Telomere shortening in breast cancer correlates with the pathological features of tumor progression

  • Authors:
    • Makoto Kammori
    • Yoshiyuki Sugishita
    • Takahiro Okamoto
    • Makio Kobayashi
    • Kazuko Yamazaki
    • Emiko Yamada
    • Tetsu Yamada
  • View Affiliations

  • Published online on: June 15, 2015     https://doi.org/10.3892/or.2015.4063
  • Pages: 627-632
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Telomeres are involved in the maintenance of genomic stability. Telomere alteration has been observed in most human cancer types, and is known to be a feature of malignancy. The aim of the present study was to evaluate whether the telomere length of breast cancer cells correlates with TNM stage and several pathological features. We investigated a total of 44 breast cancers, including 17 scirrhous, 15 papillotubular and 12 solid-tubular carcinomas. Telomere lengths were determined by tissue quantitative fluorescence in situ hybridization (Q-FISH), and compared according to the TNM stage, histological tumor size, lymph node metastases, vascular invasion and immunohistochemical status (ER, PR, HER2 status and Ki67 labeling index). In all histological types, telomeres of cancer cells were significantly shorter than those of normal epithelial cells. Mean telomere length was significantly less in patients with TNM stage III, and in those with large tumors, lymph node metastases and vascular invasion. Our results suggest that the telomere length of cancer cells is strongly correlated with the degree of cancer progression.

Introduction

Telomeres are protective structures that cap the ends of eukaryotic chromosomes (1), comprising multiple 5′-TTAGGG-3′ repeats and ending in a single-stranded overhang of the G-rich sequence (2). Telomeres protect chromosome ends from end-to-end fusion, nucleolytic decay, degradation and atypical recombination (3). The telomere repeat sequence becomes shortened by each cell division, DNA damage due to oxidative stress or through changes in telomere-associated proteins (4,5). It has been proposed that telomere shortening is an important biological factor involved in carcinogenesis, cell senescence, cell replication, cell immortality and aging (68). Accumulating evidence has led to the hypothesis that telo-mere dysfunction contributes to genetic changes intrinsic to the development and progression of tumors (911), and that telomere length can be considered as a biological index of malignant potential (12). It has been reported that telomere shortening occurs in breast cancer cells (13) and contributes to tumor progression in numerous cancer types including breast cancer (14,15).

Breast cancer is the most frequent malignant disease and the leading cause of cancer-related death among women worldwide. Globally, 1.4 million new cases of breast cancer are diagnosed annually of which approximately one third are fatal (16). It is well established that telomere shortening is present in the majority of in situ and invasive carcinomas (14) including breast cancers (17,18). In addition to their role in tumor initiation, short dysfunctional telomeres affect disease progression. Previous studies have shown that telomeres are shorter in grade III tumors (19), that telomere shortening is correlated with aneuploidy and lymph node metastasis (20), and that shorter telomeres are associated with higher stage and histological grade (21). Moreover, it has been reported that short telomeres are associated with tumor size, nodal involvement and TNM stage (22). However, another study found no correlation between telomere length and tumor volume, grading, node status or expression of estrogen receptor (ER) and progesterone receptor (PR) (23).

The aim of the present study was to clarify whether the telomeres in three histological types of breast cancer (scirrhous, papillotubular and solid-tubular carcinomas) are shorter than those of normal epithelial cells, and whether telomere length is correlated with TNM stage and several pathologic factors. For this purpose, we measured telomere lengths in breast cancer using quantitative fluorescence in situ hybridization (Q-FISH), which allowed us to estimate the telomere lengths of individual cells in each section.

Materials and methods

Tissue specimens

We examined a total of 44 breast cancers, including 17 scirrhous, 15 papillotubular and 12 solid-tubular carcinomas. Both tumor and adjacent normal tissues were obtained from each individual patient and embedded in paraffin using standard processing procedures. Sections 4-µm thick were prepared for tissue Q-FISH and immunohistochemistry (IHC) analysis. All samples were collected at the Division of Surgical Endocrinology of Tokyo Women’s Medical University, Tokyo, Japan, after obtaining informed consent from all of the patients. Table I summarizes the clinicopathological results. All pathologic examinations were conducted by one of the authors who was a trained pathologist (M.K.).

Table I

Clinicopathological characteristics of the study patients.

Table I

Clinicopathological characteristics of the study patients.

Histological typeScirrhous carcinoma (n=17)Papillotubular carcinoma (n=15)Solid-tubular carcinoma (n=12)
Clinical characteristics
 Age (years)a55.5±11.052.0±17.056.1±14.3
 TNM stage
  0 (DCIS)110
  I556
  II985
  III211
Pathological characteristics
 Tumor size (mm)a22.2±12.023.3±15.921.4±12.1
 Lymph nodemetastasesb
  pN09139
  pN1313
  pN3200
 Vascular invasionb
  +1087
  −674
 ER statusb
  +14116
  −136
 PR statusb
  +1495
  −157
 HER2 statusb
  0–111128
  2322
  3112
 Ki67 LI (%)a6.8±11.613.0±14.411.4±9.7

a Data are expressed as mean ± standard deviation.

b Including unknown cases. LI, labeling index.

Tissue Q-FISH

Tissue Q-FISH was performed as previously described (2426). In brief, tissue sections were deparaffinized and treated with 0.2 N HCl and 1 M sodium thiocyanate at 80°C, 1% pepsin at 37°C and 10 mg/ml RNase at 37°C. A peptide nucleic acid (PNA) telomere probe conjugated to Cy3 (telo C Cy3 probe, 5′-CCCTAACCCTAACCCTAA-3′); and a PNA centromere probe conjugated to fluorescein isothiocyanate (FITC) (Cenp 1 probe, 5′-CTTCGTTGGAAACGGGGT-3′) (both from Fasmac, Kanagawa, Japan) were applied to each section. The nuclei were stained with 4′,6-diamidino-2-phenyl-indole (DAPI) (Sigma-Aldrich, St, Louis, MO, USA).

FISH images were captured by a CCD camera attached to an epifluorescence microscope (Eclipse 90i; Nikon, Tokyo, Japan) equipped with a triple band-pass filter set for DAPI/FITC/Cy3 (61000v2m; Chroma Technology Corp., Rockingham, VT, USA) and a ×40 objective lens (Plan Fluor ×40/0.75; Nikon). Microscope control and image recording were performed using Image-Pro Plus software (version 6.3; Media Cybernetics, Bethesda, MD, USA). The recorded images were analyzed as previously described using original software prepared by our colleague (S.P.) (2426), ‘Tissue Telo Version 3.2′, which allows manual identification of nuclear regions from the composite color image: DAPI (blue channel), FITC (green) and Cy3 (red). Fluorescence intensities of telomere (Cy3) and centromere signals (FITC) for each nucleus were measured, and then the telomere-centromere ratio (TCR) was calculated, since there is no guarantee that all information on telomere signals will be acquired within any given tissue section.

IHC

The expression of ER, PR, human epidermal growth factor receptor 2 (HER2) and Ki67 (Dako Japan, Tokyo, Japan) protein was determined by IHC staining. For ER, PR and Ki67, the tissue sections were pretreated with Tris-EDTA buffer solution (pH 9.0) at 95°C, and for HER2 with citrate buffer solution (pH 6.0) at 95°C. After incubation with the primary antibody for 2 h, visualization was performed using a polymer IHC detection system (EnVision kit; Dako Japan). The Ki67 labeling index (LI) was calculated by counting at least 500 cells in each specimen.

Statistical analysis

The significance of differences in mean TCR was examined by Welch’s t-test. When three groups were compared, we used one-way ANOVA and the Tukey-Kramer post hoc test. For analyzing correlations between continuous variables (pathological tumor size and Ki67 LI) and TCR, Pearson’s correlation coefficient was calculated. Differences at P<0.05 were considered to indicate a statistically significant result.

Results

Telomere length (TCR distribution) in cancer and adjacent normal breast tissues

Telomere signals were evident within nuclei as small red spots in normal epithelial cells (Fig. 1a-1). Telomere signals of tumor cells in scirrhous carcinomas (Fig. 1b-1 and b-2) were weaker than those in adjacent n ormal epithelial cells (Fig. 1a-1 and a-2). Cancer cells most frequently had very short telomeres, yet a few cells had long telomeres such as TCR >4, and the peak frequency of TCR was in <1 (Fig. 2a-2, b-2 and c-2). On the other hand, normal epithelial cells had relatively long telomeres (TCR >4) and the TCR had a very wide distribution (Fig. 2a-1, b-1 and c-1). The mean TCR of cancer cells was significantly lower than that of normal epithelial cells in all histological types (Fig. 2, P<0.05).

Relationship between telomere length and several pathological and immunohistochemical factors

In the present study, associations between mean TCR and clinical TNM stage, histological tumor size, pathologically proven lymph node metastasis, vascular invasion, ER, PR, HER2 status and Ki67 LI were analyzed (Fig. 3). Tumor size was divided into three groups (≤20, 20–50 and >50 mm) in accordance with the UICC criteria. For Ki67 LI, though cut-off values have varied among previous studies (2730), a nuclear LI of ≥20% was considered high and one of <20% was considered low for the purposes of the present study. Telomere shortening was associated with TNM stage III, a large tumor size, presence of a large number of lymph node metastases, presence of vascular invasion, ER positivity and PR positivity (Fig. 3). However, HER2 status did not correlate with telomere length (Fig. 3). Telomeres lengths of Ki67 LI ≥20% patients were shorter than those of LI <20% patients (Fig. 3). However, there was no significant correlation between Ki67 LI and mean TCR when we analyzed the Pearson’s correlation coefficient (P=0.806, Fig. 4).

We analyzed the correlation between histological tumor size and mean TCR by calculation of Pearson’s correlation coefficient. Although correlation coefficient was not so strong (R2=0.102), statistically significant inverse correlation was observed (P=0.037, Fig. 4).

Discussion

Generally, the cells of malignant tumors including breast cancer have shorter telomeres than the corresponding cells in normal tissue (13,21), and telomere dysfunction or shortening has been considered as a negative prognostic indicator in patients with solid tumors (31,32), including breast cancer (33,34). Furthermore, it has been reported that telomere shortening contributes to tumor progression (14,15). In the present study, using tissue Q-FISH technique, we evaluated telomere length in breast cancer tissues and also confirmed that cancer cells had shorter telomeres than the adjacent normal epithelial cells in three histological types of carcinoma (scirrhous, papillotubular and solid-tubular carcinomas). Tissue Q-FISH effectively estimates telomere length in different cell types using separate PNA probes for telomeres and centromeres, thus allowing specific evaluation for cancer cells. We obtained the telomere-centromere ratio (TCR) as a parameter representative of telomere length, and many previous studies have verified its accuracy for this purpose (2426). Moreover, we evaluated whether telomere length was correlated with several pathological features, indicating tumor progression.

The principal conclusion emerging from the present study was that telomere shortening is associated with some parameters of cancer progression. Mean telomere length was significantly less in patients with TNM stage III disease, a large tumor size, a large number of lymph node metastases and vascular invasion. As previously described, several studies of breast cancer have revealed that telomere length is associated with TNM stage, tumor size, nodal involvement and prognosis (21,22,35). On the other hand, some studies have found no correlation between telomere length and tumor volume, grading, nodal or ER and PR status (20,23). Our present findings are in agreement with the results of some of these studies (21,22,35), and suggest that telomere length may be a useful index of tumor aggressiveness in breast cancer. Although tumor size has been the traditional prognostic factor, it has been regarded as one of the most powerful predictors of tumor behavior in breast cancer (3638). Since the present study demonstrated an inverse relationship between mean TCR and histologically evident tumor size, our results reinforce the assumption that telomere length reflects the prognosis.

