Skip to main content
Top
Published in: Advances in Therapy 4/2020

Open Access 01-04-2020 | Review

Cellular Senescence as a Therapeutic Target for Age-Related Diseases: A Review

Authors: Mateo Amaya-Montoya, Agustín Pérez-Londoño, Valentina Guatibonza-García, Andrea Vargas-Villanueva, Carlos O. Mendivil

Published in: Advances in Therapy | Issue 4/2020

Login to get access

Abstract

Life expectancy has increased substantially over the last few decades, leading to a worldwide increase in the prevalence and burden of aging-associated diseases. Recent evidence has proven that cellular senescence contributes substantially to the development of these disorders. Cellular senescence is a state of cell cycle arrest with suppressed apoptosis and concomitant secretion of multiple bioactive factors (the senescence-associated secretory phenotype—SASP) that plays a physiological role in embryonic development and healing processes. However, DNA damage and oxidative stress that occur during aging cause the accumulation of senescent cells, which through their SASP bring about deleterious effects on multiple organ and systemic functions. Ablation of senescent cells through genetic or pharmacological means leads to improved life span and health span in animal models, and preliminary evidence suggests it may also have a positive impact on human health. Thus, strategies to reduce or eliminate the burden of senescent cells or their products have the potential to impact multiple clinical outcomes with a single intervention. In this review, we touch upon the basics of cell senescence and summarize the current state of development of therapies against cell senescence for human use.
Literature
2.
go back to reference Mason A, Lee R, Abrigo M, Lee SH. Support ratios and demographic dividens: estimates for the world. United Nations, Department of Economic and Social Affairs, Population Division. Technical paper no. 2017/1. New York; 2017. Mason A, Lee R, Abrigo M, Lee SH. Support ratios and demographic dividens: estimates for the world. United Nations, Department of Economic and Social Affairs, Population Division. Technical paper no. 2017/1. New York; 2017.
3.
go back to reference World Health Organization. World health statistics overview 2019: monitoring health for SDG, sustainable development goals. License: CC BY-NC-SA 3.0 IGO. Geneva; 2019. World Health Organization. World health statistics overview 2019: monitoring health for SDG, sustainable development goals. License: CC BY-NC-SA 3.0 IGO. Geneva; 2019.
5.
go back to reference Institute for Health and Evaluation (IHME). Findings from the global burden of disease study 2017. Seattle: IHME; 2018. Institute for Health and Evaluation (IHME). Findings from the global burden of disease study 2017. Seattle: IHME; 2018.
7.
go back to reference Shay JW, Wright WE. Hayflick, his limit, and cellular ageing. Nat Rev Mol Cell Biol. 2000;1:72–6.PubMed Shay JW, Wright WE. Hayflick, his limit, and cellular ageing. Nat Rev Mol Cell Biol. 2000;1:72–6.PubMed
8.
go back to reference Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.PubMed Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.PubMed
9.
go back to reference Chen Q, Fischer A, Reagan JD, Yan LJ, Ames BN. Oxidative DNA damage and senescence of human diploid fibroblast cells. Proc Natl Acad Sci USA. 1995;92:4337–411.PubMedPubMedCentral Chen Q, Fischer A, Reagan JD, Yan LJ, Ames BN. Oxidative DNA damage and senescence of human diploid fibroblast cells. Proc Natl Acad Sci USA. 1995;92:4337–411.PubMedPubMedCentral
10.
go back to reference Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell. 1997;88:593–602.PubMed Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell. 1997;88:593–602.PubMed
11.
go back to reference Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature. 2004;432:307–15.PubMed Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature. 2004;432:307–15.PubMed
13.
go back to reference Kang C. Senolytics and senostatics: a two-pronged approach to target cellular senescence for delaying aging and age-related diseases. Mol Cells. 2019;42:821–7.PubMedPubMedCentral Kang C. Senolytics and senostatics: a two-pronged approach to target cellular senescence for delaying aging and age-related diseases. Mol Cells. 2019;42:821–7.PubMedPubMedCentral
14.
go back to reference Di Leonardo A, Linke SP, Clarkin K, et al. DNA damage triggers a prolonged p53-dependent G1 arrest and long-term induction of Cip1 in normal human fibroblasts. Genes Dev. 1994;8:2540–51.PubMed Di Leonardo A, Linke SP, Clarkin K, et al. DNA damage triggers a prolonged p53-dependent G1 arrest and long-term induction of Cip1 in normal human fibroblasts. Genes Dev. 1994;8:2540–51.PubMed
15.
go back to reference Ogryzko VV, Hirai TH, Russanova VR, et al. Human fibroblast commitment to a senescence-like state in response to histone deacetylase inhibitors is cell cycle dependent. Mol Cell Biol. 1996;16:5210–8.PubMedPubMedCentral Ogryzko VV, Hirai TH, Russanova VR, et al. Human fibroblast commitment to a senescence-like state in response to histone deacetylase inhibitors is cell cycle dependent. Mol Cell Biol. 1996;16:5210–8.PubMedPubMedCentral
16.
go back to reference Terzi MY, Izmirli M, Gogebakan B. The cell fate: senescence or quiescence. Mol Biol Rep. 2016;43:1213–20.PubMed Terzi MY, Izmirli M, Gogebakan B. The cell fate: senescence or quiescence. Mol Biol Rep. 2016;43:1213–20.PubMed
17.
go back to reference Patel PL, Suram A, Mirani N, et al. Derepression of hTERT gene expression promotes escape from oncogene-induced cellular senescence. Proc Natl Acad Sci USA. 2016;113:E5024–E50335033.PubMedPubMedCentral Patel PL, Suram A, Mirani N, et al. Derepression of hTERT gene expression promotes escape from oncogene-induced cellular senescence. Proc Natl Acad Sci USA. 2016;113:E5024–E50335033.PubMedPubMedCentral
18.
go back to reference Milanovic M, Fan DNY, Belenki D. Senescence-associated reprogramming promotes cancer stemness. Nature. 2018;553:96–100.PubMed Milanovic M, Fan DNY, Belenki D. Senescence-associated reprogramming promotes cancer stemness. Nature. 2018;553:96–100.PubMed
19.
go back to reference Chen W, Kang J, Xia J, et al. p53-related apoptosis resistance and tumor suppression activity in UVB-induced premature senescent human skin fibroblasts. Int J Mol Med. 2008;21:645–53.PubMed Chen W, Kang J, Xia J, et al. p53-related apoptosis resistance and tumor suppression activity in UVB-induced premature senescent human skin fibroblasts. Int J Mol Med. 2008;21:645–53.PubMed
20.
go back to reference Childs BG, Baker DJ, Kirkland JL. Senescence and apoptosis: dueling or complementary cell fates? EMBO Rep. 2014;15:1139–53.PubMedPubMedCentral Childs BG, Baker DJ, Kirkland JL. Senescence and apoptosis: dueling or complementary cell fates? EMBO Rep. 2014;15:1139–53.PubMedPubMedCentral
21.
go back to reference Myrianthopoulos V, Evangelou K, Vasileiou PVS, et al. Senescence and senotherapeutics: a new field in cancer therapy. Pharmacol Ther. 2019;193:31–49.PubMed Myrianthopoulos V, Evangelou K, Vasileiou PVS, et al. Senescence and senotherapeutics: a new field in cancer therapy. Pharmacol Ther. 2019;193:31–49.PubMed
22.
go back to reference Salotti J, Johnson PF. Regulation of senescence and the SASP by the transcription factor C/EBPβ. Exp Gerontol. 2019;128:110752.PubMed Salotti J, Johnson PF. Regulation of senescence and the SASP by the transcription factor C/EBPβ. Exp Gerontol. 2019;128:110752.PubMed
23.
go back to reference Da Silva-Álvarez S, Picallos-Rabina P, Antelo-Iglesias L. The development of cell senescence. Exp Gerontol. 2019;128:110742.PubMed Da Silva-Álvarez S, Picallos-Rabina P, Antelo-Iglesias L. The development of cell senescence. Exp Gerontol. 2019;128:110742.PubMed
24.
go back to reference Acosta JC, O'Loghlen A, Banito A, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133:1006–188.PubMed Acosta JC, O'Loghlen A, Banito A, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133:1006–188.PubMed
25.
go back to reference Acosta JC, Banito A, Wuestefeld T, et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol. 2013;15:978–90.PubMedPubMedCentral Acosta JC, Banito A, Wuestefeld T, et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol. 2013;15:978–90.PubMedPubMedCentral
26.
go back to reference Demaria M, Ohtani N, Youssef SA, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell. 2014;31:722–33.PubMedPubMedCentral Demaria M, Ohtani N, Youssef SA, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev Cell. 2014;31:722–33.PubMedPubMedCentral
27.
go back to reference Mosteiro L, Pantoja C, Alcazar N, et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science. 2016;354:aaf.4445. Mosteiro L, Pantoja C, Alcazar N, et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science. 2016;354:aaf.4445.
28.
go back to reference Storer M, Mas A, Robert-Moreno A, et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell. 2013;155:1119–30.PubMed Storer M, Mas A, Robert-Moreno A, et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell. 2013;155:1119–30.PubMed
29.
go back to reference Franceschi C, Bonafè M, Valensin S, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244–54.PubMed Franceschi C, Bonafè M, Valensin S, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244–54.PubMed
31.
go back to reference Valenzuela CA, Quintanilla R, Moore-Carrasco R. The potential role of senescence as a modulator of platelets and tumorigenesis. Front Oncol. 2017;7:188.PubMedPubMedCentral Valenzuela CA, Quintanilla R, Moore-Carrasco R. The potential role of senescence as a modulator of platelets and tumorigenesis. Front Oncol. 2017;7:188.PubMedPubMedCentral
32.
go back to reference Parrinello S, Coppe JP, Krtolica A, et al. Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J Cell Sci. 2005;118:485–96.PubMed Parrinello S, Coppe JP, Krtolica A, et al. Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J Cell Sci. 2005;118:485–96.PubMed
33.
go back to reference Angelini PD, Zacarias Fluck MF, Pedersen K, et al. Constitutive HER2 signaling promotes breast cancer metastasis through cellular senescence. Cancer Res. 2013;73:450–8.PubMed Angelini PD, Zacarias Fluck MF, Pedersen K, et al. Constitutive HER2 signaling promotes breast cancer metastasis through cellular senescence. Cancer Res. 2013;73:450–8.PubMed
36.
go back to reference Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019;19:439–53.PubMed Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019;19:439–53.PubMed
37.
go back to reference Galbiati A, Beauséjour C, d’Adda di Fagagna F. A novel single-cell method provides direct evidence persistent DNA damage in senescent cells and aged mammalian tissues. Aging Cell. 2017;16:422–7.PubMedPubMedCentral Galbiati A, Beauséjour C, d’Adda di Fagagna F. A novel single-cell method provides direct evidence persistent DNA damage in senescent cells and aged mammalian tissues. Aging Cell. 2017;16:422–7.PubMedPubMedCentral
39.
go back to reference Rogakou E, Boon C, Redon C, et al. Megabase chromatin domains involved in DNA double-strand breaks in vivo. J Cell Biol. 1999;146:905–15.PubMedPubMedCentral Rogakou E, Boon C, Redon C, et al. Megabase chromatin domains involved in DNA double-strand breaks in vivo. J Cell Biol. 1999;146:905–15.PubMedPubMedCentral
40.
go back to reference Sedelnikova OA, Horikawa I, Zimonjic DB, et al. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double-strand breaks. Nat Cell Biol. 2004;6:168–71.PubMed Sedelnikova OA, Horikawa I, Zimonjic DB, et al. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double-strand breaks. Nat Cell Biol. 2004;6:168–71.PubMed
41.
go back to reference Alimonti A, Nardella C, Chen Z. A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis. J Clin Investig. 2010;120:681–93.PubMedPubMedCentral Alimonti A, Nardella C, Chen Z. A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis. J Clin Investig. 2010;120:681–93.PubMedPubMedCentral
42.
go back to reference Demidenko Z, Blagosklonny M. Growth stimulation leads to cellular senescence when the cell cycle is blocked. Cell Cycle. 2008;7:3355–61.PubMed Demidenko Z, Blagosklonny M. Growth stimulation leads to cellular senescence when the cell cycle is blocked. Cell Cycle. 2008;7:3355–61.PubMed
43.
go back to reference Shenghui H, Sharpless N. Senescence in health and disease. Cell. 2017;169:1000–111. Shenghui H, Sharpless N. Senescence in health and disease. Cell. 2017;169:1000–111.
44.
go back to reference Pluquet O, Pourtier A, Abbadie C. The unfolded protein response and cellular senescence. A review in the theme: cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. Am J Physiol Cell Physiol. 2014;308:C415–C425425.PubMed Pluquet O, Pourtier A, Abbadie C. The unfolded protein response and cellular senescence. A review in the theme: cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. Am J Physiol Cell Physiol. 2014;308:C415–C425425.PubMed
45.
go back to reference Burton D, Faragher R. Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology. 2018;19:447–59.PubMedPubMedCentral Burton D, Faragher R. Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology. 2018;19:447–59.PubMedPubMedCentral
46.
go back to reference Minamino T, Orimo M, Shimizu I, et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med. 2009;15:1082–8.PubMed Minamino T, Orimo M, Shimizu I, et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat Med. 2009;15:1082–8.PubMed
47.
go back to reference Higuchi M, Dusting G, Peshavariya H, et al. Differentiation of human adipose-derived stem cells into fat involves reactive oxygen species and forkhead box-O1 mediated upregulation of antioxidant enzymes. Stem Cells Dev. 2013;22:878–88.PubMed Higuchi M, Dusting G, Peshavariya H, et al. Differentiation of human adipose-derived stem cells into fat involves reactive oxygen species and forkhead box-O1 mediated upregulation of antioxidant enzymes. Stem Cells Dev. 2013;22:878–88.PubMed
48.
go back to reference Dodig S, Cepelak I, Pavic I. Hallmarks of senescence and aging. Biochem Med (Zagreb). 2019;29:1–15. Dodig S, Cepelak I, Pavic I. Hallmarks of senescence and aging. Biochem Med (Zagreb). 2019;29:1–15.
49.
go back to reference Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15:482–96.PubMed Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15:482–96.PubMed
50.
go back to reference Wang E, Gundersen D. Increased organization of cytoskeleton accompanying the aging of human fibroblasts in vitro. Exp Cell Res. 1984;154:191–202.PubMed Wang E, Gundersen D. Increased organization of cytoskeleton accompanying the aging of human fibroblasts in vitro. Exp Cell Res. 1984;154:191–202.PubMed
51.
go back to reference Nishio K, Inoue A, Qiao S, et al. Senescence and cytoskeleton: overproduction of vimentin induces senescent-like morphology in human fibroblasts. Histochem Cell Biol. 2001;116:321–7.PubMed Nishio K, Inoue A, Qiao S, et al. Senescence and cytoskeleton: overproduction of vimentin induces senescent-like morphology in human fibroblasts. Histochem Cell Biol. 2001;116:321–7.PubMed
52.
go back to reference Ogrodnik M, Salmonowicz H, Jurk D. Expansion and cell-cycle arrest: common denominators of cellular senescence. Trends Biochem Sci. 2019;44:996–1008.PubMed Ogrodnik M, Salmonowicz H, Jurk D. Expansion and cell-cycle arrest: common denominators of cellular senescence. Trends Biochem Sci. 2019;44:996–1008.PubMed
53.
go back to reference Kurz DJ, Decary S, Hong Y, et al. Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells. J Cell Sci. 2000;113:3613–22.PubMed Kurz DJ, Decary S, Hong Y, et al. Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells. J Cell Sci. 2000;113:3613–22.PubMed
55.
go back to reference Jones SA, Scheller J, Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J Clin Investig. 2011;121:3375–83.PubMedPubMedCentral Jones SA, Scheller J, Rose-John S. Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling. J Clin Investig. 2011;121:3375–83.PubMedPubMedCentral
56.
go back to reference Kuilman T, Michaloglou C, Vredeveld LC, et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133:1019–31.PubMed Kuilman T, Michaloglou C, Vredeveld LC, et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008;133:1019–31.PubMed
57.
go back to reference Kumar S, Millis AJ, Baglioni C. Expression of interleukin 1-inducible genes and production of interleukin 1 by aging human fibroblasts. Proc Natl Acad Sci USA. 1992;89:4683–7.PubMedPubMedCentral Kumar S, Millis AJ, Baglioni C. Expression of interleukin 1-inducible genes and production of interleukin 1 by aging human fibroblasts. Proc Natl Acad Sci USA. 1992;89:4683–7.PubMedPubMedCentral
58.
go back to reference Orjalo AV, Bhaumik D, Gengler BK, et al. Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc Natl Acad Sci USA. 2009;106:17031–6.PubMedPubMedCentral Orjalo AV, Bhaumik D, Gengler BK, et al. Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc Natl Acad Sci USA. 2009;106:17031–6.PubMedPubMedCentral
59.
go back to reference Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity. 2000;12:121–7.PubMed Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity. 2000;12:121–7.PubMed
60.
go back to reference Coppé JP, Desprez PY, Krtolica A, et al. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.PubMedPubMedCentral Coppé JP, Desprez PY, Krtolica A, et al. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.PubMedPubMedCentral
61.
go back to reference Chen J, Mo R, Lescure PA, et al. Aging is associated with increased T-cell chemokine expression in C57BL/6 mice. J Gerontol A Biol Sci Med Sci. 2003;58:975–83.PubMed Chen J, Mo R, Lescure PA, et al. Aging is associated with increased T-cell chemokine expression in C57BL/6 mice. J Gerontol A Biol Sci Med Sci. 2003;58:975–83.PubMed
62.
go back to reference Sasaki M, Miyakoshi M, Sato Y, et al. Chemokine–chemokine receptor CCL2–CCR2 and CX3CL1–CX3CR1 axis may play a role in the aggravated inflammation in primary biliary cirrhosis. Dig Dis Sci. 2014;59:358–64.PubMed Sasaki M, Miyakoshi M, Sato Y, et al. Chemokine–chemokine receptor CCL2–CCR2 and CX3CL1–CX3CR1 axis may play a role in the aggravated inflammation in primary biliary cirrhosis. Dig Dis Sci. 2014;59:358–64.PubMed
63.
go back to reference Bachstetter AD, Morganti JM, Jernberg J, et al. Fractalkine and CX 3 CR1 regulate hippocampal neurogenesis in adult and aged rats. Neurobiol Aging. 2011;32:2030–44.PubMed Bachstetter AD, Morganti JM, Jernberg J, et al. Fractalkine and CX 3 CR1 regulate hippocampal neurogenesis in adult and aged rats. Neurobiol Aging. 2011;32:2030–44.PubMed
64.
go back to reference Mecca C, Giambanco I, Donato R. Microglia and aging: the role of the TREM2–DAP12 and CX3CL1–CX3CR1 axes. Int J Mol Sci. 2018;19:E318.PubMed Mecca C, Giambanco I, Donato R. Microglia and aging: the role of the TREM2–DAP12 and CX3CL1–CX3CR1 axes. Int J Mol Sci. 2018;19:E318.PubMed
65.
go back to reference Pollak M. Insulin and insulin-like growth factor signaling in neoplasia. Nat Rev Cancer. 2008;8:915–28.PubMed Pollak M. Insulin and insulin-like growth factor signaling in neoplasia. Nat Rev Cancer. 2008;8:915–28.PubMed
66.
go back to reference Tran D, Bergholz J, Zhang H, et al. Insulin-like growth factor-1 regulates the SIRT1-p53 pathway in cellular senescence. Aging Cell. 2014;13:669–78.PubMedPubMedCentral Tran D, Bergholz J, Zhang H, et al. Insulin-like growth factor-1 regulates the SIRT1-p53 pathway in cellular senescence. Aging Cell. 2014;13:669–78.PubMedPubMedCentral
67.
go back to reference Lopes-Paciencia S, Saint-Germain E, Rowell MC, et al. The senescence-associated secretory phenotype and its regulation. Cytokine. 2019;117:15–22.PubMed Lopes-Paciencia S, Saint-Germain E, Rowell MC, et al. The senescence-associated secretory phenotype and its regulation. Cytokine. 2019;117:15–22.PubMed
68.
go back to reference Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 2014;15:786–801.PubMedPubMedCentral Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 2014;15:786–801.PubMedPubMedCentral
69.
go back to reference West MD, Pereira-Smith OM, Smith JR. Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Exp Cell Res. 1989;184:138–47.PubMed West MD, Pereira-Smith OM, Smith JR. Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Exp Cell Res. 1989;184:138–47.PubMed
70.
go back to reference Millis AJ, Hoyle M, McCue HM, et al. Differential expression of metalloproteinase and tissue inhibitor of metalloproteinase genes in aged human fibroblasts. Exp Cell Res. 1992;201:373–9.PubMed Millis AJ, Hoyle M, McCue HM, et al. Differential expression of metalloproteinase and tissue inhibitor of metalloproteinase genes in aged human fibroblasts. Exp Cell Res. 1992;201:373–9.PubMed
71.
go back to reference Hassona Y, Cirillo N, Prime HK, et al. Senescent cancer-associated fibroblasts secrete active MMP-2 that promotes keratinocyte dis-cohesion and invasion. Br J Cancer. 2014;111:1230–7.PubMedPubMedCentral Hassona Y, Cirillo N, Prime HK, et al. Senescent cancer-associated fibroblasts secrete active MMP-2 that promotes keratinocyte dis-cohesion and invasion. Br J Cancer. 2014;111:1230–7.PubMedPubMedCentral
72.
go back to reference Özcan S, Alessio N, Acar MB, et al. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging (Albany NY). 2016;8:1316–29. Özcan S, Alessio N, Acar MB, et al. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging (Albany NY). 2016;8:1316–29.
73.
go back to reference Zeng G, Millis AJ. Differential regulation of collagenase and stromelysin mRNA in late passage cultures of human fibroblasts. Exp Cell Res. 1996;222:150–6.PubMed Zeng G, Millis AJ. Differential regulation of collagenase and stromelysin mRNA in late passage cultures of human fibroblasts. Exp Cell Res. 1996;222:150–6.PubMed
74.
go back to reference Quillard T, Araújo HA, Franck G. Matrix metalloproteinase-13 predominates over matrix metalloproteinase-8 as the functional interstitial collagenase in mouse atheromata. Arterioscler Thromb Vasc Biol. 2014;34:1179–86.PubMedPubMedCentral Quillard T, Araújo HA, Franck G. Matrix metalloproteinase-13 predominates over matrix metalloproteinase-8 as the functional interstitial collagenase in mouse atheromata. Arterioscler Thromb Vasc Biol. 2014;34:1179–86.PubMedPubMedCentral
75.
go back to reference McQuibban GA, Gong JH, Wong JP. Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood. 2002;100:1160–7.PubMed McQuibban GA, Gong JH, Wong JP. Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood. 2002;100:1160–7.PubMed
76.
go back to reference Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol. 2018;28:436–56.PubMed Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol. 2018;28:436–56.PubMed
77.
78.
go back to reference Karimian A, Ahmadi Y, Yousefi B. Multiple functions of P21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair (Amst). 2016;42:63–71. Karimian A, Ahmadi Y, Yousefi B. Multiple functions of P21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair (Amst). 2016;42:63–71.
80.
go back to reference Kobbe V. Cellular senescence: a view throughout organismal life. Cell Mol Life Sci. 2018;75:3553–677. Kobbe V. Cellular senescence: a view throughout organismal life. Cell Mol Life Sci. 2018;75:3553–677.
81.
go back to reference Muñoz-Espín D, Cañamero M, Maraver A, et al. Programmed cell senescence during mammalian embryonic development. Cell. 2013;155:1104–18.PubMed Muñoz-Espín D, Cañamero M, Maraver A, et al. Programmed cell senescence during mammalian embryonic development. Cell. 2013;155:1104–18.PubMed
82.
go back to reference Vicente R, Mausset-Bonnefont A, Jorgensen C, et al. Cellular senescence impact on immune cell fate and function. Aging Cell. 2016;15:400–6.PubMedPubMedCentral Vicente R, Mausset-Bonnefont A, Jorgensen C, et al. Cellular senescence impact on immune cell fate and function. Aging Cell. 2016;15:400–6.PubMedPubMedCentral
83.
go back to reference Rajagopalan S. HLA-G-mediated NK cell senescence promotes vascular remodeling: implications for reproduction. Cell Mol Immunol. 2014;11:460–6.PubMedPubMedCentral Rajagopalan S. HLA-G-mediated NK cell senescence promotes vascular remodeling: implications for reproduction. Cell Mol Immunol. 2014;11:460–6.PubMedPubMedCentral
84.
go back to reference Rajagopalan S, Long E. Cellular senescence induced by CD158d reprograms natural killer cells to promote vascular remodeling. Proc Natl Acad Sci USA. 2012;109:20596–601.PubMedPubMedCentral Rajagopalan S, Long E. Cellular senescence induced by CD158d reprograms natural killer cells to promote vascular remodeling. Proc Natl Acad Sci USA. 2012;109:20596–601.PubMedPubMedCentral
85.
go back to reference Stein GH, Drullinger LF, Soulard A, et al. Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. Mol Cell Biol. 1999;19:2109–17.PubMedPubMedCentral Stein GH, Drullinger LF, Soulard A, et al. Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. Mol Cell Biol. 1999;19:2109–17.PubMedPubMedCentral
86.
go back to reference Jun J-I, Lau L. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat Cell Biol. 2010;12:676–94.PubMedPubMedCentral Jun J-I, Lau L. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat Cell Biol. 2010;12:676–94.PubMedPubMedCentral
87.
go back to reference Baker D, Wijshake T, Tchkonia T. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–7.PubMedPubMedCentral Baker D, Wijshake T, Tchkonia T. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–7.PubMedPubMedCentral
88.
go back to reference Childs B, Baker D, Wijshake T. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354:472–7.PubMedPubMedCentral Childs B, Baker D, Wijshake T. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354:472–7.PubMedPubMedCentral
89.
go back to reference Pal A, Potjer T, Thomsen S, et al. Loss of function mutations in the cell-cycle control gene CDKN2A impact on glucose homeostasis in humans. Diabetes. 2016;65:527–33.PubMed Pal A, Potjer T, Thomsen S, et al. Loss of function mutations in the cell-cycle control gene CDKN2A impact on glucose homeostasis in humans. Diabetes. 2016;65:527–33.PubMed
90.
go back to reference González-Navarro H, Vinue A, Sanz MJ, et al. Increased dosage of Ink4/Arf protects against glucose intolerance and insulin resistance associated with aging. Aging Cell. 2013;12:102–11.PubMed González-Navarro H, Vinue A, Sanz MJ, et al. Increased dosage of Ink4/Arf protects against glucose intolerance and insulin resistance associated with aging. Aging Cell. 2013;12:102–11.PubMed
91.
go back to reference Demaria M, O’Leary MN, Chang J, et al. Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov. 2017;7:165–76.PubMed Demaria M, O’Leary MN, Chang J, et al. Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov. 2017;7:165–76.PubMed
92.
go back to reference Calcinotto A, Kohli J, Zagato E, et al. Cellular senescence: aging, cancer, and injury. Physiol Rev. 2019;99:1047–78.PubMed Calcinotto A, Kohli J, Zagato E, et al. Cellular senescence: aging, cancer, and injury. Physiol Rev. 2019;99:1047–78.PubMed
93.
go back to reference Bhat R, Crowe E, Bitto A, et al. Astrocyte senescence as a component of Alzheimer’s disease. PLoS One. 2012;7:e45069.PubMedPubMedCentral Bhat R, Crowe E, Bitto A, et al. Astrocyte senescence as a component of Alzheimer’s disease. PLoS One. 2012;7:e45069.PubMedPubMedCentral
94.
go back to reference Chinta SJ, Woods G, Demaria M, et al. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell Rep. 2017;22:930–40. Chinta SJ, Woods G, Demaria M, et al. Cellular senescence is induced by the environmental neurotoxin paraquat and contributes to neuropathology linked to Parkinson’s disease. Cell Rep. 2017;22:930–40.
95.
go back to reference Courtois-Cox S, Jones SL, Cichowski K. Many roads lead to oncogene-induced senescence. Oncogene. 2008;27:2801–9.PubMed Courtois-Cox S, Jones SL, Cichowski K. Many roads lead to oncogene-induced senescence. Oncogene. 2008;27:2801–9.PubMed
96.
go back to reference Childs BG, Gluscevic M, Baker DJ, et al. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov. 2017;16:718–35.PubMedPubMedCentral Childs BG, Gluscevic M, Baker DJ, et al. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov. 2017;16:718–35.PubMedPubMedCentral
97.
go back to reference Trujillo M, Pajvani U, Scherer P. Apoptosis through targeted activation of caspase (“ATTAC-mice”): novel mouse models of inducible and reversible tissue ablation. Cell Cycle. 2005;4:1141–5.PubMed Trujillo M, Pajvani U, Scherer P. Apoptosis through targeted activation of caspase (“ATTAC-mice”): novel mouse models of inducible and reversible tissue ablation. Cell Cycle. 2005;4:1141–5.PubMed
98.
go back to reference Clackson T, Yang W, Rozamus L. Redesigning an FKBP–ligand interface to generate chemical dimerizers with novel specificity. Proc Natl Acad Sci USA. 1998;95:10437–42.PubMedPubMedCentral Clackson T, Yang W, Rozamus L. Redesigning an FKBP–ligand interface to generate chemical dimerizers with novel specificity. Proc Natl Acad Sci USA. 1998;95:10437–42.PubMedPubMedCentral
99.
go back to reference Wang W, Wu J, Zhang Z, Tong T. Characterization of regulatory elements on the promoter region of p16INK4a that contribute to overexpression of p16 in senescent fibroblasts. J Biol Chem. 2001;276:48655–61.PubMed Wang W, Wu J, Zhang Z, Tong T. Characterization of regulatory elements on the promoter region of p16INK4a that contribute to overexpression of p16 in senescent fibroblasts. J Biol Chem. 2001;276:48655–61.PubMed
100.
go back to reference Baker DJ, Childs B, Durik M, et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature. 2016;530:184–8.PubMedPubMedCentral Baker DJ, Childs B, Durik M, et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature. 2016;530:184–8.PubMedPubMedCentral
101.
go back to reference Zhu Y, Tchkonia T, Pirtskhalava T, et al. The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14:644–58.PubMedPubMedCentral Zhu Y, Tchkonia T, Pirtskhalava T, et al. The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14:644–58.PubMedPubMedCentral
102.
go back to reference Chang Q, Jorgensen C, Pawson T. Hedley DW Effects of dasatinib on EphA2 receptor tyrosine kinase activity and downstream signalling in pancreatic cancer. Br J Cancer. 2008;99:1074–82.PubMedPubMedCentral Chang Q, Jorgensen C, Pawson T. Hedley DW Effects of dasatinib on EphA2 receptor tyrosine kinase activity and downstream signalling in pancreatic cancer. Br J Cancer. 2008;99:1074–82.PubMedPubMedCentral
103.
go back to reference Boly R, Gras T, Lamkami T, et al. Quercetin inhibits a large panel of kinases implicated in cell biology. Int J Oncol. 2011;38:833–42.PubMed Boly R, Gras T, Lamkami T, et al. Quercetin inhibits a large panel of kinases implicated in cell biology. Int J Oncol. 2011;38:833–42.PubMed
104.
go back to reference Wang C, Maddick M, Miwa S, et al. Adult-onset, short-term dietary restriction reduces cell senescence in mice. Aging (Albany NY). 2010;2:555–66. Wang C, Maddick M, Miwa S, et al. Adult-onset, short-term dietary restriction reduces cell senescence in mice. Aging (Albany NY). 2010;2:555–66.
105.
go back to reference Fontana L, Mitchell SE, Wang B, et al. The effects of graded caloric restriction: XII. Comparison of mouse to human impact on cellular senescence in the colon. Aging Cell. 2018;17:e12746.PubMedPubMedCentral Fontana L, Mitchell SE, Wang B, et al. The effects of graded caloric restriction: XII. Comparison of mouse to human impact on cellular senescence in the colon. Aging Cell. 2018;17:e12746.PubMedPubMedCentral
106.
go back to reference Ogrodnik M, Miwa S, Tchkonia T, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017;8:15691.PubMedPubMedCentral Ogrodnik M, Miwa S, Tchkonia T, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017;8:15691.PubMedPubMedCentral
107.
go back to reference Jurk D, Wang C, Miwa S, et al. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell. 2012;11:996–1004.PubMed Jurk D, Wang C, Miwa S, et al. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell. 2012;11:996–1004.PubMed
108.
go back to reference Fontana L, Nehme J, Demaria M. Caloric restriction and cellular senescence. Mech Ageing Dev. 2018;176:19–23.PubMed Fontana L, Nehme J, Demaria M. Caloric restriction and cellular senescence. Mech Ageing Dev. 2018;176:19–23.PubMed
109.
go back to reference Fontana L, Villareal DT, Das SK, et al. Effects of 2-year calorie restriction on circulating levels of IGF-1, IGF-binding proteins and cortisol in nonobese men and women: a randomized clinical trial. Aging Cell. 2016;15:22–7.PubMed Fontana L, Villareal DT, Das SK, et al. Effects of 2-year calorie restriction on circulating levels of IGF-1, IGF-binding proteins and cortisol in nonobese men and women: a randomized clinical trial. Aging Cell. 2016;15:22–7.PubMed
110.
go back to reference Moiseeva O, Deschênes-Simard X, St-Germain E, et al. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell. 2013;12:489–98.PubMed Moiseeva O, Deschênes-Simard X, St-Germain E, et al. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell. 2013;12:489–98.PubMed
111.
go back to reference Campbell JM, Bellman SM, Stephenson MD, et al. 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;40:31–44.PubMed Campbell JM, Bellman SM, Stephenson MD, et al. 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;40:31–44.PubMed
112.
go back to reference Wang R, Yu Z, Sunchu B, Shoaf J, et al. Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism. Aging Cell. 2017;16:564–74.PubMedPubMedCentral Wang R, Yu Z, Sunchu B, Shoaf J, et al. Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism. Aging Cell. 2017;16:564–74.PubMedPubMedCentral
113.
go back to reference Gurău F, Baldoni S, Prattichizzo F, et al. Anti-senescence compounds: a potential nutraceutical approach to healthy aging. Ageing Res Rev. 2018;46:14–311.PubMed Gurău F, Baldoni S, Prattichizzo F, et al. Anti-senescence compounds: a potential nutraceutical approach to healthy aging. Ageing Res Rev. 2018;46:14–311.PubMed
114.
go back to reference Xu M, Tchkonia T, Ding H, et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci USA. 2015;112:E6301–E63106310.PubMedPubMedCentral Xu M, Tchkonia T, Ding H, et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci USA. 2015;112:E6301–E63106310.PubMedPubMedCentral
115.
go back to reference Roskoski R Jr. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol Res. 2016;111:784–803.PubMed Roskoski R Jr. Janus kinase (JAK) inhibitors in the treatment of inflammatory and neoplastic diseases. Pharmacol Res. 2016;111:784–803.PubMed
116.
go back to reference Palmer AK, Xu M, Zhu Y, et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell. 2019;18:e12950.PubMedPubMedCentral Palmer AK, Xu M, Zhu Y, et al. Targeting senescent cells alleviates obesity-induced metabolic dysfunction. Aging Cell. 2019;18:e12950.PubMedPubMedCentral
117.
go back to reference Xu M, Pirtskhalava T, Farr JN, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018;24:1246–56.PubMedPubMedCentral Xu M, Pirtskhalava T, Farr JN, et al. Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018;24:1246–56.PubMedPubMedCentral
118.
go back to reference Hickson LJ, Langhi Prata LGP, Bobart SA, 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;47:446–56.PubMedPubMedCentral Hickson LJ, Langhi Prata LGP, Bobart SA, 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;47:446–56.PubMedPubMedCentral
119.
go back to reference Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell. 2016;15:428–35.PubMedPubMedCentral Zhu Y, Tchkonia T, Fuhrmann-Stroissnigg H, et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell. 2016;15:428–35.PubMedPubMedCentral
120.
go back to reference Chang J, Wang Y, Shao L, et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med. 2016;22:78–83.PubMed Chang J, Wang Y, Shao L, et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med. 2016;22:78–83.PubMed
121.
go back to reference Fuhrmann-Stroissnigg H, Ling YY, Zhao J, et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun. 2017;8:422.PubMedPubMedCentral Fuhrmann-Stroissnigg H, Ling YY, Zhao J, et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun. 2017;8:422.PubMedPubMedCentral
122.
123.
go back to reference Baar MP, Brandt RM, Putavet DA, et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell. 2017;169:132–47.PubMedPubMedCentral Baar MP, Brandt RM, Putavet DA, et al. Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell. 2017;169:132–47.PubMedPubMedCentral
124.
go back to reference Yousefzadeh MJ, Zhu Y, McGowan SJ, et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018;36:18–28.PubMedPubMedCentral Yousefzadeh MJ, Zhu Y, McGowan SJ, et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018;36:18–28.PubMedPubMedCentral
125.
go back to reference Prata LGPL, Ovsyannikova IG, Tchkonia T, Kirkland JL. Senescent cell clearance by the immune system: emerging therapeutic opportunities. Semin Immunol. 2018;40:101275.PubMed Prata LGPL, Ovsyannikova IG, Tchkonia T, Kirkland JL. Senescent cell clearance by the immune system: emerging therapeutic opportunities. Semin Immunol. 2018;40:101275.PubMed
126.
go back to reference Khosla S, Farr JN, Kirkland JL. Inhibiting cellular senescence: a new therapeutic paradigm for age-related osteoporosis. J Clin Endocrinol Metab. 2018;103:1282–90.PubMedPubMedCentral Khosla S, Farr JN, Kirkland JL. Inhibiting cellular senescence: a new therapeutic paradigm for age-related osteoporosis. J Clin Endocrinol Metab. 2018;103:1282–90.PubMedPubMedCentral
127.
go back to reference Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC, Verdin E. From discoveries in ageing research to therapeutics for healthy ageing. Nature. 2019;571:183–92.PubMedPubMedCentral Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC, Verdin E. From discoveries in ageing research to therapeutics for healthy ageing. Nature. 2019;571:183–92.PubMedPubMedCentral
128.
go back to reference Sikora E, Bielak-Zmijewska A, Mosieniak G. Targeting normal and cancer senescent cells as a strategy of senotherapy. Ageing Res Rev. 2019;55:100941.PubMed Sikora E, Bielak-Zmijewska A, Mosieniak G. Targeting normal and cancer senescent cells as a strategy of senotherapy. Ageing Res Rev. 2019;55:100941.PubMed
Metadata
Title
Cellular Senescence as a Therapeutic Target for Age-Related Diseases: A Review
Authors
Mateo Amaya-Montoya
Agustín Pérez-Londoño
Valentina Guatibonza-García
Andrea Vargas-Villanueva
Carlos O. Mendivil
Publication date
01-04-2020
Publisher
Springer Healthcare
Published in
Advances in Therapy / Issue 4/2020
Print ISSN: 0741-238X
Electronic ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-020-01287-0

Other articles of this Issue 4/2020

Advances in Therapy 4/2020 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine