Abstract
Cellular senescence and the hallmarks of aging contribute to age-related disease and dysfunction. The Unitary Theory of Fundamental Aging Mechanisms highlights the interdependence among the hallmarks of aging and suggests that by intervening in one fundamental aging process, most or all of the other processes could be impacted. Accumulation of senescent cells is associated with frailty, cardiovascular disease, obesity, diabetes, cognitive decline, and other age- and/or chronic disease-related disorders, suggesting that senescent cells are a target for intervention. Early preclinical data using senolytics, agents that target senescent cells, show promising results in several aging and disease models. The first in-human trials using the senolytic combination of Dasatinib and Quercetin indicated reduced senescent cell burden in adipose tissue of diabetic kidney disease patients and improved physical function in patients with idiopathic pulmonary fibrosis. Clinical trials with other senolytics, including the flavonoid Fisetin and BCL-xL inhibitors, are underway. These results from preclinical and early clinical trials illustrate the potential of senolytics to alleviate age-related dysfunction and diseases. However, multiple clinical trials across different aging and disease models are desperately needed. Parallel trials across institutions through the Translational Geroscience Network are facilitating testing to determine whether senolytics can be translated into clinical application.
Keywords: Senolytics, Fisetin, Dasatinib, Quercetin, Unitary Theory of Fundamental Aging Processes, Translational Geroscience Network
1. Introduction
1.1. Impact of aging/disease: Need for senolytics
As the aging population steadily increases, age-related diseases and disorders are becoming a major subset of health problems [1]. Since 2002, the leading causes of death in individuals aged 65 or older has been age-related diseases, with cardiovascular disease, cancer, and chronic lower respiratory disease accounting for over half of these deaths [2]. Frailty, neurodegenerative diseases, cardiac dysfunction, respiratory diseases, cancers, and other chronic diseases are directly impacted by senescent cell accumulation [3]. Additionally, senescent cell burden negatively impacts healthspan and lifespan [4]. As more people suffer from these chronic conditions, the burden on the healthcare system grows. Current treatment options for many of these conditions are focused on treating them one-disease-at-a-time. This is as opposed to intervening against multiple disorders together, since multimorbidity often occurs in older people, by targeting root causal fundamental aging processes shared across these conditions. Targeting cellular senescence using senolytics, agents that selectively clear senescent cells, could be a potential therapeutic option to delay, prevent, or treat multiple age-related disorders.
2. Geroscience Hypothesis
2.1. Hallmarks of Aging
Fundamental aging mechanisms can be grouped into hallmarks or pillars of aging. One such grouping includes nine hallmarks: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, dysregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication [5, 6]. Each of these pillars can contribute to age-related disease and dysfunction (Figure 1).
Figure 1:
The hallmarks of aging may underpin a variety of age-related diseases and disorders, as well as chronic disease in younger individuals.
2.2. Unitary Theory of Fundamental Aging Processes
The nine hallmarks of aging may not be independent processes, but rather interdependent [7]. Our Unitary Theory of Fundamental Aging Processes posits that by targeting one fundamental aging process, it may be feasible to impact several or all others [7][8]. Cellular senescence is bi-directionally linked to other hallmarks of aging, including epigenetic alterations [5], mitochondrial dysfunction [9][10][11][12], dysfunctional nutrient processing [13–15], stem cell exhaustion [16][17][18], altered cellular communication [19][19][5], genomic instability [20][21], telomere attrition [22][23][24][25], and loss of proteostasis [26][27]. Because of its known and well documented interactions with other aging hallmarks, cellular senescence appears to be an appropriate target for intervention.
2.3. Cellular senescence as a target
Cellular senescence is a cell fate that entails cell cycle arrest first observed in human fibroblasts by Hayflick and Moorehead in 1961 [28]. Senescent cells resist apoptosis but can be removed by the immune system [29][30]. Our ‘Threshold Theory of Senescent Cell Burden’ posits that there is a point beyond which senescent cell accumulation exceeds capacity of the immune system to clear senescent cells, leading to rising senescent cell burden and associated acceleration of age- and disease-related dysfunction [31][7][13].
