Abstract
Purpose of review.
To summarizes the literature on cellular senescence and frailty in solid-organ transplantation and highlight the emerging role of senotherapeutics as a treatment for cellular senescence.
Recent findings.
Solid-organ transplant patients are aging. Many factors contribute to aging acceleration in this population, including cellular senescence. Senescent cells accumulate in tissues and secrete proinflammatory and profibrotic proteins which result in tissue damage. Cellular senescence contributes to age-related diseases and frailty. Our understanding of the role cellular senescence plays in transplant-specific complications such as allograft immunogenicity and infections is expanding. Promising treatments, including senolytics, senomorphics, cell-based regenerative therapies, and behavioral interventions, may reduce cellular senescence abundance and frailty in patients with solid-organ transplants.
Summary.
Cellular senescence and frailty contribute to adverse outcomes in solid-organ transplantation. Continued pursuit of understanding the role cellular senescence plays in transplantation may lead to improved senotherapeutic approaches and better graft and patient outcomes.
Keywords: Frailty, Senescence, Biomarkers, Aging, Transplantation, Senolytics
Introduction.
The world is aging. According to the World Health Organization, the number of people aged 60 or over is expected to grow to 2 billion, or 22% of the global population, by 2050.1 With increased aging comes increased disease burden. Age is a risk factor for numerous chronic illnesses, such as nonalcoholic fatty liver disease,2 chronic obstructive pulmonary disease,3 heart failure,4 and end-stage kidney disease.5 Given that many patients with these diseases progress to needing organ replacement therapy, this epidemic of aging and age-related comorbidities directly impacts the field of transplantation where the average age and of liver,6 lung,7 heart,8 kidney9 transplant recipients is steadily increasing.
Our understanding of the aging process has evolved dramatically over the past fifty years. We now know that aging has many pathophysiologic drivers, including shortening of telomeres, genomic instability, stem cell exhaustion, and cellular senescence abundance.10 Cellular senescence refers to the process in which cells enter a state of irreversible growth arrest in response to perturbations, including DNA damage and oxidative stress.11 Senescent cells are resistant to apoptosis,12 accumulate with age,13 and contribute to tissue dysfunction, age-related disease,14 and frailty.15 The purpose of this review is to outline recent advances in our understanding of cellular senescence abundance and frailty in solid-organ transplantation and discuss potential therapeutics.
Cellular Senescence.
Although senescent cells have may identifying characteristics, they display significant heterogeneity, and no one single marker identifies them all.14 Morphologically, senescent cells tend to be flatter and larger than healthy cells and are often multinucleated. Histochemically, they stain for senescence-associated β-galactosidase (SA-β-gal), a lysosomal enzyme that catalyzes the hydrolysis of carbohydrates. They exhibit increased levels of DNA damage (e.g. p53)16 and cell cycle inhibitors (e.g. p16 and p21).14 Senescent cells can also be indirectly identified by the products they secrete into the circulation. Senescent cells upregulate production of various cytokines, growth factors, chemokines, and other proteins referred to as the senescence-associated secretory phenotype (SASP), levels of which can be detected in the blood (Table 1).17 In a recent study, Saul et al.18 sought to develop a more accurate way of identifying senescent cells. Their team developed a panel of genes capable of identifying senescent cells across tissues and species which may be useful in future studies evaluating senescent cell burden.
Table 1.
Examples of Senescence-Associated Secretory Phenotype (SASP) components (adapted from Kirkland et al.22)
Cytokines |
Interleukin-6 (IL-6) Tumor necrosis factor-α (TNF-α) Transforming growth factor-β (TGF-β) Vascular endothelial growth factor-A (VEGF-A) Platelet-derived growth factor-AA (PDGF-AA) |
Chemokines |
Monocyte chemoattract protein-1 (MCP-1/CCL-2) Chemokine (C-X-C motif) ligand 2 (CXCL2) Macrophage inflammatory protein-1α (MIP-1α) Eotaxin |
Extracellular matrix proteases and remodeling factors |
Matrix metalloproteinases (MMPs) Tissue inhibitor of metallopeptidase (TIMPs) |
Receptors |
Tumor necrosis factor receptor-1 (sTNFR1) Tumor necrosis factor receptor-2 (sTNFR2) Urokinase plasminogen activator receptor (uPAR) |
Reactive metabolites |
Reactive oxygen species |
Senescent cell accumulation contributes to aging and chronic disease throughout the body.14 Senescent cells accrue in diseased tissue, such as the lungs in chronic obstructive pulmonary disease, adipose and kidney tissue in type 2 diabetes mellitus, and the heart in cardiovascular disease. Cellular senescence is abundant in the transplanted organ19 and reduces allograft survival in mouse models.20 Senescent cells induce the formation of other senescent cells locally and systemically via the SASP and contribute to tissue dysfunction.14 SASP proteins, such as interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and interferon gamma (IFN-γ), are proinflammatory and can induce matrix remodelling, fibrosis, and apoptosis.21,22 These cytokines and chemokines attract and activate immune cells within tissue.21 The resulting low-grade, chronic inflammation associated with the SASP is often referred to as ‘inflamm-aging.’23 In adipose tissue, senescent cells can decrease replication of preadipocytes and decrease insulin sensitivity.24 In the liver, they can induce hepatic steatosis.25 In bone, they can lead to decreased fracture repair.26 In the immune system, they can cause dysregulation by increasing the number of immunosuppressive cells (i.e. regulatory T cells) and decreasing the efficacy of CD4 and CD8 T cells, NK cells, and B cells, a process referred to as immunosenescence.27,28 Senescent cell abundance can also promote malignant transformation in tissues throughout the body.29 In organ transplantation, senescent cells accumulate due to immune activation and reduced immune clearance from ongoing immunosuppressive therapy.30–32
Cellular senescence and frailty.
Cellular senescence has emerged as a potential driver of frailty. The concept of frailty was initially described by Linda Fried and colleagues. In her landmark paper published in 2001, Fried defined frailty as a syndrome of decreased physiologic reserve and increased susceptibility to stress.33 The Fried frailty phenotype, also known as the physical frailty phenotype, consists of five criteria: slow gait speed, unintentional weight loss, exhaustion, low self-reported physical activity, and weak grip strength. Patients meeting three or more of the criteria are classified as frail, while patients meeting one or two of the criteria are classified as pre-frail.33 Other measures of frailty include tests of physical performance, the cumulative deficit model, and surveys (Table 2).33 Emerging digital measures of frailty include pendant and upper extremity sensors and accelerometers.34–37 These digital measures of frailty have shown to predict falls,38, functional mobility,39 and cardiovascular events after vascular surgery.40 These digital measures have the advantage of being able to be assessed using telemedicine, monitored remotely, and do not require weight bearing.41,42
Table 2.
Commonly utilized measures of frailty in solid-organ transplantation
Category | Description | Examples |
---|---|---|
Performance-based | Tests of physical performance | |
Phenotype model | Combination of tests of physical performance and self-report |
|
Cumulative deficit model | Comorbidities, disabilities, and symptoms determined from the medical record | |
Self-report | Questions answered by the patient | |
Provider-based | Assessed by healthcare provider |
Frailty is associated with numerous adverse health outcomes across patient populations, including disability, falls, hospitalization, and death.43,44 Frailty reflects derangements in multiple physiologic systems, including the cardiopulmonary system, the nervous system, and the musculoskeletal system.45 Cellular senescence is believed to contribute to the muscle dysfunction observed in frailty in several different ways. Senescent cells accumulate in muscle fibers,46 reduce muscle hypertrophy after exercise,47 contribute to muscle atrophy and decreased contractility,48 and lead to sarcopenia.49 In support of the causal role cellular senescence plays in muscle dysfunction and frailty, studies have shown that circulating levels of SASP are associated with frailty,50 including worse grip strength, gait speed, chair rise time, and balance.51 In fact, transplanting senescent cells into mice has been shown to decrease hanging endurance and gait speed.48
Treating cellular senescence and frailty.
The growing understanding of adverse implications of cellular senescence has led to extensive research on how best to halt or reverse the process. Promising pharmacologic treatments, termed senotherapeutics, are emerging. Senotherapeutics include senolytics which selectively deplete senescent cells and senomorphics which inhibit the SASP. One of the first studied senolytics involves a combination of dasatinib (a tyrosine kinase inhibitor) plus quercetin (a naturally occurring flavanoid).22 Together dasatinib and quercetin have been shown to improve cardiac ejection fraction and pulmonary function in older mice.52,53 In the first-in-human study, intermittent oral administration of dasatinib plus quercetin in patients with idiopathic pulmonary fibrosis, a cellular senescence-associated disease, was associated with improved 6-minute walk distance, gait speed, and chair stand time.54 The first evidence that senolytics cleared senescent cell abundance in humans was demonstrated in a phase I trial applying dasatinib plus quercetin in older patients with diabetic kidney disease.55 In addition to reducing senescent cell burden, senolytics also improve grip strength, muscle endurance, and exercise capacity in mice.48
Senomorphic drugs that suppress the SASP without eliminating senescent cells require continuous administration and are another potential avenue of therapy.22 One such drug, rapamycin, an mTOR inhibitor, is commonly used as an immunosuppressive agent in the transplant population. Rapamycin has been shown to improve lifespan in mice.56,57 Moreover, Hoff et al.58 demonstrated in a rat model of kidney transplantation that early post-transplant rapamycin therapy prevented accumulation of p16 positive cells in kidney compartments (tubules, interstitium, and glomeruli). However, the effects of rapamycin on cellular senescence or SASP in solid-organ transplant patients is unclear. Metformin, an antidiabetic medication, is another promising SASP suppressor and the most commonly studied senomorphic.59,60 Similar to senolytics, SASP suppressors have been shown to improve muscle performance and physical function in animal models.61,62
A recent addition to the armamentarium of senotherapeutic agents are stem cells and the extracellular vesicles that they produce. Mesenchymal stem cell derived-extracellular vesicles reduce senescence burden and extend lifespan in aging mice.63 Another study by Sahu et al.64 demonstrated the anti-geronic effect of extracellular vesicles. They showed that extracellular vesicles within the serum of young animals contribute to the regeneration of aged skeletal muscle in older animals.
The study of senotherapeutics in humans and particularly in solid organ transplant recipients remains in its infancy. Special consideration must be given to the safety of using these agents in immunocompromised patients. For example, adverse effects of the senolytic drug dasatinib include myelosuppression and infection.65,66 The use of senotherapeutics could promote malignant transformation given that the SASP has been shown to play a physiological role in tumor suppression.14 This issue is especially relevant to solid-organ transplant recipients who are already at an increased risk of malignancy.
In addition to pharmacologic therapy, behavioral interventions can play a role in treating cellular senescence and frailty. The most commonly studied behavioral intervention is exercise.15,67 Englund et al. demonstrated that a 12-week exercise intervention not only improved grip strength, sit-to-stand time, and the timed up and go test but also decreased expression of the senescent markers (i.e. p16 and p21) and decreased circulating levels of SASP.68 Furthermore, several pilot studies have examined the impact of exercise on frailty in patients with advanced liver and kidney disease, including transplant candidates. These studies have demonstrated that exercise is safe and associated with improvements in frailty parameters.15,69–72 Further studies examining the impact of prehabilitation, or exercise prior to surgery, on quality of life, healthcare utilization, and mortality are needed.69
Cellular senescence, frailty, and transplantation.
Cellular senescence is believed not only to contribute to frailty in transplantation but also to other adverse outcomes such as organ dysfunction, decreased quality of life, and death (Figure 1). Our emerging understanding of the transplant-specific impact of cellular senescence will hopefully lead to better interventions capable of improving transplant outcomes. The following paragraphs will highlight data regarding frailty and cellular senescence in liver, lung, heart, and kidney transplantation.
Figure 1.
Potential impact of cellular senescence in solid-organ transplantation
In liver transplantation, frailty associated with waitlist mortality,73,74 longer hospital length of stay,75 rehospitalization after transplantation,76 acute cellular rejection,77 worse post-transplant global functional and physical health,78 and post-transplant mortality.76 A recent study examined markers of cellular senescence in liver allograft biopsies with rejection and found increased expression of senescent cell markers, including p16 and p21, in biopsies showing late acute cellular rejection and chronic rejection.79
In lung transplantation, frailty has been associated with increased mortality in transplant candidates,80 post-operative delirium,81 rehospitalizations,82,83 lower quality of life,84 disability,85 and post-transplant mortality. Few studies have examined cellular senescence in patients with lung transplants. However, senescent cells have been found in the airway epithelium of lung transplants.19
In heart transplantation, frailty has been associated with lower rates of waitlisting, transplantation, and mortality in transplant candidates.86 Candidates who are frail are also more likely to have depression and anemia and require more frequent hospitalizations.87 Lastly, frailty has been associated with longer hospital length of stay and mortality after heart transplantation.88 Cellular senescence is now being recognized as a potential explanation for why older allografts are associated with more inflammation, alloreactivity, and adverse outcomes than younger allografts.89,90 Specifically, a study by Iske et al.91 demonstrated that cardiac allografts from older donors contain higher numbers of senescent cells. These senescent cells produce cell-free DNA that promotes immunogenicity after heart transplantation. Promisingly, treating older mice with the senolytics dasatinib and quercetin led to fewer senescent cells within the allograft, decreased levels of cell-free DNA, and better graft survival after subsequent cardiac transplantation.91 Research into whether older donor organs can transfer senescent cells into the recipient is ongoing.92
In kidney transplantation, frailty has been associated with many adverse outcomes both before and after transplantation.15 Specifically, frailty has been associated with waitlist mortality,93,94 delayed graft function,95 delirium,96 longer hospital length of stay,97,98 and rehospitalizations.97,99 It has also been associated with immunosuppression intolerance,100 cognitive decline,101 lower quality of life,102 and post-transplant mortality.103
Numerous studies have investigated the impact of cellular senescence in kidney transplantation. First, cellular senescence may contribute to mortality. In kidney transplant candidates, SASP components, including IL-6, TNF-α, and tumor necrosis factor receptor-1 (sTNFR1) were associated with increased risk of waitlist and post-transplant mortality.94,104 Second, cellular senescence may contribute to donor organ quality. An animal study by He et al.105 found that transplanted kidneys from older donors have different immunophenotypes than kidneys from younger donors, including increased levels of CD8 T cells. Older allografts also are associated with more inflammatory cytokine production during the first week post-transplant. However, pretreating older murine donors with senolytics prior to organ harvest appears to reduce expression of inflammatory cytokines in the older allografts suggesting a potential role for senolytics in improving donor organ function similar to the above studies in heart transplantation. Third, cellular senescence may adversely impact kidney transplant function. Transplanting senescent cells into mice has been shown to be associated with increased creatinine levels, renal tissue hypoxia, and fibrosis.106 Fourth, cellular senescence may be involved in renal allograft rejection. Extracellular vesicles (miRNA) from kidney transplant recipients with antibody-mediated rejection have been shown to induce renal tubular senescence in vitro via complement activation and fibrosis.107 Lastly, immunosenescence may increase infection risk in kidney transplant recipients. T cell senescence and impaired-cytomegalovirus (CMV) specific response have been associated with increased vulnerability to infections after kidney transplantation.108 Fortunately, a recent study demonstrated that early post-transplant administration of rapamycin protects against cellular senescence in a murine model of kidney transplantation.58
Conclusions.
Our understanding of the role cellular senescence plays in aging and frailty is expanding. The study of frailty is complicated by the numerous ways used to measure it. Digital measurements of frailty may allow for better remote diagnosis and monitoring of frailty. Senotherapeutics and exercise have been shown to decrease cellular senescence abundance and improve frailty in non-transplant patients. While numerous studies have highlighted the relationship between frailty and adverse health outcomes before and after solid-organ transplantation, less is known about the role and impact of cellular senescence in transplant outcomes. Recent studies suggest that cellular senescence may play a role in the alloreactivity seen with older allografts and in rejection. Future research examining how to safely and effectively reverse cellular senescence and frailty in solid-organ transplant patients before and after transplantation is needed. As donor age boundaries are pushed, further research into the promising role of senotherapeutics in expanding the organ pool by improving the function of older organs would also be beneficial.109,110
Disclosure.
E.C.L. and L.J.H. are supported in part by the National Institute of Diabetes and Digestive and Kidney Diseases (DK 123313, DK 109134, DK 123492). C.C.K. is supported in part by the National Heart, Lung, and Blood Institute (HL 128859, HL 158811).
Footnotes
Conflict of Interest. The authors declare that they have no conflict of interest.
Human and Animal Rights. All reported studies/experiments with human or animal subjects performed by the authors have been previously published and complied with all applicable ethical standards (including the Helsinki declaration and its amendments, institutional/national research committee standards, and international/national/institutional guidelines).
References
- 1.10 Facts on Ageing and Health. Accessed December 21, 2022, from https://www.who.int/news-room/fact-sheets/detail/10-facts-on-ageing-and-health.
- 2.He Y et al. Emerging role of aging in the progression of NAFLD to HCC. Ageing Res Rev 84, 101833, doi: 10.1016/j.arr.2022.101833 (2023). [DOI] [PubMed] [Google Scholar]
- 3.MacNee W, Rabinovich RA & Choudhury G Ageing and the border between health and disease. Eur Respir J 44, 1332–1352, doi: 10.1183/09031936.00134014 (2014). [DOI] [PubMed] [Google Scholar]
- 4.Bleumink GS et al. Quantifying the heart failure epidemic: prevalence, incidence rate, lifetime risk and prognosis of heart failure The Rotterdam Study. Eur Heart J 25, 1614–1619, doi: 10.1016/j.ehj.2004.06.038 (2004). [DOI] [PubMed] [Google Scholar]
- 5.McCullough KP, Morgenstern H, Saran R, Herman WH & Robinson BM Projecting ESRD Incidence and Prevalence in the United States through 2030. J Am Soc Nephrol 30, 127–135, doi: 10.1681/ASN.2018050531 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Durand F et al. Age and liver transplantation. J Hepatol 70, 745–758, doi: 10.1016/j.jhep.2018.12.009 (2019). [DOI] [PubMed] [Google Scholar]
- 7●.Schaenman JM et al. Frailty and aging-associated syndromes in lung transplant candidates and recipients. Am J Transplant 21, 2018–2024, doi: 10.1111/ajt.16439 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]; A review of recent advances in our understanding of frailty in lung transplantation.
- 8.Colvin M et al. OPTN/SRTR 2016 Annual Data Report: Heart. Am J Transplant 18 Suppl 1, 291–362, doi: 10.1111/ajt.14561 (2018). [DOI] [PubMed] [Google Scholar]
- 9.Lentine KL et al. OPTN/SRTR 2020 Annual Data Report: Kidney. Am J Transplant 22 Suppl 2, 21–136, doi: 10.1111/ajt.16982 (2022). [DOI] [PubMed] [Google Scholar]
- 10.Li Z et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology 22, 165–187, doi: 10.1007/s10522-021-09910-5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Rhinn M, Ritschka B & Keyes WM Cellular senescence in development, regeneration and disease. Development 146, doi: 10.1242/dev.151837 (2019). [DOI] [PubMed] [Google Scholar]
- 12.Yosef R et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun 7, 11190, doi: 10.1038/ncomms11190 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Tchkonia T, Palmer AK & Kirkland JL New Horizons: Novel Approaches to Enhance Healthspan Through Targeting Cellular Senescence and Related Aging Mechanisms. J Clin Endocrinol Metab 106, e1481–e1487, doi: 10.1210/clinem/dgaa728 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14●●.Huang W, Hickson LJ, Eirin A, Kirkland JL & Lerman LO Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol 18, 611–627, doi: 10.1038/s41581-022-00601-z (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; An overview of cellular senescence and its impact on the kidney.
- 15.Lorenz EC et al. Frailty in CKD and Transplantation. Kidney Int Rep 6, 2270–2280, doi: 10.1016/j.ekir.2021.05.025 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sharpless NE & Sherr CJ Forging a signature of in vivo senescence. Nat Rev Cancer 15, 397–408, doi: 10.1038/nrc3960 (2015). [DOI] [PubMed] [Google Scholar]
- 17.LeBrasseur NK et al. Identifying Biomarkers for Biological Age: Geroscience and the ICFSR Task Force. J Frailty Aging 10, 196–201, doi: 10.14283/jfa.2021.5 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Saul D et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat Commun 13, 4827, doi: 10.1038/s41467-022-32552-1 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Parker SM et al. Airway epithelial cell senescence in the lung allograft. Am J Transplant 8, 1544–1549, doi: 10.1111/j.1600-6143.2008.02284.x (2008). [DOI] [PubMed] [Google Scholar]
- 20.Braun H et al. Cellular senescence limits regenerative capacity and allograft survival. J Am Soc Nephrol 23, 1467–1473, doi: 10.1681/ASN.2011100967 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Palmer AK, Tchkonia T & Kirkland JL Targeting cellular senescence in metabolic disease. Mol Metab 66, 101601, doi: 10.1016/j.molmet.2022.101601 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22●.Chaib S, Tchkonia T & Kirkland JL Cellular senescence and senolytics: the path to the clinic. Nat Med 28, 1556–1568, doi: 10.1038/s41591-022-01923-y (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; A review describing the role of senotherapeutics in treating cellular senescence and age-related diseases.
- 23.Franceschi C & Campisi J Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci 69 Suppl 1, S4–9, doi: 10.1093/gerona/glu057 (2014). [DOI] [PubMed] [Google Scholar]
- 24.Palmer AK et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell 18, e12950, doi: 10.1111/acel.12950 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ogrodnik M & Jurk D Senescence explains age- and obesity-related liver steatosis. Cell Stress 1, 70–72, doi: 10.15698/cst2017.10.108 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Saul D et al. Modulation of fracture healing by the transient accumulation of senescent cells. Elife 10, doi: 10.7554/eLife.69958 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Li MO, Wan YY, Sanjabi S, Robertson AK & Flavell RA Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol 24, 99–146, doi: 10.1146/annurev.immunol.24.021605.090737 (2006). [DOI] [PubMed] [Google Scholar]
- 28.Wang Y, Dong C, Han Y, Gu Z & Sun C Immunosenescence, aging and successful aging. Front Immunol 13, 942796, doi: 10.3389/fimmu.2022.942796 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Krtolica A, Parrinello S, Lockett S, Desprez PY & Campisi J Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci U S A 98, 12072–12077, doi: 10.1073/pnas.211053698 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Docherty MH, O’Sullivan ED, Bonventre JV & Ferenbach DA Cellular Senescence in the Kidney. J Am Soc Nephrol 30, 726–736, doi: 10.1681/ASN.2018121251 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hoffmann U et al. NK Cells of Kidney Transplant Recipients Display an Activated Phenotype that Is Influenced by Immunosuppression and Pathological Staging. PLoS One 10, e0132484, doi: 10.1371/journal.pone.0132484 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sagiv A & Krizhanovsky V Immunosurveillance of senescent cells: the bright side of the senescence program. Biogerontology 14, 617–628, doi: 10.1007/s10522-013-9473-0 (2013). [DOI] [PubMed] [Google Scholar]
- 33.Fried LP et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci 56, M146–156, doi: 10.1093/gerona/56.3.m146 (2001). [DOI] [PubMed] [Google Scholar]
- 34.Park C, Mishra R, Golledge J & Najafi B Digital Biomarkers of Physical Frailty and Frailty Phenotypes Using Sensor-Based Physical Activity and Machine Learning. Sensors (Basel) 21, doi: 10.3390/s21165289 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Park C et al. Digital Biomarker Representing Frailty Phenotypes: The Use of Machine Learning and Sensor-Based Sit-to-Stand Test. Sensors (Basel) 21, doi: 10.3390/s21093258 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Razjouyan J et al. Toward Using Wearables to Remotely Monitor Cognitive Frailty in Community-Living Older Adults: An Observational Study. Sensors (Basel) 20, doi: 10.3390/s20082218 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhou H et al. Harnessing Digital Health to Objectively Assess Functional Performance in Veterans with Chronic Obstructive Pulmonary Disease. Gerontology 68, 829–839, doi: 10.1159/000520401 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schwenk M et al. Sensor-derived physical activity parameters can predict future falls in people with dementia. Gerontology 60, 483–492, doi: 10.1159/000363136 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kang GE, Naik AD, Ghanta RK, Rosengart TK & Najafi B A Wrist-Worn Sensor-Derived Frailty Index Based on an Upper-Extremity Functional Test in Predicting Functional Mobility in Older Adults. Gerontology 67, 753–761, doi: 10.1159/000515078 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Najafi B et al. Association Between Wearable Device-Based Measures of Physical Frailty and Major Adverse Events Following Lower Extremity Revascularization. JAMA Netw Open 3, e2020161, doi: 10.1001/jamanetworkopen.2020.20161 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Park C et al. Toward Remote Assessment of Physical Frailty Using Sensor-based Sit-to-stand Test. J Surg Res 263, 130–139, doi: 10.1016/j.jss.2021.01.023 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zahiri M et al. Remote Physical Frailty Monitoring-The Application of Deep Learning-Based Image Processing in Tele-Health. IEEE Access 8, 219391–219399, doi: 10.1109/access.2020.3042451 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Fernando ME et al. Frailty in People with Chronic Limb Threatening Ischemia and Diabetes-Related Foot Ulcers: A Systematic Review. Ann Vasc Surg 89, 322–337, doi: 10.1016/j.avsg.2022.09.057 (2023). [DOI] [PubMed] [Google Scholar]
- 44.Mohler MJ, Fain MJ, Wertheimer AM, Najafi B & Nikolich-Zugich J The Frailty syndrome: clinical measurements and basic underpinnings in humans and animals. Exp Gerontol 54, 6–13, doi: 10.1016/j.exger.2014.01.024 (2014). [DOI] [PubMed] [Google Scholar]
- 45.Thillainadesan J, Scott IA & Le Couteur DG Frailty, a multisystem ageing syndrome. Age Ageing 49, 758–763, doi: 10.1093/ageing/afaa112 (2020). [DOI] [PubMed] [Google Scholar]
- 46.Englund DA, Zhang X, Aversa Z & LeBrasseur NK Skeletal muscle aging, cellular senescence, and senotherapeutics: Current knowledge and future directions. Mech Ageing Dev 200, 111595, doi: 10.1016/j.mad.2021.111595 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dungan CM et al. Senolytic treatment rescues blunted muscle hypertrophy in old mice. Geroscience 44, 1925–1940, doi: 10.1007/s11357-022-00542-2 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xu M et al. Senolytics improve physical function and increase lifespan in old age. Nat Med 24, 1246–1256, doi: 10.1038/s41591-018-0092-9 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Mankhong S et al. Experimental Models of Sarcopenia: Bridging Molecular Mechanism and Therapeutic Strategy. Cells 9, doi: 10.3390/cells9061385 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Schafer MJ et al. The senescence-associated secretome as an indicator of age and medical risk. JCI Insight 5, doi: 10.1172/jci.insight.133668 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Fielding RA et al. Associations between biomarkers of cellular senescence and physical function in humans: observations from the lifestyle interventions for elders (LIFE) study. Geroscience 44, 2757–2770, doi: 10.1007/s11357-022-00685-2 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ & Robbins PD The Clinical Potential of Senolytic Drugs. J Am Geriatr Soc 65, 2297–2301, doi: 10.1111/jgs.14969 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Schafer MJ et al. Cellular senescence mediates fibrotic pulmonary disease. Nat Commun 8, 14532, doi: 10.1038/ncomms14532 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Justice JN et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 40, 554–563, doi: 10.1016/j.ebiom.2018.12.052 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hickson LJ et al. Corrigendum to ‘Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease’ EBioMedicine 47 (2019) 446–456. EBioMedicine 52, 102595, doi: 10.1016/j.ebiom.2019.12.004 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Harrison DE et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460, 392–395, doi: 10.1038/nature08221 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lamming DW, Ye L, Sabatini DM & Baur JA Rapalogs and mTOR inhibitors as anti-aging therapeutics. J Clin Invest 123, 980–989, doi: 10.1172/JCI64099 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Hoff U et al. The mTOR inhibitor Rapamycin protects from premature cellular senescence early after experimental kidney transplantation. PLoS One 17, e0266319, doi: 10.1371/journal.pone.0266319 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hansel C et al. Metformin Protects against Radiation-Induced Acute Effects by Limiting Senescence of Bronchial-Epithelial Cells. Int J Mol Sci 22, doi: 10.3390/ijms22137064 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kulkarni AS, Gubbi S & Barzilai N Benefits of Metformin in Attenuating the Hallmarks of Aging. Cell Metab 32, 15–30, doi: 10.1016/j.cmet.2020.04.001 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Xu M et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife 4, e12997, doi: 10.7554/eLife.12997 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhang L et al. Novel small molecule inhibition of IKK/NF-kappaB activation reduces markers of senescence and improves healthspan in mouse models of aging. Aging Cell 20, e13486, doi: 10.1111/acel.13486 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Dorronsoro A et al. Mesenchymal stem cell-derived extracellular vesicles reduce senescence and extend health span in mouse models of aging. Aging Cell 20, e13337, doi: 10.1111/acel.13337 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sahu A et al. Regulation of aged skeletal muscle regeneration by circulating extracellular vesicles. Nat Aging 1, 1148–1161, doi: 10.1038/s43587-021-00143-2 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Brave M et al. Sprycel for chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia resistant to or intolerant of imatinib mesylate. Clin Cancer Res 14, 352–359, doi: 10.1158/1078-0432.CCR-07-4175 (2008). [DOI] [PubMed] [Google Scholar]
- 66.Rodriguez GH, Ahmed SI, Al-akhrass F, Rallapalli V & Safdar A Characteristics of, and risk factors for, infections in patients with cancer treated with dasatinib and a brief review of other complications. Leuk Lymphoma 53, 1530–1535, doi: 10.3109/10428194.2012.656626 (2012). [DOI] [PubMed] [Google Scholar]
- 67.Kidd T et al. What are the most effective interventions to improve physical performance in pre-frail and frail adults? A systematic review of randomised control trials. BMC Geriatr 19, 184, doi: 10.1186/s12877-019-1196-x (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Englund DA et al. Exercise reduces circulating biomarkers of cellular senescence in humans. Aging Cell 20, e13415, doi: 10.1111/acel.13415 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69●●.Kobashigawa J et al. Report from the American Society of Transplantation on frailty in solid organ transplantation. Am J Transplant 19, 984–994, doi: 10.1111/ajt.15198 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]; A consensus statement from the American Society of Transplantation on frailty in solid-organ transplantation.
- 70.Lai JC et al. A Multicenter Pilot Randomized Clinical Trial of a Home-Based Exercise Program for Patients With Cirrhosis: The Strength Training Intervention (STRIVE). Am J Gastroenterol 116, 717–722, doi: 10.14309/ajg.0000000000001113 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lorenz EC et al. Protocolized exercise improves frailty parameters and lower extremity impairment: A promising prehabilitation strategy for kidney transplant candidates. Clin Transplant, e14017, doi: 10.1111/ctr.14017 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.McAdams-DeMarco MA et al. Prehabilitation prior to kidney transplantation: Results from a pilot study. Clin Transplant 33, e13450, doi: 10.1111/ctr.13450 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73●.Haugen CE et al. Multicenter Study of Age, Frailty, and Waitlist Mortality Among Liver Transplant Candidates. Ann Surg 271, 1132–1136, doi: 10.1097/SLA.0000000000003207 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; A study examining the relationship between frailty and mortality in liver transplant candidates.
- 74.Lai JC et al. Frailty predicts waitlist mortality in liver transplant candidates. Am J Transplant 14, 1870–1879, doi: 10.1111/ajt.12762 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.DeMaria S Jr., Khromava M, Schiano TD, Lin HM & Kim S Standardized measures of frailty predict hospital length of stay following orthotopic liver transplantation for hepatocellular carcinoma. Clin Transplant 33, e13746, doi: 10.1111/ctr.13746 (2019). [DOI] [PubMed] [Google Scholar]
- 76.Ferreira AP & Machado MV Impact of pretransplant frailty and sarcopenia on the post-transplant prognosis of patients with liver cirrhosis: a systematic review. Eur J Gastroenterol Hepatol 33, e883–e897, doi: 10.1097/MEG.0000000000002291 (2021). [DOI] [PubMed] [Google Scholar]
- 77.Fozouni L et al. Frailty Is Associated With Increased Rates of Acute Cellular Rejection Within 3 Months After Liver Transplantation. Liver Transpl 26, 390–396, doi: 10.1002/lt.25669 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lai JC et al. Association of Frailty With Health-Related Quality of Life in Liver Transplant Recipients. JAMA Surg, doi: 10.1001/jamasurg.2022.6387 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rastogi A, Nigam N, Gayatri R, Bihari C & Pamecha V Biliary Epithelial Senescence in Cellular Rejection Following Live Donor Liver Transplantation. J Clin Exp Hepatol 12, 1420–1427, doi: 10.1016/j.jceh.2022.08.004 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Montgomery E et al. Frailty as a Predictor of Mortality in Patients With Interstitial Lung Disease Referred for Lung Transplantation. Transplantation 104, 864–872, doi: 10.1097/TP.0000000000002901 (2020). [DOI] [PubMed] [Google Scholar]
- 81.Dalton T, Snyder LD, Buckley EB & Smith PJ Pretransplant physical frailty, postoperative delirium, and short-term outcomes among older lung transplant recipients. Exp Gerontol 163, 111781, doi: 10.1016/j.exger.2022.111781 (2022). [DOI] [PubMed] [Google Scholar]
- 82.Courtwright AM et al. Causes, Preventability, and Cost of Unplanned Rehospitalizations Within 30 Days of Discharge After Lung Transplantation. Transplantation 102, 838–844, doi: 10.1097/TP.0000000000002101 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Swaminathan AC et al. Evaluation of Frailty Measures and Short-Term Outcomes after Lung Transplantation. Chest, doi: 10.1016/j.chest.2023.01.017 (2023). [DOI] [PubMed] [Google Scholar]
- 84.Venado A et al. Frailty after lung transplantation is associated with impaired health-related quality of life and mortality. Thorax 75, 669–678, doi: 10.1136/thoraxjnl-2019-213988 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Singer JP et al. Frailty Phenotypes, Disability, and Outcomes in Adult Candidates for Lung Transplantation. Am J Respir Crit Care Med 192, 1325–1334, doi: 10.1164/rccm.201506-1150OC (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lee YK et al. Benefits of both physical assessment and electronic health record review to assess frailty prior to heart transplant. Clin Transplant 36, e14559, doi: 10.1111/ctr.14559 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ayesta A et al. Prevalence and characterization of frailty, depression, and cognitive impairment in patients listed for heart transplantation: Results of the FELICITAR prospective registry. Clin Transplant 35, e14391, doi: 10.1111/ctr.14391 (2021). [DOI] [PubMed] [Google Scholar]
- 88.Macdonald PS et al. The impact of frailty on mortality after heart transplantation. J Heart Lung Transplant 40, 87–94, doi: 10.1016/j.healun.2020.11.007 (2021). [DOI] [PubMed] [Google Scholar]
- 89.Dayoub JC, Cortese F, Anzic A, Grum T & de Magalhaes JP The effects of donor age on organ transplants: A review and implications for aging research. Exp Gerontol 110, 230–240, doi: 10.1016/j.exger.2018.06.019 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Dixon W, Feng S & Gardner J Transplantation From Older Donors: Can Senolytics Turn Back the Clock? Transplantation 105, 681–682, doi: 10.1097/TP.0000000000003538 (2021). [DOI] [PubMed] [Google Scholar]
- 91●.Iske J et al. Senolytics prevent mt-DNA-induced inflammation and promote the survival of aged organs following transplantation. Nat Commun 11, 4289, doi: 10.1038/s41467-020-18039-x (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; A study investigating the impact of inflamm-aging and senolytics on graft survival following transplantation with organs from older donors.
- 92.Iske J, Matsunaga T, Zhou H & Tullius SG Donor and Recipient Age-Mismatches: The Potential of Transferring Senescence. Front Immunol 12, 671479, doi: 10.3389/fimmu.2021.671479 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Lorenz EC et al. The Relationship Between Frailty and Decreased Physical Performance With Death on the Kidney Transplant Waiting List. Prog Transplant 29, 108–114, doi: 10.1177/1526924819835803 (2019). [DOI] [PubMed] [Google Scholar]
- 94.McAdams-DeMarco MA et al. Frailty, Inflammatory Markers, and Waitlist Mortality Among Patients With End-stage Renal Disease in a Prospective Cohort Study. Transplantation 102, 1740–1746, doi: 10.1097/TP.0000000000002213 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Garonzik-Wang JM et al. Frailty and delayed graft function in kidney transplant recipients. Arch Surg 147, 190–193, doi: 10.1001/archsurg.2011.1229 (2012). [DOI] [PubMed] [Google Scholar]
- 96.Haugen CE et al. Incidence, Risk Factors, and Sequelae of Post-kidney Transplant Delirium. J Am Soc Nephrol 29, 1752–1759, doi: 10.1681/ASN.2018010064 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lorenz EC et al. Relationship between pre-transplant physical function and outcomes after kidney transplant. Clin Transplant 31, doi: 10.1111/ctr.12952 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.McAdams-DeMarco MA et al. Frailty, Length of Stay, and Mortality in Kidney Transplant Recipients: A National Registry and Prospective Cohort Study. Ann Surg 266, 1084–1090, doi: 10.1097/SLA.0000000000002025 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.McAdams-DeMarco MA et al. Frailty and early hospital readmission after kidney transplantation. Am J Transplant 13, 2091–2095, doi: 10.1111/ajt.12300 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.McAdams-DeMarco MA et al. Frailty, mycophenolate reduction, and graft loss in kidney transplant recipients. Transplantation 99, 805–810, doi: 10.1097/TP.0000000000000444 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chu NM et al. Frailty and Changes in Cognitive Function after Kidney Transplantation. J Am Soc Nephrol 30, 336–345, doi: 10.1681/ASN.2018070726 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.McAdams-DeMarco MA et al. Frailty and Postkidney Transplant Health-Related Quality of Life. Transplantation 102, 291–299, doi: 10.1097/TP.0000000000001943 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.McAdams-DeMarco MA et al. Frailty and mortality in kidney transplant recipients. Am J Transplant 15, 149–154, doi: 10.1111/ajt.12992 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Haugen CE et al. Development and Validation of an Inflammatory-Frailty Index for Kidney Transplantation. J Gerontol A Biol Sci Med Sci 76, 470–477, doi: 10.1093/gerona/glaa167 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105●.He A et al. Renal inflamm-aging provokes intra-graft inflammation following experimental kidney transplantation. Am J Transplant 22, 2529–2547, doi: 10.1111/ajt.17154 (2022). [DOI] [PubMed] [Google Scholar]; A study examining the interplay between allograft senescent cell burden and the recipient immune system and the impact of senolytic therapy on allograft survival.
- 106.Kim SR et al. Transplanted senescent renal scattered tubular-like cells induce injury in the mouse kidney. Am J Physiol Renal Physiol 318, F1167–F1176, doi: 10.1152/ajprenal.00535.2019 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Franzin R et al. Extracellular vesicles derived from patients with antibody-mediated rejection induce tubular senescence and endothelial to mesenchymal transition in renal cells. Am J Transplant 22, 2139–2157, doi: 10.1111/ajt.17097 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Pickering H et al. T cell senescence and impaired CMV-specific response are associated with infection risk in kidney transplant recipients. Hum Immunol 83, 273–280, doi: 10.1016/j.humimm.2022.01.016 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Matsunaga T et al. The potential of Senolytics in transplantation. Mech Ageing Dev 200, 111582, doi: 10.1016/j.mad.2021.111582 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Matsunaga T et al. Preserving and rejuvenating old organs for transplantation: novel treatments including the potential of senolytics. Curr Opin Organ Transplant 27, 481–487, doi: 10.1097/MOT.0000000000001019 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Guralnik JM et al. A short physical performance battery assessing lower extremity function: association with self-reported disability and prediction of mortality and nursing home admission. J Gerontol 49, M85–94, doi: 10.1093/geronj/49.2.m85 (1994). [DOI] [PubMed] [Google Scholar]
- 112.Brooks D, Solway S & Gibbons WJ ATS statement on six-minute walk test. Am J Respir Crit Care Med 167, 1287, doi: 10.1164/ajrccm.167.9.950 (2003). [DOI] [PubMed] [Google Scholar]
- 113.Podsiadlo D & Richardson S The timed “Up & Go”: a test of basic functional mobility for frail elderly persons. J Am Geriatr Soc 39, 142–148, doi: 10.1111/j.1532-5415.1991.tb01616.x (1991). [DOI] [PubMed] [Google Scholar]
- 114.Brito LB et al. Ability to sit and rise from the floor as a predictor of all-cause mortality. Eur J Prev Cardiol 21, 892–898, doi: 10.1177/2047487312471759 (2014). [DOI] [PubMed] [Google Scholar]
- 115.Lai JC et al. Development of a novel frailty index to predict mortality in patients with end-stage liver disease. Hepatology 66, 564–574, doi: 10.1002/hep.29219 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Mitnitski AB, Mogilner AJ & Rockwood K Accumulation of deficits as a proxy measure of aging. ScientificWorldJournal 1, 323–336, doi: 10.1100/tsw.2001.58 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mitnitski AB, Song X & Rockwood K The estimation of relative fitness and frailty in community-dwelling older adults using self-report data. J Gerontol A Biol Sci Med Sci 59, M627–632, doi: 10.1093/gerona/59.6.m627 (2004). [DOI] [PubMed] [Google Scholar]
- 118.Gandek B et al. Cross-validation of item selection and scoring for the SF-12 Health Survey in nine countries: results from the IQOLA Project. International Quality of Life Assessment. J Clin Epidemiol 51, 1171–1178, doi: 10.1016/s0895-4356(98)00109-7 (1998). [DOI] [PubMed] [Google Scholar]
- 119.Rose M, Bjorner JB, Becker J, Fries JF & Ware JE Evaluation of a preliminary physical function item bank supported the expected advantages of the Patient-Reported Outcomes Measurement Information System (PROMIS). J Clin Epidemiol 61, 17–33, doi: 10.1016/j.jclinepi.2006.06.025 (2008). [DOI] [PubMed] [Google Scholar]
- 120.Rose M et al. The PROMIS Physical Function item bank was calibrated to a standardized metric and shown to improve measurement efficiency. J Clin Epidemiol 67, 516–526, doi: 10.1016/j.jclinepi.2013.10.024 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Rockwood K et al. A global clinical measure of fitness and frailty in elderly people. Cmaj 173, 489–495, doi: 10.1503/cmaj.050051 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Karnofsky DA, B. J The Clinical Evaluation of Chemotherapeutic Agents in Cancer. 196 (Columbia Univ Press, 1949). [Google Scholar]