Senotherapeutics in Cancer and HIV
Abstract
:1. Biology and Molecular Mechanisms of Senescence
1.1. Definition and Induction of Cellular Senescence
1.2. Characteristics of Senescent Cells
2. Bimodal Role of Cellular Senescence
2.1. Senescent Cell Clearance by the Immune System
2.2. Senescence and Cancer
3. Implications of Cellular Senescence in Diseases
4. Therapeutic Difficulties
5. Senotherapeutics
5.1. First Senolytics: Dasatinib and Quercetin
5.2. Inhibitors of BCL2
5.3. Piperlongumine
5.4. Inhibitors of HSP90
5.5. Panobinostat
5.6. Cardiac Glycosides
5.7. FOXO4-TP53 Disrupting Peptides
5.8. Other Senolytics
- Fenofibrate (FN), a PPARα agonist used for dyslipidaemias in humans, was identified as a senolytic. This agent induced the apoptosis of senescent cells, increased autophagy and protected against cartilage degradation. Fibrate treatment improved osteoarthritis in patients from the Osteoarthritis Initiative (OAI) cohort in a retrospective study [99].
- UBX0101 is a p53/MDM2 interaction inhibitor. However, the mechanism by which senescent cell apoptosis is induced has not been fully elucidated. Intra-articular injection of UBX0101 selectively caused the clearance of senescent cells that are accumulated in the articular cartilage. More beneficial effects were the reduction in the development of post-traumatic osteoarthritis and the increase in the chondrogenesis [66]. Some clinical trials are investigating this inhibitor in knee arthritis (NCT04129944, NCT04349956, NCT04229225, NCT03513016).
5.9. Inhibitors of mTOR Kinase
5.10. JAK Inhibitors
5.11. Metformin
5.12. Resveratrol
5.13. ATM Inhibitor
Agent | Mechanism of Action | Clinical Status | Clinical Indication | Clinical Trial | Effect | Side Effects | Doses | |
---|---|---|---|---|---|---|---|---|
Senolytic | Dasatinib | (-) Src family tyrosine kinase | Approved to (1) Experimental to (2) and (3) (phase 2) | (1) Chronic myelogenous leukemia (2) Epigenetic aging (3) Frailty | NCT04946383 NCT04733534 | ↑ Lifespan ↓ Osteoporosis Atherosclerosis | Blood counts Diarrhea Bleeding Fever | 100–140 mg/day orally |
Quercetin | PI3K antagonist | Experimental (phase 2) | Epigenetic aging Frailty | NCT04946383 NCT04733534 | Nausea Kidney damage | 420–1400 mg/m2 intravenous (IV) bolus once/week | ||
Navitoclax (ABT-263) | BCL-2 antagonist | Experimental (Phase 3) | Myelofibrosis | [65] NCT04472598 NCT04468984 | ↑ Hematopoitetic stem cell function ↓ Atherosclerosis | Diarrhea Nausea Thrombocytopenia Lymphocitopenia | 150 mg/7-day/325 mg on continuous (21/21) schedule (Clin trial) | |
Fisetin | (-) BCL-xL, HIF-1α and other SCAP network components | Experimental (Phase 2) | Osteoarthritis Frailty COVID-19 | NCT04210986 NCT04733534 NCT04476953 NCT04537299 NCT04771611 | Not described | No evidence in animal studies | 20 mg/kg for two consecutive days (Clinical trial) | |
Piperlongumine | GSTP1 antagonist | Experimental | - | [70] | Not described | Not described | Not described | |
Geldanamycin | (-) HSP90 | Experimental | - | [73,74,75] | ↑ Lifespan ↓ Age related symptoms | Not described | Not described | |
Tanespimycin (17-AAG) | Experimental | Solid tumors Multiple myeloma | [76] | Diarrhea Nausea Hepatotoxicity Anemia Thrombocytopenia | 56–450mg/m2 on different dosing schedules (no established) | |||
Panobinostat | (-) nonselective histone deacetylase | Approved to (1) | (1) Multiple myeloma | [77] | Not described | Diarrhea Severe and fatal cardiac events | 20 mg/3 doses/week in combination with Bortezomib and dexamethasone | |
Senostatic | Rapamycin | (-) mTOR kinase | Approved to (1) | (1) Immunesuppression | [80,81] NCT04488601 | ↑ Lifespan | Headache Diarrhea Nausea Joint pain | 16–24 mg/µL for prophylaxis of Renal Transplant Rejection |
Ruxolitinib | (-) JAK | Approved to (1) | (1) Myeloproliferative diseases | [82] | Not described | Anemia Thrombocytopenia Bruising Neutropenia | 5–20 mg orally twice daily | |
Metformin | AMPK agonist (-) mGPD | Approved to (1) | (1) Diabetes type II | [84,85] | ↑ Lifespan in mice | Lactic Acidosis Vitamin B12 deficiency Hypoglycemia | 500–2000 mg orally once daily | |
Resveratrol | (-) Histone deacetylase | Experimental | - | [91,92] | ↑ Lifespan in cerevisae | No evidence | 250-1000 mg daily for up to 3 months. |
6. Senotherapeutics for HIV Infection
7. Conclusions and Future Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef]
- Harley, C.B.; Futcher, A.B.; Greider, C.W. Telomeres shorten during ageing of human fibroblasts. Nature 1990, 345, 458–460. [Google Scholar] [CrossRef] [PubMed]