Cellular senescence presents a unique target for intervention at two points: 1) clearance of senescent cells and 2) inhibition of production of senescence-associated secretory phenotype (SASP) factors. The accumulation of senescent cells can cause immune system dysfunction (inducing further senescent cell accumulation), inflammation, tissue damage, and cell impairment due to the SASP [32]. The SASP, secreted by some, but not all, senescent cells, can lead to stem and progenitor cell damage, as well as tissue and systemic dysfunction [33][34][7]. The SASP includes a variety of cytokines, chemokines, proteases, activins and inhibins, growth factors, bioactive lipids, micro-RNAs, cell-free mitochondrial DNA, other non-coding nucleotides, and microsomes and exosomes [32][35][7][36]. Senescent cells and their SASP are found at etiological sites in a host of age-related diseases and disorders including frailty, cardiovascular disease, osteoporosis, cancers, diabetes/obesity, and other chronic diseases, even in children and younger adults [37][38][39][40]. Senotherapeutics include agents that target both of these processes: senolytics selectively target senescent cells and senomorphics (e.g., metformin or rapamycin), act as SASP inhibitors [41].
Senomorphics can suppress effects of senescence in one of two ways, (i) controlling the regulatory network of the SASP, and (ii) by targeting a specific SASP component. Although senomorphics may be beneficial to individuals who take them regularly, it is unclear if intermittent dosing would provide the same benefits [42]. More research is needed to determine the efficacy of senomorphics.
3. Discovery of senolytic drugs
Prompted by the finding that senescent cells are linked to declines in healthspan [4], which suggested senescent cells a potential target for intervention, our team began the search for potential senolytic interventions. We turned to a hypothesis-driven, mechanism-based drug discovery approach for identifying possible senolytics. Using the principles that senescent cells resist their own removal through Senescent Cell Anti-Apoptotic Pathway (SCAP) networks, we targeted several different SASP pathways by RNA interference to identify key nodes in the SCAP network [43]. Once identified, we used bioinformatics approaches to identify compounds that target these nodes and could possibly be effective in enabling apoptosis of those senescent cells that have a pro-apoptotic SASP [44][43]. Eventually, 46 compounds were so identified as being potentially senolytic. We focused on advancing those compounds that are natural products or drugs already used in humans to expedite translation to clinical application.
The first generation of senolytics were mostly agents or combinations of agents with multiple SCAP targets, rather than being agents with a single molecular target that follow a traditional one target-one drug-one disease model [7]. Initially Dasatinib (D), an anti-cancer agent, was identified as senolytic against human senescent adipocyte progenitors, and Quercetin (Q), a naturally occurring flavonoid, senolytic against umbilical vein endothelial cells, with the combination, D+Q, being senolytic against a broader range of senescent cell types than either alone [45]. Fisetin, a naturally occurring flavanoid related to Q, was found to also be senolytic [46]. Later, we and another group identified Navitoclax, which has several BCL-2-related targets, as being a potential senolytic [7, 27, 45]. However, Navitoclax induces apoptosis in HUVECs but not human preadipocytes, remains to date an investigational agent that is not yet on the formulary, and can have severe off-target effects on non-senescent cells, including severe neutropenia and platelet deficiency (although there has been progress in devising a solution for the latter) [46][45][47][7]. After the first senolytics were identified using the mechanism-based approach, a second approach for identifying senolytics, using high-throughput drug library screens, is currently being developed and appears to be successful [7][48]. The first FDA-registered clinical trial of the combination of D+Q was a cancer trial [42]. This 2006 trial included patients who had B lymphoma as well as other certain types of leukemia. While senolytics specifically clear senescent cells, if the Unitary Theory of Fundamental Aging Processes is correct, senolytics could act to alleviate adverse effects of other fundamental aging mechanisms.
4. Preclinical trials
4.1. Senolytics alleviate multiple disorders in mouse models
Senolytics have been tested in a variety of different preclinical models [7]. In naturally-aged mice, treatment with D+Q improved cardiac function; in radiation-exposed mice, D+Q improved exercise capacity; in progeroid Ercc1(−/Δ) (accelerated aging) mice, D+Q delayed age-related dysfunction [45][39]. Idiopathic pulmonary fibrosis (IPF) is characterized by senescent cell accumulation in the lungs, and in bleomycin-injured mice, D+Q effectively killed senescent fibroblasts, leading to improvement in physical and pulmonary function [49]. In obese mice, D+Q alleviated metabolic and adipose tissue dysfunction [15]. D+Q also helped decrease senescent cell burden in atherosclerosis mouse models [39]. Furthermore, in osteoporosis mouse models, treatment with D+Q increased bone mass and strength [40].
