• Open Access
    Perspective

    Clinical studies with drugs and biologics aimed at slowing or reversing normal aging processes—emerging results and future perspectives

    Ricardo P. Garay 1,2*

    Explor Drug Sci. 2024;2:144–153 DOI: https://doi.org/10.37349/eds.2024.00040

    Received: August 03, 2023 Accepted: January 05, 2024 Published: April 10, 2024

    Academic Editor: Jean-Marc Sabatier, Aix-Marseille University, France

    This article belongs to the special issue Recent Advances with Investigational Compounds and Strategies to Delay or Reverse Normal Aging Processes

    Abstract

    Five families of investigational products are in clinical investigation to slow or reverse normal aging processes [longevity candidates, mesenchymal stem cells, senolytics drugs, sirtuin activators, and nicotinamide adenine dinucleotide (NAD)+ precursors]. The longevity candidates, vitamin D and metformin, appear to significantly reduce all-cause mortality and prolong life expectancy. This should be confirmed by interventional studies. The mesenchymal stem cell family is the most advanced in clinical trial development [phase 2b randomized controlled trial (RCT)]. An allogeneic bone marrow stem cell preparation (Lomecel-B) reduced locomotor frailty in older people. The improvement in locomotion was modest. In the future, attempts could be made to improve potency through a precondition or genetic modification of naive bone marrow stem cells. Autologous adipose stem cell-assisted fat grafting increased graft survival, facial volume, and skin quality. The association of the senolytic drugs dasatinib and quercetin was well tolerated, with low brain penetration of dasatinib and undetectable levels of quercetin. The sirtuin-1 activator resveratrol (combined with physical exercise) improved physical function in older adults with physical limitations. The NAD+ precursor nicotinamide riboside improved physical exercise performance. In conclusion, Lomecel-B is the most advanced agent in clinical trial development for normal aging processes (phase 2b for locomotion frailty), followed by resveratrol and nicotinamide riboside.

    Keywords

    Aging, clinical trials, frailty, longevity, rejuvenation, senolytics, stem cells, vitamin D

    Introduction

    Aging is characterized by a slow loss of organ function, reducing vitality, resilience, and healthy life expectancy. The International Conference on Frailty and Sarcopenia Research (ICFSR) clinical practice guidelines (CPGs) specify that there is no specific medical or biological treatment for these normal conditions of aging. The ICFSR does not recommend any of the non-specific pharmacological treatments currently available (only therapeutic interventions based on exercise and nutritional supplements are recommended) [1].

    A substantial effort is currently deployed to find specific drugs and biologics intended to slow or reverse normal aging processes. Currently, several literature reviews are available that identify and analyze clinical studies with the following specific families of compounds (Table 1): (i) longevity candidates [2, 3], (ii) mesenchymal stem cells [46], (iii) senolytics drugs [79], (iv) sirtuin activators [1013], and (v) nicotinamide adenine dinucleotide (NAD)+ precursors [1418].

    Drugs and biologics that have shown positive results in clinical research to slow or reverse normal aging processes

    FamilyIndicationLeading agentStudy typeReference
    Longevity candidatesLongevityVitamin DObservational[34, 36]
    MetforminObservational[40]
    Mesenchymal stem cellsLocomotion frailtyLomecel-BRCT (phase 2)[47]
    Facial rejuvenationSVFRCT[54]
    Senolytics drugsCognitive declineD + QOL (phase 1)[58]
    Sirtuin activatorsAging frailtyResveratrolRCT (phase 2)[61]
    NAD+ precursorsPhysical performanceNRSRCT[65]
    Display full size

    SVF: stromal vascular fraction; D + Q: dasatinib and quercetin; NRS: nicotinamide riboside supplements; OL: open-label

    An overview of emerging results and future perspectives from clinical studies conducted with the five compound families mentioned above is presented here. Preclinical data and details about ongoing clinical trials (without results) can be found in the literature reviews cited above [212, 14, 15]. The reader interested in pharmacological mechanisms and biomarkers can consult the reviews by Nielsen et al. [16], Tchkonia et al. [19], Moskalev et al. [20], and Fraser et al. [21].

    Longevity candidates

    Twenty years ago, the National Institute on Aging (NIA, USA) launched an Intervention Testing Program (ITP) dedicated to identifying longevity candidates in a genetically heterogeneous mouse model (candidates are evaluated in three independent laboratories) [22]. The ITP has identified two longevity drug candidates, the immunosuppressant rapamycin [23] and the antidiabetic acarbose [24] that are currently under clinical investigation [3]. Vitamin D and the antidiabetic drug metformin have been identified in other animal models and are also under clinical investigation [3, 25].

    The mechanism by which these longevity candidates prolong animal lifespan is unclear. Rapamycin is a [mammalian target of rapamycin (mTOR)] kinase inhibitor, which possesses immunosuppressive and antiproliferative properties [25]. Ehninger et al. [26] have suggested that rapamycin extends lifespan in mice by suppressing cancerogenesis. Acarbose is an inhibitor of alpha-glucosidases, a class of intestinal enzymes necessary to digest carbohydrates [27]. Acarbose exerts anti-inflammatory effects on human monocytic THP-1 cells [28], and Sadagurski et al. [29] suggested that acarbose may prolong lifespan in mice by inhibiting age-related hypothalamic inflammation. Metformin appears to exert antiaging actions via multiple mechanisms, including nutrient sensing, DNA repair, oxidative stress, telomere attrition, inflammation, cellular senescence, stem cell decline, and autophagy [30]. Finally, vitamin D is a gene expression modifier (acting on more than 200 genes) and has pleiotropic biological actions [31].

    Emerging results with vitamin D

    Vitamin D is mainly synthesized in skin exposed to sunlight, and strong sunlight at equatorial latitudes leads to high mean serum concentrations of 25-hydroxy vitamin D [25(OH)D = around 115 nmol/L] [32]. On the contrary, low levels of 25(OH)D (< 50 nmol/L) have been found in a considerable proportion of individuals from temperate countries, 40% in Europe and 24% in the USA [33]. In China, the prevalence of vitamin D deficiency in older adults was 68.4% (in 2014) [34, 35].

    A recent observational and Mendelian analysis in middle-aged European persons [36] suggested a causal relationship between low levels of 25(OH)D (< 40 nmol/L) and all-cause mortality. In China, all-cause mortality was significantly higher in subjects with low vitamin D status, and the change from vitamin D deficiency to no deficiency was associated with a lower risk of all‐cause mortality [34].

    Emerging results with metformin

    Kulkarni et al. [37] conducted a phase 4, crossover design RCT (ClinicalTrials.gov identifier: NCT02432287) to investigate the effect of metformin on metabolic and nonmetabolic regulation pathways in skeletal muscle and subcutaneous adipose tissue from 14 older adults (around 70 years old). Participants received oral metformin (1,700 mg/day) or placebo for 6 weeks. Metformin was found to significantly modify gene expression (RNA sequencing) in biopsies from skeletal muscle (647 genes) and subcutaneous adipose tissue (146 genes) [37]. These transcriptomic changes included several well-known anti-aging genes from the GenAge database [37, 38]. In addition, a Mendelian randomization study using UK Biobank data [39] showed that metformin use was associated with younger phenotypic age.

    Campbell et al. [40] reviewed observational studies comparing all-cause mortality in patients with diabetes taking metformin with non-diabetics, or with diabetics receiving non-metformin therapies. The results suggested that metformin reduces all-cause mortality independently of its antidiabetic effect [40].

    Several clinical trials have been conducted with metformin for aging-related diseases, but they are outside the scope of this article for a recent review, see [41].

    Perspectives of clinical research with longevity candidates

    All-cause mortality is a marker of longevity [3]. The above results from observational studies suggest that vitamin D supplementation [in persons with low 25(OH)D serum levels] [34, 36] as well as metformin [40] can significantly reduce all-cause mortality and prolong life expectancy (Table 1). This prolongevity efficacy should be confirmed by interventional studies.

