Abstract
The improvement in survival of childhood cancer observed across the past 50 years has resulted in a growing acknowledgment that simply extending the lifespan of survivors is not enough. It is incumbent on both the cancer research and the clinical care communities to also improve the health span of survivors. It is well established that aging adult survivors of childhood cancer are at increased risk of chronic health conditions, relative to the general population. However, as the first generation of survivors age into their 50s and 60s, it has become increasingly evident that this population is also at risk of early onset of physiologic aging. Geriatric measures have uncovered evidence of reduced strength and speed and increased fatigue, all components of frailty, among survivors with a median age of 33 years, which is similar to adults older than 65 years of age in the general population. Furthermore, frailty in survivors independently increased the risk of morbidity and mortality. Although there has been a paucity of research investigating the underlying biologic mechanisms for advanced physiologic age in survivors, results from geriatric populations suggest five biologically plausible mechanisms that may be potentiated by exposure to cancer therapies: increased cellular senescence, reduced telomere length, epigenetic modifications, somatic mutations, and mitochondrial DNA infidelity. There is now a critical need for research to elucidate the biologic mechanisms of premature aging in survivors of childhood cancer. This research could pave the way for new frontiers in the prevention of these life-changing outcomes.
INTRODUCTION
Advances in treatment and supportive care over the past five decades have resulted in improved survival among children diagnosed with malignancies. More than 80% will survive at least 10 years after diagnosis.1 Unfortunately, a cure is not without life-long consequences, including the early development of adverse health outcomes. Adult survivors of childhood cancer develop chronic health conditions typically seen among persons decades older,2 suggesting early onset of physiologic aging and/or frailty. Frailty is a loss of physiologic capacity that interferes with normal function, most commonly described in older adults. In the general population, it identifies individuals highly vulnerable to adverse health outcomes and is a predictor of early mortality.3 In childhood cancer survivors, cancer and treatment exposures may influence the biologic mechanisms of aging, changing the trajectory of normal loss of physiologic capacity, and increasing the risk of frailty (Fig 1).
Fig 1.
Extrapolated figure hypothesizing that molecular integrity and associated physiologic capacity provoked by exposure to chemotherapy and/or radiation may be associated with excess risk and advanced onset of age-related diseases and frailty.
FRAILTY IN ADULT SURVIVORS OF CHILDHOOD CANCER
Data from the St. Jude Lifetime Cohort Study support this hypothesis (Fig 2), documenting frailty among nearly 8% of survivors at a median age of 33 years (range, 18 to 50 years).4 This compares with rates among age-matched peers of 0%, and among adults older than 65 years of age of 7.2%.3 Frailty is characterized by three or more of (1) low lean muscle mass, (2) reduced strength, (3) slow walking speed, (4) low energy expenditure, and (5) fatigue.3 The overall loss of physiologic reserve among survivors is substantial; an additional 22.2% of survivors have two of these impairments, and so meet the definition of pre-frail. As is observed in elderly populations, survivors who were frail had a 2.2-fold increased risk of a new-onset chronic condition and a 2.6-fold increased risk of death, independent of treatment exposures and other chronic health conditions.4 This is important because reversal and/or prevention of frailty may reduce the burden of disease, improve quality of life, and extend lifespan in this ever-growing population.5
Fig 2.
Percentage of survivors in the St. Jude Lifetime Cohort Study (SJLIFE) who meet the criteria for frailty compared with participants in the Cardiovascular Health Study and normal controls. N = 1,922 (50.3% male); mean time since diagnosis, 25.5±7.7 years; mean age at diagnosis, 8.2±5.6 years; 43% leukemia; 33% with cranial radiation exposure.
Components of the frailty phenotype are amenable to intervention in animal models,6 and they show promise among the elderly.7 However, interventions to remediate or prevent frailty in childhood cancer survivors have not been tested. In addition, although there is some evidence that radiation exposure contributes to the development of frailty among childhood cancer survivors,4 those not exposed are also at risk. Associations between chemotherapy and frailty have not been explored fully.4 To effectively design interventions, there is a need to gain a better understanding of the mechanisms that result in frail health in childhood cancer survivors. This review describes age-related chronic health conditions, summarizes plausible biologic mechanisms of aging that may be responsible for frailty, and suggests potential interventions that might be tailored to and tested among childhood cancer survivors to prevent or remediate frail health.
AGE-RELATED HEALTH CONDITIONS AMONG CHILDHOOD CANCER SURVIVORS
It is well established that adults who were treated for cancer during childhood are at an elevated risk of early onset of multiple age-related conditions. Data from clinically evaluated participants in the St. Jude Lifetime Cohort indicate that by age 50 years, each survivor, on average, experiences 4.7 grade 3 to 5 chronic conditions.8 Condition-specific results from the Childhood Cancer Survivor Study (CCSS) estimate the cumulative incidence of grade 3 to 5 chronic conditions by age 45 years to be 5.3% for coronary artery disease and 4.8% for heart failure.9 The incidence is lower in siblings: 0.9% for coronary artery disease and 0.3% for heart failure.9 Hypertension requiring medication is prevalent among 14.9% of CCSS cohort members and exponentially escalates the risk of a major cardiac event, including stroke.9,10 The age-adjusted rate for stroke among childhood cancer survivors is 77 of 100,000 compared with 9.3 of 100,000 person-years among siblings.10 Incident second neoplasms among childhood cancer survivors are 5.4 times (95% CI, 5.1 times to 5.7 times) higher than expected,11 a median of 17 years after diagnosis. Cataracts are prevalent in > 40% after significant radiotherapy exposure,12 low bone mineral density in 7.6%,13 and metabolic syndrome in nearly a third of survivors14 at a median age of 32 years. Survivors demonstrate a 1.8-fold (95% CI, 1.4-fold to 2.3-fold) greater risk of developing diabetes mellitus than their siblings.15 Primary hypogonadism is also prevalent, noted among 8.5% of male and 7.8% of female survivors who are in their 30s.13 Cognitive decline, one of the most concerning late effects of childhood cancer, and an age-related phenomenon in the general population, is experienced by survivors, 2% to 13% reporting problems with memory and organization and 22% with task efficiency.16 Despite reductions in late mortality (≥ 5 years from diagnosis) among survivors treated more recently (15-year cumulative incidence of 10.7% among those treated in the 1970s v 5.8% among those treated in the 1990s), 41% of deaths in the CCSS cohort are attributable to health-related causes rather than to progression or relapse of the primary cancer.17
Although there is evidence that cancer treatment exposures (Table 1),13 and in some cases genetic susceptibility,18-20 increase the risk of chronic health conditions2 and frailty,21,22 little information is available regarding the underlying biologic and molecular processes driving these outcomes. Although much of what is known about the normal aging process comes from the evaluation of aging in the general and geriatric populations, it is unclear whether these established processes are at work in survivors of childhood cancer. This population was exposed to potentially damaging agents that affected tissue integrity, cellular function, and DNA structure during key developmental periods, which may have prematurely accelerated the molecular processes seen in older adults.
