Reduced expression of TRF1 is associated with tumor progression and poor prognosis in oral squamous cell carcinoma

  • Authors:
    • Hui-Ching Chuang
    • Chang-Han Chen
    • Chao-Cheng Huang
    • Fu-Min Fang
    • Hsin-Ting Tsai
    • Chih-Yen Chien
  • View Affiliations

  • Published online on: December 2, 2010     https://doi.org/10.3892/etm.2010.168
  • Pages: 63-67
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The functions of telomeric repeat-binding factor 1 (TRF1) and 2 (TRF2) in oral carcinogenesis are largely unexplored. This study examined the relationship between the expression of TRF1 and TRF2 and clinicopathological variables and survival in oral cavity squamous cell carcinoma (OCSCC). Western blotting and immunohistochemistry were used to evaluate the protein expression of TRF1 and TRF2 in paired OCSCC patient specimens. Expression of TRF1 and TRF2 was assessed by immunohistochemistry in 256 OCSCC patients who underwent tumor resection without previous radiotherapy. The results were analyzed using Fisher's exact test. Protein expression of TRF1 and TRF2 was significantly lower in the OCSCC than in the adjacent non-tumor tissue. Reduced TRF1 and TRF2 levels in 256 patients, as revealed by immunohistochemistry, were significantly associated with aggressive clinicopathological features, such as advanced tumor stage (p<0.001) and advanced tumor node metastasis stage (p<0.001). According to Kaplan-Meier analysis, reduced TRF1 expression was significantly correlated with an unfavorable cumulative 5-year overall survival rate (p<0.001). In conclusion, decreased expression of TRF1 was significantly associated with tumor progression and poor prognosis in OCSCC patients.

Introduction

Oral cavity squamous cell carcinoma (OCSCC) accounts for at least 90% of all oral malignancies. It is a multifactorial condition with etiological links to a wide variety of external causes of cancer, including alcohol, tobacco and betel nut use, and certain viral infections. The high and increasing prevalence of OCSCC in Taiwan has been attributed to the popularity of betel nut chewing. It was estimated that, in 2006, more than 4,000 people in Taiwan were diagnosed with oral cancer. This represents 5.49% of all newly diagnosed malignancies. Despite advances in technology and the implementation of multidisciplinary treatment programs, only modest improvements in survival rates have been achieved, and these are primarily due to earlier diagnosis, rather than improved therapeutic interventions (1). Moreover, the rate of recurrence of advanced tumors remains relatively high. Salvage outcomes are unsatisfactory, although they depend on the stage of the recurrent tumors (2). Investigation of OCSCC progression from a genetic perspective has identified distinct patterns and timings of genetic alterations (3). The most important prognostic factors in OCSCC are those that form part of the grading system, including tumor stage and lymph node status (46). The identification of new prognostic factors linked to OCSCC initiation and progression may aid in the development of new diagnostic tools and treatment strategies.

Among the various molecular factors implicated in carcinogenesis, telomere dysfunction has emerged as an early event associated with genetic instability. Telomeres stabilize the ends of chromosomes, protect them from end-to-end fusion and mediate chromosome pairing during cell division (710). Recently, telomere-associated proteins, such as telomeric repeat-binding factor 1 (TRF1) and 2 (TRF2), have been identified as putative modulators of telomerase activity and have been suggested to play key roles in the maintenance of the telomere function (8,9,11,12). Several reports have indicated that the altered expression of TRF1 and TRF2 proteins is associated with tumor progression in various human carcinomas, including lung, stomach, adrenal and pancreatic cancer; the altered expression has also been identified in malignant hematopoietic cells and colorectal pre-neoplastic lesions (1320). However, the relationship between TRF1 and TRF2 and OCSCC remains unclear. The aim of the present study was to examine TRF1 and TRF2 expression in OCSCC and to determine its relationship with clinicopathological variables and survival.

