PDF
Abstract
Background: The intricate relationship between cellular ageing processes and cancer development represents one of the most significant challenges in contemporary oncology. As populations worldwide experience unprecedented demographic shifts towards advanced age, understanding the molecular mechanisms that link ageing to cancer initiation, progression, and therapeutic response has become essential for developing effective precision medicine approaches.
Main body: This review examines the fundamental molecular pathways through which ageing influences cancer biology, including telomere dysfunction, cellular senescence, DNA damage accumulation, and epigenetic alterations. These age-related changes create a permissive environment for oncogenesis while simultaneously affecting therapeutic efficacy and treatment tolerance. Key ageing-associated molecular signatures include p16^INK4a^ upregulation, shortened telomeres, increased DNA damage response activation, and altered chromatin structure. The accumulation of senescent cells with age contributes to chronic inflammation and tissue dysfunction that promotes tumour development. Additionally, age-related changes in drug metabolism, DNA repair capacity, and immune function significantly impact therapeutic outcomes. Recent advances in molecular ageing biomarkers, including transcriptomic ageing clocks and protein-based signatures, offer promising approaches for personalizing cancer treatment strategies. The integration of ageing biology into precision oncology frameworks presents opportunities for developing age-informed therapeutic protocols that optimize efficacy while minimizing toxicity. Emerging technologies, including artificial intelligence-driven molecular analysis and advanced imaging techniques, enable more precise characterization of ageing-cancer interactions at the cellular and tissue levels.
Conclusion: The molecular mechanisms underlying ageing-cancer relationships provide critical insights for advancing precision oncology approaches. Understanding these pathways enables the development of targeted interventions that account for age-related biological changes, ultimately improving therapeutic outcomes for older cancer patients. Future research must focus on translating molecular ageing discoveries into clinically actionable tools that enhance treatment personalization and optimize care delivery across the cancer continuum.
Keywords
ageing mechanisms
/
biomarkers
/
cancer biology
/
cellular ageing
/
DNA damage
/
epigenetic alterations
/
molecular senescence
/
precision oncology
/
therapeutic response
/
translational medicine
Cite this article
Download citation ▾
Laiba Husain.
Molecular mechanisms of ageing in cancer development and therapeutic response: Translational implications for precision oncology.
Clinical and Translational Discovery, 2025, 5(3): e70065 DOI:10.1002/ctd2.70065
| [1] |
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023; 186(2): 243-278.
|
| [2] |
Swanton C, Bernard E, Abbosh C, et al. Embracing cancer complexity: hallmarks of systemic disease. Cell. 2024; 187(7): 1589-1616.
|
| [3] |
Li Y, Liang M, Ji X, et al. Molecular mechanisms of aging and anti-aging strategies. Cell Commun Signal. 2024; 22(1): 285.
|
| [4] |
Zhavoronkov A, Mamoshina P, Vanhaelen Q, Scheibye-Knudsen M, Moskalev A, Aliper A. Artificial intelligence for aging and longevity research: recent advances and perspectives. Ageing Res Rev. 2019; 49: 49-66.
|
| [5] |
Lyu YX, Zhavoronkov A, Scheibye-Knudsen M, Bakula D. Longevity biotechnology: bridging AI, biomarkers, geroscience and clinical applications for healthy longevity. Aging (Albany NY). 2024; 16(20): 13474-13499.
|
| [6] |
Niehues JM, Quirke P, West NP, et al. Generalizable biomarker prediction from cancer pathology slides with self-supervised deep learning: a retrospective multi-centric study. Cell Rep Med. 2023; 4(4):100980.
|
| [7] |
Al-Danakh A, Safi M, Jian Y, et al. Aging-related biomarker discovery in the era of immune checkpoint inhibitors for cancer patients. Front Immunol. 2024; 15:1348189.
|
| [8] |
Kaur A, Webster MR, Marchbank K, et al. sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature. 2016; 532(7598): 250-254.
|
| [9] |
Cesario A, D'Oria M, Calvani R, et al. The Role of Artificial Intelligence in Managing Multimorbidity and Cancer. J Pers Med. 2021; 11(4): 314.
|
| [10] |
Yang J, Chen X, Wang Y, Liu Z, Zhang L, Li M. Progress in Understanding Oxidative Stress, Aging, and Aging-Related Diseases. Antioxidants. 2024; 13(4): 394.
|
| [11] |
Zapatería B, Arias E. Aging, cancer, and autophagy: connections and therapeutic perspectives. Front Mol Biosci. 2025; 11:1516789.
|
| [12] |
Tartiere AG, Freije JMP, López-Otín C. The hallmarks of aging as a conceptual framework for health and longevity research. Front Aging. 2024; 5:1334261.
|
| [13] |
Haghani A, Li CZ, Robeck TR, et al. DNA methylation networks underlying mammalian traits. Science. 2023; 381(6658):eabq5693.
|
| [14] |
Boccardi V, Marano L. The telomere connection between aging and cancer: the burden of replication stress and dysfunction. Mech Ageing Dev. 2025; 223:112026.
|
| [15] |
Boccardi V, Marano L. Aging, Cancer, and Inflammation: the Telomerase Connection. Int J Mol Sci. 2024; 25(15): 8542.
|
| [16] |
Jia Q, Chen C, Xu A, et al. A biological age model based on physical examination data to predict mortality in a Chinese population. iScience. 2024; 27(3):108891.
|
| [17] |
Hernandez-Segura A, de Jong TV, Melov S, Guryev V, Campisi J, Demaria M. Unmasking transcriptional heterogeneity in senescent cells. Curr Biol. 2017; 27(17): 2652-2660.
|
| [18] |
Xu M, Pirtskhalava T, Farr JN, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018; 24(8): 1246-1256.
|
| [19] |
Saul D, Kosinsky RL, Atkinson EJ, et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat Commun. 2022; 13(1): 4827.
|
| [20] |
Guarente L, Sinclair DA, Kroemer G. Human trials exploring anti-aging medicines. Cell Metab. 2024; 36(2): 354-376.
|
| [21] |
Ehrlich M. DNA hypomethylation in cancer cells. Epigenomics. 2009; 1(2): 239-259.
|
| [22] |
Baylin SB, Jones PA. Epigenetic determinants of cancer. Cold Spring Harb Perspect Biol. 2016; 8(9):a019505.
|
| [23] |
Fraga MF, Esteller M. Epigenetics and aging: the targets and the marks. Trends Genet. 2007; 23(8): 413-418.
|
| [24] |
Sen P, Shah PP, Nativio R, et al. Epigenetic mechanisms of longevity and aging. Cell. 2016; 166(4): 822-839.
|
| [25] |
Puts MTE, Alibhai SMH, Szumacher E, et al. Evaluation of geriatric assessment and management on the toxic effects of cancer treatment (GAP70+): a cluster-randomised study. Lancet. 2021; 398(10314): 1894-1904.
|
| [26] |
Zhavoronkov A, Mamoshina P. Deep aging clocks: the emergence of AI-based biomarkers of aging and longevity. Trends Pharmacol Sci. 2019; 40(8): 546-549.
|
| [27] |
Liu Z, Liang Q, Ren Y, Guo C, Ge X, Wang L, et al. Immunosenescence: molecular mechanisms and diseases. Signal Transduct Target Ther. 2023; 8(1): 200.
|
| [28] |
Sebastiani P, Thyagarajan B, Sun F, et al. Biomarker signatures of aging. Aging Cell. 2017; 16(2): 329-338.
|
| [29] |
Lu AT, Quach A, Wilson JG, et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging. 2019; 11(2): 303-327.
|
| [30] |
Horvath S, Raj K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat Rev Genet. 2018; 19(6): 371-384.
|
| [31] |
Zhang B, Lee DE, Trapp A, et al. Multi-omic rejuvenation and life span extension on exposure to youthful circulation. Nat Aging. 2023; 3(2): 157-168.
|
| [32] |
Belsky DW, Caspi A, Houts R, et al. Quantification of biological aging in young adults. Proc Natl Acad Sci USA. 2015; 112(30): E4104-10.
|
| [33] |
Lewis-Israeli YR, Volmert BD, Gadue P. Molecular imaging of the aging heart. Curr Cardiol Rep. 2021; 23(8): 108.
|
| [34] |
Gillies RJ, Kinahan PE, Hricak H. Radiomics: images are more than pictures, they are data. Radiology. 2016; 278(2): 563-577.
|
| [35] |
Ferrucci L, Gonzalez-Freire M, Fabbri E, et al. Measuring biological aging in humans: a quest. Aging Cell. 2020; 19(2):e13080.
|
| [36] |
Sedrak MS, Gilmore NJ, Carroll JE, et al. Measuring biological age in the CARG score: development of novel aging measures in older adults with cancer. J Geriatr Oncol. 2021; 12(8): 1215-1222.
|
| [37] |
Galvin A, Helmer Q, Cheng MP, et al. Biological age is a predictor of mortality in ischemic stroke. Sci Rep. 2018; 8(1): 4148.
|
| [38] |
Moskalev A, Stambler I, Caruso C. Innate and adaptive immunity in aging and longevity: the foundation of resilience. Aging Dis. 2020; 11(6): 1363-1373.
|
| [39] |
Bell CG, Lowe R, Adams PD, et al. DNA methylation aging clocks: challenges and recommendations. Genome Biol. 2019; 20(1): 249.
|
| [40] |
Lowe D, Horvath S, Raj K. Epigenetic clock analyses of cellular senescence and ageing. Nat Rev Mol Cell Biol. 2022; 23(7): 467-480.
|
| [41] |
Belsky DW, Caspi A, Arseneault L, et al. Etiological features of the Dunedin pace of aging derived from 20 years of cohort data. eLife. 2022; 11:e73420.
|
| [42] |
Cohen HJ, Smith D, Sun CL, et al. Frailty as determined by a comprehensive geriatric assessment-derived deficit-accumulation index in older patients with cancer who receive chemotherapy. Cancer. 2016; 122(24): 3865-3872.
|
| [43] |
Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric Oncology consensus on geriatric assessment in older patients with cancer. J Clin Oncol. 2014; 32(24): 2595-2603.
|
| [44] |
Ness KK, Krull KR, Jones KE, et al. Physiologic frailty as a sign of accelerated aging among adult survivors of childhood cancer: a report from the St Jude Lifetime Cohort Study. J Clin Oncol. 2013; 31(36): 4496-4503.
|
| [45] |
Williams GR, Dunham L, Chang Y, et al. Geriatric assessment as an aide to understanding falls and cognition in older adults with cancer. Support Care Cancer. 2019; 27(4): 1365-1372.
|
| [46] |
Ferrara R, Mezquita L, Texier M, et al. Hyperprogressive disease in patients with advanced non-small cell lung cancer treated with PD-1/PD-L1 inhibitors or with single-agent chemotherapy. JAMA Oncol. 2018; 4(11): 1543-1552.
|
| [47] |
van Holstein Y, Kapiteijn E, Bastiaannet E, et al. Efficacy and adverse events of immunotherapy with checkpoint inhibitors in older patients with cancer. Drugs Aging. 2019; 36(10): 927-938.
|
| [48] |
Friedlaender A, Banna GL, Buffoni L, et al. Poor-performance status assessment of patients with non-small-cell lung cancer remains vague: an ancillary analysis from the SAFIR02-Lung/UNICANCER trial. Ann Oncol. 2019; 30(10): 1665-1670.
|
| [49] |
Aerts HJ, Mak RH, Chang TG, et al. FaceAge: examining facial features to predict cancer prognosis using artificial intelligence. Lancet Digit Health. 2024; 6(12): e891-9.
|
| [50] |
Chang TG, Cao Y, Sfreddo HJ, et al. LORIS robustly predicts patient outcomes with immune checkpoint blockade therapy using common clinical, pathologic and genomic features. Nat Cancer. 2024; 5(6): 881-892.
|
| [51] |
Soubeyran P, Bellera C, Goyard J, et al. Screening for vulnerability in older cancer patients: the ONCODAGE prospective multicenter cohort study. PLoS One. 2014; 9(12):e115060.
|
| [52] |
Rugo HS, Hurvitz SA, Yardley DA, et al. Alliance A171601: a phase II study of palbociclib plus letrozole in women ≥70 years with hormone receptor-positive, HER2-negative advanced breast cancer. J Geriatr Oncol. 2024; 15(1):101672.
|
| [53] |
Mohile SG, Dale W, Somerfield MR, et al. Practical assessment and management of vulnerabilities in older patients receiving systemic cancer therapy: ASCO Guideline Update. J Clin Oncol. 2018; 36(22): 2326-2347.
|
| [54] |
Duan Q, Reid SP, Clark NR, et al. L1000CDS2: lINCS L1000 characteristic direction signatures search engine. NPJ Syst Biol Appl. 2016; 2:16015.
|
| [55] |
Hamaker ME, Jonker JM, de Rooij SE, et al. Frailty screening methods for predicting outcome of a comprehensive geriatric assessment in elderly patients with cancer: a systematic review. Lancet Oncol. 2012; 13(4): e437-44.
|
| [56] |
Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020; 288(5): 518-536.
|
| [57] |
Kocak B, Durmaz ES, Ates E, et al. Radiogenomics in clear cell renal cell carcinoma: machine learning-based high-dimensional quantitative CT texture analysis in predicting PBRM1 mutation status. AJR Am J Roentgenol. 2019; 212(3): W55-63.
|
| [58] |
Zhavoronkov A, Mamoshina P, Vanhaelen Q, et al. Artificial intelligence for aging and longevity research: recent advances and perspectives. Ageing Res Rev. 2019; 49: 49-66.
|
RIGHTS & PERMISSIONS
2025 The Author(s). Clinical and Translational Discovery published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.