Fisetin has also had favorable preclinical results. Fisetin induced apoptosis in senescent human umbilical vein endothelial cells (HUVECs) [49]. In wild-type mice, Fisetin decreased age-related pathology and increased lifespan [50]. There is also growing preclinical data about the effectiveness of Fisetin in alleviating neurodegenerative disorders, including Alzheimer’s disease, Parkinson’s disease, ALS, and Huntington’s disease [51].
4.2. Translation to humans
Because of the promising preclinical data showing the efficacy of senolytics on a variety of different disease models, senolytics were considered for clinical trials. Due to the novel nature of senolytic agents and the unknown risk/benefit ratio, senolytics were first considered for clinical trials in patients with serious conditions including diabetes, IPF, and osteoporosis [7]. Fisetin and Quercetin, naturally occurring flavanoids, have favorable safety profiles and therefore have been considered for a variety of clinical trials, including with healthy older adults. Dasatinib has been used in clinical practice since 2006 and has a well-established safety profile. With approval from the Food and Drug Administration (FDA), the first clinical trials using Fisetin and D+Q have begun and many more are in the planning stages.
5. Current and Planned Clinical Trials
5.1. Active trials, completed trials, and early results
The first in-human trial of senolytics showed improved physical function in idiopathic pulmonary fibrosis (IPF) patients after treatment with D+Q [52]. Another early clinical trial showed treatment with D+Q reduces senescent cell burden in adipose tissue of diabetic kidney disease patients [53]. Recently, an open label pilot phase 1 trial of D+Q for Alzheimer’s disease, SToMP-AD [54], has been completed with results that are about to be submitted for publication and the Phase 2 component of this trial is about to begin. Currently active trials with senolytics include, but are not limited to, those listed in Table 1.
Table 1.
Active Translational Geroscience Network (TGN) Trials with Senolytics.
| Dasatinib + Quercetin (D+Q) Population/Disease |
Lead Site | ClinicalTrials.gov # |
|---|---|---|
| Senescent Cell Burden in Hematopoietic Stem Cell Transplant Survivors | • Mayo Clinic | NCT02652052 |
| Senolytic Therapy to Modulate Progression of Alzheimer’s Disease | • The University of Texas Health Science Center at San Antonio | NCT04063124 |
| Senescence, Frailty, and Mesenchymal Stem Cell Functionality in Chronic Kidney Disease: Effect of Senolytic Agents | • Mayo Clinic | NCT02848131 |
| Fisetin (F) | ||
| Use of Senolytic and Anti-Fibrotic Agents to Improve the Beneficial Effect of Bone Marrow Stem Cells for Osteoarthritis | • The Steadman Clinic | NCT04815902 |
| Senolytic Drugs Attenuate Osteoarthritis-Related Articular Cartilage Degeneration: A Clinical Trial | • The Steadman Clinic | NCT04210986 |
| COVID-FISETIN: Pilot in SaRS-CoV-2 of Fisetin to Alleviate Dysfunction and Inflammation | • Mayo Clinic | NCT04476953 |
| Alleviation by Fisetin of Frailty, Inflammation, and Related Measures in Older Adults (AFFIRM-LITE) | • Mayo Clinic | NCT03675724 |
| Alleviation by Fisetin of Frailty, Inflammation, and Related Measures in Older Women (AFFIRM) | • Mayo Clinic | NCT03430037 |
| Inflammation and Stem Cells in Diabetic and Chronic Kidney Disease | • Mayo Clinic | NCT03325322 |
| Both Arms | ||
| Targeting Cellular Senescence with Senolytics to Improve Skeletal Health in Older Humans | • Mayo Clinic | NCT04313634 |
5.2. Planned trials
The first clinical trials provided safety and early efficacy data [52, 53]. Since the initial data was published, many trials using senolytics have been planned (Table 2). Most recently, our team found application of senolytics (both Fisetin and D+Q) effective on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mouse models [55]. With the COVID-19 pandemic still very much present, several trials for COVID-19 application are in the planning stages, with one ongoing for hospitalized patients (COVID-FISETIN: Pilot in SARS-CoV-2 of Fisetin to Alleviate Dysfunction and Inflammation, NCT04476953).
Table 2.
Planned Translational Geroscience Network (TGN) Trials with Senolytics.
| Dasatinib + Quercetin (D+Q) Population/Disease |
Lead Site | ClinicalTrials.gov # |
|---|---|---|
| ALSENLITE: An Open-Label Pilot Study of Senolytics for Alzheimer’s Disease | • Mayo Clinic | NCT04785300 |
| Senolytic Therapy to Modulate the Progression of Alzheimer’s Disease (SToMP-AD) Study | • Wake Forest Health Sciences | NCT04685590 |
| Fisetin (F) | ||
| COVFIS-HOME: COVID-19 Pilot Study of Fisetin to Alleviate Dysfunction and Decrease Complications | • Mayo Clinic | NCT04771611 |
| COVID-FIS: Pilot in COVID-19 (SARS-CoV-2) of Fisetin in Older Adults in Nursing Homes | • Mayo Clinic | NCT04537299 |
| Both Arms | ||
| An Open-Label Intervention Trial to Reduce Senescence and Improve Frailty in Adult Survivors of Childhood Cancer | • St. Jude Children’s Research Hospital | NCT04733534 |
6. Translational Geroscience Network
Due to the need for effective interventions and the slow nature of preclinical and clinical trials, a Translational Geroscience Network (TGN) was formed to conduct clinical trials of interventions targeting fundamental aging mechanisms in parallel (Figure 2). The TGN comprises 8 institutions: Mayo Clinic, Harvard, John Hopkins, Wake Forest, Universities of Minnesota, Michigan, and Connecticut, and University of Texas Health Sciences Center at San Antonio, as well as partners, including St. Jude Children’s Cancer Hospital and the Steadman Clinic. There are several subcommittees in the TGN including a Data Management and Statistical Analysis Subcommittee, the Diversity, Equity, and Inclusion Subcommittee, and Regulatory, Website, Biobanking, and Facility for Geroscience Analysis (FGA) Subcommittees.
Figure 2:
The Translational Geroscience Network (TGN) has subcommittees to achieve the four aims of the network.
The FGA is a laboratory core that develops and conducts innovative new assays for aging and disease markers. These tests are currently being used to assess target engagement and efficacy of senolytics in preclinical and early phase clinical trials. The FGA is exploring new markers of fundamental aging mechanisms and related diseases. With unified infrastructure and resources across multiple institutions, the TGN may be able to quickly navigate obstacles of initiating and completing trials of interventions targeting basic aging mechanisms, including senolytics.
7. Conclusions and Future Directions
The hallmarks of aging may be a mechanistic contributor to age-related phenotypes and multiple diseases and disorders. If the Unitary Theory of Fundamental Aging Mechanisms is true, intervening against one pillar will affect most, if not all, of the other pillars of aging. Cellular senescence may be a good interventional target and it is hoped that senotherapeutics, including senolytics and senomorphics, will prove to be efficacious. In future trials, the impact of combining multiple aging therapeutics to treat different aging processes needs to be determined. These combinations may have reduced efficacy or result in additive or synergistic effects compared to individual interventions for targeting multiple age-related or chronic conditions and diseases. As the number of people suffering from age-related disease and disorders continues to increase, it is becoming even more important to establish collaborations and infrastructure like the TGN to facilitate and accelerate translation of agents that target fundamental aging mechanisms into clinical practice.
Highlights.
Accumulation of senescent cells is associated with age-related diseases and disorders
Senolytics, including senolytics and senomorphics, are potential interventions for delaying, preventing, or treating age-related dysfunction
Senolytics show efficacy in pre-clinical and early clinical trials
Running trials in parallel across several institutions as modeled by the Translational Geroscience Network (TGN) could fast-track clinical application of senolytics
Acknowledgements
This work was supported by the National Institutes of Health (grants R37 AG13925, R33 AG61456, R01 AG072301, R01 AG61414, P01 AG62413, and UH3 AG56933), Robert and Arlene Kogod, the Connor Fund, Robert J. and Theresa W. Ryan, and the Noaber Foundation.
Abbreviations
- D
Dasatinib
- D+Q
Dasatinib and Quercetin
- FDA
Food and Drug Administration
- FGA
Facility for Geroscience Analysis
- HUVECs
Human Umbilical Vein Endothelial Cells
- IPF
Idiopathic Pulmonary Fibrosis
- Q
Quercetin
- SARS-CoV-2
Severe Acute Respiratory Syndrome Coronavirus-2
- SASP
Senescence-Associated Secretory Phenotype
- SCAP
Senescent Cell Anti-Apoptotic Pathway
- TGN
Translational Geroscience Network
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Niccoli T and Partridge L, Ageing as a risk factor for disease. Curr Biol, 2012. 22(17): p. R741–52. [DOI] [PubMed] [Google Scholar]
- [2].Gorina Y, et al. , Trends in causes of death among older persons in the United States. Aging Trends, 2005(6): p. 1–12. [PubMed] [Google Scholar]
- [3].McHugh D and Gil J, Senescence and aging: Causes, consequences, and therapeutic avenues. J Cell Biol, 2018. 217(1): p. 65–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Krishnamurthy J, et al. , Ink4a/Arf expression is a biomarker of aging. J Clin Invest, 2004. 114(9): p. 1299–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Lopez-Otin C, et al. , The hallmarks of aging. Cell, 2013. 153(6): p. 1194–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Kennedy BK, et al. , Geroscience: linking aging to chronic disease. Cell, 2014. 159(4): p. 709–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Kirkland JL and Tchkonia T, Senolytic drugs: from discovery to translation. J Intern Med, 2020. 288(5): p. 518–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Tchkonia T, Palmer AK, and Kirkland JL, New Horizons: Novel Approaches to Enhance Healthspan Through Targeting Cellular Senescence and Related Aging Mechanisms. J Clin Endocrinol Metab, 2021. 106(3): p. e1481–e1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Childs BG, et al. , Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov, 2017. 16(10): p. 718–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Yoon YS, et al. , Complex II defect via down-regulation of iron-sulfur subunit induces mitochondrial dysfunction and cell cycle delay in iron chelation-induced senescence-associated growth arrest. J Biol Chem, 2003. 278(51): p. 51577–86. [DOI] [PubMed] [Google Scholar]
- [11].Moiseeva O, et al. , Mitochondrial dysfunction contributes to oncogene-induced senescence. Mol Cell Biol, 2009. 29(16): p. 4495–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Stockl P, et al. , Sustained inhibition of oxidative phosphorylation impairs cell proliferation and induces premature senescence in human fibroblasts. Exp Gerontol, 2006. 41(7): p. 674–82. [DOI] [PubMed] [Google Scholar]
- [13].Palmer AK, Tchkonia T, and Kirkland JL, Senolytics: Potential for Alleviating Diabetes and Its Complications. Endocrinology, 2021. 162(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Chini CCS, et al. , CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD(+) and NMN levels. Nat Metab, 2020. 2(11): p. 1284–1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Palmer AK, et al. , Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell, 2019. 18(3): p. e12950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Childs BG, et al. , Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat Med, 2015. 21(12): p. 1424–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Molofsky AV, et al. , Increasing p16INK4a expression decreases forebrain progenitors and neurogenesis during ageing. Nature, 2006. 443(7110): p. 448–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Conboy IM and Rando TA, Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches. Cell Cycle, 2012. 11(12): p. 2260–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Salminen A and Kaarniranta K, AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res Rev, 2012. 11(2): p. 230–41. [DOI] [PubMed] [Google Scholar]
- [20].Gorgoulis VG and Halazonetis TD, Oncogene-induced senescence: the bright and dark side of the response. Curr Opin Cell Biol, 2010. 22(6): p. 816–27. [DOI] [PubMed] [Google Scholar]
- [21].Lidzbarsky G, et al. , Genomic Instabilities, Cellular Senescence, and Aging: In Vitro, In Vivo and Aging-Like Human Syndromes. Front Med (Lausanne), 2018. 5: p. 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Allsopp RC, et al. , Telomere length predicts replicative capacity of human fibroblasts. Proc Natl Acad Sci U S A, 1992. 89(21): p. 10114–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Greider CW, Telomerase activity, cell proliferation, and cancer. Proc Natl Acad Sci U S A, 1998. 95(1): p. 90–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Herbig U, et al. , Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol Cell, 2004. 14(4): p. 501–13. [DOI] [PubMed] [Google Scholar]
- [25].Bodnar AG, et al. , Extension of life-span by introduction of telomerase into normal human cells. Science, 1998. 279(5349): p. 349–52. [DOI] [PubMed] [Google Scholar]
- [26].Cuervo AM and Macian F, Autophagy and the immune function in aging. Curr Opin Immunol, 2014. 29: p. 97–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Chang J, et al. , Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med, 2016. 22(1): p. 78–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Hayflick L and Moorhead PS, The serial cultivation of human diploid cell strains. Exp Cell Res, 1961. 25: p. 585–621. [DOI] [PubMed] [Google Scholar]
- [29].Wang E, Senescent human fibroblasts resist programmed cell death, and failure to suppress bcl2 is involved. Cancer Res, 1995. 55(11): p. 2284–92. [PubMed] [Google Scholar]
- [30].Calcinotto A, et al. , Cellular Senescence: Aging, Cancer, and Injury. Physiol Rev, 2019. 99(2): p. 1047–1078. [DOI] [PubMed] [Google Scholar]
- [31].Wissler Gerdes EO, et al. , Discovery, development, and future application of senolytics: theories and predictions. FEBS J, 2020. 287(12): p. 2418–2427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Kirkland JL and Tchkonia T, Cellular Senescence: A Translational Perspective. EBioMedicine, 2017. 21: p. 21–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Lewis-McDougall FC, et al. , Aged-senescent cells contribute to impaired heart regeneration. Aging Cell, 2019. 18(3): p. e12931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Prata L, et al. , Senescent cell clearance by the immune system: Emerging therapeutic opportunities. Semin Immunol, 2018. 40: p. 101275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Tchkonia T, et al. , Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest, 2013. 123(3): p. 966–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Iske J, et al. , Senolytics prevent mt-DNA-induced inflammation and promote the survival of aged organs following transplantation. Nat Commun, 2020. 11(1): p. 4289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Xu M, et al. , Senolytics improve physical function and increase lifespan in old age. Nat Med, 2018. 24(8): p. 1246–1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Xu M, et al. , Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife, 2015. 4: p. e12997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Roos CM, et al. , Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell, 2016. 15(5): p. 973–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Farr JN, et al. , Targeting cellular senescence prevents age-related bone loss in mice. Nat Med, 2017. 23(9): p. 1072–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Birch J and Gil J, Senescence and the SASP: many therapeutic avenues. Genes Dev, 2020. 34(23–24): p. 1565–1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Song S, et al. , Targeting Senescent Cells for a Healthier Aging: Challenges and Opportunities. Adv Sci (Weinh), 2020. 7(23): p. 2002611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Zhu Y, et al. , The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell, 2015. 14(4): p. 644–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Subramanian A, et al. , GSEA-P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics, 2007. 23(23): p. 3251–3. [DOI] [PubMed] [Google Scholar]
- [45].Zhu Y, et al. , Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell, 2016. 15(3): p. 428–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Zhu Y, et al. , New agents that target senescent cells: the flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging (Albany NY), 2017. 9(3): p. 955–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].He Y, et al. , Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity. Nat Commun, 2020. 11(1): p. 1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Munoz-Espin D, et al. , A versatile drug delivery system targeting senescent cells. EMBO Mol Med, 2018. 10(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Schafer MJ, et al. , Cellular senescence mediates fibrotic pulmonary disease. Nat Commun, 2017. 8: p. 14532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Yousefzadeh MJ, et al. , Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine, 2018. 36: p. 18–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Maher P, Preventing and Treating Neurological Disorders with the Flavonol Fisetin. Brain Plast, 2021. 6(2): p. 155–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Justice JN, et al. , Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine, 2019. 40: p. 554–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Hickson LJ, et al. , Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine, 2019. 47: p. 446–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Gonzales MM, Garbarino VR, Zilli EM, Peterson RC, Kirkland JL, Tchkonia T, Musi N, Seshadri S, Craft S, Orr ME, Senolytic therapy to modulate the progression of Alzheimer’s disease (SToMP-AD): A pilot clinical trial. Journal of Prevention of Alzheimer’s Disease, [in press]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Camell CD, et al. , Senolytics reduce coronavirus-related mortality in old mice. Science, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]