Materials and methods

Patients and tumor samples

The study population included 256 OCSCC patients who underwent primary surgical resection without previous radiotherapy and/or chemotherapy between October 1996 and August 2005. Clinicopathological information for each subject, including gender, age, tumor (T) stage, nodal (N) status, tumor node metastasis (TNM) stage and overall survival, was obtained retrospectively from clinical records and pathological reports. TNM status was classified according to the 1997 American Joint Committee on Cancer (AJCC) system. The study was approved by the Medical Ethics and Human Clinical Trial Committee at Chang Gung Memorial Hospital, Taipei, Taiwan. The patient group comprised 17 women and 239 men, with an average age of 50.9 years (range, 26–87 years). Thirty-nine patients were diagnosed with T1 tumors, 55 with T2, 64 with T3 and 98 with T4. A total of 153 patients had an N status of N0, 38 had N1, 48 had N2b, 13 had N2c and 4 had N3. Thirty-four patients had stage I tumors, 38 stage II, 61 stage III and 123 stage IV.

Immunoblot analysis

For tissue protein extraction, frozen samples (adjacent non-tumor and tumor tissues) were homogenized in RIPA lysis buffer (50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 1% NP-40, 0.5% Na-deoxycholate and 0.1% SDS), and the protein concentrations were quantified using a Bio-Rad Protein assay (Bio-Rad, Hercules, CA, USA). Immunoblotting was performed according to standard procedures. Anti-TRF1 and -TRF2 polyclonal antibodies and the anti-GAPDH monoclonal antibody (Santa Cruz Biotechnology Santa Cruz, CA, USA) were used. The bound primary antibody was detected by incubation with HRP-conjugated secondary antibody (Bio/Can Scientific, Mississauga, ON, Canada). Blots were developed using the Western Lighting reagent, and protein bands were visualized using X-ray film.

Immunohistochemical analysis

OCSCC and adjacent non-cancerous tissue samples were identified by a pathologist based on diagnosis and microscopic morphology. Tumor tissues were fixed in 10% buffered formalin, embedded in paraffin and decalcified in 10% EDTA. Formalin-fixed paraffin-embedded tissue was sectioned to a thickness of 4 μm, and the sections were deparaffinized in xylene and rehydrated in a graded series of ethanol (100, 90, 80 and 70%). The sections were washed in phosphate-buffered saline (PBS) and treated with 3% H2O2 for 30 min to block the endogenous peroxidase activity. Antigens were retrieved by microwaving the sections in 10 mM citrate buffer, pH 6.0. The sections were then incubated with anti-TRF1 and -TRF2 antibodies (diluted 1:100) for 1 h, washed in PBS and incubated for 30 min with a horseradish peroxidase-Fab polymer conjugate (PicTure™-Plus kit; Zymed, South San Francisco, CA, USA). After the sections were washed in PBS, the immunoreactive bands were visualized by incubation with 3,3′-diaminobenzidine for 5 min. As a negative control, the primary antibody was omitted. Two pathologists blinded to the subjects’ clinical information independently evaluated the reactivity level of the immunostained tissues in 15–20 high-power fields. Criteria were developed for quantitating the immunoreactivity of the TRF1 and TRF2 staining in the adjacent non-tumor and tumor sections using a score range of 0 to +3. A percentage value of 0 indicated 0–25% of the area stained; +1, 25–50%; +2, 50–75% and +3, >75% stained. Similarly, the staining intensity was graded as +0, +1, +2 or +3. High expression of TRF1 and TRF2 was defined as ≥+2 in both scoring methods. Low expression of TRF1 and TRF2 was defined as ≤+1 in both scoring methods.

Statistical analysis

Fisher’s exact test was used to evaluate the correlation between TRF1 and TRF2 expression and various clinicopathological variables, including gender, age, N status, T stage and TNM stage. A p-value <0.05 was considered to indicate statistical significance. TRF1 and TRF2 expression and the clinicopathological variables were used in a Kaplan-Meier analysis of survival, and statistical significance (p<0.05) was assessed by the log-rank test. To determine the effects of specific prognostic factors on survival, a multivariate analysis was performed using Cox’s regression model.

Results

Down-regulation of TRF1 and TRF2 expression in OCSCC tissues

To investigate the potential roles of TRF1 and TRF2 in the pathogenesis of OCSCC, their expression was assessed in representative and paired tumor and adjacent non-cancerous tissue samples by Western blot analysis using anti-human TRF1 and TRF2 polyclonal antibodies. The protein expression levels of TRF1 and TRF2 were lower in the tumor samples compared to those in the paired non-cancerous tissues (Fig. 1).

TRF1 and TRF2 expression in the tumor and adjacent non-cancerous tissues from the 256 OCSCC patients was also examined immunohistochemically. Representative results of TRF1 and TRF2 immunostaining are presented in Fig. 2A. Staining was stronger in the adjacent non-cancerous tissues (Fig. 2A-a and -f) than the tumor tissues (Fig. 2A-b and -c, TRF1; Fig. 2A-g and -h, TRF2). Moreover, TRF1 and TRF2 expression levels in the tumor samples were negatively correlated with T stage (Fig. 2A-b and -c, TRF1; Fig. 2A-g and -h, TRF2) and N stage (Fig. 2A-d and -e, TRF1; Fig. 2A-i and -j, TRF2). Notably, TRF1 and TRF2 were focally expressed in the nuclei of both tumor and non-cancerous cells (Fig. 2A-b and -c, TRF1; Fig. 2A-g and -h, TRF2).

To investigate whether the expression of TRF1 and TRF2 is associated with various prognostic factors, including age, gender and TNM pathologic classification, we classified the patients into two groups based on the immunohistochemical analysis: low (−/+) and high (++/+++) TRF1 or TRF2 expression. Low TRF1 expression was correlated with advanced T stage (p<0.001) and advanced TNM stage (p<0.001). Low TRF2 expression was correlated with advanced T stage (p<0.001) and advanced TNM stage (p<0.001), as well as positive lymph node metastasis (p=0.022). Neither TRF1 nor TRF2 expression was correlated with age or gender (Table I). These findings suggest that TRF1 and TRF2 expression levels may be linked to tumor progression in OCSCC.

Table I.

Correlation between the clinicopathological features and expression of TRF1 and TRF2 in the oral squamous cell carcinoma cases.

Table I.

Correlation between the clinicopathological features and expression of TRF1 and TRF2 in the oral squamous cell carcinoma cases.

VariablesNo. of patientsTRF1 expression
P-valueTRF2 expression
P-value
LowHighLowHigh
Gender0.4520.434
  Male2399514483156
  Female17512413
Age (years)0.2720.333
  <602027512772130
  ≥605425291539
Tumor stage<0.001a<0.001a
  T1 and T29415791183
  T3 and T416285777686
Nodal stage0.0900.022a
  Negative1535310043110
  Positive10347564459
TNM stage<0.001a<0.001a
  I and II66561165
  III and IV190959586104

a Statistically significant

TRF1 and TRF2 expression and OCSCC patient survival

In view of the finding that TRF1 and TRF2 expression levels were associated with T stage, we investigated whether TRF1 and TRF2 expression was correlated with patient prognosis. As shown in Fig. 2B, Kaplan-Meier overall survival analysis revealed that the prognosis of patients with low (−/+) tumor expression of TRF1 and TRF2 was significantly poorer than that of patients displaying higher (++/+++) expression (p<0.001). Univariate analysis revealed that advanced T stage (p<0.001), positive N stage (p<0.001), advanced TNM stage (p<0.001), low TRF1 expression and low TRF2 expression each predicted a significantly worse prognosis for OCSCC patients (Table II). The prognosis was not associated with age or gender. Cox regression analysis revealed that T stage (95% CI, 1.585–5.037; RR=2.826; p<0.001), N status (95% CI, 1.966–4.681; RR=3.034; p<0.001) and TRF1 expression (95% CI, 0.391–0.924; RR=0.601; p=0.02) were independent prognostic factors for survival. These results clearly indicate that the clinical prognosis for OCSCC patients is affected by the tumor expression of TRF1 and TRF2, and suggest that TRF1 and TRF2 may be good prognostic indicators in OCSCC.

Table II.

Cumulative 5-year overall survival rate according to clinicopathological features.

Table II.

Cumulative 5-year overall survival rate according to clinicopathological features.

VariablesNo. of patientsCumulative 5-year overall survival rate (%)P-value
Gender0.1360
  Male23962.5
  Female1781.9
TRF1 expression<0.0010a
  Low10073.3
  High15649.1
TRF2 expression0.0003a
  Low8770.1
  High16950.3
Age (years)0.1300
  <6020266.2
  ≥605455.1
Tumor stage<0.0010a
  T1 and T29484.0
  T3 and T416256.5
Nodal stage<0.0010a
  Negative15377.0
  Positive10344.7
TNM stage<0.0010a
  I and II6692.4
  III and IV19053.9

a Statistically significant.

Discussion

To our knowledge, this is the first investigation of TRF1 and TRF2 expression in primary OCSCC specimens from a large cohort of patients. Our results indicate that the reduced expression of TRF1 and TRF2 is associated with increased tumor aggressiveness and poor prognosis in OCSCC patients. Low expression of TRF1 and TRF2 and advanced T stage and N stage were correlated with a poor prognosis. As it is difficult to determine a prognosis for these patients, TRF1 and TRF2 staining of oral cancer cells may be helpful in selecting an appropriate therapeutic strategy following surgery. Our findings suggest that TRF1 is a good prognostic indicator in OCSCC and a candidate molecular target for oral cancer therapy.

Semi-quantitative RT-PCR analysis of TRF1 and TRF2 mRNA expression in total RNA from tumor samples and matched adjacent non-cancerous tissues from 5 OCSCC patients demonstrated that TRF1 and TRF2 mRNA levels did not differ significantly between the tumor and adjacent non-cancerous tissues (data not shown). This observation suggests that altered TRF1 and TRF2 protein expression during the development of OCSCC may be realized post-transcriptionally.

Telomere-binding proteins have attracted increasing interest due to their essential roles in regulating the length of telomeric DNA tracts and protecting against chromosomal end-to-end fusion (21). In cancer, telomeres become dysfunctional due to the loss or alteration of telomere-binding proteins involved in telomere maintenance or to DNA damage caused by oxidative stress (22). The telomere-binding proteins TRF1 and TRF2 are crucial for the protection and maintenance of telomeres in mammalian cells (8,23). TRF1 and TRF2 contain a Myb-like helix-turn-helix domain in the C-terminus of the protein, and a conserved central domain that is responsible for the formation of homodimers (24). Previous studies have indicated that TRF1 and TRF2 are down-regulated in malignant tissues (13,14,2528). To our knowledge, the present study is the first to report not only that TRF1 and TRF2 are down-regulated in OCSCC, but also that their expression levels are correlated with the clinical characteristics of tumors. The correlation of TRF1 and TRF2 expression with clinical T stage may be explained at the cellular level by the roles of TRF1 and TRF2 in regulating the growth of cancer cells, while the correlation with N stage may reflect the participation of TRF1 and TRF2 in the control of metastasis. In contrast to our results, other studies have revealed that TRF1 and TRF2 are up-regulated in aggressive adenocarcinoma (2932). This apparent disparity may be the result of differences in the tumors examined and in their microenvironments.

In the present study, decreased expression of TRF1 and TRF2 was detected in OCSCC patients based on Western blotting and immunohistochemistry. TRF1 and TRF2 were strongly expressed in the nucleus of adjacent non-cancerous tissues, and weakly expressed in human OCSCC specimens. Additionally, expression of TRF1 and TRF2 was correlated with 5-year overall survival and clinical prognosis. Notably, TRF1 expression was an independent prognostic indicator for OCSCC in this cohort. These results indicate that TRF1 and TRF2 may be critical regulators of disease progression in OCSCC, making them potential therapeutic targets. Future studies of the physiological targets of TRF1 and TRF2 and their potential roles in the pathogenesis of OCSCC may facilitate the development of novel therapeutic strategies.

Acknowledgements

This study was supported by Chang Gung Memorial Hospital, Taiwan (grant nos. CMRPG870411, CMRPG870412 and CMRPG860513).

References

1. 

Shah JP and Singh B: Why the lack of progress for oral cancer? Lancet Oncol. 7:356–357. 2006. View Article : Google Scholar : PubMed/NCBI

2. 

Chien CY, Su CY, Chuang HC, et al: Angiopoietin-1 and -2 expression in recurrent squamous cell carcinoma of the oral cavity. J Surg Oncol. 97:273–277. 2008. View Article : Google Scholar : PubMed/NCBI

3. 

Chen PH, Ko YC, Yang YH, et al: Important prognostic factors of long-term oropharyngeal carcinoma survivors in Taiwan. Oral Oncol. 40:847–855. 2004. View Article : Google Scholar : PubMed/NCBI

4. 

Matsuo JM, Patel SG, Singh B, et al: Clinical nodal stage is an independently significant predictor of distant failure in patients with squamous cell carcinoma of the larynx. Ann Surg. 238:412–421. 2003.PubMed/NCBI

5. 

Chien CY, Su CY, Chuang HC, et al: Comprehensive study on the prognostic role of osteopontin expression in oral squamous cell carcinoma. Oral Oncol. 45:798–802. 2009. View Article : Google Scholar : PubMed/NCBI

6. 

Chen CH, Chien CY, Huang CC, et al: Expression of FLJ10540 is correlated with aggressiveness of oral cavity squamous cell carcinoma by stimulating cell migration and invasion through increased FOXM1 and MMP-2 activity. Oncogene. 28:2723–2737. 2009. View Article : Google Scholar : PubMed/NCBI

7. 

Vaziri H, Schachter F, Uchida I, et al: Loss of telomeric DNA during aging of normal and trisomy 21 human lymphocytes. Am J Hum Genet. 52:661–667. 1993.PubMed/NCBI

8. 

Van Steensel B and de Lange T: Control of telomere length by the human telomeric protein TRF1. Nature. 385:740–743. 1997.

9. 

Smogorzewska A, van Steensel B, Bianchi A, et al: Control of human telomere length by TRF1 and TRF2. Mol Cell Biol. 20:1659–1668. 2000. View Article : Google Scholar : PubMed/NCBI

10. 

Sfeir A, Kosiyatrakul ST, Hockemeyer D, et al: Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell. 138:90–103. 2009. View Article : Google Scholar : PubMed/NCBI

11. 

Van Steensel B, Smogorzewska A and de Lange T: TRF2 protects human telomeres from end-to-end fusions. Cell. 92:401–413. 1998.PubMed/NCBI

12. 

Ancelin K, Brunori M, Bauwens S, et al: Targeting assay to study the cis functions of human telomeric proteins: evidence for inhibition of telomerase by TRF1 and for activation of telomere degradation by TRF2. Mol Cell Biol. 22:3474–3487. 2002. View Article : Google Scholar : PubMed/NCBI

13. 

Yamada K, Yagihashi A, Yamada M, et al: Decreased gene expression for telomeric-repeat binding factors and TIN2 in malignant hematopoietic cells. Anticancer Res. 22:1315–1320. 2002.PubMed/NCBI

14. 

Yamada M, Tsuji N, Nakamura M, et al: Down-regulation of TRF1, TRF2 and TIN2 genes is important to maintain telomeric DNA for gastric cancers. Anticancer Res. 22:3303–3307. 2002.PubMed/NCBI

15. 

Miyachi K, Fujita M, Tanaka N, Sasaki K and Sunagawa M: Correlation between telomerase activity and telomeric-repeat binding factors in gastric cancer. J Exp Clin Cancer Res. 21:269–275. 2002.PubMed/NCBI

16. 

Lin X, Gu J, Lu C, Spitz MR and Wu X: Expression of telomere-associated genes as prognostic markers for overall survival in patients with non-small cell lung cancer. Clin Cancer Res. 12:5720–5725. 2006. View Article : Google Scholar : PubMed/NCBI

17. 

Raynaud CM, Jang SJ, Nuciforo P, et al: Telomere shortening is correlated with the DNA damage response and telomeric protein down-regulation in colorectal preneoplastic lesions. Ann Oncol. 19:1875–1881. 2008. View Article : Google Scholar : PubMed/NCBI

18. 

Frias C, Garcia-Aranda C, De Juan C, et al: Telomere shortening is associated with poor prognosis and telomerase activity correlates with DNA repair impairment in non-small cell lung cancer. Lung Cancer. 60:416–425. 2008. View Article : Google Scholar : PubMed/NCBI

19. 

d’Adda di Fagagna F, Reaper PM, Clay-Farrace L, et al: A DNA damage checkpoint response in telomere-initiated senescence. Nature. 426:194–198. 2003.PubMed/NCBI

20. 

Yajima T, Yagihashi A, Kameshima H, Kobayashi D, Hirata K and Watanabe N: Telomerase reverse transcriptase and telomeric-repeat binding factor protein 1 as regulators of telomerase activity in pancreatic cancer cells. Br J Cancer. 85:752–757. 2001. View Article : Google Scholar

21. 

Salhab M, Jiang WG, Newbold RF and Mokbel K: The expression of gene transcripts of telomere-associated genes in human breast cancer: correlation with clinico-pathological parameters and clinical outcome. Breast Cancer Res Treat. 109:35–46. 2008. View Article : Google Scholar

22. 

Feldser DM, Hackett JA and Greider CW: Telomere dysfunction and the initiation of genome instability. Nat Rev Cancer. 3:623–627. 2003. View Article : Google Scholar : PubMed/NCBI

23. 

Chong L, van Steensel B, Broccoli D, et al: A human telomeric protein. Science. 270:1663–1667. 1995. View Article : Google Scholar : PubMed/NCBI

24. 

Broccoli D, Smogorzewska A, Chong L and de Lange T: Human telomeres contain two distinct Myb-related proteins, TRF1 and TRF2. Nat Genet. 17:231–235. 1997. View Article : Google Scholar : PubMed/NCBI

25. 

Saito K, Yagihashi A, Nasu S, Izawa Y, et al: Gene expression for suppressors of telomerase activity (telomeric-repeat binding factors) in breast cancer. Jpn J Cancer Res. 93:253–258. 2002. View Article : Google Scholar : PubMed/NCBI

26. 

Yamada K, Yajima T, Yagihashi A, et al: Role of human telomerase reverse transcriptase and telomeric-repeat binding factor proteins 1 and 2 in human hematopoietic cells. Jpn J Cancer Res. 91:1278–1284. 2000. View Article : Google Scholar : PubMed/NCBI

27. 

La Torre D, de Divitiis O, Conti A, et al: Expression of telomeric repeat binding factor-1 in astroglial brain tumors. Neurosurgery. 56:802–810. 2005.PubMed/NCBI

28. 

Kishi S, Wulf G, Nakamura M and Lu KP: Telomeric protein Pin2/TRF1 induces mitotic entry and apoptosis in cells with short telomeres and is down-regulated in human breast tumors. Oncogene. 20:1497–1508. 2001. View Article : Google Scholar : PubMed/NCBI

29. 

Oh BK, Kim YJ, Park C and Park YN: Up-regulation of telomere-binding proteins, TRF1, TRF2, and TIN2 is related to telomere shortening during human multistep hepatocarcinogenesis. Am J Pathol. 166:73–80. 2005. View Article : Google Scholar : PubMed/NCBI

30. 

Klapper W, Qian W, Schulte C and Parwaresch R: DNA damage transiently increases TRF2 mRNA expression and telomerase activity. Leukemia. 17:2007–2015. 2003. View Article : Google Scholar : PubMed/NCBI

31. 

Klapper W, Krams M, Qian W, Janssen D and Parwaresch R: Telomerase activity in B-cell non-Hodgkin lymphomas is regulated by hTERT transcription and correlated with telomere-binding protein expression but uncoupled from proliferation. Br J Cancer. 89:713–719. 2003. View Article : Google Scholar

32. 

Nakanishi K, Kawai T, Kumaki F, et al: Expression of mRNAs for telomeric repeat binding factor (TRF)-1 and TRF2 in atypical adenomatous hyperplasia and adenocarcinoma of the lung. Clin Cancer Res. 9:1105–1111. 2003.PubMed/NCBI

Related Articles

Journal Cover

January-February 2011
Volume 2 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chuang H, Chen C, Huang C, Fang F, Tsai H and Chien C: Reduced expression of TRF1 is associated with tumor progression and poor prognosis in oral squamous cell carcinoma. Exp Ther Med 2: 63-67, 2011
APA
Chuang, H., Chen, C., Huang, C., Fang, F., Tsai, H., & Chien, C. (2011). Reduced expression of TRF1 is associated with tumor progression and poor prognosis in oral squamous cell carcinoma. Experimental and Therapeutic Medicine, 2, 63-67. https://doi.org/10.3892/etm.2010.168
MLA
Chuang, H., Chen, C., Huang, C., Fang, F., Tsai, H., Chien, C."Reduced expression of TRF1 is associated with tumor progression and poor prognosis in oral squamous cell carcinoma". Experimental and Therapeutic Medicine 2.1 (2011): 63-67.
Chicago
Chuang, H., Chen, C., Huang, C., Fang, F., Tsai, H., Chien, C."Reduced expression of TRF1 is associated with tumor progression and poor prognosis in oral squamous cell carcinoma". Experimental and Therapeutic Medicine 2, no. 1 (2011): 63-67. https://doi.org/10.3892/etm.2010.168