    A phase 3 clinical trial with metformin [Targeting Aging with Metformin (TAME)] was planned to assess the time to new onset of a composite outcome including cardiovascular events, cancer, dementia, and mortality [42, 43]. TAME plans to include 3,000 participants, aged 65–79 years, at 14 centers across the US. In 2015, TAME started a discussion with the Food and Drug Administration (FDA, USA), but to this day it is not included in the ClinicalTrials.gov database.

    According to Barzilai et al. [43], the goal of TAME was to demonstrate “that metformin modulates aging and its diseases, beyond an isolated impact on diabetes”. According to Glossmann and Lutz [44]: “The acronym chosen and the intention behind it – namely, that aging is a ‘disorder’ that can be treated like any other disease – was a clear provocation.”.

    It is interesting to mention that two other longevity candidates (rapamycin and acarbose) are currently in clinical trials, but the results have not yet been reported to ClinicalTrials.gov [3].

    Mesenchymal stem cell preparations

    Mesenchymal stem cell preparations and multipotential stromal cells were in development for locomotion frailty and facial skin aging [4].

    Positive results on locomotion frailty

    Physical frailty in the elderly is characterized by reduced locomotor activity [1], and locomotion frailty is associated with an increased risk of falls, disability, and hospitalization [45].

    Lomecel-B (Longeveron, USA) is an allogeneic bone marrow stem cell preparation expanded in culture [46]. The [allogeneiC human mesenchymal stem cells in patients with aging fRAilTy via intravenoUS delivery (CRATUS)] trial [46, 47] included: (i) a phase 1 safety study in 15 frail aging patients followed for 1 month [46], and (ii) a phase 2 RCT [47] investigating the efficacy of intravenous Lomecel-B to reduce physical frailty in 30 elderly subjects with mild to moderate locomotion frailty (treatment period: 6 months). The phase 1 study showed that Lomecel-B was safe and immunologically tolerated [46]. Participants in the phase 2 RCT [47] received 100 million cells (n = 10), 200 million cells (n = 10), or placebo (n = 10). The 6-min walk distance (6-MWD) increased significantly in the 100 m group (from 345.9 m to 410.5 m, mean values at baseline and 6 months, respectively). Immunotolerability was acceptable. Tumor necrosis factor (TNF)-alpha levels decreased significantly.

    Lomecel-B is a candidate for further development in phase 3 trials.

    Perspectives of clinical research with stem cell preparations for physical frailty

    Locomotion frailty improvement with intravenous Lomecel-B was modest [48]. Preconditioning of naive bone marrow stem cells (with growth factors, drugs, or other agents), as well as genetic modification, can improve their therapeutic efficacy [49]. On the other hand, intravenous administration risks trapping the stem cells in the lungs [50]. In such a case, (smaller) exosomes may be an option to increase efficacy.

    Several other phase 1 and 2 clinical trials with stem cell preparations are currently underway [4]. In particular, a clinical trial (NCT04314011) using an allogeneic preparation of umbilical cord-derived stem cells was recently completed, but the results have not yet been reported to ClinicalTrials.gov.

    The use of allogeneic stem cell preparations may limit its clinical application. Predictive markers of graft rejection are being developed [51], markers that could help identify patients at risk of tissue rejection before administering stem cells.

    Positive results on facial skin aging

    Facial skin aging is due to natural causes, as well as extrinsic factors (especially sun exposure: photoaging). The [stromal vascular fraction (SVF)] is a preparation of human adipose stem cells obtained by liposuction, followed by collagenase digestion and centrifugation [4, 52, 53]. Yin et al. [54] conducted an RCT to investigate autologous SVF-assisted fat grafting for facial rejuvenation. Fifty patients were randomly assigned into two groups: an intervention group (n = 25) and a control group (n = 25, fat grafting only). SVF-assisted autologous fat grafting increased graft survival, facial volume, and skin quality (Table 1).

    Perspectives of clinical research with stem cell preparations for facial rejuvenation

    Several clinical trials for facial skin aging are currently underway in Egypt, Indonesia, Cuba, Iran, and Malaysia [4]. Autologous SVF preparations are regulated as biological products by the FDA (USA) because they require mechanical processing [4, 41], and very numerous clinical trials with autologous SVF preparations for facial rejuvenation [53] are not registered on ClinicalTrials.gov. This was not the case with the study of Yin et al. [54], which was registered in ClinicalTrials.gov (NCT02923219) as well as other clinical trials conducted in Egypt (NCT03928444) and Indonesia (NCT05508191) [4]. Results from trials NCT03928444 and NCT05508191 have not yet been posted to ClinicalTrials.gov. The application of similar RCT protocols could pave the way for autologous SVF preparations (in the USA) to be registered on ClinicalTrials.gov and comply with FDA regulations.

    Senolytic drugs

    Cellular senescence stops the proliferation of damaged or dysfunctional cells [7]. Aging is associated with the accumulation of senescent cells in various tissues and organs. In 2011, the group of Baker et al. [55] showed that killing senescent cells using a transgenic suicide gene is beneficial in preventing or delaying tissue dysfunction and prolonging lifespan in mouse models. Subsequently, the same group reported the discovery of senolytic agents in animal models: (i) the combination of D + Q in 2015 [56], and (ii) fisetin in 2017 [57].

    Results

    Gonzales et al. [58] recently reported the results of a 12 week, phase 1 clinical trial investigating the safety and cerebrospinal fluid (CSF) penetrance of D + Q in 5 participants (aged 70 years to 82 years) with early-stage symptomatic Alzheimer’s disease (Table 1). D + Q (100 mg and 1,000 mg respectively) were orally given for two days, followed by a treatment interruption of 13 days to 15 days, for a total of 6 cycles. Treatment was well tolerated and was not discontinued prematurely [58]. Low levels of dasatinib were detected in the CSF of 4 participants. Quercetin was not detectable in the CSF.

    Perspectives

    Several senolytics, including D + Q and fisetin, are in development for cognitive decline [59], aging frailty, and skeletal health in normal postmenopausal women [7]. A clear advantage of some senolytics (quercetin, fisetin) is that they are natural products [60], but it seems too early to draw conclusions about clinical research with senolytic agents.

    Sirtuin activators

    Human sirtuins are a family of 7 signaling NAD+-dependent protein deacetylases that are involved in metabolic regulation, resistance to stress, and cellular processes, including aging and cell death [10, 11]. The natural polyphenol resveratrol is a powerful activator of sirtuin-1 (SIRT1) [12, 13]. Curcumin (another natural polyphenol) is a non-specific activator/upregulator of sirtuins (mainly influencing SIRT1 and SIRT3) [13].

    Positive results

    Harper et al. [61] have recently conducted a pilot clinical trial showing that resveratrol and exercise combined improve aging frailty (Table 1).

    Perspectives

    The positive results of the pilot trial by Harper et al. [61] deserve confirmation in larger-scale trials.

    NAD+ precursors

    A large body of evidence clearly shows that aging is associated with a reduction in cellular NAD+ levels [1418]. In particular, Mouchiroud et al. [62] found that NAD+ levels are reduced in aged worms (the nematode Caenorhabditis elegans) and aged mice and that decreasing NAD+ levels reduced worm lifespan. Zhu et al. [63] reported a decrease in intracellular NAD+ levels in the healthy aged human brain in vivo. Massudi et al. [64] found a strong negative correlation between age and NAD+ levels in human pelvic skin samples.

    Barker et al. [14] published a systematic review of 26 trials that investigated the effect of NAD+ precursors on physical frailty outcomes. Only four of these trials enrolled participants with a mean age of 60 years or more [14]. Of these, only two trials included healthy participants. Dolopikou et al. [65] found that NRS significantly improved exercise performance (isometric peak torque and fatigue index) in the elderly. Martens et al. [66] reported that NRS elevates NAD+ levels in circulating peripheral blood mononuclear cells of healthy middle-aged and older adults.

    Concluding remarks

    Clinical research with drugs and biologics intended to slow or reverse normal aging processes has only recently begun [218]. Positive results are beginning to emerge from the very numerous clinical trials (and observational studies) launched worldwide (Table 1).

    Research highlights and perspectives:

    • Vitamin D and metformin are leaders in the clinical development of longevity candidates (Table 1). Observational studies strongly suggested that these compounds can significantly reduce all-cause mortality (and prolong life expectancy) (Table 1). This prolongevity efficacy should be confirmed by interventional studies.

    • Intravenous Lomecel-B (a mesenchymal stem cell preparation) showed efficacy in reducing locomotor frailty in older people. However, the frailty improvement was modest. Preconditioning and/or genetic modification of naive mesenchymal stem cells can improve their therapeutic efficacy. The use of allogeneic stem cell preparations may limit its clinical application. Predictive markers of graft rejection are being developed, markers that could help identify patients at risk of tissue rejection before administering stem cells.

    • An RCT on facial rejuvenation [54] was registered on ClinicalTrials.gov (NCT02923219) and showed that an autologous fat graft enriched with human adipose tissue stem cells improves graft survival, facial volume, and skin quality. The application of a similar RCT protocol could pave the way for autologous SVF preparations to be registered on ClinicalTrials.gov and comply with FDA regulations.

    • Senolytic drugs (including D + Q and fisetin), the SIRT1 activator resveratrol, and the NAD+ precursor nicotinamide riboside (NR) are being developed to slow normal aging processes, but it seems too early to draw conclusions about the current clinical research with these compounds.

    Abbreviations

    25(OH)D:

    25-hydroxy vitamin D

    CSF:

    cerebrospinal fluid

    D + Q:

    dasatinib and quercetin

    FDA:

    Food and Drug Administration

    NAD:

    nicotinamide adenine dinucleotide

    NRS:

    nicotinamide riboside supplements

    RCT:

    randomized controlled trial

    SIRT1:

    sirtuin-1

    SVF:

    stromal vascular fraction

    TAME:

    Targeting Aging with Metformin

    Declarations

    Author contributions

    RPG: Conceptualization, Writing—review & editing.

    Conflicts of interest

    The author declares that he has no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2024.

    References

    Dent E, Morley JE, Cruz-Jentoft AJ, Woodhouse L, Rodríguez-Mañas L, Fried LP, et al. Physical frailty: ICFSR international clinical practice guidelines for identification and management. J Nutr Health Aging. 2019;23:77187. [DOI] [PubMed] [PMC]
    Laskovs M, Partridge L, Slack C. Molecular inhibition of RAS signalling to target ageing and age-related health. Dis Model Mech. 2022;15:dmm049627. [DOI] [PubMed] [PMC]
    Garay RP. Investigational drugs and nutrients for human longevity. Recent clinical trials registered in ClinicalTrials.gov and clinicaltrialsregister.eu. Expert Opin Investig Drugs. 2021;30:74958. [DOI] [PubMed]
    Garay RP. Recent clinical trials with stem cells to slow or reverse normal aging processes. Front Aging. 2023;4:1148926. [DOI] [PubMed] [PMC]
    Sun XL, Hao QK, Tang RJ, Xiao C, Ge ML, Dong BR. Frailty and rejuvenation with stem cells: therapeutic opportunities and clinical challenges. Rejuvenation Res. 2019;22:48497. [DOI] [PubMed] [PMC]
    Zhu Y, Ge J, Huang C, Liu H, Jiang H. Application of mesenchymal stem cell therapy for aging frailty: from mechanisms to therapeutics. Theranostics. 2021;11:567585. [DOI] [PubMed] [PMC]
    Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: the path to the clinic. Nat Med. 2022;28:155668. [DOI] [PubMed] [PMC]
    Wyles SP, Tchkonia T, Kirkland JL. Targeting cellular senescence for age-related diseases: path to clinical translation. Plast Reconstr Surg. 2022;150. [DOI] [PubMed] [PMC]
    Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J. 2023;290:136283. [DOI] [PubMed]
    Dai H, Sinclair DA, Ellis JL, Steegborn C. Sirtuin activators and inhibitors: promises, achievements, and challenges. Pharmacol Ther. 2018;188:14054. [DOI] [PubMed] [PMC]
    Watroba M, Szukiewicz D. Sirtuins at the service of healthy longevity. Front Physiol. 2021;12:724506. [DOI] [PubMed] [PMC]
    Toniolo L, Concato M, Giacomello E. Resveratrol, a multitasking molecule that improves skeletal muscle health. Nutrients. 2023;15:3413. [DOI] [PubMed] [PMC]
    Wiciński M, Erdmann J, Nowacka A, Kuźmiński O, Michalak K, Janowski K, et al. Natural phytochemicals as SIRT activators—focus on potential biochemical mechanisms. Nutrients. 2023;15:3578. [DOI] [PubMed] [PMC]
    Barker FJ, Hart A, Sayer AA, Witham MD. Effects of nicotinamide adenine dinucleotide precursors on measures of physical performance and physical frailty: a systematic review. JCSM Clin Rep. 2022;7:93106. [DOI]
    Reiten OK, Wilvang MA, Mitchell SJ, Hu Z, Fang EF. Preclinical and clinical evidence of NAD+ precursors in health, disease, and ageing. Mech Ageing Dev. 2021;199:111567. [DOI] [PubMed]
    Nielsen JL, Bakula D, Scheibye-Knudsen M. Clinical trials targeting aging. Front Aging. 2022;3:820215. [DOI]
    Fang EF, Lautrup S, Hou Y, Demarest TG, Croteau DL, Mattson MP, et al. NAD+ in aging: molecular mechanisms and translational implications. Trends Mol Med. 2017;23:899916. [DOI] [PubMed] [PMC]
    Yoshino J, Baur JA, Imai SI. NAD+ intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 2018;27:51328. [DOI] [PubMed] [PMC]
    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. 2021;106:e14817. [DOI] [PubMed] [PMC]
    Moskalev A, Guvatova Z, Lopes IA, Beckett CW, Kennedy BK, De Magalhaes JP, et al. Targeting aging mechanisms: pharmacological perspectives. Trends Endocrinol Metab. 2022;33:26680. [DOI] [PubMed]
    Fraser HC, Kuan V, Johnen R, Zwierzyna M, Hingorani AD, Beyer A, et al. Biological mechanisms of aging predict age-related disease co-occurrence in patients. Aging Cell. 2022;21:e13524. [DOI] [PubMed] [PMC]
    ITP [Internet]. [cited 2023 Jul 28]. Available from: https://www.nia.nih.gov/research/dab/interventions-testing-program-itp
    Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:3925. [DOI]
    Harrison DE, Strong R, Allison DB, Ames BN, Astle CM, Atamna H, et al. Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell. 2014;13:27382. [DOI] [PubMed] [PMC]
    Huggins B, Farris M. Vitamin D3 promotes longevity in Caenorhabditis elegans. Geroscience. 2023;45:34558. [DOI]
    Ehninger D, Neff F, Xie K. Longevity, aging and rapamycin. Cell Mol Life Sci. 2014;71:432546. [DOI] [PubMed] [PMC]
    DiNicolantonio JJ, Bhutani J, O’Keefe JH. Acarbose: safe and effective for lowering postprandial hyperglycaemia and improving cardiovascular outcomes. Open Heart. 2015;2:e000327. [DOI] [PubMed] [PMC]
    Lin YC, Chen YC, Hsiao HP, Kuo CH, Chen BH, Chen YT, et al. The effects of acarbose on chemokine and cytokine production in human monocytic THP-1 cells. Hormones (Athens). 2019;18:17987. [DOI] [PubMed]
    Sadagurski M, Cady G, Miller RA. Anti-aging drugs reduce hypothalamic inflammation in a sex-specific manner. Aging Cell. 2017;16:65260. [DOI] [PubMed] [PMC]
    Cheng FF, Liu YL, Du J, Lin JT. Metformin’s mechanisms in attenuating hallmarks of aging and age-related disease. Aging Dis. 2022;13:97086. [DOI] [PubMed] [PMC]
    Ramagopalan SV, Heger A, Berlanga AJ, Maugeri NJ, Lincoln MR, Burrell A, et al. A ChIP-seq defined genome-wide map of vitamin D receptor binding: associations with disease and evolution. Genome Res. 2010;20:135260. [DOI] [PubMed] [PMC]
    Luxwolda MF, Kuipers RS, Kema IP, Dijck-Brouwer DA, Muskiet FA. Traditionally living populations in East Africa have a mean serum 25-hydroxyvitamin D concentration of 115 nmol/l. Br J Nutr. 2012;108:155761. [DOI] [PubMed]
    Amrein K, Scherkl M, Hoffmann M, Neuwersch-Sommeregger S, Köstenberger M, Tmava Berisha A, et al. Vitamin D deficiency 2.0: an update on the current status worldwide. Eur J Clin Nutr. 2020;74:1498513. [DOI] [PubMed] [PMC]
    Zeng J, Li T, Sun B, Miao X, Wang L, Ma LC, et al. Change of vitamin D status and all-cause mortality among Chinese older adults: a population-based cohort study. BMC Geriatr. 2022;22:245. [DOI] [PubMed] [PMC]
    Chinese longitudinal healthy longevity survey (CLHLS), 1998-2014 [Internet]. The Regents of the University of Michigan; c2024 [cited 2023 Aug 2]. Available from: https://doi.org/10.3886/ICPSR36692.v1
    Emerging Risk Factors Collaboration/EPIC-CVD/Vitamin D Studies Collaboration. Estimating dose-response relationships for vitamin D with coronary heart disease, stroke, and all-cause mortality: observational and Mendelian randomisation analyses. Lancet Diabetes Endocrinol. 2024;12:e211. [DOI] [PubMed] [PMC]
    Kulkarni AS, Brutsaert EF, Anghel V, Zhang K, Bloomgarden N, Pollak M, et al. Metformin regulates metabolic and nonmetabolic pathways in skeletal muscle and subcutaneous adipose tissues of older adults. Aging Cell. 2018;17:e12723. [DOI] [PubMed] [PMC]
    Tacutu R, Craig T, Budovsky A, Wuttke D, Lehmann G, Taranukha D, et al. Human ageing genomic resources: integrated databases and tools for the biology and genetics of ageing. Nucleic Acids Res. 2013;41:D102733. [DOI] [PubMed] [PMC]
    Luo S, Wong ICK, Chui CSL, Zheng J, Huang Y, Schooling CM, et al. Effects of putative metformin targets on phenotypic age and leukocyte telomere length: a mendelian randomisation study using data from the UK Biobank. Lancet Healthy Longev. 2023;4:e33744. [DOI] [PubMed]
    Campbell JM, Bellman SM, Stephenson MD, Lisy K. Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: a systematic review and meta-analysis. Ageing Res Rev. 2017;40:3144. [DOI] [PubMed]
    Overview on the regulation of cellular therapies in aesthetic medicine [Internet]. [cited 2023 Aug 2]. Available from: https://www.aslms.org/docs/default-source/for-professionals/resources/task-force-whitepaper-2019-final-4-9-21.pdf
    Padki MM, Stambler I. Targeting aging with metformin (TAME). In: Gu D, Dupre ME, editors. Encyclopedia of gerontology and population aging. Cham: Springer International Publishing; 2021. pp. 4908–10.
    Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin as a tool to target aging. Cell Metab. 2016;23:10605. [DOI] [PubMed] [PMC]
    Glossmann HH, Lutz OMD. Metformin and aging: a review. Gerontology. 2019;65:58190. [DOI] [PubMed]
    Xue QL. The frailty syndrome: definition and natural history. Clin Geriatr Med. 2011;27:115. [DOI] [PubMed] [PMC]
    Golpanian S, DiFede DL, Khan A, Schulman IH, Landin AM, Tompkins BA, et al. Allogeneic human mesenchymal stem cell infusions for aging frailty. J Gerontol A Biol Sci Med Sci. 2017;72:150512. [DOI] [PubMed] [PMC]
    Tompkins BA, DiFede DL, Khan A, Landin AM, Schulman IH, Pujol MV, et al. Allogeneic mesenchymal stem cells ameliorate aging frailty: a phase II randomized, double-blind, placebo-controlled clinical trial. J Gerontol A Biol Sci Med Sci. 2017;72:151322. [DOI] [PubMed] [PMC]
    Larrick JW, Mendelsohn AR. Mesenchymal stem cells for frailty? Rejuvenation Res. 2017;20:5259. [DOI] [PubMed]
    Ocansey DKW, Pei B, Yan Y, Qian H, Zhang X, Xu W, et al. Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med. 2020;18:42. [DOI] [PubMed] [PMC]
    Salvadori M, Cesari N, Murgia A, Puccini P, Riccardi B, Dominici M. Dissecting the pharmacodynamics and pharmacokinetics of mscs to overcome limitations in their clinical translation. Mol Ther Methods Clin Dev. 2019;14:115. [DOI] [PubMed] [PMC]
    Kadri N, Amu S, Iacobaeus E, Boberg E, Le Blanc K. Current perspectives on mesenchymal stromal cell therapy for graft versus host disease. Cell Mol Immunol. 2023;20:61325. [DOI] [PubMed] [PMC]
    Coleman SR. The technique of periorbital lipoinfiltration. Oper Tech Plast Reconstr Surg. 1994;1:1206. [DOI]
    Surowiecka A, Strużyna J. Adipose-derived stem cells for facial rejuvenation. J Pers Med. 2022;12:117. [DOI] [PubMed] [PMC]
    Yin Y, Li J, Li Q, Zhang A, Jin P. Autologous fat graft assisted by stromal vascular fraction improves facial skin quality: a randomized controlled trial. J Plast Reconstr Aesthet Surg. 2020;73:116673. [DOI] [PubMed]
    Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:2326. [DOI] [PubMed] [PMC]
    Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14:64458. [DOI] [PubMed] [PMC]
    Zhu Y, Doornebal EJ, Pirtskhalava T, Giorgadze N, Wentworth M, Fuhrmann-Stroissnigg H, et al. New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging (Albany NY). 2017;9:95563. [DOI] [PubMed] [PMC]
    Gonzales MM, Garbarino VR, Kautz TF, Palavicini JP, Lopez-Cruzan M, Dehkordi SK, et al. Senolytic therapy in mild Alzheimer’s disease: a phase 1 feasibility trial. Nat Med. 2023;29:24818. [DOI] [PubMed] [PMC]
    Orr M, Gonzales M, Garbarino V, Kautz T, Palavicini J, Lopez-Cruzan M, et al. Senolytic therapy to modulate the progression of Alzheimer’s Disease (SToMP-AD) – outcomes from the first clinical trial of senolytic therapy for Alzheimer’s disease. [Preprint]. 2023 [cited 2023 Aug 2]. Available from: https://www.researchsquare.com/article/rs-2809973/v1
    Barrera-Vázquez OS, Magos-Guerrero GA, Escobar-Ramírez JL, Gomez-Verjan JC. Natural products as a major source of candidates for potential senolytic compounds obtained by in silico screening. Med Chem. 2023;19:65368. [DOI] [PubMed]
    Harper SA, Bassler JR, Peramsetty S, Yang Y, Roberts LM, Drummer D, et al. Resveratrol and exercise combined to treat functional limitations in late life: a pilot randomized controlled trial. Exp Gerontol. 2021;143:111111. [DOI] [PubMed] [PMC]
    Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Cantó C, et al. The NAD+/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell. 2013;154:43041. [DOI] [PubMed] [PMC]
    Zhu XH, Lu M, Lee BY, Ugurbil K, Chen W. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc Natl Acad Sci U S A. 2015;112:287681. [DOI] [PubMed] [PMC]
    Massudi H, Grant R, Braidy N, Guest J, Farnsworth B, Guillemin GJ. Age-associated changes in oxidative stress and NAD+ metabolism in human tissue. PLoS One. 2012;7:e42357. [DOI] [PubMed] [PMC]
    Dolopikou CF, Kourtzidis IA, Margaritelis NV, Vrabas IS, Koidou I, Kyparos A, et al. Acute nicotinamide riboside supplementation improves redox homeostasis and exercise performance in old individuals: a double-blind cross-over study. Eur J Nutr. 2020;59:50515. [DOI] [PubMed]
    Martens CR, Denman BA, Mazzo MR, Armstrong ML, Reisdorph N, McQueen MB, et al. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat Commun. 2018;9:1286. [DOI] [PubMed] [PMC]