Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Decelerated DNA methylation age predicts poor prognosis of breast cancer

Authors: Jun-Ting Ren, Mei-Xia Wang, Yi Su, Lu-Ying Tang, Ze-Fang Ren

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

DNA methylation (DNAm) age was found to be an indicator for all-cause mortality, cancer incidence, and longevity, but no study has involved in the associations of DNAm age with the prognosis of breast cancer.

Methods

We retrieved information of 1076 breast cancer patients from Genomic Data Commons (GDC) data portal on March 30, 2017, including breast cancer DNAm profiling, demographic features, clinicopathological parameters, recurrence, and all-cause fatality. Horvath’s method was applied to calculate the DNAm age. Cox proportional hazards regression models were used to test the associations between DNAm age of the cancerous tissues and the prognosis (recurrence of breast cancer and all-cause fatality) with or without adjusting for chronological age and clinicopathological parameters.

Results

The DNAm age was markedly decelerated in the patients who were premenopausal, ER or PR negative, HER2-enriched or basal-like than their counterparts. In the first five-year follow-up dataset for survival, every ten-year increase in DNAm age was associated with a 15% decrease in fatality; subjects with DNAm age in the second (HR: 0.52; 95%CI: 0.29–0.92), the third (HR: 0.49; 95%CI: 0.27–0.87) and the fourth quartile (HR: 0.38; 95%CI: 0.20–0.72) had significant longer survival time than those in the first quartile. In the first five-year follow-up dataset for recurrence, every ten-year increase in DNAm age was associated with a 14% decrease of the recurrence; in the categorical analysis, a clear dose-response was shown (P for trend =0.02) and the fourth quartile was associated with a longer recurrence free survival (HR: 0.32; 95%CI: 0.14–0.74). In the full follow-up dataset, similar results were obtained.

Conclusions

DNAm age of breast cancer tissue, which associated with menopausal status and pathological features, was a strong independent predictor of the prognosis. It was suggested that the prognosis of breast cancer was related to intrinsic biological changes and specific molecular targets for treatment of breast cancer may be implicit.
Literature
2.
go back to reference Latorre E, Harries LW. Splicing regulatory factors, ageing and age-related disease. Ageing Res Rev. 2017;36:165–70.CrossRef Latorre E, Harries LW. Splicing regulatory factors, ageing and age-related disease. Ageing Res Rev. 2017;36:165–70.CrossRef
3.
go back to reference Teschendorff AE, West J, Beck S. Age-associated epigenetic drift: implications, and a case of epigenetic thrift? Hum Mol Genet. 2013;22(R1):R7–R15.CrossRef Teschendorff AE, West J, Beck S. Age-associated epigenetic drift: implications, and a case of epigenetic thrift? Hum Mol Genet. 2013;22(R1):R7–R15.CrossRef
4.
go back to reference Genovese G, Kahler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, Chambert K, Mick E, Neale BM, Fromer M, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–87.CrossRef Genovese G, Kahler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, Chambert K, Mick E, Neale BM, Fromer M, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med. 2014;371(26):2477–87.CrossRef
5.
go back to reference Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan JB, Gao Y, et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013;49(2):359–67.CrossRef Hannum G, Guinney J, Zhao L, Zhang L, Hughes G, Sadda S, Klotzle B, Bibikova M, Fan JB, Gao Y, et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol Cell. 2013;49(2):359–67.CrossRef
6.
go back to reference Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(10):R115.CrossRef Horvath S. DNA methylation age of human tissues and cell types. Genome Biol. 2013;14(10):R115.CrossRef
7.
go back to reference Marioni RE, Shah S, McRae AF, Chen BH, Colicino E, Harris SE, Gibson J, Henders AK, Redmond P, Cox SR, et al. DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol. 2015;16:25.CrossRef Marioni RE, Shah S, McRae AF, Chen BH, Colicino E, Harris SE, Gibson J, Henders AK, Redmond P, Cox SR, et al. DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol. 2015;16:25.CrossRef
8.
go back to reference Zhang Y, Wilson R, Heiss J, Breitling LP, Saum KU, Schottker B, Holleczek B, Waldenberger M, Peters A, Brenner H. DNA methylation signatures in peripheral blood strongly predict all-cause mortality. Nat Commun. 2017;8:14617.CrossRef Zhang Y, Wilson R, Heiss J, Breitling LP, Saum KU, Schottker B, Holleczek B, Waldenberger M, Peters A, Brenner H. DNA methylation signatures in peripheral blood strongly predict all-cause mortality. Nat Commun. 2017;8:14617.CrossRef
9.
go back to reference Christiansen L, Lenart A, Tan Q, Vaupel JW, Aviv A, McGue M, Christensen K. DNA methylation age is associated with mortality in a longitudinal Danish twin study. Aging Cell. 2016;15(1):149–54.CrossRef Christiansen L, Lenart A, Tan Q, Vaupel JW, Aviv A, McGue M, Christensen K. DNA methylation age is associated with mortality in a longitudinal Danish twin study. Aging Cell. 2016;15(1):149–54.CrossRef
10.
go back to reference Lin Q, Weidner CI, Costa IG, Marioni RE, Ferreira MR, Deary IJ, Wagner W. DNA methylation levels at individual age-associated CpG sites can be indicative for life expectancy. Aging (Albany NY). 2016;8(2):394–401.CrossRef Lin Q, Weidner CI, Costa IG, Marioni RE, Ferreira MR, Deary IJ, Wagner W. DNA methylation levels at individual age-associated CpG sites can be indicative for life expectancy. Aging (Albany NY). 2016;8(2):394–401.CrossRef
11.
go back to reference Moore AZ, Hernandez DG, Tanaka T, Pilling LC, Nalls MA, Bandinelli S, Singleton AB, Ferrucci L. Change in epigenome-wide DNA methylation over 9 years and subsequent mortality: results from the InCHIANTI study. J Gerontol A Biol Sci Med Sci. 2016;71(8):1029–35.CrossRef Moore AZ, Hernandez DG, Tanaka T, Pilling LC, Nalls MA, Bandinelli S, Singleton AB, Ferrucci L. Change in epigenome-wide DNA methylation over 9 years and subsequent mortality: results from the InCHIANTI study. J Gerontol A Biol Sci Med Sci. 2016;71(8):1029–35.CrossRef
12.
go back to reference Zheng Y, Joyce BT, Colicino E, Liu L, Zhang W, Dai Q, Shrubsole MJ, Kibbe WA, Gao T, Zhang Z, et al. Blood epigenetic age may predict Cancer incidence and mortality. EBioMedicine. 2016;5:68–73.CrossRef Zheng Y, Joyce BT, Colicino E, Liu L, Zhang W, Dai Q, Shrubsole MJ, Kibbe WA, Gao T, Zhang Z, et al. Blood epigenetic age may predict Cancer incidence and mortality. EBioMedicine. 2016;5:68–73.CrossRef
13.
go back to reference Perna L, Zhang Y, Mons U, Holleczek B, Saum KU, Brenner H. Epigenetic age acceleration predicts cancer, cardiovascular, and all-cause mortality in a German case cohort. Clin Epigenetics. 2016;8:64.CrossRef Perna L, Zhang Y, Mons U, Holleczek B, Saum KU, Brenner H. Epigenetic age acceleration predicts cancer, cardiovascular, and all-cause mortality in a German case cohort. Clin Epigenetics. 2016;8:64.CrossRef
14.
go back to reference Yang Z, Wong A, Kuh D, Paul DS, Rakyan VK, Leslie RD, Zheng SC, Widschwendter M, Beck S, Teschendorff AE. Correlation of an epigenetic mitotic clock with cancer risk. Genome Biol. 2016;17(1):205.CrossRef Yang Z, Wong A, Kuh D, Paul DS, Rakyan VK, Leslie RD, Zheng SC, Widschwendter M, Beck S, Teschendorff AE. Correlation of an epigenetic mitotic clock with cancer risk. Genome Biol. 2016;17(1):205.CrossRef
15.
go back to reference Zheng SC, Widschwendter M, Teschendorff AE. Epigenetic drift, epigenetic clocks and cancer risk. Epigenomics. 2016;8(5):705–19.CrossRef Zheng SC, Widschwendter M, Teschendorff AE. Epigenetic drift, epigenetic clocks and cancer risk. Epigenomics. 2016;8(5):705–19.CrossRef
16.
go back to reference Durso DF, Bacalini MG, Sala C, Pirazzini C, Marasco E, Bonafe M, do Valle IF, Gentilini D, Castellani G, AMC F, et al. Acceleration of leukocytes’ epigenetic age as an early tumor and sex-specific marker of breast and colorectal cancer. Oncotarget. 2017;8(14):23237–45.CrossRef Durso DF, Bacalini MG, Sala C, Pirazzini C, Marasco E, Bonafe M, do Valle IF, Gentilini D, Castellani G, AMC F, et al. Acceleration of leukocytes’ epigenetic age as an early tumor and sex-specific marker of breast and colorectal cancer. Oncotarget. 2017;8(14):23237–45.CrossRef
17.
go back to reference Levine ME, Hosgood HD, Chen B, Absher D, Assimes T, Horvath S. DNA methylation age of blood predicts future onset of lung cancer in the women's health initiative. Aging (Albany NY). 2015;7(9):690–700.CrossRef Levine ME, Hosgood HD, Chen B, Absher D, Assimes T, Horvath S. DNA methylation age of blood predicts future onset of lung cancer in the women's health initiative. Aging (Albany NY). 2015;7(9):690–700.CrossRef
18.
go back to reference Lin Q, Wagner W. Epigenetic aging signatures are coherently modified in Cancer. PLoS Genet. 2015;11(6):e1005334.CrossRef Lin Q, Wagner W. Epigenetic aging signatures are coherently modified in Cancer. PLoS Genet. 2015;11(6):e1005334.CrossRef
19.
go back to reference Hassan C, Afshinnekoo E, Li S, Wu S, Mason CE. Genetic and epigenetic heterogeneity and the impact on cancer relapse. Exp Hematol. 2017;54:26–30.CrossRef Hassan C, Afshinnekoo E, Li S, Wu S, Mason CE. Genetic and epigenetic heterogeneity and the impact on cancer relapse. Exp Hematol. 2017;54:26–30.CrossRef
20.
go back to reference Silva TC, Colaprico A, Olsen C, D'Angelo F, Bontempi G, Ceccarelli M, Noushmehr H. TCGA workflow: analyze cancer genomics and epigenomics data using Bioconductor packages. F1000Research. 2016;5:1542.CrossRef Silva TC, Colaprico A, Olsen C, D'Angelo F, Bontempi G, Ceccarelli M, Noushmehr H. TCGA workflow: analyze cancer genomics and epigenomics data using Bioconductor packages. F1000Research. 2016;5:1542.CrossRef
21.
go back to reference Jylhava J, Pedersen NL, Hagg S. Biological age predictors. EBioMedicine. 2017;21:29–36.CrossRef Jylhava J, Pedersen NL, Hagg S. Biological age predictors. EBioMedicine. 2017;21:29–36.CrossRef
22.
go back to reference van Niekerk G, Davids LM, Hattingh SM, Engelbrecht AM. Cancer stem cells: a product of clonal evolution? Int J Cancer. 2017;140(5):993–9.CrossRef van Niekerk G, Davids LM, Hattingh SM, Engelbrecht AM. Cancer stem cells: a product of clonal evolution? Int J Cancer. 2017;140(5):993–9.CrossRef
23.
go back to reference Chen H, Lin F, Xing K, He X. The reverse evolution from multicellularity to unicellularity during carcinogenesis. Nat Commun. 2015;6:6367.CrossRef Chen H, Lin F, Xing K, He X. The reverse evolution from multicellularity to unicellularity during carcinogenesis. Nat Commun. 2015;6:6367.CrossRef
24.
go back to reference Huo D, Hu H, Rhie SK, Gamazon ER, Cherniack AD, Liu J, Yoshimatsu TF, Pitt JJ, Hoadley KA, Troester M, et al. Comparison of breast Cancer molecular features and survival by African and European ancestry in the Cancer genome atlas. JAMA Oncol. 2017;3:1654–62.CrossRef Huo D, Hu H, Rhie SK, Gamazon ER, Cherniack AD, Liu J, Yoshimatsu TF, Pitt JJ, Hoadley KA, Troester M, et al. Comparison of breast Cancer molecular features and survival by African and European ancestry in the Cancer genome atlas. JAMA Oncol. 2017;3:1654–62.CrossRef
25.
go back to reference Ambatipudi S, Horvath S, Perrier F, Cuenin C, Hernandez-Vargas H, Le Calvez-Kelm F, Durand G, Byrnes G, Ferrari P, Bouaoun L, et al. DNA methylome analysis identifies accelerated epigenetic ageing associated with postmenopausal breast cancer susceptibility. Eur J Cancer. 2017;75:299–307.CrossRef Ambatipudi S, Horvath S, Perrier F, Cuenin C, Hernandez-Vargas H, Le Calvez-Kelm F, Durand G, Byrnes G, Ferrari P, Bouaoun L, et al. DNA methylome analysis identifies accelerated epigenetic ageing associated with postmenopausal breast cancer susceptibility. Eur J Cancer. 2017;75:299–307.CrossRef
26.
go back to reference Sehl ME, Henry JE, Storniolo AM, Ganz PA, Horvath S. DNA methylation age is elevated in breast tissue of healthy women. Breast Cancer Res Treat. 2017;164(1):209–19.CrossRef Sehl ME, Henry JE, Storniolo AM, Ganz PA, Horvath S. DNA methylation age is elevated in breast tissue of healthy women. Breast Cancer Res Treat. 2017;164(1):209–19.CrossRef
27.
go back to reference Benz CC. Impact of aging on the biology of breast cancer. Crit Rev Oncol Hematol. 2008;66(1):65–74.CrossRef Benz CC. Impact of aging on the biology of breast cancer. Crit Rev Oncol Hematol. 2008;66(1):65–74.CrossRef
28.
go back to reference Brandt J, Garne JP, Tengrup I, Manjer J. Age at diagnosis in relation to survival following breast cancer: a cohort study. World J Surg Oncol. 2015;13:33.CrossRef Brandt J, Garne JP, Tengrup I, Manjer J. Age at diagnosis in relation to survival following breast cancer: a cohort study. World J Surg Oncol. 2015;13:33.CrossRef
29.
go back to reference Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–217.CrossRef Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–217.CrossRef
Metadata
Title
Decelerated DNA methylation age predicts poor prognosis of breast cancer
Authors
Jun-Ting Ren
Mei-Xia Wang
Yi Su
Lu-Ying Tang
Ze-Fang Ren
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4884-6

Other articles of this Issue 1/2018

BMC Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine