invasion.

Analysis of prognostic factors in all patients

Univariate Cox regression analysis for overall survival in all patients showed that age, tumor size, histological type, M factor, curability of the surgery, and expression level of BRM as well as BAF180 were significant predictors (Table III). On multivariate Cox proportional hazard analysis, histology, expression level of BRM and BAF180 were significant independent predictors of overall survival in patients with pancreatic cancer (Table IV).

Table III.

Univariate analysis for overall survival in pancreatic cancer.

Table III.

Univariate analysis for overall survival in pancreatic cancer.

FactorsHR (95% CI)p-value
Age (years)0.035
  <651.0
  ≥650.533 (0.293–0.967)
Sex0.632
  Male1.0
  Female0.865 (0.478–1.565)
Tumor size (cm)0.035
  <41.0
  ≥41.979 (1.048–3.739)
Histology0.002
  Well/mod1.0
  Poor2.744 (1.429–5.271)
T0.071
  T1–31.0
  T41.733 (0.955–3.146)
N0.602
  N01.0
  N11.208 (0.594–2.458)
M0.010
  M01.0
  M12.329 (1.222–4.439)
Curability of surgery0.020
  R01.0
  R12.068 (1.121–3.815)
BRM0.011
  Low1.0
  High2.225 (1.199–4.129)
BRG10.601
  Low1.0
  High0.853 (0.471–1.546)
BAF250a0.479
  Low1.0
  High0.807 (0.446–1.461)
BAF1800.007
  Low1.0
  High0.428 (0.231–0.793)
BAF470.226
  Low1.0
  High0.690 (0.378–1.258)

[i] HR, hazard ratio; 95% CI, 95% confidence interval; well, well differentiated adenocarcinoma; mod, moderately differentiated adenocarcinoma; poor, poorly differentiated adenocarcinoma.

Table IV.

Multivariate analysis for overall survival in pancreatic cancer.

Table IV.

Multivariate analysis for overall survival in pancreatic cancer.

FactorsHR (95% CI)p-value
Age0.169
  <651.0
  ≥650.633 (0.330–1.214)
Tumor size (cm)0.755
  <41.0
  ≥41.122 (0.543–2.318)
Histology0.011
  Well/Mod1.0
  Poor2.702 (1.253–5.830)
M0.486
  M01.0
  M11.381 (0.557–3.424)
Curability of surgery0.076
  R01.0
  R11.981 (0.932–4.214)
BRM0.032
  Low1.0
  High2.144 (1.066–4.311)
BAF1800.041
  Low1.0
  High0.501 (0.258–0.971)

[i] HR, hazard ratio; 95% CI, 95% confidence interval; well, well differentiated adenocarcinoma; mod, moderately differentiated adenocarcinoma; poor, poorly differentiated adenocarcinoma.

Comparison of survival by the status of BRM and BAF180

The 5-year survival rate of high BRM patients was 9.8%, which was significantly worse than that of low BRM patients (43.8%) (Fig. 3). Also, the 5-year survival rate of low BAF180 (8.1%) was significantly worse than that of high BAF180 patients (40.8%) (Fig. 3).

Hazard analysis of SWI/SNF components in the patients treated with adjuvant gemcitabine

Multivariate analysis (Table V) and survival analysis (Fig. 4) showed that BRM-high and BAF180-low were independent prognostic factors for overall survival in the patients treated with adjuvant gemcitabine.

Table V.

Multivariate analysis for overall survival in patients with adjuvant gemcitabine.

Table V.

Multivariate analysis for overall survival in patients with adjuvant gemcitabine.

FactorsHR (95% CI)p-value
Age0.002
  <651.0
  ≥650.227 (0.089–0.580)
Tumor size (cm)0.280
  <41.0
  ≥40.593 (0.230–1.531)
Histology0.267
  Well/Mod1.0
  Poor1.907 (0.610–5.964)
M0.923
  M01.0
  M10.947 (0.315–2.847)
Curability of surgery0.784
  R01.0
  R11.145 (0.433–3.029)
BRM0.017
  Low1.0
  High3.411 (1.251–9.305)
BAF1800.016
  Low1.0
  High0.336 (0.138–0.819)

[i] HR, hazard ratio; 95% CI, 95% confidence interval; well, well differentiated adenocarcinoma; mod, moderately differentiated adenocarcinoma; poor, poorly differentiated adenocarcinoma.

Discussion

Chromatin remodeling factors have been the subject of great interest in oncology. However, little is known about their role in pancreatic cancer.

The SWI/SNF complexes are large, multi-subunit complexes containing 10 or more subunits, serving as a master switch that directs and limits the execution of specific cellular programs, such as differentiation and growth control (21). Each complex has one of the two different ATPase as core motor; BRM or BRG1, and subunits which are referred to as BAFs (BRM- or BRG1-associated factors). The BRM-containing complex is termed BRM/BAF. The BRG1-containing complexes are further divided into those that contain the BAF250a (termed BRG1/BAF) or the BAF180 (termed PBAF). These three types of complexes are believed to have different molecular functions (22).

There are several studies reporting that the subunit of SWI/SNF complex was decreased in cancer tissues. They revealed the mutation of ARID1A, which codes BAF250a protein, in about half of ovarian clear cell carcinomas (23,24), and PBRM1, which codes BAF180, in approximately 40% of renal cell carcinomas (25). Another study identified the SWI/SNF chromatin remodeling complex as tumor suppressor, by mediating retinoblastoma protein (RB)-derived regulation of the cell cycle (22,26,27). However, the roles of these subunits in pancreatic cancers are poorly understood.

In this study, we investigated the expression levels of 5 key subunits; BRM, BRG1, BAF250a, BAF180, which are the key subunits when subdividing complex types, and BAF47. There is established evidence that BAF47 is a tumor suppressor in rhabdoid tumors (28).

In the analysis of expression level and clinicopahological features, high BRM was related to worse clinicopathological features in general, including larger tumor size, T4 disease, other organ metastasis, lymphatic invasion, and stage IV disease. Stage IV disease was also correlated to high BRG1, which is reported to have similar biological function as BRM. On the other hand, better clinicopathological features were related to high BAF expression. High BAF180 was related to smaller tumor size, and high BAF47 was associated with negative lymphatic invasion.

In addition, our multivariate analysis revealed both high BRM and low BAF180 were independent prognostic indicators for poor survival, whereas the expression level of BRG1, BAF250a, and BAF47 were not related to overall survival.

As a next step, we investigated the prognostic significance of these factors in the patients with adjuvant gemcitabine. Gemcitabine remains standard therapy in the adjuvant and palliative settings for pancreatic cancer (29,30). However, the response rate of gemcitabine is very low, with only 18% of 1-year survival rate (31). Developing a novel biomarker, which predicts the response for gemcitabine, is urgently needed. In the analysis of the patients with gemcitabine, we reached the same result; both high BRM and low BAF180 were independent prognostic indicators for poor survival.

A previous study showed that BRM or BRG1 is lost in 10–20% of the bladder, colon, breast, esophageal, pancreatic and ovarian cancers by immunohistochemical staining of tissue microarrays (32). Another study reported BRM was lost in approximately 15–20% of primary non-small lung cancers, and silencing of BRM was a prognostic factor for poor outcome (33,34). Although BRM is supposed to be involved in many biological functions, these data showed BRM-containing complexes (BRM/BAF) as tumor suppressor in cancer tissue.

It is also reported that BRM has a role in trans cription of CD44 (35), which is important in the process of tumor-endothelium interactions, cell migration, cell adhesion, tumor progression and metastasis (36).

Our result showed that the patient with high BRM had a significantly worse survival than those without (5-year OS: 9.8 vs. 43.8%, p=0.009), suggesting BRM/BAF in pancreatic cancer may contribute to tumor progression.

We also revealed the significant relationship between high BAF180 expression and smaller-sized tumor, and identified BAF180 as an independent prognostic factor for better survival in pancreatic cancer.

BAF180 maps to the 3p12 region (37) where allele loss is frequent and homozygous deletion have been detected in lung and breast cancer cell lines (38,39). Thus, genes located on this region have been thought as candidates for tumor suppressors. Actually, it is reported that BAF180 mutation is associated with carcinogenesis of breast cancer, and BAF180 suppresses tumorigenesis through its ability to regulate p21 (40), which controls the cell cycle (41). Recent research also clarified BAF180 mutation in clear cell renal cell carcinoma (42). These results suggest the idea that BAF180-containing complexes (PBAF) suppress tumor progression, which does not contradict our present results.

BAF250a-containing SWI/SNF complexes (BRG1/BAF) are reported to have different structure and biological properties from PBAF (43,44). A previous study showed that BAF250a was deleted in as many as 30% of renal cell carcinoma and 10% of breast carcinoma (19,45). These results lead to the concept that BRG1/BAF appear to have antagonistic effect on cell cycle progression (46). However, our data did not show the relationship of BAF250a expression to clinicopathological features or overall survival in pancreatic cancer.

Based on this study, we reached the conclusion that high BRM, and low BAF180 are useful biomarker not only for the patients with curative resection, but also for those with adjuvant gemcitabine. Future investigation into biological functions of SWI/SNF components could lead to better management in pancreatic cancer.

References

1. 

Parkin DM, Bray FI and Devesa SS: Cancer burden in the year 2000. The global picture. Eur J Cancer. 37(Suppl 8): S4–S66. 2001.PubMed/NCBI

2. 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar

3. 

Hidalgo M: Pancreatic cancer. N Engl J Med. 362:1605–1617. 2010. View Article : Google Scholar

4. 

Reisman DN, Strobeck MW, Betz BL, et al: Concomitant down-regulation of BRM and BRG1 in human tumor cell lines: differential effects on RB-mediated growth arrest vs CD44 expression. Oncogene. 21:1196–1207. 2002. View Article : Google Scholar : PubMed/NCBI

5. 

Wade PA: Transcriptional control at regulatory checkpoints by histone deacetylases: molecular connections between cancer and chromatin. Hum Mol Genet. 10:693–698. 2001. View Article : Google Scholar

6. 

Neely KE and Workman JL: The complexity of chromatin remodeling and its links to cancer. Biochim Biophys Acta. 1603:19–29. 2002.PubMed/NCBI

7. 

Klochendler-Yeivin A, Muchardt C and Yaniv M: SWI/SNF chromatin remodeling and cancer. Curr Opin Genet Dev. 12:73–79. 2002. View Article : Google Scholar

8. 

Davis PK and Brackmann RK: Chromatin remodeling and cancer. Cancer Biol Ther. 2:22–29. 2003. View Article : Google Scholar

9. 

Narlikar GJ, Fan HY and Kingston RE: Cooperation between complexes that regulate chromatin structure and transcription. Cell. 108:475–487. 2002. View Article : Google Scholar : PubMed/NCBI

10. 

Langst G and Becker PB: Nucleosome mobilization and positioning by ISWI-containing chromatin-remodeling factors. J Cell Sci. 114:2561–2568. 2001.PubMed/NCBI

11. 

Kingston RE and Narlikar GJ: ATP-dependent remodeling and acetylation as regulators of chromatin fluidity. Genes Dev. 13:2339–2352. 1999. View Article : Google Scholar : PubMed/NCBI

12. 

Burns LG and Peterson CL: The yeast SWI-SNF complex facilitates binding of a transcriptional activator to nucleosomal sites in vivo. Mol Cell Biol. 17:4811–4819. 1997.PubMed/NCBI

13. 

Wu C: Chromatin remodeling and the control of gene expression. J Biol Chem. 272:28171–28174. 1997. View Article : Google Scholar : PubMed/NCBI

14. 

Breeden L and Nasmyth K: Cell cycle control of the yeast HO gene: cis- and trans-acting regulators. Cell. 48:389–397. 1987. View Article : Google Scholar : PubMed/NCBI

15. 

Recht J and Osley MA: Mutations in both the structured domain and N-terminus of histone H2B bypass the requirement for Swi-Snf in yeast. EMBO J. 18:229–240. 1999. View Article : Google Scholar : PubMed/NCBI

16. 

Phelan ML, Sif S, Narlikar GJ and Kingston RE: Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits. Mol Cell. 3:247–253. 1999. View Article : Google Scholar : PubMed/NCBI

17. 

Tyler JK and Kadonaga JT: The ‘dark side’ of chromatin remodeling: repressive effects on transcription. Cell. 99:443–446. 1999.

18. 

Wang W, Cote J, Xue Y, et al: Purification and biochemical heterogeneity of the mammalian SWI-SNF complex. EMBO J. 15:5370–5382. 1996.PubMed/NCBI

19. 

Decristofaro MF, Betz BL, Rorie CJ, Reisman DN, Wang W and Weissman BE: Characterization of SWI/SNF protein expression in human breast cancer cell lines and other malignancies. J Cell Physiol. 186:136–145. 2001. View Article : Google Scholar : PubMed/NCBI

20. 

McCarty KS Jr, Miller LS, Cox EB, Konrath J and McCarty KS Sr: Estrogen receptor analyses. Correlation of biochemical and immunohistochemical methods using monoclonal antireceptor antibodies. Arch Pathol Lab Med. 109:716–721. 1985.

21. 

Bourgo RJ, Siddiqui H, Fox S, et al: SWI/SNF deficiency results in aberrant chromatin organization, mitotic failure, and diminished proliferative capacity. Mol Biol Cell. 20:3192–3199. 2009. View Article : Google Scholar : PubMed/NCBI

22. 

Reisman D, Glaros S and Thompson EA: The SWI/SNF complex and cancer. Oncogene. 28:1653–1668. 2009. View Article : Google Scholar : PubMed/NCBI

23. 

Wiegand KC, Shah SP, Al-Agha OM, et al: ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 363:1532–1543. 2010. View Article : Google Scholar : PubMed/NCBI

24. 

Jones S, Wang TL, Shih Ie M, et al: Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science. 330:228–231. 2010. View Article : Google Scholar : PubMed/NCBI

25. 

Varela I, Tarpey P, Raine K, et al: Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 469:539–542. 2011. View Article : Google Scholar : PubMed/NCBI

26. 

Weissman B and Knudsen KE: Hijacking the chromatin remodeling machinery: impact of SWI/SNF perturbations in cancer. Cancer Res. 69:8223–8230. 2009. View Article : Google Scholar : PubMed/NCBI

27. 

Roberts CW and Orkin SH: The SWI/SNF complex - chromatin and cancer. Nat Rev Cancer. 4:133–142. 2004. View Article : Google Scholar : PubMed/NCBI

28. 

Biegel JA, Kalpana G, Knudsen ES, et al: The role of INI1 and the SWI/SNF complex in the development of rhabdoid tumors: meeting summary from the workshop on childhood atypical teratoid/rhabdoid tumors. Cancer Res. 62:323–328. 2002.PubMed/NCBI

29. 

Heinemann V, Boeck S, Hinke A, Labianca R and Louvet C: Meta-analysis of randomized trials: evaluation of benefit from gemcitabine-based combination chemotherapy applied in advanced pancreatic cancer. BMC Cancer. 8:822008. View Article : Google Scholar

30. 

El-Rayes BF and Philip PA: A review of systemic therapy for advanced pancreatic cancer. Clin Adv Hematol Oncol. 1:430–434. 2003.PubMed/NCBI

31. 

Moore MJ, Goldstein D, Hamm J, et al: Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 25:1960–1966. 2007. View Article : Google Scholar

32. 

Glaros S, Cirrincione GM, Muchardt C, Kleer CG, Michael CW and Reisman D: The reversible epigenetic silencing of BRM: implications for clinical targeted therapy. Oncogene. 26:7058–7066. 2007. View Article : Google Scholar : PubMed/NCBI

33. 

Reisman DN, Sciarrotta J, Wang W, Funkhouser WK and Weissman BE: Loss of BRG1/BRM in human lung cancer cell lines and primary lung cancers: correlation with poor prognosis. Cancer Res. 63:560–566. 2003.PubMed/NCBI

34. 

Fukuoka J, Fujii T, Shih JH, et al: Chromatin remodeling factors and BRM/BRG1 expression as prognostic indicators in non-small cell lung cancer. Clin Cancer Res. 10:4314–4324. 2004. View Article : Google Scholar : PubMed/NCBI

35. 

Batsche E, Yaniv M and Muchardt C: The human SWI/SNF subunit Brm is a regulator of alternative splicing. Nat Struct Mol Biol. 13:22–29. 2006. View Article : Google Scholar : PubMed/NCBI

36. 

Martin TA, Harrison G, Mansel RE and Jiang WG: The role of the CD44/ezrin complex in cancer metastasis. Crit Rev Oncol Hematol. 46:165–186. 2003. View Article : Google Scholar : PubMed/NCBI

37. 

Xue Y, Canman JC, Lee CS, et al: The human SWI/SNF-B chromatin-remodeling complex is related to yeast rsc and localizes at kinetochores of mitotic chromosomes. Proc Natl Acad Sci USA. 97:13015–13020. 2000. View Article : Google Scholar : PubMed/NCBI

38. 

Maitra A, Wistuba II, Washington C, et al: High-resolution chromosome 3p allelotyping of breast carcinomas and precursor lesions demonstrates frequent loss of heterozygosity and a discontinuous pattern of allele loss. Am J Pathol. 159:119–130. 2001. View Article : Google Scholar

39. 

Zabarovsky ER, Lerman MI and Minna JD: Tumor suppressor genes on chromosome 3p involved in the pathogenesis of lung and other cancers. Oncogene. 21:6915–6935. 2002. View Article : Google Scholar : PubMed/NCBI

40. 

Xia W, Nagase S, Montia AG, et al: BAF180 is a critical regulator of p21 induction and a tumor suppressor mutated in breast cancer. Cancer Res. 68:1667–1674. 2008. View Article : Google Scholar : PubMed/NCBI

41. 

Harper JW, Adami GR, Wei N, Keyomarsi K and Elledge SJ: The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell. 75:805–816. 1993. View Article : Google Scholar : PubMed/NCBI

42. 

Duns G, Hofstra RM, Sietzema JG, et al: Targeted exome sequencing in clear cell renal cell carcinoma tumors suggests aberrant chromatin regulation as a crucial step in ccRCC development. Hum Mutat. 33:1059–1062. 2012. View Article : Google Scholar : PubMed/NCBI

43. 

Nie Z, Xue Y, Yang D, et al: A specificity and targeting subunit of a human SWI/SNF family-related chromatin-remodeling complex. Mol Cell Biol. 20:8879–8888. 2000. View Article : Google Scholar : PubMed/NCBI

44. 

Sekine I, Sato M, Sunaga N, et al: The 3p21 candidate tumor suppressor gene BAF180 is normally expressed in human lung cancer. Oncogene. 24:2735–2738. 2005. View Article : Google Scholar : PubMed/NCBI

45. 

Wang X, Nagl NG Jr, Flowers S, Zweitzig D, Dallas PB and Moran E: Expression of p270 (ARID1A), a component of human SWI/SNF complexes, in human tumors. Int J Cancer. 112:6362004. View Article : Google Scholar : PubMed/NCBI

46. 

Nagl NG Jr, Zweitzig DR, Thimmapaya B, Beck GR Jr and Moran E: The c-myc gene is a direct target of mammalian SWI/SNF-related complexes during differentiation-associated cell cycle arrest. Cancer Res. 66:1289–1293. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February 2013
Volume 42 Issue 2

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
Numata M, Morinaga S, Watanabe T, Tamagawa H, Yamamoto N, Shiozawa M, Nakamura Y, Kameda Y, Okawa S, Rino Y, Rino Y, et al: The clinical significance of SWI/SNF complex in pancreatic cancer. Int J Oncol 42: 403-410, 2013
APA
Numata, M., Morinaga, S., Watanabe, T., Tamagawa, H., Yamamoto, N., Shiozawa, M. ... Miyagi, Y. (2013). The clinical significance of SWI/SNF complex in pancreatic cancer. International Journal of Oncology, 42, 403-410. https://doi.org/10.3892/ijo.2012.1723
MLA
Numata, M., Morinaga, S., Watanabe, T., Tamagawa, H., Yamamoto, N., Shiozawa, M., Nakamura, Y., Kameda, Y., Okawa, S., Rino, Y., Akaike, M., Masuda, M., Miyagi, Y."The clinical significance of SWI/SNF complex in pancreatic cancer". International Journal of Oncology 42.2 (2013): 403-410.
Chicago
Numata, M., Morinaga, S., Watanabe, T., Tamagawa, H., Yamamoto, N., Shiozawa, M., Nakamura, Y., Kameda, Y., Okawa, S., Rino, Y., Akaike, M., Masuda, M., Miyagi, Y."The clinical significance of SWI/SNF complex in pancreatic cancer". International Journal of Oncology 42, no. 2 (2013): 403-410. https://doi.org/10.3892/ijo.2012.1723