Recently, breast cancer has been classified into different molecular subtypes with different biological features, clinical outcome and response to therapy (3942). These subtypes can be distinguished on the basis of ER, PR and HER2 status: luminal A (ER+ and/or PR+, HER2), luminal B (ER+ and/or PR+, HER2+), HER2 (ER and PR, HER2+) and basal-like (ER, PR, HER2). Various studies have indicated the importance of using the proliferation index (Ki67 LI) to distinguish between the luminal A and B subtypes (27,43). In addition, telomere shortening is reportedly associated with specific breast cancer subtypes; some studies have indicated that ER- and/or PR-negative cancers have shorter telomeres than ER- and/or PR-positive cases (44,45). In the present study, both ER- and PR-positive tumors had shorter telomeres than ER- and PR-negative tumors, respectively, being contradictory to results obtained in previous studies. This may have been due to the fact that both the ER- and PR-positive groups in the present study included pN3 cases with very short telomeres. However, due to the limited number of patients with ER- and/or PR-negative tumors, we considered that these results should be viewed with caution, and that no clear conclusion can yet to be drawn. Furthermore, telomere length did not differ significantly according to the HER2 status, and no correlation between Ki67 LI and telomere length was evident. Accordingly, the data obtained in the present study were considered insufficient for discussing their relationship with molecular subtypes such as the luminal classification. However, as previously described, it is thought that telomere metabolism is a very important factor impacting on tumor behavior, and that tissue Q-FISH provides an accurate estimation of telomere length. We considered that further studies will be needed to evaluate telomere length using the tissue Q-FISH method in relation to luminal molecular subtypes.

In summary, we have demonstrated a significant correlation between telomere length and TNM stage, tumor size and lymph node metastasis in breast cancer. Our present results suggest that the telomere length of cancer cells is strongly correlated with cancer progression.

Acknowledgments

This study was partly supported by a Grant-in-Aid for Scientific Research from the Japanese Ministry of Education, Culture, Sports, Science and Technology (#20591555). We would like to thank Dr Steven S.S. Poon (Terry Fox Laboratory, British Columbia Cancer Research Center) for preparation of the software for measurement of telomere length. We would also like to thank Miyoko Matsumoto (Kanaji Thyroid Hospital) for assistance with the manuscript preparation.

References

1 

Deng Y, Chan SS and Chang S: Telomere dysfunction and tumour suppression: The senescence connection. Nat Rev Cancer. 8:450–458. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Palm W and de Lange T: How shelterin protects mammalian telomeres. Annu Rev Genet. 42:301–334. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Stewart SA and Weinberg RA: Telomeres: Cancer to human aging. Annu Rev Cell Dev Biol. 22:531–557. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Harley CB, Futcher AB and Greider CW: Telomeres shorten during ageing of human fibroblasts. Nature. 345:458–460. 1990. View Article : Google Scholar : PubMed/NCBI

5 

von Zglinicki T: Oxidative stress shortens telomeres. Trends Biochem Sci. 27:339–344. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Harley CB and Villeponteau B: Telomeres and telomerase in aging and cancer. Curr Opin Genet Dev. 5:249–255. 1995. View Article : Google Scholar : PubMed/NCBI

7 

de Lange T: Telomeres and senescence: Ending the debate. Science. 279:334–335. 1998. View Article : Google Scholar : PubMed/NCBI

8 

DePinho RA: The age of cancer. Nature. 408:248–254. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Artandi SE and DePinho RA: Telomeres and telomerase in cancer. Carcinogenesis. 31:9–18. 2010. View Article : Google Scholar :

10 

Blasco MA: Telomeres and human disease: Ageing, cancer and beyond. Nat Rev Genet. 6:611–622. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Prescott J, Wentzensen IM, Savage SA and De Vivo I: Epidemiologic evidence for a role of telomere dysfunction in cancer etiology. Mutat Res. 730:75–84. 2012. View Article : Google Scholar

12 

Svenson U and Roos G: Telomere length as a biological marker in malignancy. Biochim Biophys Acta. 1792:317–323. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Kurabayashi R, Takubo K, Aida J, Honma N, Poon SS, Kammori M, Izumiyama-Shimomura N, Nakamura K, Tsuji E, Matsuura M, et al: Luminal and cancer cells in the breast show more rapid telomere shortening than myoepithelial cells and fibroblasts. Hum Pathol. 39:1647–1655. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Meeker AK, Hicks JL, Iacobuzio-Donahue CA, Montgomery EA, Westra WH, Chan TY, Ronnett BM and De Marzo AM: Telomere length abnormalities occur early in the initiation of epithelial carcinogenesis. Clin Cancer Res. 10:3317–3326. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Meeker AK and Argani P: Telomere shortening occurs early during breast tumorigenesis: A cause of chromosome destabilization underlying malignant transformation? J Mammary Gland Biol Neoplasia. 9:285–296. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Meeker AK, Hicks JL, Gabrielson E, Strauss WM, De Marzo AM and Argani P: Telomere shortening occurs in subsets of normal breast epithelium as well as in situ and invasive carcinoma. Am J Pathol. 164:925–935. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Chin K, de Solorzano CO, Knowles D, Jones A, Chou W, Rodriguez EG, Kuo WL, Ljung BM, Chew K, Myambo K, et al: In situ analyses of genome instability in breast cancer. Nat Genet. 36:984–988. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Odagiri E, Kanada N, Jibiki K, Demura R, Aikawa E and Demura H: Reduction of telomeric length and c-erbB-2 gene amplification in human breast cancer, fibroadenoma, and gynecomastia. Relationship to histologic grade and clinical parameters. Cancer. 73:2978–2984. 1994. View Article : Google Scholar : PubMed/NCBI

20 

Griffith JK, Bryant JE, Fordyce CA, Gilliland FD, Joste NE and Moyzis RK: Reduced telomere DNA content is correlated with genomic instability and metastasis in invasive human breast carcinoma. Breast Cancer Res Treat. 54:59–64. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Radpour R, Barekati Z, Haghighi MM, Kohler C, Asadollahi R, Torbati PM, Holzgreve W and Zhong XY: Correlation of telomere length shortening with promoter methylation profile of p16/Rb and p53/p21 pathways in breast cancer. Mod Pathol. 23:763–772. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Fordyce CA, Heaphy CM, Bisoffi M, Wyaco JL, Joste NE, Mangalik A, Baumgartner KB, Baumgartner RN, Hunt WC and Griffith JK: Telomere content correlates with stage and prognosis in breast cancer. Breast Cancer Res Treat. 99:193–202. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Rogalla P, Rohen C, Bonk U and Bullerdiek J: Telomeric repeat fragment lengths are not correlated to histological grading in 85 breast cancers. Cancer Lett. 106:155–161. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Sugishita Y, Kammori M, Yamada O, Yamazaki K, Ito K, Fukumori T, Yoshikawa K and Yamada T: Biological differential diagnosis of follicular thyroid tumor and Hürthle cell tumor on the basis of telomere length and hTERT expression. Ann Surg Oncol. 21:2318–2325. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Aida J, Izumiyama-Shimomura N, Nakamura K, Ishii A, Ishikawa N, Honma N, Kurabayashi R, Kammori M, Poon SS, Arai T, et al: Telomere length variations in 6 mucosal cell types of gastric tissue observed using a novel quantitative fluorescence in situ hybridization method. Hum Pathol. 38:1192–1200. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Kammori M, Izumiyama N, Nakamura K, Kurabayashi R, Kashio M, Aida J, Poon SS and Kaminishi M: Telomere metabolism and diagnostic demonstration of telomere measurement in the human esophagus for distinguishing benign from malignant tissue by tissue quantitative fluorescence in situ hybridization. Oncology. 71:430–436. 2006. View Article : Google Scholar

27 

Ahlin C, Aaltonen K, Amini RM, Nevanlinna H, Fjällskog ML and Blomqvist C: Ki67 and cyclin A as prognostic factors in early breast cancer. What are the optimal cut-off values? Histopathology. 51:491–498. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J, Watson M, Davies S, Bernard PS, Parker JS, et al: Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst. 101:736–750. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thürlimann B and Senn HJ; Panel members: Strategies for subtypes - dealing with the diversity of breast cancer: Highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol. 22:1736–1747. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Knutsvik G, Stefansson IM, Aziz S, Arnes J, Eide J, Collett K and Akslen LA: Evaluation of Ki67 expression across distinct categories of breast cancer specimens: A population-based study of matched surgical specimens, core needle biopsies and tissue microarrays. PLoS One. 9:e1121212014. View Article : Google Scholar : PubMed/NCBI

31 

Donaldson L, Fordyce C, Gilliland F, Smith A, Feddersen R, Joste N, Moyzis R and Griffith J: Association between outcome and telomere DNA content in prostate cancer. J Urol. 162:1788–1792. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Frías C, García-Aranda C, De Juan C, Morán A, Ortega P, Gómez A, Hernando F, López-Asenjo JA, Torres AJ, Benito M, 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

33 

Bisoffi M, Heaphy CM and Griffith JK: Telomeres: Prognostic markers for solid tumors. Int J Cancer. 119:2255–2260. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Heaphy CM, Baumgartner KB, Bisoffi M, Baumgartner RN and Griffith JK: Telomere DNA content predicts breast cancer-free survival interval. Clin Cancer Res. 13:7037–7043. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Diehl MC, Idowu MO, Kimmelshue KN, York TP, Jackson-Cook CK, Turner KC, Holt SE and Elmore LW: Elevated TRF2 in advanced breast cancers with short telomeres. Breast Cancer Res Treat. 127:623–630. 2011. View Article : Google Scholar

36 

Carter CL, Allen C and Henson DE: Relation of tumor size, lymph node status, and survival in 24,740 breast cancer cases. Cancer. 63:181–187. 1989. View Article : Google Scholar : PubMed/NCBI

37 

Leitner SP, Swern AS, Weinberger D, Duncan LJ and Hutter RV: Predictors of recurrence for patients with small (one centimeter or less) localized breast cancer (T1a,b N0 M0). Cancer. 76:2266–2274. 1995. View Article : Google Scholar : PubMed/NCBI

38 

Gebauer G, Fehm T, Lang N and Jäger W: Tumor size, axillary lymph node status and steroid receptor expression in breast cancer: Prognostic relevance 5 years after surgery. Breast Cancer Res Treat. 75:167–173. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Sørlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M, Jeffrey SS, et al: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 98:10869–10874. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R, Geisler S, et al: Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc Natl Acad Sci USA. 100:8418–8423. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Sotiriou C and Pusztai L: Gene-expression signatures in breast cancer. N Engl J Med. 360:790–800. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Voduc KD, Cheang MC, Tyldesley S, Gelmon K, Nielsen TO and Kennecke H: Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol. 28:1684–1691. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Heaphy CM, Subhawong AP, Gross AL, Konishi Y, Kouprina N, Argani P, Visvanathan K and Meeker AK: Shorter telomeres in luminal B, HER-2 and triple-negative breast cancer subtypes. Mod Pathol. 24:194–200. 2011. View Article : Google Scholar

45 

Martinez-Delgado B, Gallardo M, Tanic M, Yanowsky K, Inglada-Perez L, Barroso A, Rodriguez-Pinilla M, Cañamero M, Blasco MA and Benitez J: Short telomeres are frequent in hereditary breast tumors and are associated with high tumor grade. Breast Cancer Res Treat. 141:231–242. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2015
Volume 34 Issue 2

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
Kammori M, Sugishita Y, Okamoto T, Kobayashi M, Yamazaki K, Yamada E and Yamada T: Telomere shortening in breast cancer correlates with the pathological features of tumor progression. Oncol Rep 34: 627-632, 2015
APA
Kammori, M., Sugishita, Y., Okamoto, T., Kobayashi, M., Yamazaki, K., Yamada, E., & Yamada, T. (2015). Telomere shortening in breast cancer correlates with the pathological features of tumor progression. Oncology Reports, 34, 627-632. https://doi.org/10.3892/or.2015.4063
MLA
Kammori, M., Sugishita, Y., Okamoto, T., Kobayashi, M., Yamazaki, K., Yamada, E., Yamada, T."Telomere shortening in breast cancer correlates with the pathological features of tumor progression". Oncology Reports 34.2 (2015): 627-632.
Chicago
Kammori, M., Sugishita, Y., Okamoto, T., Kobayashi, M., Yamazaki, K., Yamada, E., Yamada, T."Telomere shortening in breast cancer correlates with the pathological features of tumor progression". Oncology Reports 34, no. 2 (2015): 627-632. https://doi.org/10.3892/or.2015.4063