Table 1.
Proportion of Chronic Health Conditions Attributable to Specific Treatment Exposures Among Survivors of Childhood Cancer
MOLECULAR MECHANISMS OF AGING
Aging in the general population is characterized by a loss of physiologic capacity, resulting in impaired function and eventually death (Fig 1). Among childhood cancer survivors, mechanisms responsible for premature loss of physiologic capacity are likely related to treatment-induced damage to normal, nonmalignant cells (neurons, cardiomyocytes, skeletal muscle, and supporting cells) after exposure to radiation and/or chemotherapy. Initial growth arrest at the molecular level may be accompanied by permanent alterations in DNA structure and function, together with interference with normal repair mechanisms at the cellular level, leaving survivors vulnerable to a lifetime of environmental exposures known to further affect the genome. Here we summarize five molecular mechanisms associated with aging in the general population, where evidence suggests a potential contribution to accelerated aging among survivors. These include cellular senescence, telomere attrition, changes in DNA methylation patterns, accumulation of somatic DNA mutations, and loss of mitochondrial fidelity.23
Cellular Senescence
Cellular senescence (Fig 3), a quiescent state representing the loss of a cell’s ability to replicate or grow, is a fate induced by DNA damage from chemotherapy or radiation, oncogene expression, mutations, repeated cell replication, cell exposure to danger-associated metabolic patterns such as nucleotides or other intracellular constituents, reactive oxygen species, and other damaging metabolites.24,25 Progression into senescence is mediated by transcription regulator cascades involving the p16INK4A/retinoblastoma protein, p53/ p21CIP1, and other factors24 and is accompanied by extensive changes in gene expression, reorganization of chromatin, and ultimately, irreversible replicative arrest, resistance to apoptosis, and sometimes a senescence-associated secretory phenotype (SASP).24,26-29 The SASP (Fig 2) entails upregulated secretion of hundreds of proteins, including pro-inflammatory cytokines, chemokines, and proteases, among others. Through the SASP, senescence spreads to other cells locally and distantly, contributing to the damaging effects senescent cells have on adjacent tissue and systemic function.30,31 Senescent cells increase in abundance with age24,32 and accumulate after DNA-damaging chemotherapy or radiation,33 in association with frailty,34,35 and at the sites of pathogenesis of chronic disorders and diseases,31 such as bone in osteoporosis,36,37 vessel walls in arterial sclerosis,38 joints in osteoarthritis,39 the lungs in pulmonary fibrosis,40 adipose tissue in diabetes or obesity,32,41,42 or the liver in hepatic steatosis or cirrhosis.43
Fig 3.
Inducers, mediators, senescent cell anti-apoptotic pathways (SCAPs), the senescence-associated secretory phenotype (SASP), and effects of senescent cells. BCL-2, B-cell lymphoma 2; C/EBPβ, Ccaat/enhancer binding protein beta; Dep, density-enhanced phosphatase; HIF, hypoxia-inducible factor; HSP, heat shock protein; IGF, insulin-like growth factor; IL, interleukin; mTOR, mammalian target of rapamycin; NFκB, nuclear factor kappa-light-chain-enhancer of activated B cells; ROS, reactive oxygen species; TGFβ, transforming growth factor beta.
Many of the inducers and consequences of senescence are relevant to childhood cancer treatment and survivors.44 The chemotherapy and radiation administered to treat childhood cancer can induce cellular senescence, as evidenced by increased expression of p16INK4a in scalp biopsy specimens of children with acute lymphoblastic leukemia exposed to cranial radiation.45 In addition, metabolic dysfunction among childhood cancer survivors may be both a consequence of the SASP and a driving factor for additional generation of senescent cells. Muscle weakness, frailty, metabolic and cardiovascular dysfunction, proclivity to develop second cancers, pulmonary fibrosis, an accelerated aging-like state, and early death all have documented associations with or are the consequence of cellular senescence in other populations.24,25,31,32,34,38,40,46 However, more evidence is needed to characterize this association among survivors.44
Telomeres and Inflammation
Telomeres are nucleoprotein structures capping chromosome ends that protect genome integrity by preventing chromosome end-to-end fusions, nucleolytic processing, and homologous recombination. The length of telomeric DNA, which varies among individuals and cell populations, is heritable and is subject to epigenetic modification.47-49 Telomere length (TL) shortens with age in replicative tissues lacking telomerase, a specialized reverse transcription that replenishes terminal telomere repeats lost during genome duplication (Fig 4).50-52 Moreover, TL is strongly correlated across different tissue types from the same individual, with similar rates of attrition over time.53 Once telomeres become critically short, they lose their ability to protect chromosome ends, triggering a DNA damage response and cellular senescence or apoptosis.54 Thus, TL predicts cellular replicative capacity.55 Mean leukocyte TL (mLTL) shortening is associated with aging-related diseases56 including type 2 diabetes,57 cardiovascular disease,58 dementia,59 and increased cancer risk in the general population.60,61 In addition, shorter mLTL is observed in individuals with impaired grip strength, reduced lean muscle mass, and lower cardiorespiratory capacity,62-64 all factors characteristic of frailty. Given the accelerated mLTL attrition noted after exposure to chemotherapy and radiation,65-71 cancer survivors may be at particularly high risk of premature, accelerated aging (Fig 3). In aging populations, loss of TL is likely related to the SASP, because telomere attrition is accompanied by an increase in systemic inflammation,72,73 characterized by a two- to four-fold increase in the expression of pro-inflammatory cytokines.74,75 This phenomenon occurs even among healthy individuals lacking comorbidities,76 predicting reduced muscle mass and muscle strength and a higher incidence of a frailty phenotype.77-82 Higher plasma concentrations of interleukin (IL)-6 and tumor necrosis factor-α have been observed in elderly individuals with lower muscle mass, reduced grip strength, and slower walking speed.83-86 Plasma C-reactive protein, transforming growth factor-β, IL-6, tumor necrosis factor alpha receptor (TNFR) 1, and TNFR2 levels are associated with frailty, in some cases more predictive of frailty than even chronologic age.87-89 Finally, both shortened TL and systemic inflammation have been shown to predict increased mortality risk in elderly populations.90-93
Fig 4.
In cells lacking telomerase expression, telomere length shortens with each cell division because of the inability of the lagging strand to replicate terminal nucleotides at the 5′ end. Progressive shortening continues until telomeres become critically short, triggering either cellular senescence or apoptosis.
A few studies have examined the impact of telomere attrition and systemic inflammation on late effects of cancer therapy, and there is some evidence to suggest that these biomarkers are related to therapeutic intensity. For example, the rate of accumulation of comorbidities observed over time in women treated for breast cancer is associated with higher levels of IL-6 and tumor necrosis factor-α, findings that are significantly associated with exposure to multimodal therapy.94 Systemic inflammation among adult-diagnosed cancer survivors has also been associated with adverse neurocognitive outcomes and fatigue.95-97 Similar to studies in adult cancer survivors, female childhood leukemia survivors with higher serum IL-6 and IL-1β levels demonstrate reduced executive function and processing speed.98 Shorter mLTL predicts the risk of second cancers,20 and together with elevated plasma inflammatory cytokines, distinguishes survivors of childhood acute leukemia with cranial irradiation from noncancer controls.99 In one study specifically designed to investigate frailty, neuroblastoma survivors treated with autologous stem-cell transplant had increased frailty scores compared with age- and sex-matched controls, as well as significantly shorter mLTL and increased C-reactive protein.100
Methylation
Chromatin has several functions, including control of gene expression and DNA replication. Basic repeating units of chromatin, or nucleosomes, include approximately 150 base pairs of DNA wrapped around eight histone proteins.101 Chromatin structure is regulated by methylation of the 5′-cytosine phosphate guanine-3′ (CpG) dinucleotide, resulting in transcriptional silencing.102 Although most CpGs in the human genome are methylated, there are large regions of concentrated CpGs (CpG islands) that are hypomethylated. These regions are found in promoter-rich areas and tend to become hypermethylated with age.103 The consequences of DNA methylation with age are not completely clear.104 However, in aging hematopoietic stem cells, regions with altered DNA methylation contribute to dysregulation of gene expression during aging.105 Although DNA methylation is closely linked to chronologic age,106 there are deviations from the linear fit of this correlation, indicating that methylation patterns may also reflect biologic age.104 Methylation is influenced by chronic inflammation107 and is associated with TL in both adolescents108 and adults in the general population.109 Importantly, DNA methylation of blood cells independently predicts all-cause mortality in older adults, even after accounting for traditional risk factors.110 DNA methylation is affected by lifestyle, environmental exposures, and cancer therapies. The acute effects of radiation and some chemotherapy exposures on cancer cells, and in murine models, demonstrate dose-specific differential methylation of genes in pathways consistent with classic biologic responses,111,112 suggesting that children treated with these agents may experience alterations in DNA methylation. A small study among pediatric medulloblastoma or primitive neuroectodermal tumor survivors indicates that methylation at least influences outcomes, because differential methylation at cg14010619 (PAK4 gene) is associated with ototoxicity.113
Somatic Mutations
Somatic mutations accumulate throughout life, resulting in tissues that contain a mosaic of cells with different genotypes.114 Mutations occur in all tissues and organs115 but are particularly evident in the aging hematopoietic system and are tied to other biologic markers of aging. For example, recurrent Tet methylcytosine dioxygenase 2 somatic mutations, evident in whole-exome sequencing studies of peripheral blood cells among normal elderly individuals, are associated with alterations in DNA methylation.116 Among older adults, clonal hematopoiesis driver genes are mutated the most frequently and are associated with all-cause mortality and risk of hematologic cancer.117,118 Recent data indicate that clonal hematopoiesis driver genes may not be responsible for this process in survivors of childhood cancer.119 Nevertheless, because defects in genome maintenance pathways in human progeroid syndromes and in murine models lead to both cancer and the tissue degeneration normally seen in old age,120,121 we speculate that the accumulation of somatic mutations may explain the increased risk of both cancer and accelerated aging among childhood cancer survivors whose underlying genetics and early life exposures to DNA-damaging agents potentially disrupted genome maintenance pathways. Additional studies exploring this hypothesis in childhood cancer survivors are needed.
Mitochondrial DNA Fidelity
Mitochondrial genomes exhibit higher rates of mutation than do somatic genomes,122 rendering mitochondrial DNA (mtDNA) particularly vulnerable to the genotoxic effects of cancer-related therapies. Alkylator-based regimens, for example, are associated with significantly enriched de novo mtDNA mutations in human primary chronic lymphocytic leukemia cells,123 whereas cisplatin preferentially accumulates in mitochondria and structurally disrupts mtDNA synthesis through the formation of mtDNA adducts.124,125 In addition, acute doxorubicin exposure provokes mtDNA alterations that persist and promote delayed onset of cardiomyopathy in mice.126,127 In murine models, induced mtDNA pathology is associated with reduced physiologic reserve and signs of premature aging, including kyphosis, loss of bone and muscle mass, weight loss, and anemia (Fig 5).128,129 Defective mitochondria, and the quality control mechanisms that regulate their accrual, are also tied to the pathobiology of frailty in humans.130-135 Mitochondrial diseases are characterized by myopathies and neuromuscular disorders,136,137 and human diseases with muscle-intrinsic pathologies exhibit secondary mitochondrial dysfunction. The accumulation of damaged mitochondria limits the ability of muscle stem cells to effectively sustain or regenerate tissue, resulting in additional loss of muscle, exacerbating existing frail health.138-143 As with somatic mutations, there is a need for an investigation into the associations between mitochondrial infidelity and accelerated aging in childhood cancer survivors.
Fig 5.
Premature aging phenotype in mitochondrial DNA mutator mice. (A) A representative photograph of 10-month-old mice depicts the smaller size caused by weight loss, alopecia, and graying hair in mice harboring proof-reading defective POLG (Polgmt/mt). (B) Representative radiographs demonstrate the kyphosis in mutant mice. (C) Representative low-magnification images of hematoxylin and eosin–stained sections highlight the loss of skeletal muscle mass (ie, sarcopenia) in the mutant mice. The mitochondrial DNA mutator mice also develop other age-related disorders, including macrocytic anemia and lymphopenia, osteoporosis, hearing loss, testicular atrophy, and cardiac hypertrophy or dysfunction. Data adapted.129,130
INTERVENTIONS FOR FRAIL HEALTH
An improved understanding of the molecular mechanisms related to accelerated aging among childhood cancer survivors should facilitate the development and implementation of interventions that target the cells or processes responsible for diminished physiologic reserve. Evidence from other populations and from animal models6 indicates that components of the frailty phenotype, and the biomarkers presented here, are responsive to exercise. In the general population, exercise interventions reduce chronic systemic inflammation, mitigate age-related telomere shortening,144-148 influence methylation patterns,107 and increase mtDNA abundance in peripheral blood.149 Although published literature describing effective exercise intervention among children with, or who have survived, cancer is limited, data indicate that survivors can gain lean mass, strength, and walking speed.40,150-155 However, adherence to exercise is problematic,156 even in healthy populations. Additional research will require strong behavioral strategies, will need to determine the most appropriate timing of an intervention (during therapy, immediately after a therapy, or years later), and should incorporate outcomes that characterize frailty and that capture the effects of exercise on biomarkers of aging.
Senolytics, soon ready for clinical trials in humans, are pharmaceutical or natural products that selectively eliminate senescent cells.25,33,157-160 These agents transiently disable senescent cell anti-apoptotic pathways, resulting in apoptosis of senescent cells, with sparing of nonsenescent cells.25,159 At least six senescent cell anti-apoptotic pathways have been identified: a dependence receptor/tyrosine kinase pathway, B-cell lymphoma 2–related factors including B-cell lymphoma-xL, a PI3K/AKT/ceramide metabolic network, p53/p21CIP1/serpine elements, a hypoxia-inducible factor 1α pathway,159 and a heat shock protein-90 pathway.161 Remarkably, these pathways resemble those that defend cancer cells against apoptosis. In fact, the senolytics identified so far induce cancer cell apoptosis in vitro or in vivo and include dasatinib, quercetin, navitoclax, fisetin, A1331852, and A1155463, among others.25,33,158-161 Because senescent cells do not divide and take weeks to months to reaccumulate if the stimuli inducing senescence remain operative, replication-related drug resistance is unlikely to develop. These agents are also attractive because they can be administered intermittently (eg, once a month) because they act by transiently downregulating prosurvival pathways. They do not need to be present continuously in the circulation. Certainly, they may be easier for survivors to adhere to than exercise. In animal models, senolytic drugs have efficacy for treating chronic diseases in animal models, including cardiac,159 vascular,38,159 metabolic dysfunction,32 pulmonary fibrosis,40,162 liver steatosis,43 osteoporosis,36,163 frailty,159 and radiation-induced muscle wasting.159,160 Because childhood cancer survivors have many of the disorders responsive to senolytic drugs, these agents may be effective in alleviating the multimorbidity and accelerated aging phenotypes that occur in these patients.25,44 Research exploring the use of these agents also must determine the most appropriate timing of their application, either immediately after therapy soon after acute cellular or physiologic injury, or years later when the health conditions typically manifest.
In summary, it is well established that survivors of childhood cancers embark on a lifetime of increased risk of adverse health consequences as a result of cancer and cancer therapy; simultaneously, as they age, many will experience early onset of physiologic frailty, independently increasing the risk of morbidity and mortality. Although there is strong biologic plausibility that age-related molecular pathways identified in the general population may be potentiated by exposure to chemotherapy and radiotherapy during important periods of development, there is a critical need for research in these areas. Better understanding of the underlying biologic mechanisms of premature aging in survivors of childhood cancer could pave the way for new frontiers in the prevention of these life-changing outcomes.
ACKNOWLEDGMENT
We thank Peter Vogel, director of the Veterinary Pathology Core at St. Jude Children’s Research Hospital, for characterizing the phenotype of the POLG mice.
AUTHOR CONTRIBUTIONS
Conception and design: Kirsten K. Ness, James L. Kirkland, Maria Monica Gramatges, Mondira Kundu, Xiujie Li-Harms, Tamar Tchkonia, Saskia Martine Francesca Pluijm, Gregory T. Armstrong
Collection and assembly of data: Kirsten K. Ness, James L. Kirkland, Maria Monica Gramatges, Zhaoming Wang, Mondira Kundu, Xiujie Li-Harms, Jinghui Zhang, Saskia Martine Francesca Pluijm, Gregory T. Armstrong
Data analysis and interpretation: All authors
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Premature Physiologic Aging as a Paradigm for Understanding Increased Risk of Adverse Health Across the Lifespan of Survivors of Childhood Cancer
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/site/ifc.
Kirsten K. Ness
No relationship to disclose
James L. Kirkland
Patents, Royalties, Other Intellectual Property: Unity Biotechnology
Maria Monica Gramatges
Research Funding: Bristol-Myers Squibb
Zhaoming Wang
No relationship to disclose
Mondira Kundu
No relationship to disclose
Kelly McCastlain
No relationship to disclose
Xiujie Li-Harms
No relationship to disclose
Jinghui Zhang
No relationship to disclose
Tamar Tchkonia
Patents, Royalties, Other Intellectual Property: Methods and Compositions for Killing Senescent Cells and for Treating Senescence-Associated Diseases and Disorders (Inst)
Saskia Martine Francesca Pluijm
No relationship to disclose
Gregory T. Armstrong
No relationship to disclose
REFERENCES
- 1. National Cancer Institute: SEER cancer statistics review (CSR) 1975-2014 https://seer.cancer.gov/csr/1975_2014/
- 2.Armstrong GT, Kawashima T, Leisenring W, et al. : Aging and risk of severe, disabling, life-threatening, and fatal events in the childhood cancer survivor study. J Clin Oncol 32:1218-1227, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Fried LP, Tangen CM, Walston J, et al. :Frailty in older adults: Evidence for a phenotype. J Gerontol A Biol Sci Med Sci 56:M146-M156, 2001 [DOI] [PubMed] [Google Scholar]
- 4.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 31:4496-4503, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ness KK, Howell CR, Bjornard KL: Frailty and quality of life in adult survivors of childhood cancer. Expert Rev Qual Life Cancer Care 2:79-85, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Schafer MJ, White TA, Evans G, et al. : Exercise prevents diet-induced cellular senescence in adipose tissue. Diabetes 65:1606-1615, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Puts MTE, Toubasi S, Andrew MK, et al. : Interventions to prevent or reduce the level of frailty in community-dwelling older adults: A scoping review of the literature and international policies. Age Ageing 46:383-392, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bhakta N, Liu Q, Ness KK, et al. : The cumulative burden of surviving childhood cancer: An initial report from the St Jude Lifetime Cohort Study (SJLIFE). Lancet 390:2569-2582, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Armstrong GT, Oeffinger KC, Chen Y, et al. : Modifiable risk factors and major cardiac events among adult survivors of childhood cancer. J Clin Oncol 31:3673-3680, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mueller S, Fullerton HJ, Stratton K, et al. : Radiation, atherosclerotic risk factors, and stroke risk in survivors of pediatric cancer: A report from the Childhood Cancer Survivor Study. Int J Radiat Oncol Biol Phys 86:649-655, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Turcotte LM, Liu Q, Yasui Y, et al. : Temporal trends in treatment and subsequent neoplasm risk among 5-year survivors of childhood cancer, 1970-2015. JAMA 317:814-824, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chodick G, Sigurdson AJ, Kleinerman RA, et al. : The risk of cataract among survivors of childhood and adolescent cancer: A report from the Childhood Cancer Survivor Study. Radiat Res 185:366-374, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hudson MM, Ness KK, Gurney JG, et al. : Clinical ascertainment of health outcomes among adults treated for childhood cancer. JAMA 309:2371-2381, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Smith WA, Li C, Nottage KA, et al. : Lifestyle and metabolic syndrome in adult survivors of childhood cancer: A report from the St. Jude Lifetime Cohort Study. Cancer 120:2742-2750, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mostoufi-Moab S, Seidel K, Leisenring WM, et al. : Endocrine abnormalities in aging survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. J Clin Oncol 34:3240-3247, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Krull KR, Annett RD, Pan Z, et al. : Neurocognitive functioning and health-related behaviours in adult survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. Eur J Cancer 47:1380-1388, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Armstrong GT, Yasui Y, Robison LL: Reduction in late mortality after childhood cancer. N Engl J Med 375:290-292, 2016 [DOI] [PubMed] [Google Scholar]
- 18.Blanco JG, Sun CL, Landier W, et al. : Anthracycline-related cardiomyopathy after childhood cancer: Role of polymorphisms in carbonyl reductase genes--A report from the Children’s Oncology Group. J Clin Oncol 30:1415-1421, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rajić V, Aplenc R, Debeljak M, et al. : Influence of the polymorphism in candidate genes on late cardiac damage in patients treated due to acute leukemia in childhood. Leuk Lymphoma 50:1693-1698, 2009 [DOI] [PubMed] [Google Scholar]
- 20.Gramatges MM, Liu Q, Yasui Y, et al. : Telomere content and risk of second malignant neoplasm in survivors of childhood cancer: A report from the Childhood Cancer Survivor Study. Clin Cancer Res 20:904-911, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ojha RP, Oancea SC, Ness KK, et al. : Assessment of potential bias from non-participation in a dynamic clinical cohort of long-term childhood cancer survivors: Results from the St. Jude Lifetime Cohort Study. Pediatr Blood Cancer 60:856-864, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wilson CL, Chemaitilly W, Jones KE, et al. : Modifiable factors associated with aging phenotypes among adult survivors of childhood acute lymphoblastic leukemia. J Clin Oncol 34:2509-2515, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.López-Otín C, Blasco MA, Partridge L, et al. : The hallmarks of aging. Cell 153:1194-1217, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tchkonia T, Zhu Y, van Deursen J, et al. : Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J Clin Invest 123:966-972, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kirkland JL, Tchkonia T: Cellular senescence: A translational perspective. EBioMedicine 21:21-28, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Coppé JP, Patil CK, Rodier F, et al. : Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol 6:2853-2868, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kuilman T, Michaloglou C, Vredeveld LC, et al. : Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 133:1019-1031, 2008 [DOI] [PubMed] [Google Scholar]
- 28.Kuilman T, Peeper DS: Senescence-messaging secretome: SMS-ing cellular stress. Nat Rev Cancer 9:81-94, 2009 [DOI] [PubMed] [Google Scholar]
- 29.Wang E: Senescent human fibroblasts resist programmed cell death, and failure to suppress bcl2 is involved. Cancer Res 55:2284-2292, 1995 [PubMed] [Google Scholar]
- 30.Nelson G, Wordsworth J, Wang C, et al. : A senescent cell bystander effect: Senescence-induced senescence. Aging Cell 11:345-349, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhu Y, Armstrong JL, Tchkonia T, et al. : Cellular senescence and the senescent secretory phenotype in age-related chronic diseases. Curr Opin Clin Nutr Metab Care 17:324-328, 2014 [DOI] [PubMed] [Google Scholar]
- 32.Xu M, Palmer AK, Ding H, et al. : Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife 4:e12997, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, et al. : Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 15:428-435, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Justice JN, Gregory H, Tchkonia T, et al. : Cellular senescence biomarker p16INK4a+ cell burden in thigh adipose is associated with poor physical function in older women. J Gerontol A Biol Sci Med Sci 10.1093/gerona/glx134 [epub ahead of print on June 27, 2017] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Xu M, Tchkonia T, Ding H, et al. : JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci USA 112:E6301-E6310, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Farr JN, Xu M, Weivoda MM, et al. : Targeting cellular senescence prevents age-related bone loss in mice. Nat Med 23:1072-1079, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Farr JN, Fraser DG, Wang H, et al. : Identification of senescent cells in the bone microenvironment. J Bone Miner Res 31:1920-1929, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Roos CM, Zhang B, Palmer AK, et al. : Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 15:973-977, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Xu M, Bradley EW, Weivoda MM, et al. : Transplanted senescent cells induce an osteoarthritis-like condition in mice. J Gerontol A Biol Sci Med Sci 72:780-785, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Schafer MJ, White TA, Iijima K, et al. : Cellular senescence mediates fibrotic pulmonary disease. Nat Commun 8:14532, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tchkonia T, Morbeck DE, Von Zglinicki T, et al. : Fat tissue, aging, and cellular senescence. Aging Cell 9:667-684, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Minamino T, Orimo M, Shimizu I, et al. : A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med 15:1082-1087, 2009 [DOI] [PubMed] [Google Scholar]
- 43.Ogrodnik M, Miwa S, Tchkonia T, et al. : Cellular senescence drives age-dependent hepatic steatosis. Nat Commun 8:15691, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ness KK, Armstrong GT, Kundu M, et al. : Frailty in childhood cancer survivors. Cancer 121:1540-1547, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Marcoux S, Le ON, Langlois-Pelletier C, et al. : Expression of the senescence marker p16INK4a in skin biopsies of acute lymphoblastic leukemia survivors: A pilot study. Radiat Oncol 8:252, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Stout MB, Tchkonia T, Pirtskhalava T, et al. : Growth hormone action predicts age-related white adipose tissue dysfunction and senescent cell burden in mice. Aging (Albany NY) 6:575-586, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.De Meyer T, Vandepitte K, Denil S, et al. : A non-genetic, epigenetic-like mechanism of telomere length inheritance? Eur J Hum Genet 22:10-11, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Blasco MA: Carcinogenesis Young Investigator Award. Telomere epigenetics: A higher-order control of telomere length in mammalian cells. Carcinogenesis 25:1083-1087, 2004 [DOI] [PubMed] [Google Scholar]
- 49.Gadalla SM, Cawthon R, Giri N, et al. : Telomere length in blood, buccal cells, and fibroblasts from patients with inherited bone marrow failure syndromes. Aging (Albany NY) 2:867-874, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Vaziri H, Dragowska W, Allsopp RC, et al. : Evidence for a mitotic clock in human hematopoietic stem cells: Loss of telomeric DNA with age. Proc Natl Acad Sci USA 91:9857-9860, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chiang YJ, Calado RT, Hathcock KS, et al. : Telomere length is inherited with resetting of the telomere set-point. Proc Natl Acad Sci USA 107:10148-10153, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Müezzinler A, Zaineddin AK, Brenner H: A systematic review of leukocyte telomere length and age in adults. Ageing Res Rev 12:509-519, 2013 [DOI] [PubMed] [Google Scholar]
- 53.Daniali L, Benetos A, Susser E, et al. : Telomeres shorten at equivalent rates in somatic tissues of adults. Nat Commun 4:1597, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Artandi SE, Attardi LD: Pathways connecting telomeres and p53 in senescence, apoptosis, and cancer. Biochem Biophys Res Commun 331:881-890, 2005 [DOI] [PubMed] [Google Scholar]
- 55.Allsopp RC, Vaziri H, Patterson C, et al. : Telomere length predicts replicative capacity of human fibroblasts. Proc Natl Acad Sci USA 89:10114-10118, 1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Blackburn EH, Epel ES, Lin J: Human telomere biology: A contributory and interactive factor in aging, disease risks, and protection. Science 350:1193-1198, 2015 [DOI] [PubMed] [Google Scholar]
- 57.Zhao J, Miao K, Wang H, et al. : Association between telomere length and type 2 diabetes mellitus: A meta-analysis. PLoS One 8:e79993, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Haycock PC, Heydon EE, Kaptoge S, et al. : Leucocyte telomere length and risk of cardiovascular disease: Systematic review and meta-analysis. BMJ 349:g4227, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Honig LS, Kang MS, Schupf N, et al. : Association of shorter leukocyte telomere repeat length with dementia and mortality. Arch Neurol 69:1332-1339, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ma H, Zhou Z, Wei S, et al. : Shortened telomere length is associated with increased risk of cancer: A meta-analysis. PLoS One 6:e20466, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wentzensen IM, Mirabello L, Pfeiffer RM, et al. : The association of telomere length and cancer: A meta-analysis. Cancer Epidemiol Biomarkers Prev 20:1238-1250, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Woo J, Yu R, Tang N, et al. : Telomere length is associated with decline in grip strength in older persons aged 65 years and over. Age (Dordr) 36:9711, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Loprinzi PD: Cardiorespiratory capacity and leukocyte telomere length among adults in the United States. Am J Epidemiol 182:198-201, 2015 [DOI] [PubMed] [Google Scholar]
- 64.Meyer A, Salewsky B, Spira D, et al. : Leukocyte telomere length is related to appendicular lean mass: Cross-sectional data from the Berlin Aging Study II (BASE-II). Am J Clin Nutr 103:178-183, 2016 [DOI] [PubMed] [Google Scholar]
- 65.Li P, Hou M, Lou F, et al. : Telomere dysfunction induced by chemotherapeutic agents and radiation in normal human cells. Int J Biochem Cell Biol 44:1531-1540, 2012 [DOI] [PubMed] [Google Scholar]
- 66.Fumagalli M, Rossiello F, Clerici M, et al. : Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat Cell Biol 14:355-365, 2012. [Erratum: Nat Cell Biol 14:555, 2012] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Schröder CP, Wisman GB, de Jong S, et al. : Telomere length in breast cancer patients before and after chemotherapy with or without stem cell transplantation. Br J Cancer 84:1348-1353, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ayouaz A, Raynaud C, Heride C, et al. : Telomeres: Hallmarks of radiosensitivity. Biochimie 90:60-72, 2008 [DOI] [PubMed] [Google Scholar]
- 69.Zhong YH, Liao ZK, Zhou FX, et al. : Telomere length inversely correlates with radiosensitivity in human carcinoma cells with the same tissue background. Biochem Biophys Res Commun 367:84-89, 2008 [DOI] [PubMed] [Google Scholar]
- 70.Diker-Cohen T, Uziel O, Szyper-Kravitz M, et al. : The effect of chemotherapy on telomere dynamics: Clinical results and possible mechanisms. Leuk Lymphoma 54:2023-2029, 2013 [DOI] [PubMed] [Google Scholar]
- 71.Lee JJ, Nam CE, Cho SH, et al. : Telomere length shortening in non-Hodgkin’s lymphoma patients undergoing chemotherapy. Ann Hematol 82:492-495, 2003 [DOI] [PubMed] [Google Scholar]
- 72.Wong JY, De Vivo I, Lin X, et al. : The relationship between inflammatory biomarkers and telomere length in an occupational prospective cohort study. PLoS One 9:e87348, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.O’Donovan A, Pantell MS, Puterman E, et al. : Cumulative inflammatory load is associated with short leukocyte telomere length in the Health, Aging and Body Composition Study. PLoS One 6:e19687, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Franceschi C, Bonafè M, Valensin S, et al. : Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci 908:244-254, 2000 [DOI] [PubMed] [Google Scholar]
- 75.Baylis D, Bartlett DB, Patel HP, et al. : Understanding how we age: Insights into inflammaging. Longev Healthspan 2:8, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Puzianowska-Kuźnicka M, Owczarz M, Wieczorowska-Tobis K, et al. : Interleukin-6 and C-reactive protein, successful aging, and mortality: The PolSenior study. Immun Ageing 13:21, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Soysal P, Stubbs B, Lucato P, et al. : Inflammation and frailty in the elderly: A systematic review and meta-analysis. Ageing Res Rev 31:1-8, 2016 [DOI] [PubMed] [Google Scholar]
- 78.Mekli K, Nazroo JY, Marshall AD, et al. : Proinflammatory genotype is associated with the frailty phenotype in the English Longitudinal Study of Ageing. Aging Clin Exp Res 28:413-421, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Schaap LA, Pluijm SM, Deeg DJ, et al. : Higher inflammatory marker levels in older persons: Associations with 5-year change in muscle mass and muscle strength. J Gerontol A Biol Sci Med Sci 64:1183-1189, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hubbard RE, O’Mahony MS, Savva GM, et al. : Inflammation and frailty measures in older people. J Cell Mol Med 13:3103-3109, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Barzilay JI, Blaum C, Moore T, et al. : Insulin resistance and inflammation as precursors of frailty: The Cardiovascular Health Study. Arch Intern Med 167:635-641, 2007 [DOI] [PubMed] [Google Scholar]
- 82.Darvin K, Randolph A, Ovalles S, et al. : Plasma protein biomarkers of the geriatric syndrome of frailty. J Gerontol A Biol Sci Med Sci 69:182-186, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Visser M, Pahor M, Taaffe DR, et al. : Relationship of interleukin-6 and tumor necrosis factor-alpha with muscle mass and muscle strength in elderly men and women: The Health ABC Study. J Gerontol A Biol Sci Med Sci 57:M326-M332, 2002 [DOI] [PubMed] [Google Scholar]
- 84.Schaap LA, Pluijm SM, Deeg DJ, et al: Inflammatory markers and loss of muscle mass (sarcopenia) and strength. Am J Med 119:526.e9-17, 2006. [DOI] [PubMed]
- 85.Taaffe DR, Harris TB, Ferrucci L, et al. : Cross-sectional and prospective relationships of interleukin-6 and C-reactive protein with physical performance in elderly persons: MacArthur studies of successful aging. J Gerontol A Biol Sci Med Sci 55:M709-M715, 2000 [DOI] [PubMed] [Google Scholar]
- 86.Taekema DG, Westendorp RG, Frölich M, et al. : High innate production capacity of tumor necrosis factor-alpha and decline of handgrip strength in old age. Mech Ageing Dev 128:517-521, 2007 [DOI] [PubMed] [Google Scholar]
- 87.Van Epps P, Oswald D, Higgins PA, et al. : Frailty has a stronger association with inflammation than age in older veterans. Immun Ageing 13:27, 2016. [Erratum: Immun Ageing 14:4, 2017] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Collerton J, Martin-Ruiz C, Davies K, et al. : Frailty and the role of inflammation, immunosenescence and cellular ageing in the very old: Cross-sectional findings from the Newcastle 85+ Study. Mech Ageing Dev 133:456-466, 2012 [DOI] [PubMed] [Google Scholar]
- 89.Gale CR, Baylis D, Cooper C, et al. : Inflammatory markers and incident frailty in men and women: The English Longitudinal Study of Ageing. Age (Dordr) 35:2493-2501, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Barron E, Lara J, White M, et al. : Blood-borne biomarkers of mortality risk: Systematic review of cohort studies. PLoS One 10:e0127550, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Needham BL, Rehkopf D, Adler N, et al. : Leukocyte telomere length and mortality in the National Health and Nutrition Examination Survey, 1999-2002. Epidemiology 26:528-535, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Rode L, Nordestgaard BG, Bojesen SE: Peripheral blood leukocyte telomere length and mortality among 64,637 individuals from the general population. J Natl Cancer Inst 107:djv074, 2015 [DOI] [PubMed] [Google Scholar]
- 93.Proctor MJ, McMillan DC, Horgan PG, et al. : Systemic inflammation predicts all-cause mortality: A Glasgow inflammation outcome study. PLoS One 10:e0116206, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Alfano CM, Peng J, Andridge RR, et al. : Inflammatory cytokines and comorbidity development in breast cancer survivors versus noncancer controls: Evidence for accelerated aging? J Clin Oncol 35:149-156, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Cheung YT, Ng T, Shwe M, et al. : Association of proinflammatory cytokines and chemotherapy-associated cognitive impairment in breast cancer patients: A multi-centered, prospective, cohort study. Ann Oncol 26:1446-1451, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Lyon DE, Cohen R, Chen H, et al. : Relationship of systemic cytokine concentrations to cognitive function over two years in women with early stage breast cancer. J Neuroimmunol 301:74-82, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Orre IJ, Reinertsen KV, Aukrust P, et al. : Higher levels of fatigue are associated with higher CRP levels in disease-free breast cancer survivors. J Psychosom Res 71:136-141, 2011 [DOI] [PubMed] [Google Scholar]
- 98.Cheung YT, Brinkman TM, Mulrooney DA, et al. : Impact of sleep, fatigue, and systemic inflammation on neurocognitive and behavioral outcomes in long-term survivors of childhood acute lymphoblastic leukemia. Cancer 123:3410-3419, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ariffin H, Azanan MS, Abd Ghafar SS, et al. : Young adult survivors of childhood acute lymphoblastic leukemia show evidence of chronic inflammation and cellular aging. Cancer 123:4207-4214, 2017 [DOI] [PubMed] [Google Scholar]
- 100.Vatanen A, Hou M, Huang T, et al. : Clinical and biological markers of premature aging after autologous SCT in childhood cancer. Bone Marrow Transplant 52:600-605, 2017 [DOI] [PubMed] [Google Scholar]
- 101.Goldberg AD, Allis CD, Bernstein E: Epigenetics: A landscape takes shape. Cell 128:635-638, 2007 [DOI] [PubMed] [Google Scholar]
- 102.Schübeler D: Function and information content of DNA methylation. Nature 517:321-326, 2015 [DOI] [PubMed] [Google Scholar]
- 103.Day K, Waite LL, Thalacker-Mercer A, et al. : Differential DNA methylation with age displays both common and dynamic features across human tissues that are influenced by CpG landscape. Genome Biol 14:R102, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Benayoun BA, Pollina EA, Brunet A: Epigenetic regulation of ageing: Linking environmental inputs to genomic stability. Nat Rev Mol Cell Biol 16:593-610, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Sun D, Luo M, Jeong M, et al. : Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14:673-688, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Horvath S: DNA methylation age of human tissues and cell types. Genome Biol 14:R115, 2013 [Erratum: Genome Biol 16:96, 2015] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Horsburgh S, Robson-Ansley P, Adams R, et al. : Exercise and inflammation-related epigenetic modifications: Focus on DNA methylation. Exerc Immunol Rev 21:26-41, 2015 [PubMed] [Google Scholar]
- 108.Dong Y, Huang Y, Gutin B, et al. : Associations between global DNA methylation and telomere length in healthy adolescents. Sci Rep 7:4210, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Wong JY, De Vivo I, Lin X, et al. : The association between global DNA methylation and telomere length in a longitudinal study of boilermakers. Genet Epidemiol 38:254-264, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Marioni RE, Shah S, McRae AF, et al. : DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol 16:25, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Antwih DA, Gabbara KM, Lancaster WD, et al. : Radiation-induced epigenetic DNA methylation modification of radiation-response pathways. Epigenetics 8:839-848, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kovalchuk A, Rodriguez-Juarez R, Ilnytskyy Y, et al. : Sex-specific effects of cytotoxic chemotherapy agents cyclophosphamide and mitomycin C on gene expression, oxidative DNA damage, and epigenetic alterations in the prefrontal cortex and hippocampus - an aging connection. Aging (Albany NY) 8:697-711, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Brown AL, Foster KL, Lupo PJ, et al. : DNA methylation of a novel PAK4 locus influences ototoxicity susceptibility following cisplatin and radiation therapy for pediatric embryonal tumors. Neuro-oncol 19:1372-1379, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Vijg J: Somatic mutations, genome mosaicism, cancer and aging. Curr Opin Genet Dev 26:141-149, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Blokzijl F, de Ligt J, Jager M, et al. : Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538:260-264, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Busque L, Patel JP, Figueroa ME, et al. : Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat Genet 44:1179-1181, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Genovese G, Kähler AK, Handsaker RE, et al. : Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 371:2477-2487, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Jaiswal S, Fontanillas P, Flannick J, et al. : Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 371:2488-2498, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Collord G, Park N, Podestà M, et al. : Clonal haematopoiesis is not prevalent in survivors of childhood cancer. Br J Haematol, 2017. [epub ahead of print on April 3, 2017] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Hasty P, Campisi J, Hoeijmakers J, et al. : Aging and genome maintenance: Lessons from the mouse? Science 299:1355-1359, 2003 [DOI] [PubMed] [Google Scholar]
- 121.Vijg J, Calder RB: Transcripts of aging. Trends Genet 20:221-224, 2004 [DOI] [PubMed] [Google Scholar]
- 122.Yakes FM, Van Houten B: Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci USA 94:514-519, 1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Carew JS, Zhou Y, Albitar M, et al. : Mitochondrial DNA mutations in primary leukemia cells after chemotherapy: Clinical significance and therapeutic implications. Leukemia 17:1437-1447, 2003 [DOI] [PubMed] [Google Scholar]
- 124.Garrido N, Pérez-Martos A, Faro M, et al. : Cisplatin-mediated impairment of mitochondrial DNA metabolism inversely correlates with glutathione levels. Biochem J 414:93-102, 2008 [DOI] [PubMed] [Google Scholar]
- 125.Podratz JL, Knight AM, Ta LE, et al. : Cisplatin induced mitochondrial DNA damage in dorsal root ganglion neurons. Neurobiol Dis 41:661-668, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Adachi K, Fujiura Y, Mayumi F, et al. : A deletion of mitochondrial DNA in murine doxorubicin-induced cardiotoxicity. Biochem Biophys Res Commun 195:945-951, 1993 [DOI] [PubMed] [Google Scholar]
- 127.Lebrecht D, Setzer B, Ketelsen UP, et al. : Time-dependent and tissue-specific accumulation of mtDNA and respiratory chain defects in chronic doxorubicin cardiomyopathy. Circulation 108:2423-2429, 2003 [DOI] [PubMed] [Google Scholar]
- 128.Kujoth GC, Hiona A, Pugh TD, et al. : Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 309:481-484, 2005 [DOI] [PubMed] [Google Scholar]
- 129.Trifunovic A, Wredenberg A, Falkenberg M, et al. : Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 429:417-423, 2004 [DOI] [PubMed] [Google Scholar]
- 130.Reznik E, Miller ML, Şenbabaoğlu Y, et al. : Mitochondrial DNA copy number variation across human cancers. eLife 5:e10769, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zong WX, Rabinowitz JD, White E: Mitochondria and cancer. Mol Cell 61:667-676, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Pinto M, Moraes CT: Mechanisms linking mtDNA damage and aging. Free Radic Biol Med 85:250-258, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Gaziev AI, Abdullaev S, Podlutsky A: Mitochondrial function and mitochondrial DNA maintenance with advancing age. Biogerontology 15:417-438, 2014 [DOI] [PubMed] [Google Scholar]
- 134.D’Erchia AM, Atlante A, Gadaleta G, et al. : Tissue-specific mtDNA abundance from exome data and its correlation with mitochondrial transcription, mass and respiratory activity. Mitochondrion 20:13-21, 2015 [DOI] [PubMed] [Google Scholar]
- 135.Toyama EQ, Herzig S, Courchet J, et al. : Metabolism. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science 351:275-281, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Taylor RW, Turnbull DM: Mitochondrial DNA mutations in human disease. Nat Rev Genet 6:389-402, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Mkaouar-Rebai E, Felhi R, Tabebi M, et al. : Mitochondrial DNA triplication and punctual mutations in patients with mitochondrial neuromuscular disorders. Biochem Biophys Res Commun 473:578-585, 2016 [DOI] [PubMed] [Google Scholar]
- 138.Chen H, Vermulst M, Wang YE, et al. : Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141:280-289, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.García-Prat L, Martínez-Vicente M, Perdiguero E, et al. : Autophagy maintains stemness by preventing senescence. Nature 529:37-42, 2016 [DOI] [PubMed] [Google Scholar]
- 140.Kang C, Ji LL: PGC-1α overexpression via local transfection attenuates mitophagy pathway in muscle disuse atrophy. Free Radic Biol Med 93:32-40, 2016 [DOI] [PubMed] [Google Scholar]
- 141.Romanello V, Sandri M: Mitochondrial quality control and muscle mass maintenance. Front Physiol 6:422, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Rygiel KA, Picard M, Turnbull DM: The ageing neuromuscular system and sarcopenia: A mitochondrial perspective. J Physiol 594:4499-4512, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Sunitha B, Gayathri N, Kumar M, et al. : Muscle biopsies from human muscle diseases with myopathic pathology reveal common alterations in mitochondrial function. J Neurochem 138:174-191, 2016 [DOI] [PubMed] [Google Scholar]
- 144.Gondim OS, de Camargo VT, Gutierrez FA, et al. : Benefits of regular exercise on inflammatory and cardiovascular risk markers in normal weight, overweight and obese adults. PLoS One 10:e0140596, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Beavers KM, Ambrosius WT, Nicklas BJ, et al. : Independent and combined effects of physical activity and weight loss on inflammatory biomarkers in overweight and obese older adults. J Am Geriatr Soc 61:1089-1094, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Imayama I, Ulrich CM, Alfano CM, et al. : Effects of a caloric restriction weight loss diet and exercise on inflammatory biomarkers in overweight/obese postmenopausal women: A randomized controlled trial. Cancer Res 72:2314-2326, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Kasapis C, Thompson PD: The effects of physical activity on serum C-reactive protein and inflammatory markers: A systematic review. J Am Coll Cardiol 45:1563-1569, 2005 [DOI] [PubMed] [Google Scholar]
- 148.Mundstock E, Zatti H, Louzada FM, et al. : Effects of physical activity in telomere length: Systematic review and meta-analysis. Ageing Res Rev 22:72-80, 2015 [DOI] [PubMed] [Google Scholar]
- 149.Lim S, Kim SK, Park KS, et al. : Effect of exercise on the mitochondrial DNA content of peripheral blood in healthy women. Eur J Appl Physiol 82:407-412, 2000 [DOI] [PubMed] [Google Scholar]
- 150.Beulertz J, Prokop A, Rustler V, et al. : Effects of a 6-month, group-based, therapeutic exercise program for childhood cancer outpatients on motor performance, level of activity, and quality of life. Pediatr Blood Cancer 63:127-132, 2016 [DOI] [PubMed] [Google Scholar]
- 151.Braam KI, van der Torre P, Takken T, et al. : Physical exercise training interventions for children and young adults during and after treatment for childhood cancer. Cochrane Database Syst Rev 3:CD008796, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Esbenshade AJ, Friedman DL, Smith WA, et al. : Feasibility and initial effectiveness of home exercise during maintenance therapy for childhood acute lymphoblastic leukemia. Pediatr Phys Ther 26:301-307, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Madzima TA, Ormsbee MJ, Schleicher EA, et al. : Effects of resistance training and protein supplementation in breast cancer survivors. Med Sci Sports Exerc 49:1283-1292, 2017 [DOI] [PubMed] [Google Scholar]
- 154.Padilha CS, Marinello PC, Galvão DA, et al. : Evaluation of resistance training to improve muscular strength and body composition in cancer patients undergoing neoadjuvant and adjuvant therapy: A meta-analysis. J Cancer Surviv 11:339-349, 2017 [DOI] [PubMed] [Google Scholar]
- 155.Smith WA, Ness KK, Joshi V, et al. : Exercise training in childhood cancer survivors with subclinical cardiomyopathy who were treated with anthracyclines. Pediatr Blood Cancer 10.1002/pbc.24850 [epub ahead of print on November 6, 2013] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Adams RN, Mosher CE, Blair CK, et al. : Cancer survivors’ uptake and adherence in diet and exercise intervention trials: An integrative data analysis. Cancer 121:77-83, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Kirkland JL, Tchkonia T: Clinical strategies and animal models for developing senolytic agents. Exp Gerontol 68:19-25, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Zhu Y, Doornebal EJ, Pirtskhalava T, et al: New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging (Albany NY) 9:955-963, 2017. [DOI] [PMC free article] [PubMed]
- 159.Zhu Y, Tchkonia T, Pirtskhalava T, et al. : The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 14:644-658, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Yosef R, Pilpel N, Tokarsky-Amiel R, et al. : Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun 7:11190, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Fuhrmann-Stroissnigg H, Ling YY, Zhao J, et al. : Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun 8:422, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Lehmann M, Korfei M, Mutze K, et al. : Senolytic drugs target alveolar epithelial cell function and attenuate experimental lung fibrosis ex vivo. Eur Respir J 50:1602367, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Zhu H, Guariglia S, Li W, et al. : Role of extracellular signal-regulated kinase 5 in adipocyte signaling. J Biol Chem 289:6311-6322, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]