Therapy-Induced Senescence: Opportunities to Improve Anticancer Therapy

Pataje G Prasanna, Deborah E Citrin, Jeffrey Hildesheim, Mansoor M Ahmed, Sundar Venkatachalam, Gabriela Riscuta, Dan Xi, Guangrong Zheng, Jan van Deursen, Jorg Goronzy, Stephen J Kron, Mitchell S Anscher, Norman E Sharpless, Judith Campisi, Stephen L Brown, Laura J Niedernhofer, Ana O'Loghlen, Alexandros G Georgakilas, Francois Paris, David Gius, David A Gewirtz, Clemens A Schmitt, Mohamed E Abazeed, James L Kirkland, Ann Richmond, Paul B Romesser, Scott W Lowe, Jesus Gil, Marc S Mendonca, Sandeep Burma, Daohong Zhou, C Norman Coleman, Pataje G Prasanna, Deborah E Citrin, Jeffrey Hildesheim, Mansoor M Ahmed, Sundar Venkatachalam, Gabriela Riscuta, Dan Xi, Guangrong Zheng, Jan van Deursen, Jorg Goronzy, Stephen J Kron, Mitchell S Anscher, Norman E Sharpless, Judith Campisi, Stephen L Brown, Laura J Niedernhofer, Ana O'Loghlen, Alexandros G Georgakilas, Francois Paris, David Gius, David A Gewirtz, Clemens A Schmitt, Mohamed E Abazeed, James L Kirkland, Ann Richmond, Paul B Romesser, Scott W Lowe, Jesus Gil, Marc S Mendonca, Sandeep Burma, Daohong Zhou, C Norman Coleman

Abstract

Cellular senescence is an essential tumor suppressive mechanism that prevents the propagation of oncogenically activated, genetically unstable, and/or damaged cells. Induction of tumor cell senescence is also one of the underlying mechanisms by which cancer therapies exert antitumor activity. However, an increasing body of evidence from preclinical studies demonstrates that radiation and chemotherapy cause accumulation of senescent cells (SnCs) both in tumor and normal tissue. SnCs in tumors can, paradoxically, promote tumor relapse, metastasis, and resistance to therapy, in part, through expression of the senescence-associated secretory phenotype. In addition, SnCs in normal tissue can contribute to certain radiation- and chemotherapy-induced side effects. Because of its multiple roles, cellular senescence could serve as an important target in the fight against cancer. This commentary provides a summary of the discussion at the National Cancer Institute Workshop on Radiation, Senescence, and Cancer (August 10-11, 2020, National Cancer Institute, Bethesda, MD) regarding the current status of senescence research, heterogeneity of therapy-induced senescence, current status of senotherapeutics and molecular biomarkers, a concept of "one-two punch" cancer therapy (consisting of therapeutics to induce tumor cell senescence followed by selective clearance of SnCs), and its integration with personalized adaptive tumor therapy. It also identifies key knowledge gaps and outlines future directions in this emerging field to improve treatment outcomes for cancer patients.

© The Author(s) 2021. Published by Oxford University Press.

Figures

Figure 1.
Figure 1.
Senescent cell as a target in one-two punch cancer therapy. A) Key knowledge gaps and future directions to advance one-two punch cancer therapy. B) Therapy-induced senescence (TIS) and one-two punch cancer therapy. Cancer therapies (first punch) induce senescence both in tumor and normal tissue. SnCs are normally cleared by immune surveillance but can accumulate after cancer therapy. Therapy-induced SnCs are heterogeneous and dynamic, which is also reflected in biomarkers, cellular plasticity, expression of SASPs and SCAPs, tissue of origin, and cell lineage. Selective clearance of SnCs with a serotherapeutic (second punch) in tumors will prevent tumor relapse, metastasis, and development of resistance to treatment. Similarly, selective clearance of SnCs in normal tissue in a spatiotemporal dynamic environment will prevent, mitigate, and treat therapy-induced side effects and restore tissue homeostasis. However, time of administration of the second punch therapy will be important to improve efficacy. The figure was created with BioRender.com. SnCs = senescent cells; SASP = senescence-associated secretory phenotype; SCAPs = senescent cell anti-apoptotic pathways.
Figure 2.
Figure 2.
Schematic diagram of an approach to integrate one-two punch cancer therapy with personalized adaptive tumor therapy. To therapeutically exploit and benefit from the differences in response to treatment between tumor and normal tissue for the best patient outcome, factors that should be considered for pretreatment planning include tumor molecular profiling, tumor heterogeneity, imaging, identification of target(s), metabolic status, and planned integrated biomarkers for tumor diagnosis and treatment matching (136). Similar profiling of normal tissue response to treatment may include determination of genetic susceptibility, immune status, stromal tissue subsets, the impact of the anatomical location of the tumor on normal tissue, metabolic status, and biomarkers that predict response and adverse effects. In a one-two punch therapy, punch 1 may include spatially targeted radiotherapy (eg, dose-boost to hypoxic regions), molecularly targeted drugs, and/or immune therapy to the tumor, which will induce TIS in the tumor, stroma, and bystander tissue. Thus, tumor, stroma, and bystander tissue all need to be evaluated for TIS for the second punch to be successful. Biomarker-driven TIS evaluation will be essential to optimize immune modulation, dose, and schedule of the second punch with a suitable senolytic. Along with dynamic adaptive tumor targeting (with drugs, immune modulators, and radiation), the use of different types of senolytics may be necessary to address spatial, temporal, and tissue heterogeneity among tumors and senescent cells. Repeat treatment courses (punch #n) with senotherapeutics (senolytics or senomorphics) may be necessary to prevent tumor recurrence, drug resistance, plasticity, and normal tissue injury and mitigate and/or treat adverse effects months to years after completing the one-two punch therapy for optimal tissue remodeling and tissue function restoration. Dotted boxes represent current biomarkers and future opportunities to develop diagnostics or therapeutics for precision medicine in TIS. Tissues are indicated by the colors red (tumor), green (normal tissue), blue (stroma and immune related to tumor), and brown (bystander tissues). The figure was created with BioRender.com. Rx = prescription; TIS = therapy-induced senescence.

References

    1. Bernadotte A, Mikhelson VM, Spivak IM.. Markers of cellular senescence. Telomere shortening as a marker of cellular senescence. Aging. 2016;8(1):3–11.
    1. Munoz-Espin D, Serrano M.. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15(7):482–496.
    1. Baker DJ, Childs BG, Durik M, et al.Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature. 2016;530(7589):184–189.
    1. Dimri GP, Lee X, Basile G, et al.A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA. 1995;92(20):9363–9367.
    1. Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013;75:685–705.
    1. Sharpless NE, Sherr CJ.. Forging a signature of in vivo senescence. Nat Rev Cancer. 2015;15(7):397–408.
    1. Borghesan M, Fafian-Labora J, Eleftheriadou O, et al.Small extracellular vesicles are key regulators of non-cell autonomous intercellular communication in senescence via the interferon protein IFITM3. Cell Rep. 2019;27(13):3956–3971.e6.
    1. Basisty N, Kale A, Jeon OH, et al.A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 2020;18(1):e3000599.
    1. Iske J, Seyda M, Heinbokel T, et al.Senolytics prevent mt-DNA-induced inflammation and promote the survival of aged organs following transplantation. Nat Commun. 2020;11(1):4289.
    1. Colpani O, Spinetti G.. MicroRNAs orchestrating senescence of endothelial and vascular smooth muscle cells. Vasc Biol. 2019;1(1):H75–H81.
    1. Lee S, Schmitt CA.. The dynamic nature of senescence in cancer. Nat Cell Biol. 2019;21(1):94–101.
    1. Ruscetti M, Morris JPM, Mezzadra R, et al.Senescence-induced vascular remodeling creates therapeutic vulnerabilities in pancreas cancer. Cell. 2020;181(2):424–441.e21.
    1. Ewald JA, Desotelle JA, Wilding G, et al.Therapy-induced senescence in cancer. J Natl Cancer Inst. 2010;102(20):1536–1546.
    1. Shao L, Feng W, Li H, et al.Total body irradiation causes long-term mouse BM injury via induction of HSC premature senescence in an Ink4a- and Arf-independent manner. Blood. 2014;123(20):3105–3115.
    1. White RR, Vijg J.. Do DNA double-strand breaks drive aging? Mol Cell. 2016;63(5):729–738.
    1. Schmitt CA, Fridman JS, Yang M, et al.A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell. 2002;109(3):335–346.
    1. Faget DV, Ren Q, Stewart SA.. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019;19(8):439–453.
    1. Romanov SR, Kozakiewicz BK, Holst CR, et al.Normal human mammary epithelial cells spontaneously escape senescence and acquire genomic changes. Nature. 2001;409(6820):633–637.
    1. Georgakilas AG, Martin OA, Bonner WM.. p21: a two-faced genome guardian. Trends Mol Med. 2017;23(4):310–319.
    1. Milanovic M, Fan DNY, Belenki D, et al.Senescence-associated reprogramming promotes cancer stemness. Nature. 2018;553(7686):96–100.
    1. Cahu J, Bustany S, Sola B.. Senescence-associated secretory phenotype favors the emergence of cancer stem-like cells. Cell Death Dis. 2012;3:e446.
    1. Guillon J, Petit C, Moreau M, et al.Regulation of senescence escape by TSP1 and CD47 following chemotherapy treatment. Cell Death Dis. 2019;10(3):199.
    1. Xu M, Pirtskhalava T, Farr JN, et al.Senolytics improve physical function and increase lifespan in old age. Nat Med. 2018;24(8):1246–1256.
    1. Demaria M, O’Leary MN, Chang J, et al.Cellular senescence promotes adverse effects of chemotherapy and cancer relapse. Cancer Discov. 2017;7(2):165–176.
    1. Myrianthopoulos V, Evangelou K, Vasileiou PVS, et al.Senescence and senotherapeutics: a new field in cancer therapy. Pharmacol Ther. 2019;193:31–49.
    1. Robbins PD, Jurk D, Khosla S, et al.Senolytic drugs: reducing senescent cell viability to extend health span. Annu Rev Pharmacol Toxicol. 2021;61:779–803.
    1. Wang L, Leite de Oliveira R, Wang C, et al.High-throughput functional genetic and compound screens identify targets for senescence induction in cancer. Cell Rep. 2017;21(3):773–783.
    1. Leite de Oliveira R, Bernards R.. Anti-cancer therapy: senescence is the new black. EMBO J. 2018;37(10):e99386.
    1. Chakradeo S, Elmore LW, Gewirtz DA.. Is senescence reversible? Curr Drug Targets. 2016;17(4):460–466.
    1. Sapieha P, Mallette FA.. Cellular senescence in postmitotic cells: beyond growth arrest. Trends Cell Biol. 2018;28(8):595–607.
    1. Saleh T, Tyutyunyk-Massey L, Gewirtz DA.. Tumor cell escape from therapy-induced senescence as a model of disease recurrence after dormancy. Cancer Res. 2019;79(6):1044–1046.
    1. Bielak-Zmijewska A, Mosieniak G, Sikora E.. Is DNA damage indispensable for stress-induced senescence? Mech Ageing Dev. 2018;170:13–21.
    1. Wagner V, Gil J.. Senescence as a therapeutically relevant response to CDK4/6 inhibitors. Oncogene. 2020;39(29):5165–5176.
    1. Vilgelm AE, Johnson CA, Prasad N, et al.Connecting the dots: therapy-induced senescence and a tumor-suppressive immune microenvironment. J Natl Cancer Inst. 2016;108(6):djv406.
    1. Liu Y, Hawkins OE, Su Y, et al.Targeting aurora kinases limits tumour growth through DNA damage-mediated senescence and blockade of NF-kappaB impairs this drug-induced senescence. EMBO Mol Med. 2013;5(1):149–166.
    1. Petrova NV, Velichko AK, Razin SV, et al.Small molecule compounds that induce cellular senescence. Aging Cell. 2016;15(6):999–1017.
    1. Macedo JC, Vaz S, Bakker B, et al.FoxM1 repression during human aging leads to mitotic decline and aneuploidy-driven full senescence. Nat Commun. 2018;9(1):2834.
    1. Chapman J, Fielder E, Passos JF.. Mitochondrial dysfunction and cell senescence: deciphering a complex relationship. FEBS Lett. 2019;593(13):1566–1579.
    1. Wiley CD, Velarde MC, Lecot P, et al.Mitochondrial dysfunction induces senescence with a distinct secretory phenotype. Cell Metab. 2016;23(2):303–314.
    1. Correia-Melo C, Marques FD, Anderson R, et al.Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J. 2016;35(7):724–742.
    1. Lafargue A, Degorre C, Corre I, et al.Ionizing radiation induces long-term senescence in endothelial cells through mitochondrial respiratory complex II dysfunction and superoxide generation. Free Radic Biol Med. 2017;108:750–759.
    1. Wolf DA. Is reliance on mitochondrial respiration a “chink in the armor” of therapy-resistant cancer? Cancer Cell. 2014;26(6):788–795.
    1. Vizioli MG, Liu T, Miller KN, et al.Mitochondria-to-nucleus retrograde signaling drives formation of cytoplasmic chromatin and inflammation in senescence. Genes Dev. 2020;34(5-6):428–445.
    1. Wang Y, Boerma M, Zhou D.. Ionizing radiation-induced endothelial cell senescence and cardiovascular diseases. Radiat Res. 2016;186(2):153–161.
    1. Efimova EV, Takahashi S, Shamsi NA, et al.Linking cancer metabolism to DNA repair and accelerated senescence. Mol Cancer Res. 2016;14(2):173–184.
    1. Efimova EV, Appelbe OK, Ricco N, et al.O-GlcNAcylation enhances double-strand break repair, promotes cancer cell proliferation, and prevents therapy-induced senescence in irradiated tumors. Mol Cancer Res. 2019;17(6):1338–1350.
    1. Dorr JR, Yu Y, Milanovic M, et al.Synthetic lethal metabolic targeting of cellular senescence in cancer therapy. Nature. 2013;501(7467):421–425.
    1. Martinez-Zamudio RI, Roux PF, de Freitas J, et al.AP-1 imprints a reversible transcriptional programme of senescent cells. Nat Cell Biol. 2020;22(7):842–855.
    1. Reimann M, Schrezenmeier JF, Richter-Pechanska P, et al.Adaptive T-cell immunity controls senescence-prone MyD88- or CARD11-mutant B-cell lymphomas. Blood. 2020. doi: 10.1182/blood.2020005244.
    1. Gopal P, Rogacki K, Peacock CD , et al.Dynamic transdifferentiation programs in small cell lung carcinoma (Abstract nr 2897). Cancer Res 2019;79(suppl 13). doi: 10.1158/1538-7445.AM2019-2897
    1. 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;47:446–456.
    1. Kirkland JL, Tchkonia T.. Cellular senescence: a translational perspective. EBioMedicine. 2017;21:21–28.
    1. Yu Y, Schleich K, Yue B, et al.Targeting the senescence-overriding cooperative activity of structurally unrelated H3K9 demethylases in melanoma. Cancer Cell. 2018;33(4):785.
    1. Saleh T, Bloukh S, Carpenter VJ, et al.Therapy-Induced senescence: an “old” friend becomes the enemy. Cancers (Basel). 2020;12(4):822.
    1. Le Duff M, Gouju J, Jonchère B, et al.Regulation of senescence escape by the CDK4-EZH2-AP2M1 pathway in response to chemotherapy. Cell Death Dis. 2018;9(2):199.
    1. Hernandez-Segura A, de Jong TV, Melov S, et al.Unmasking transcriptional heterogeneity in senescent cells. Curr Biol. 2017;27(17):2652–2660.e4.
    1. Ozcan S, Alessio N, Acar MB, et al.Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging. 2016;8(7):1316–1329.
    1. Acosta JC, O’Loghlen A, Banito A, et al.Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008;133(6):1006–1018.
    1. Acosta JC, Banito A, Wuestefeld T, et al.A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol. 2013;15(8):978–990.
    1. Lujambio A, Akkari L, Simon J, et al.Non-cell-autonomous tumor suppression by p53. Cell. 2013;153(2):449–460.
    1. Vilgelm AE, Saleh N, Shattuck-Brandt R, et al.MDM2 antagonists overcome intrinsic resistance to CDK4/6 inhibition by inducing p21. Sci Transl Med. 2019;11(505):eaav7171. doi: 10.1126/scitranslmed.aav7171.
    1. Vilgelm AE, Pawlikowski JS, Liu Y, et al.MDM2 and aurora kinase a inhibitors synergize to block melanoma growth by driving apoptosis and immune clearance of tumor cells. Cancer Res. 2015;75(1):181–193.
    1. Ruscetti M, Leibold J, Bott MJ, et al.NK cell-mediated cytotoxicity contributes to tumor control by a cytostatic drug combination. Science. 2018;362(6421):1416–1422.
    1. Wiley CD, Flynn JM, Morrissey C, et al.Analysis of individual cells identifies cell-to-cell variability following induction of cellular senescence. Aging Cell. 2017;16(5):1043–1050.
    1. Tang H, Geng A, Zhang T, et al.Single senescent cell sequencing reveals heterogeneity in senescent cells induced by telomere erosion. Protein Cell. 2019;10(5):370–375.
    1. Robbins PD, Jurk D, Khosla S, et al.Senolytic drugs: reducing senescent cell viability to extend health span. Annu Rev Pharmacol Toxicol. 2021;61:779–803.
    1. Zhu Y, Tchkonia T, Pirtskhalava T, et al.The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell. 2015;14(4):644–658.
    1. Roy AL, Sierra F, Howcroft K, et al.A blueprint for characterizing senescence. Cell. 2020;183(5):1143–1146.
    1. Ahmed MM, Coleman CN, Mendonca M, et al.Workshop report for cancer research: defining the shades of Gy: Utilizing the biological consequences of radiotherapy in the development of new treatment approaches-meeting viewpoint. Cancer Res. 2018;78(9):2166–2170.
    1. He Y, Zheng G, Zhou D.. Senolytic drug development. In: Muñoz-Espin D, Demaria M, eds. Senolytics in Diseases, Ageing and Longevity. Healthy Ageing and Longevity. Cham: Springer; 2020.
    1. Zhu Y, Doornebal EJ, Pirtskhalava T, et al.New agents that target senescent cells: the flavone, fisetin, and the BCL-X-L inhibitors, A1331852 and A1155463. Aging. 2017;9(3):955–963.
    1. Laberge RM, Sun Y, Orjalo AV, et al.MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat Cell Biol. 2015;17(8):1049–U416.
    1. Xu M, Palmer AK, Ding H, et al.Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife. 2015;4:e12997.
    1. Tasdemir N, Banito A, Roe JS, et al.BRD4 connects enhancer remodeling to senescence immune surveillance. Cancer Discov. 2016;6(6):612–629.
    1. Carpenter VJ, Saleh T, Gewirtz DA.. Senolytics for cancer therapy: Is all that glitters really gold? Cancers. 2021;13(4):723.
    1. Pan J, Li D, Xu Y, et al.Inhibition of Bcl-2/xl with ABT-263 selectively kills senescent type II pneumocytes and reverses persistent pulmonary fibrosis induced by ionizing radiation in mice. Int J Radiat Oncol Biol Phys. 2017;99(2):353–361.
    1. Wang C, Vegna S, Jin H, et al.Inducing and exploiting vulnerabilities for the treatment of liver cancer. Nature. 2019;574(7777):268–272.
    1. Gonzalez-Gualda E, Paez-Ribes M, Lozano-Torres B, et al.Galacto-conjugation of Navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity. Aging Cell. 2020;19(4):e13142.
    1. Guerrero A, Guiho R, Herranz N, et al.Galactose-modified duocarmycin prodrugs as senolytics. Aging Cell. 2020;19(4):e13133.
    1. Cai Y, Zhou H, Zhu Y, et al.Elimination of senescent cells by beta-galactosidase-targeted prodrug attenuates inflammation and restores physical function in aged mice. Cell Res. 2020;30(7):574–589.
    1. Hall BM, Balan V, Gleiberman AS, et al.Aging of mice is associated with p16(Ink4a)- and beta-galactosidase-positive macrophage accumulation that can be induced in young mice by senescent cells. Aging. 2016;8(7):1294–1315.
    1. He Y, Zhang X, Chang J, et al.Using proteolysis-targeting chimera technology to reduce navitoclax platelet toxicity and improve its senolytic activity. Nat Commun. 2020;11(1):1996.
    1. de Vos S, Leonard JP, Friedberg JW, et al.Safety and efficacy of navitoclax, a BCL-2 and BCL-XL inhibitor, in patients with relapsed or refractory lymphoid malignancies: results from a phase 2a study. Leuk Lymphoma. 2021;62(4):810–818.
    1. Patterson CM, Balachander SB, Grant I, et al.Design and optimisation of dendrimer-conjugated Bcl-2/xL inhibitor, AZD0466, with improved therapeutic index for cancer therapy. Commun Biol. 2021;4(1):112.
    1. Dolgin E. Send in the senolytics. Nat Biotechnol. 2020;38(12):1371–1377.
    1. Prasanna PG, Narayanan D, Hallett K, et al.Radioprotectors and radiomitigators for improving radiation therapy: the Small Business Innovation Research (SBIR) Gateway for Accelerating Clinical Translation. Radiat Res. 2015;184(3):235–248.
    1. Zakeri K, Narayanan D, Prasanna PGS, et al.Development of novel radiosensitizers through the National Cancer Institute’s Small Business Innovation Research Program. Radiat Res. 2020;193(5):425–434.
    1. Zakeri K, Narayanan D, Vikram B, et al.Decreasing the toxicity of radiation therapy: radioprotectors and radiomitigators being developed by the National Cancer Institute through small business innovation research contracts. Int J Radiat Oncol Biol Phys. 2019;104(1):188–196.
    1. Sieben CJ, Sturmlechner I, van de Sluis B, et al.Two-step senescence-focused cancer therapies. Trends Cell Biol. 2018;28(9):723–737.
    1. Hoenicke L, Zender L.. Immune surveillance of senescent cells–biological significance in cancer- and non-cancer pathologies. Carcinogenesis. 2012;33(6):1123–1126.
    1. Fleury H, Malaquin N, Tu V, et al.Exploiting interconnected synthetic lethal interactions between PARP inhibition and cancer cell reversible senescence. Nat Commun. 2019;10(1):2556.
    1. Saleh T, Carpenter VJ, Tyutyunyk-Massey L, et al.Clearance of therapy-induced senescent tumor cells by the senolytic ABT-263 via interference with BCL-XL -BAX interaction. Mol Oncol. 2020;14(10):2504.
    1. Guerrero A, Herranz N, Sun B, et al.Cardiac glycosides are broad-spectrum senolytics. Nat Metab. 2019;1(11):1074–1088.
    1. Triana-Martinez F, Picallos-Rabina P, Da Silva AS, et al.Identification and characterization of cardiac glycosides as senolytic compounds. Nat Commun. 2019;10(1):4731.
    1. Amor C, Feucht J, Leibold J, et al.Senolytic CAR T cells reverse senescence-associated pathologies. Nature. 2020;583(7814):127–132.
    1. Acklin S, Zhang M, Du W, et al.Depletion of senescent-like neuronal cells alleviates cisplatin-induced peripheral neuropathy in mice. Sci Rep. 2020;10(1):14170.
    1. Chang JH, Wang YY, Shao LJ, et al.Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med. 2016;22(1):78–83.
    1. Vilgelm AE, Richmond A.. Chemokines modulate immune surveillance in tumorigenesis, metastasis, and response to immunotherapy. Front Immunol. 2019;10:article333. doi: 10.3389/fimmu.2019.00333
    1. Justice JN, Nambiar AM, Tchkonia T, et al.Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine. 2019;40:554–563.
    1. Hickson LJ, Langhi PL, Bobart SA, et al.Corrigendum to ‘Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease’ EBioMedicine 47 (2019) 446-456. EBioMedicine. 2019;52:102595.
    1. Jeon OH, Kim C, Laberge RM, et al.Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med. 2017;23(6):775–781.
    1. Kirkland JL, Tchkonia T.. Senolytic drugs: from discovery to translation. J Intern Med. 2020;288(5):518–536.
    1. Eggert T, Wolter K, Ji J, et al.Distinct functions of senescence-associated immune responses in liver tumor surveillance and tumor progression. Cancer Cell. 2016;30(4):533–547.
    1. Pare R, Shin JS, Lee CS.. Increased expression of senescence markers p14(ARF) and p16(INK4a) in breast cancer is associated with an increased risk of disease recurrence and poor survival outcome. Histopathology. 2016;69(3):479–491.
    1. Pare R, Soon PS, Shah A, et al.Differential expression of senescence tumour markers and its implications on survival outcomes of breast cancer patients. PLoS One. 2019;14(4):e0214604.
    1. Jennings CJ, Murer B, O’Grady A, et al.Differential p16/INK4A cyclin-dependent kinase inhibitor expression correlates with chemotherapy efficacy in a cohort of 88 malignant pleural mesothelioma patients. Br J Cancer. 2015;113(1):69–75.
    1. Bernardes de Jesus B, Blasco MA.. Assessing cell and organ senescence biomarkers. Circ Res. 2012;111(1):97–109.
    1. Gorgoulis V, Adams PD, Alimonti A, et al.Cellular senescence: defining a path forward. Cell. 2019;179(4):813–827.
    1. Idda ML, McClusky WG, Lodde V, et al.Survey of senescent cell markers with age in human tissues. Aging. 2020;12(5):4052–4066.
    1. Coleman CN, Higgins GS, Brown JM, et al.Improving the predictive value of preclinical studies in support of radiotherapy clinical trials. Clin Cancer Res. 2016;22(13):3138–3147.
    1. Hu JL, Todhunter ME, LaBarge MA, et al.Opportunities for organoids as new models of aging. J Cell Biol. 2018;217(1):39–50.
    1. Sanoff HK, Deal AM, Krishnamurthy J, et al.Effect of cytotoxic chemotherapy on markers of molecular age in patients with breast cancer. J Natl Cancer Inst. 2014;106(4):dju057.
    1. Smitherman AB, Wood WA, Mitin N, et al.Accelerated aging among childhood, adolescent, and young adult cancer survivors is evidenced by increased expression of p16(INK4a) and frailty. Cancer. 2020;126(22):4975–4983.
    1. Wyld L, Bellantuono I, Tchkonia T, et al.Senescence and cancer: a review of clinical implications of senescence and senotherapies. Cancers. 2020;12(8):2134.
    1. Farr JN, Xu M, Weivoda MM, et al.Targeting cellular senescence prevents age-related bone loss in mice. Nat Med. 2017;23(9):1072–1079.
    1. Johnson SM, Torrice CD, Bell JF, et al.Mitigation of hematologic radiation toxicity in mice through pharmacological quiescence induced by CDK4/6 inhibition. J Clin Invest. 2010;120(7):2528–2536.
    1. He S, Roberts PJ, Sorrentino JA, et al.Transient CDK4/6 inhibition protects hematopoietic stem cells from chemotherapy-induced exhaustion. Sci Transl Med. 2017;9(387):eaal3986.
    1. Weiss JM, Csoszi T, Maglakelidze M, et al.; G1T28-02 Study Group. Myelopreservation with the CDK4/6 inhibitor trilaciclib in patients with small-cell lung cancer receiving first-line chemotherapy: a phase Ib/randomized phase II trial. Ann Oncol. 2019;30(10):1613–1621.
    1. Tan AR, Wright GS, Thummala AR, et al.Trilaciclib plus chemotherapy versus chemotherapy alone in patients with metastatic triple-negative breast cancer: a multicentre, randomised, open-label, phase 2 trial. Lancet Oncol. 2019;20(11):1587–1601.
    1. Goel S, DeCristo MJ, McAllister SS, et al.CDK4/6 inhibition in cancer: beyond cell cycle arrest. Trends Cell Biol. 2018;28(11):911–925.
    1. Pernas S, Tolaney SM, Winer EP, et al.CDK4/6 inhibition in breast cancer: current practice and future directions. Ther Adv Med Oncol. 2018;10:1758835918786451.
    1. Hayflick L, Moorhead PS.. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.
    1. . Guida JL, Agurs-Collins T, Ahles TA, et al.Strategies to prevent or remediate cancer and treatment-related aging. J Natl Cancer Inst. 2021;113(2):112–122..
    1. Terasima T, Tolmach LJ.. Variations in several responses of HeLa cells to x-irradiation during the division cycle. Biophys J. 1963;3:11–33.
    1. Fuhrmann-Stroissnigg H, Ling YY, Zhao J, et al.Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun. 2017;8:article 422.
    1. Li W, He Y, Zhang R, et al.The curcumin analog EF24 is a novel senolytic agent. Aging. 2019;11(2):771–782.
    1. Liu X, Wang Y, Zhang X, et al.Senolytic activity of piperlongumine analogues: synthesis and biological evaluation. Bioorg Med Chem. 2018;26(14):3925–3938.
    1. Wang Y, Chang J, Liu X, et al.Discovery of piperlongumine as a potential novel lead for the development of senolytic agents. Aging. 2016;8(11):2915–2926.
    1. Zhang X, Zhang S, Liu X, et al.Oxidation resistance 1 is a novel senolytic target. Aging Cell. 2018;17(4):e12780.
    1. Yousefzadeh MJ, Zhu Y, McGowan SJ, et al.Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine. 2018;36:18–28.
    1. Yosef R, Pilpel N, Tokarsky-Amiel R, et al.Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat Commun. 2016;7:11190.
    1. He Y, Li W, Lv D, et al.Inhibition of USP7 activity selectively eliminates senescent cells in part via restoration of p53 activity. Aging Cell. 2020;19(3):e13117.
    1. Samaraweera L, Adomako A, Rodriguez-Gabin A, et al.A novel indication for panobinostat as a senolytic drug in NSCLC and HNSCC. Sci Rep. 2017;7:1900.
    1. Baar MP, Brandt RMC, Putavet DA, et al.Targeted apoptosis of senescent cells restores tissue homeostasis in response to chemotoxicity and aging. Cell. 2017;169(1):132–147.e16.
    1. Wakita M, Takahashi A, Sano O, et al.A BET family protein degrader provokes senolysis by targeting NHEJ and autophagy in senescent cells. Nat Commun. 2020;11(1):1935.
    1. Malone ER, Oliva M, Sabatini PJB, et al.Molecular profiling for precision cancer therapies. Genome Med. 2020;12(1):8.

Source: PubMed

3
订阅