- Olovnikov, A. A theory of marginotomy: The incomplete copying of template margin in enzymic synthesis of polynucleotides and biological significance of the phenomenon. J. Theor. Biol. 1973, 41, 181–190. [Google Scholar] [CrossRef]
- Shay, J.W.; Wright, W.E. Hallmarks of telomeres in ageing research. J. Pathol. 2007, 211, 114–123. [Google Scholar] [CrossRef]
- Bodnar, A.G.; Ouellette, M.; Frolkis, M.; Holt, S.E.; Chiu, C.-P.; Morin, G.B.; Harley, C.B.; Shay, J.W.; Lichtsteiner, S.; Wright, W.E. Extension of Life-Span by Introduction of Telomerase into Normal Human Cells. Science 1998, 279, 349–352. [Google Scholar] [CrossRef] [Green Version]
- Dominguez-Bautista, J.A.; Acevo-Rodríguez, P.S.; Castro-Obregón, S. Programmed Cell Senescence in the Mouse Developing Spinal Cord and Notochord. Front. Cell Dev. Biol. 2021, 9, 9. [Google Scholar] [CrossRef]
- Toussaint, O.; Royer, V.; Salmon, M.; Remacle, J. Stress-induced premature senescence and tissue ageing. Biochem. Pharmacol. 2002, 64, 1007–1009. [Google Scholar] [CrossRef]
- Sherr, C.J.; DePinho, R.A. Cellular senescence: Mitotic clock or culture shock? Cell 2000, 102, 407–410. [Google Scholar] [CrossRef] [Green Version]
- Serrano, M.; Lee, H.-W.; Chin, L.; Cordon-Cardo, C.; Beach, D.; DePinho, R. Role of the INK4a Locus in Tumor Suppression and Cell Mortality. Cell 1996, 85, 27–37. [Google Scholar] [CrossRef] [Green Version]
- Serrano, M.; Lin, A.W.; McCurrach, M.E.; Beach, D.; Lowe, S.W. Oncogenic ras Provokes Premature Cell Senescence Associated with Accumulation of p53 and p16INK4a. Cell 1997, 88, 593–602. [Google Scholar] [CrossRef] [Green Version]
- Michaloglou, C.; Vredeveld, L.C.W.; Soengas, M.S.; Denoyelle, C.; Kuilman, T.; Van Der Horst, C.M.A.M.; Majoor, D.M.; Shay, J.W.; Mooi, W.J.; Peeper, D.S. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 2005, 436, 720–724. [Google Scholar] [CrossRef]
- Bartkova, J.; Rezaei, N.; Liontos, M.; Karakaidos, P.; Kletsas, D.; Issaeva, N.; Vassiliou, L.-V.F.; Kolettas, E.; Niforou, K.; Zoumpourlis, V.C.; et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 2006, 444, 633–637. [Google Scholar] [CrossRef]
- Gorgoulis, V.G.; Pefani, D.-E.; Pateras, I.S.; Trougakos, I.P. Integrating the DNA damage and protein stress responses during cancer development and treatment. J. Pathol. 2018, 246, 12–40. [Google Scholar] [CrossRef] [Green Version]
- Wiley, C.D.; Velarde, M.C.; Lecot, P.; Liu, S.; Sarnoski, E.A.; Freund, A.; Shirakawa, K.; Lim, H.W.; Davis, S.S.; Ramanathan, A.; et al. Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. Cell Metab. 2016, 23, 303–314. [Google Scholar] [CrossRef] [Green Version]
- Campisi, J. Aging, Cellular Senescence, and Cancer. Annu. Rev. Physiol. 2013, 75, 685–705. [Google Scholar] [CrossRef] [Green Version]
- Childs, B.G.; Gluscevic, M.; Baker, D.J.; Laberge, R.-M.; Marquess, D.; Dananberg, J.; Van Deursen, J.M. Senescent cells: An emerging target for diseases of ageing. Nat. Rev. Drug Discov. 2017, 16, 718–735. [Google Scholar] [CrossRef] [Green Version]
- Ewald, J.A.; Desotelle, J.A.; Wilding, G.; Jarrard, D.F. Therapy-Induced Senescence in Cancer. JNCI J. Natl. Cancer Inst. 2010, 102, 1536–1546. [Google Scholar] [CrossRef] [Green Version]
- Evangelou, K.; Gorgoulis, V.G. Sudan Black B, the Specific Histochemical Stain for Lipofuscin: A Novel Method to Detect Senescent Cells. In Oncogene-Induced Senescence: Methods and Protocols; Nikiforov, M.A., Ed.; Springer: New York, NY, USA, 2017; pp. 111–119. [Google Scholar]
- Brenner, A.J.; Stampfer, M.R.; Aldaz, C.M. Increased p16 expression with first senescence arrest in human mammary epithelial cells and extended growth capacity with p16 inactivation. Oncogene 1998, 17, 199–205. [Google Scholar] [CrossRef] [Green Version]
- Beauséjour, C.M.; Krtolica, A.; Galimi, F.; Narita, M.; Lowe, S.W.; Yaswen, P.; Campisi, J. Reversal of human cellular senescence: Roles of the p53 and p16 pathways. EMBO J. 2003, 22, 4212–4222. [Google Scholar] [CrossRef]
- Chen, Z.; Trotman, L.C.; Shaffer, D.; Lin, H.-K.; Dotan, Z.A.; Niki, M.; Koutcher, J.A.; Scher, H.I.; Ludwig, T.; Gerald, W.; et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 2005, 436, 725–730. [Google Scholar] [CrossRef] [Green Version]
- Bielak-Zmijewska, A.; Mosieniak, G.; Sikora, E. Is DNA damage indispensable for stress-induced senescence? Mech. Ageing Dev. 2018, 170, 13–21. [Google Scholar] [CrossRef]
- Lee, S.; Cho, H.-S.; Cho, W.-C.; Kim, S.-K.; Cho, Y.; Kim, C.-H. Operational durability of three-dimensional Ni-Fe layered double hydroxide electrocatalyst for water oxidation. Electrochimica Acta 2019, 315, 94–101. [Google Scholar] [CrossRef]
- Kaplon, J.; Zheng, L.; Meissl, K.; Chaneton, B.; Selivanov, V.; Mackay, G.; Van Der Burg, S.H.; Verdegaal, E.M.E.; Cascante, M.; Shlomi, T.; et al. A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature 2013, 498, 109–112. [Google Scholar] [CrossRef]
- Hampel, B.; Malisan, F.; Niederegger, H.; Testi, R.; Jansen-Dürr, P. Differential regulation of apoptotic cell death in senescent human cells. Exp. Gerontol. 2004, 39, 1713–1721. [Google Scholar] [CrossRef]
- Ritschka, B.; Storer, M.; Mas, A.; Heinzmann, F.; Ortells, M.C.; Morton, J.P.; Sansom, O.J.; Zender, L.; Keyes, W.M. The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration. Genes Dev. 2017, 31, 172–183. [Google Scholar] [CrossRef] [Green Version]
- Coppé, J.-P.; Patil, C.K.; Rodier, F.; Sun, Y.; Muñoz, D.P.; Goldstein, J.; Nelson, P.S.; Desprez, P.-Y.; Campisi, J. Senescence-Associated Secretory Phenotypes Reveal Cell-Nonautonomous Functions of Oncogenic RAS and the p53 Tumor Suppressor. PLoS Biol. 2008, 6, e301. [Google Scholar] [CrossRef]
- Takasugi, M.; Okada, R.; Takahashi, A.; Virya Chen, D.; Watanabe, S.; Hara, E. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA. Nat. Commun. 2017, 8, 15729. [Google Scholar] [CrossRef]
- Mosteiro, L.; Pantoja, C.; Alcazar, N.; Marión, R.M.; Chondronasiou, D.; Rovira, M.; Fernandez-Marcos, P.J.; Muñoz-Martin, M.; Blanco-Aparicio, C.; Pastor, J.; et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science 2016, 354, aaf4445. [Google Scholar] [CrossRef] [PubMed]
- Xue, W.; Zender, L.; Miething, C.; Dickins, R.A.; Hernando, E.; Krizhanovsky, V.; Cordon-Cardo, C.; Lowe, S.W. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 2007, 445, 656–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eggert, T.; Wolter, K.; Ji, J.; Ma, C.; Yevsa, T.; Klotz, S.; Medina-Echeverz, J.; Longerich, T.; Forgues, M.; Reisinger, F.; et al. Distinct Functions of Senescence-Associated Immune Responses in Liver Tumor Surveillance and Tumor Progression. Cancer Cell 2016, 30, 533–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
- He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef]
- Muñoz-Espín, D.; Serrano, M. Cellular senescence: From physiology to pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496. [Google Scholar] [CrossRef]
- Herbig, U.; Ferreira, M.; Condel, L.; Carey, D.; Sedivy, J.M. Cellular Senescence in Aging Primates. Science 2006, 311, 1257. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Sanoff, H.K.; Cho, H.; Burd, C.E.; Torrice, C.; Ibrahim, J.G.; Thomas, N.E.; Sharpless, N.E. Expression of p16INK4a in peripheral blood T-cells is a biomarker of human aging. Aging Cell 2009, 8, 439–448. [Google Scholar] [CrossRef] [Green Version]
- Helman, A.; Klochendler, A.; Azazmeh, N.; Gabai, Y.; Horwitz, E.; Anzi, S.; Swisa, A.; Condiotti, R.; Granit, R.Z.; Nevo, Y.; et al. p16Ink4a-induced senescence of pancreatic beta cells enhances insulin secretion. Nat. Med. 2016, 22, 412–420. [Google Scholar] [CrossRef] [Green Version]
- Kang, T.-W.; Yevsa, T.; Woller, N.; Hoenicke, L.; Wuestefeld, T.; Dauch, D.; Hohmeyer, A.; Gereke, M.; Rudalska, R.; Potapova, A.; et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 2011, 479, 547–551. [Google Scholar] [CrossRef]
- Sanoff, H.K.; Deal, A.M.; Krishnamurthy, J.; Torrice, C.; Dillon, P.; Sorrentino, J.; Ibrahim, J.G.; Jolly, T.A.; Williams, G.; Carey, L.A.; et al. Effect of Cytotoxic Chemotherapy on Markers of Molecular Age in Patients With Breast Cancer. JNCI J. Natl. Cancer Inst. 2014, 106, dju057. [Google Scholar] [CrossRef] [Green Version]
- DeMaria, M.; O’Leary, M.N.; Chang, J.; Shao, L.; Liu, S.; Alimirah, F.; Koenig, K.; Le, C.; Mitin, N.; Deal, A.M.; et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 2017, 7, 165–176. [Google Scholar] [CrossRef] [Green Version]
- Chang, J.; Wang, Y.; Shao, L.; Laberge, R.-M.; DeMaria, M.; Campisi, J.; Janakiraman, K.; Sharpless, N.E.; Ding, S.; Feng, W.; et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 2016, 22, 78–83. [Google Scholar] [CrossRef] [Green Version]
- Hudson, M.M.; Ness, K.K.; Gurney, J.G.; Mulrooney, D.A.; Chemaitilly, W.; Krull, K.R.; Green, D.M.; Armstrong, G.T.; Nottage, K.A.; Jones, K.E.; et al. Clinical Ascertainment of Health Outcomes Among Adults Treated for Childhood Cancer. JAMA J. Am. Med Assoc. 2013, 309, 2371–2381. [Google Scholar] [CrossRef]
- Milanovic, M.; Fan, D.N.Y.; Belenki, D.; Däbritz, J.H.M.; Zhao, Z.; Yu, Y.; Dörr, J.R.; Dimitrova, L.; Lenze, D.; Monteiro Barbosa, I.A.; et al. Senescence-associated reprogramming promotes cancer stemness. Nature 2018, 553, 96–100. [Google Scholar] [CrossRef] [Green Version]
- Hoare, M.; Narita, M. Transmitting senescence to the cell neighbourhood. Nat. Cell Biol. 2013, 15, 887–889. [Google Scholar] [CrossRef]
- Campisi, J. Aging and Cancer: The Double-Edged Sword of Replicative Senescence. J. Am. Geriatr. Soc. 1997, 45, 482–488. [Google Scholar] [CrossRef]
- Mavrogonatou, E.; Pratsinis, H.; Papadopoulou, A.; Karamanos, N.; Kletsas, D. Extracellular matrix alterations in senescent cells and their significance in tissue homeostasis. Matrix Biol. 2019, 75–76, 27–42. [Google Scholar] [CrossRef]
- Halazonetis, T.D.; Gorgoulis, V.G.; Bartek, J. An Oncogene-Induced DNA Damage Model for Cancer Development. Science 2008, 319, 1352–1355. [Google Scholar] [CrossRef] [Green Version]
- Gonzalez-Meljem, J.M.; Haston, S.; Carreno, G.; Apps, J.R.; Pozzi, S.; Stache, C.; Kaushal, G.; Virasami, A.; Panousopoulos, L.; Mousavy-Gharavy, S.N.; et al. Stem cell senescence drives age-attenuated induction of pituitary tumours in mouse models of paediatric craniopharyngioma. Nat. Commun. 2017, 8, 1819. [Google Scholar] [CrossRef]
- Gonzalez-Meljem, J.M.; Apps, J.R.; Fraser, H.C.; Martinez-Barbera, J.P. Paracrine roles of cellular senescence in promoting tumourigenesis. Br. J. Cancer 2018, 118, 1283–1288. [Google Scholar] [CrossRef]
- Patel, P.L.; Suram, A.; Mirani, N.; Bischof, O.; Herbig, U. Derepression of hTERT gene expression promotes escape from oncogene-induced cellular senescence. Proc. Natl. Acad. Sci. USA 2016, 113, E5024–E5033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, B.; Lam, E.W.-F.; Sun, Y. Senescent cells: A new Achilles’ heel to exploit for cancer medicine? Aging Cell 2018, 18, e12875. [Google Scholar] [CrossRef] [Green Version]
- Ness, K.K.; Kirkland, J.L.; Gramatges, M.M.; Wang, Z.; Kundu, M.; McCastlain, K.; Li-Harms, X.; Zhang, J.; Tchkonia, T.; Pluijm, S.M.F.; et al. Premature Physiologic Aging as a Paradigm for Understanding Increased Risk of Adverse Health Across the Lifespan of Survivors of Childhood Cancer. J. Clin. Oncol. 2018, 36, 2206–2215. [Google Scholar] [CrossRef] [PubMed]
- Cupit-Link, M.C.; Kirkland, J.L.; Ness, K.K.; Armstrong, G.T.; Tchkonia, T.; LeBrasseur, N.K.; Armenian, S.H.; Ruddy, K.J.; Hashmi, S.K. Biology of premature ageing in survivors of cancer. ESMO Open 2017, 2, e000250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Myrianthopoulos, V.; Evangelou, K.; Vasileiou, P.; Cooks, T.; Vassilakopoulos, T.P.; Pangalis, G.A.; Kouloukoussa, M.; Kittas, C.; Georgakilas, A.G.; Gorgoulis, V.G. Senescence and senotherapeutics: A new field in cancer therapy. Pharmacol. Ther. 2019, 193, 31–49. [Google Scholar] [CrossRef]
- Kirkland, J.L.; Tchkonia, T.; Zhu, Y.; Niedernhofer, L.J.; Robbins, P.D. The Clinical Potential of Senolytic Drugs. J. Am. Geriatr. Soc. 2017, 65, 2297–2301. [Google Scholar] [CrossRef]
- Baker, D.J.; Perez-Terzic, C.; Jin, F.; Pitel, K.S.; Niederländer, N.J.; Jeganathan, K.; Yamada, S.; Reyes, S.; Rowe, L.; Hiddinga, H.J.; et al. Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nat. Cell Biol. 2008, 10, 825–836. [Google Scholar] [CrossRef]
- Evangelou, K.; Lougiakis, N.; Rizou, S.V.; Kotsinas, A.; Kletsas, D.; Muñoz-Espín, D.; Kastrinakis, N.G.; Pouli, N.; Marakos, P.; Townsend, P.; et al. Robust, universal biomarker assay to detect senescent cells in biological specimens. Aging Cell 2017, 16, 192–197. [Google Scholar] [CrossRef]
- Freund, A.; Laberge, R.-M.; Demaria, M.; Campisi, J. Lamin B1 loss is a senescence-associated biomarker. Mol. Biol. Cell 2012, 23, 2066–2075. [Google Scholar] [CrossRef]
- Chambers, C.R.; Ritchie, S.; Pereira, B.A.; Timpson, P. Overcoming the senescence-associated secretory phenotype (SASP): A complex mechanism of resistance in the treatment of cancer. Mol. Oncol. 2021, 15, 3242–3255. [Google Scholar] [CrossRef]
- Birch, J.; Gil, J. Senescence and the SASP: Many therapeutic avenues. Genes Dev. 2020, 34, 1565–1576. [Google Scholar] [CrossRef]
- Coppé, J.-P.; Patil, C.K.; Rodier, F.; Krtolica, A.; Beauséjour, C.M.; Parrinello, S.; Hodgson, J.G.; Chin, K.; Desprez, P.-Y.; Campisi, J. A Human-Like Senescence-Associated Secretory Phenotype Is Conserved in Mouse Cells Dependent on Physiological Oxygen. PLoS ONE 2010, 5, e9188. [Google Scholar] [CrossRef]
- Rudin, C.M.; Hann, C.L.; Garon, E.B.; De Oliveira, M.R.; Bonomi, P.D.; Camidge, D.R.; Chu, Q.; Giaccone, G.; Khaira, D.; Ramalingam, S.S.; et al. Phase II Study of Single-Agent Navitoclax (ABT-263) and Biomarker Correlates in Patients with Relapsed Small Cell Lung Cancer. Clin. Cancer Res. 2012, 18, 3163–3169. [Google Scholar] [CrossRef] [Green Version]
- Berenbaum, F. Osteoarthritis as an inflammatory disease (osteoarthritis is not osteoarthrosis!). Osteoarthr. Cartil. 2013, 21, 16–21. [Google Scholar] [CrossRef] [Green Version]
- Wieland, H.A.; Michaelis, M.; Kirschbaum, B.J.; Rudolphi, K.A. Osteoarthritis—An untreatable disease? Nat. Rev. Drug Discov. 2005, 4, 331–344. [Google Scholar] [CrossRef]
- March, L.; Amatya, B.; Osborne, R.H.; Brand, C. Developing a minimum standard of care for treating people with osteoarthritis of the hip and knee. Best Pr. Res. Clin. Rheumatol. 2010, 24, 121–145. [Google Scholar] [CrossRef]
- Jeon, O.H.; Kim, C.; Laberge, R.-M.; DeMaria, M.; Rathod, S.; Vasserot, A.P.; Chung, J.W.; Kim, D.H.; Poon, Y.; David, N.; et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat. Med. 2017, 23, 775–781. [Google Scholar] [CrossRef]
- Zhu, Y.I.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015, 14, 644–658. [Google Scholar] [CrossRef]
- Kirkland, J.L.; Tchkonia, T. Senolytic drugs: From discovery to translation. J. Intern. Med. 2020, 288, 518–536. [Google Scholar] [CrossRef]
- Keating, G.M. Dasatinib: A Review in Chronic Myeloid Leukaemia and Ph+ Acute Lymphoblastic Leukaemia. Drugs 2016, 77, 85–96. [Google Scholar] [CrossRef]
- Li, J.; Fang, B.; Kinose, F.; Bai, Y.; Kim, J.-Y.; Chen, Y.; Rix, U.; Koomen, J.M.; Haura, E.B. Target Identification in Small Cell Lung Cancer via Integrated Phenotypic Screening and Activity-Based Protein Profiling. Mol. Cancer Ther. 2016, 15, 334–342. [Google Scholar] [CrossRef] [Green Version]
- Yosef, R.; Pilpel, N.; Tokarsky-Amiel, R.; Biran, A.; Ovadya, Y.; Cohen, S.; Vadai, E.; Dassa, L.; Shahar, E.; Condiotti, R.; et al. Directed elimination of senescent cells by inhibition of BCL-W and BCL-XL. Nat. Commun. 2016, 7, 11190. [Google Scholar] [CrossRef]
- Bruning, A. Inhibition of mTOR Signaling by Quercetin in Cancer Treatment and Prevention. Anti Cancer Agents Med. Chem. 2013, 13, 1025–1031. [Google Scholar] [CrossRef] [PubMed]
- Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; Lebrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hickson, L.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.; Tchkonia, T.; Fuhrmann-Stroissnigg, H.; Dai, H.; Ling, Y.Y.; Stout, M.B.; Pirtskhalava, T.; Giorgadze, N.; Johnson, K.O.; Giles, C.B.; et al. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 2016, 15, 428–435. [Google Scholar] [CrossRef]
- Tarantini, S.; Balasubramanian, P.; Delfavero, J.; Csipo, T.; Yabluchanskiy, A.; Kiss, T.; Nyúl-Tóth, Á.; Mukli, P.; Toth, P.; Ahire, C.; et al. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in aged mice. GeroScience 2021, 43, 2427–2440. [Google Scholar] [CrossRef]
- Zhu, Y.; Doornebal, E.; Pirtskhalava, T.; Giorgadze, N.; Wentworth, M.; Fuhrmann-Stroissnigg, H.; Niedernhofer, L.; Robbins, P.; Tchkonia, T.; Kirkland, J.L. New agents that target senescent cells: The flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging 2017, 9, 955–963. [Google Scholar] [CrossRef] [Green Version]
- Yousefzadeh, M.J.; Zhu, Y.; McGowan, S.J.; Angelini, L.; Fuhrmann-Stroissnigg, H.; Xu, M.; Ling, Y.Y.; Melos, K.I.; Pirtskhalava, T.; Inman, C.L.; et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 2018, 36, 18–28. [Google Scholar] [CrossRef] [Green Version]
- Chiu, S.-J.; Yang, P.-M.; Tseng, H.-H.; Peng, C.-W.; Chen, W.-S. Dietary flavonoid fisetin targets caspase-3-deficient human breast cancer MCF-7 cells by induction of caspase-7-associated apoptosis and inhibition of autophagy. Int. J. Oncol. 2011, 40, 469–478. [Google Scholar] [CrossRef] [Green Version]
- Didelot, C.; Lanneau, D.; Brunet, M.; Joly, A.-L.; Thonel, A.; Chiosis, G.; Garrido, C. Anti-Cancer Therapeutic Approaches Based on Intracellular and Extracellular Heat Shock Proteins. Curr. Med. Chem. 2007, 14, 2839–2847. [Google Scholar] [CrossRef]
- Solit, D.B.; Rosen, N. Hsp90: A novel target for cancer therapy. Curr. Top. Med. Chem. 2006, 6, 1205–1214. [Google Scholar] [CrossRef]
- Gupta, S.D.; Pan, C.H. Recent update on discovery and development of Hsp90 inhibitors as senolytic agents. Int. J. Biol. Macromol. 2020, 161, 1086–1098. [Google Scholar] [CrossRef]
- Dimopoulos, M.-A.; Mitsiades, C.S.; Anderson, K.C.; Richardson, P.G. Tanespimycin as Antitumor Therapy. Clin. Lymphoma Myeloma Leuk. 2011, 11, 17–22. [Google Scholar] [CrossRef]
- Ochel, H.-J.; Eichhorn, K.; Gademann, G. Geldanamycin: The prototype of a class of antitumor drugs targeting the heat shock protein 90 family of molecular chaperones. Cell Stress Chaperon 2001, 6, 105–112. [Google Scholar] [CrossRef] [Green Version]
- Singh, S.B.; Genilloud, O.; Peláez, F. 2.05-Terrestrial Microorganisms—Filamentous Bacteria. In Comprehensive Natural Products II.; Liu, H.-W., Mander, L., Eds.; Elsevier: Oxford, UK, 2010; pp. 109–140. [Google Scholar]
- Sharp, S.; Jones, K.; Workman, P. Chapter 9-Exploiting Cancer Dependence on Molecular Chaperones: HSP90 Inhibitors Past, Present, and Future. In Cancer Drug Design and Discovery, 2nd ed.; Neidle, S., Ed.; Academic Press: San Diego, CA, USA, 2014; pp. 239–274. [Google Scholar]
- Giulino-Roth, L.; Van Besien, H.J.; Dalton, T.; Totonchy, J.E.; Rodina, A.; Taldone, T.; Bolaender, A.; Erdjument-Bromage, H.; Sadek, J.; Chadburn, A.; et al. Inhibition of Hsp90 Suppresses PI3K/AKT/mTOR Signaling and Has Antitumor Activity in Burkitt Lymphoma. Mol. Cancer Ther. 2017, 16, 1779–1790. [Google Scholar] [CrossRef] [Green Version]
- Joshi, P.; Maidji, E.; Stoddart, C.A. Inhibition of Heat Shock Protein 90 Prevents HIV Rebound. J. Biol. Chem. 2016, 291, 10332–10346. [Google Scholar] [CrossRef] [Green Version]
- Samaraweera, L.; Adomako, A.; Rodriguez-Gabin, A.; McDaid, H.M. A Novel Indication for Panobinostat as a Senolytic Drug in NSCLC and HNSCC. Sci. Rep. 2017, 7, 1900. [Google Scholar] [CrossRef]
- Prystowsky, M.; Feeney, K.; Kawachi, N.; Montagna, C.; Willmott, M.; Wasson, C.; Antkowiak, M.; Loudig, O.; Parish, J. Inhibition of Plk1 and Cyclin B1 Expression Results in Panobinostat-Induced G2 Delay and Mitotic Defects. Sci. Rep. 2013, 3, 2640. [Google Scholar] [CrossRef] [Green Version]
- Triana-Martínez, F.; Picallos-Rabina, P.; Alvarez, S.D.S.; Pietrocola, F.; Llanos, S.; Rodilla, V.; Soprano, E.; Pedrosa, P.; Ferreirós, A.; Barradas, M.; et al. Identification and characterization of Cardiac Glycosides as senolytic compounds. Nat. Commun. 2019, 10, 1–12. [Google Scholar] [CrossRef]
- Guerrero, A.; Herranz, N.; Sun, B.; Wagner, V.; Gallage, S.; Guiho, R.; Wolter, K.; Pombo, J.; Irvine, E.E.; Innes, A.J.; et al. Cardiac glycosides are broad-spectrum senolytics. Nat. Metab. 2019, 1, 1074–1088. [Google Scholar] [CrossRef]
- Shi, H.; Mao, X.; Zhong, Y.; Liu, Y.; Zhao, X.; Yu, K.; Zhu, R.; Wei, Y.; Zhu, J.; Sun, H.; et al. Digoxin reduces atherosclerosis in apolipoprotein E-deficient mice. J. Cereb. Blood Flow Metab. 2016, 173, 1517–1528. [Google Scholar] [CrossRef]
- Li, B.; Huang, X.; Liu, Z.; Xu, X.; Xiao, H.; Zhang, X.; Dai, H.; Wang, C. Ouabain ameliorates bleomycin induced pulmonary fibrosis by inhibiting proliferation and promoting apoptosis of lung fibroblasts. Am. J. Transl. Res. 2018, 10, 2967–2974. [Google Scholar]
- Baar, M.P.; Brandt, R.M.C.; Putavet, D.A.; Klein, J.D.D.; Derks, K.W.J.; Bourgeois, B.R.M.; Stryeck, S.; Rijksen, Y.; Van Willigenburg, H.; Feijtel, D.A.; et al. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell 2017, 169, 132–147.e16. [Google Scholar] [CrossRef] [Green Version]
- Zhang, C.; Xie, Y.; Chen, H.; Lv, L.; Yao, J.; Zhang, M.; Xia, K.; Feng, X.; Li, Y.; Liang, X.; et al. FOXO4-DRI alleviates age-related testosterone secretion insufficiency by targeting senescent Leydig cells in aged mice. Aging 2020, 12, 1272–1284. [Google Scholar] [CrossRef]
- Meng, J.; Li, Y.; Wan, C.; Sun, Y.; Dai, X.; Huang, J.; Hu, Y.; Gao, Y.; Wu, B.; Zhang, Z.; et al. Targeting senescence-like fibroblasts radiosensitizes non–small cell lung cancer and reduces radiation-induced pulmonary fibrosis. JCI Insight 2021, 6, e146334. [Google Scholar] [CrossRef]
- Le, H.H.; Cinaroglu, S.S.; Manalo, E.C.; Ors, A.; Gomes, M.M.; Sahbaz, B.D.; Bonic, K.; Marmolejo, C.A.O.; Quentel, A.; Plaut, J.S.; et al. Molecular modelling of the FOXO4-TP53 interaction to design senolytic peptides for the elimination of senescent cancer cells. EBioMedicine 2021, 73, 103646. [Google Scholar] [CrossRef]
- Nogueira-Recalde, U.; Lorenzo-Gómez, I.; Blanco, F.J.; Loza, M.I.; Grassi, D.; Shirinsky, V.; Shirinsky, I.; Lotz, M.; Robbins, P.D.; Domínguez, E.; et al. Fibrates as drugs with senolytic and autophagic activity for osteoarthritis therapy. EBioMedicine 2019, 45, 588–605. [Google Scholar] [CrossRef] [Green Version]
- Ballou, L.M.; Lin, R.Z. Rapamycin and mTOR kinase inhibitors. J. Chem. Biol. 2008, 1, 27–36. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One Drug, Many Effects. Cell Metab. 2014, 19, 373–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, R.; Yu, Z.; Sunchu, B.; Shoaf, J.; Dang, I.; Zhao, S.; Caples, K.; Bradley, L.; Beaver, L.M.; Ho, E.; et al. Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism. Aging Cell 2017, 16, 564–574. [Google Scholar] [CrossRef] [PubMed]
- Laberge, R.-M.; Sun, Y.; Orjalo, A.V.; Patil, C.K.; Freund, A.; Zhou, L.; Curran, S.C.; Davalos, A.R.; Wilson-Edell, K.A.; Liu, S.; et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol. 2015, 17, 1049–1061. [Google Scholar] [CrossRef] [PubMed]
- Walters, H.E.; Cox, L.S. mTORC Inhibitors as Broad-Spectrum Therapeutics for Age-Related Diseases. Int. J. Mol. Sci. 2018, 19, 2325. [Google Scholar] [CrossRef] [Green Version]
- Kim, L.C.; Cook, R.S.; Chen, J. mTORC1 and mTORC2 in cancer and the tumor microenvironment. Oncogene 2016, 36, 2191–2201. [Google Scholar] [CrossRef] [Green Version]
- Chen, G.Y.; Nuñez, G. Sterile inflammation: Sensing and reacting to damage. Nat. Rev. Immunol. 2010, 10, 826–837. [Google Scholar] [CrossRef] [Green Version]
- Xu, M.; Tchkonia, T.; Ding, H.; Ogrodnik, M.; Lubbers, E.R.; Pirtskhalava, T.; White, T.A.; Johnson, K.O.; Stout, M.B.; Mezera, V.; et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl. Acad. Sci. USA 2015, 112, E6301–E6310. [Google Scholar] [CrossRef] [Green Version]
- Griveau, A.; Wiel, C.; Ziegler, D.V.; Bergo, M.O.; Bernard, D. The JAK1/2 inhibitor ruxolitinib delays premature aging phenotypes. Aging Cell 2020, 19, e13122. [Google Scholar] [CrossRef] [Green Version]
- Marconi, V.C.; Moser, C.; Gavegnano, C.; Deeks, S.G.; Lederman, M.M.; Overton, E.T.; Tsibris, A.; Hunt, P.W.; Kantor, A.; Sekaly, R.-P.; et al. Randomized Trial of Ruxolitinib in Antiretroviral-Treated Adults With Human Immunodeficiency Virus. Clin. Infect. Dis. 2021, 74, 95–104. [Google Scholar] [CrossRef]
- Barzilai, N.; Crandall, J.P.; Kritchevsky, S.B.; Espeland, M.A. Metformin as a Tool to Target Aging. Cell Metab. 2016, 23, 1060–1065. [Google Scholar] [CrossRef] [Green Version]
- Moiseeva, O.; Deschênes-Simard, X.; St-Germain, E.; Igelmann, S.; Huot, G.; Cadar, A.E.; Bourdeau, V.; Pollak, M.N.; Ferbeyre, G. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell 2013, 12, 489–498. [Google Scholar] [CrossRef]
- Anisimov, V.N.; Berstein, L.M.; Popovich, I.G.; Zabezhinski, M.A.; Egormin, P.A.; Piskunova, T.S.; Semenchenko, A.V.; Tyndyk, M.L.; Yurova, M.; Kovalenko, I.G.; et al. If started early in life, metformin treatment increases life span and postpones tumors in female SHR mice. Aging 2011, 3, 148–157. [Google Scholar] [CrossRef] [Green Version]
- Xu, M.; Palmer, A.; Ding, H.; Weivoda, M.M.; Pirtskhalava, T.; White, T.A.; Sepe, A.; Johnson, K.O.; Stout, M.B.; Giorgadze, N.; et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife 2015, 4, e12997. [Google Scholar] [CrossRef]
- Barzilai, N. TARGETING AGING WITH METFORMIN (TAME). Innov. Aging 2017, 1, 743. [Google Scholar] [CrossRef] [Green Version]
- Mohammed, I.; Hollenberg, M.D.; Ding, H.; Triggle, C.R. A Critical Review of the Evidence That Metformin Is a Putative Anti-Aging Drug That Enhances Healthspan and Extends Lifespan. Front. Endocrinol. 2021, 12, 12. [Google Scholar] [CrossRef] [PubMed]
- Bhat, K.P.L.; Kosmeder, J.W., II; Pezzuto, J.M. Biological Effects of Resveratrol. Antioxid. Redox Signal. 2001, 3, 1041–1064. [Google Scholar] [CrossRef] [PubMed]
- King, R.E.; Bomser, J.A.; Min, D.B. Bioactivity of Resveratrol. Compr. Rev. Food Sci. Food Saf. 2006, 5, 65–70. [Google Scholar] [CrossRef]
- Yang, T.; Wang, L.; Zhu, M.; Zhang, L.; Yan, L. Properties and molecular mechanisms of resveratrol: A review. Die Pharmazie 2015, 70, 501–506. [Google Scholar]
- Pollack, R.M.; Crandall, J.P. Resveratrol: Therapeutic Potential for Improving Cardiometabolic Health. Am. J. Hypertens. 2013, 26, 1260–1268. [Google Scholar] [CrossRef] [Green Version]
- Sarkar, F.H.; Li, Y.; Wang, Z.; Kong, D. Cellular signaling perturbation by natural products. Cell. Signal. 2009, 21, 1541–1547. [Google Scholar] [CrossRef] [Green Version]
- Huffman, D.M.; Schafer, M.J.; LeBrasseur, N. Energetic interventions for healthspan and resiliency with aging. Exp. Gerontol. 2016, 86, 73–83. [Google Scholar] [CrossRef] [Green Version]
- Pitozzi, V.; Mocali, A.; Laurenzana, A.; Giannoni, E.; Cifola, I.; Battaglia, C.; Chiarugi, P.; Dolara, P.; Giovannelli, L. Chronic Resveratrol Treatment Ameliorates Cell Adhesion and Mitigates the Inflammatory Phenotype in Senescent Human Fibroblasts. Journals Gerontol. Ser. A 2012, 68, 371–381. [Google Scholar] [CrossRef]
- Yoshino, J.; Conte, C.; Fontana, L.; Mittendorfer, B.; Imai, S.-I.; Schechtman, K.B.; Gu, C.C.; Kunz, I.; Fanelli, F.R.; Patterson, B.W.; et al. Resveratrol Supplementation Does Not Improve Metabolic Function in Nonobese Women with Normal Glucose Tolerance. Cell Metab. 2012, 16, 658–664. [Google Scholar] [CrossRef] [Green Version]
- Poulsen, M.M.; Vestergaard, P.F.; Clasen, B.F.; Radko, Y.; Christensen, L.P.; Stødkilde-Jørgensen, H.; Møller, N.; Jessen, N.; Pedersen, S.B.; Jørgensen, J.O. High-dose resveratrol supplementation in obese men: An investigator-initiated, randomized, placebo-controlled clinical trial of substrate metabolism, insulin sensitivity, and body composition. Diabetes 2013, 62, 1186–1195. [Google Scholar] [CrossRef] [Green Version]
- Agarwal, B.; Campen, M.J.; Channell, M.M.; Wherry, S.J.; Varamini, B.; Davis, J.G.; Baur, J.A.; Smoliga, J.M. Resveratrol for primary prevention of atherosclerosis: Clinical trial evidence for improved gene expression in vascular endothelium. Int. J. Cardiol. 2013, 166, 246–248. [Google Scholar] [CrossRef] [Green Version]
- Kang, H.T.; Park, J.T.; Choi, K.; Kim, Y.; Choi, H.J.C.; Jung, C.W.; Lee, Y.-S.; Park, S.C. Chemical screening identifies ATM as a target for alleviating senescence. Nat. Chem. Biol. 2017, 13, 616–623. [Google Scholar] [CrossRef]
- Egger, M.; Johnson, L.F.; Wandeler, G. Trends in life expectancy of HIV-positive adults on antiretroviral therapy across the globe: Comparisons with general population. Curr. Opin. HIV AIDS 2016, 11, 492–500. [Google Scholar] [CrossRef]
- Finzi, D.; Hermankova, M.; Pierson, T.; Carruth, L.M.; Buck, C.; Chaisson, R.E.; Quinn, T.C.; Chadwick, K.; Margolick, J.; Brookmeyer, R.; et al. Identification of a Reservoir for HIV-1 in Patients on Highly Active Antiretroviral Therapy. Science 1997, 278, 1295–1300. [Google Scholar] [CrossRef]
- Chun, T.-W.; Stuyver, L.; Mizell, S.B.; Ehler, L.A.; Mican, J.A.M.; Baseler, M.; Lloyd, A.L.; Nowak, M.A.; Fauci, A.S. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc. Natl. Acad. Sci. 1997, 94, 13193–13197. [Google Scholar] [CrossRef] [Green Version]
- Spudich, S.; Robertson, K.R.; Bosch, R.J.; Gandhi, R.T.; Cyktor, J.C.; Mar, H.; Macatangay, B.J.; Lalama, C.M.; Rinaldo, C.; Collier, A.C.; et al. Persistent HIV-infected cells in cerebrospinal fluid are associated with poorer neurocognitive performance. J. Clin. Investig. 2019, 129, 3339–3346. [Google Scholar] [CrossRef] [Green Version]
- Blanco, J.R.; Negredo, E.; Bernal, E.; Blanco, J. Impact of HIV infection on aging and immune status. Expert Rev. Anti Infect. Ther. 2021, 19, 719–731. [Google Scholar] [CrossRef]
- Monti, D.; Ostan, R.; Borelli, V.; Castellani, G.; Franceschi, C. Inflammaging and human longevity in the omics era. Mech. Ageing Dev. 2017, 165, 129–138. [Google Scholar] [CrossRef]
- Deeks, S.G. HIV Infection, Inflammation, Immunosenescence, and Aging. Annu. Rev. Med. 2011, 62, 141–155. [Google Scholar] [CrossRef] [Green Version]
- Guaraldi, G.; Orlando, G.; Zona, S.; Menozzi, M.; Carli, F.; Garlassi, E.; Berti, A.; Rossi, E.; Roverato, A.; Palella, F. Premature Age-Related Comorbidities Among HIV-Infected Persons Compared With the General Population. Clin. Infect. Dis. 2011, 53, 1120–1126. [Google Scholar] [CrossRef] [Green Version]
- High, K.P.; Brennan-Ing, M.; Clifford, D.B.; Cohen, M.H.; Currier, J.; Deeks, S.G.; Deren, S.; Effros, R.B.; Gebo, K.; Goronzy, J.J. HIV and aging: State of knowledge and areas of critical need for research. A report to the NIH Office of AIDS Research by the HIV and Aging Working Group. J. Acquir. Immune Defic. Syndr. 2012, 60, S1–S18. [Google Scholar] [CrossRef] [Green Version]
- Song, P.; An, J.; Zou, M.-H. Immune Clearance of Senescent Cells to Combat Ageing and Chronic Diseases. Cells 2020, 9, 671. [Google Scholar] [CrossRef] [Green Version]
- Sanada, F.; Taniyama, Y.; Muratsu, J.; Otsu, R.; Shimizu, H.; Rakugi, H.; Morishita, R. Source of Chronic Inflammation in Aging. Front. Cardiovasc. Med. 2018, 5, 12. [Google Scholar] [CrossRef] [Green Version]
- Mangino, G.; Percario, Z.A.; Fiorucci, G.; Vaccari, G.; Acconcia, F.; Chiarabelli, C.; Leone, S.; Noto, A.; Horenkamp, F.A.; Manrique, S.; et al. HIV-1 Nef Induces Proinflammatory State in Macrophages through Its Acidic Cluster Domain: Involvement of TNF Alpha Receptor Associated Factor. PLoS ONE 2011, 6, e22982. [Google Scholar] [CrossRef] [Green Version]
- Christensen-Quick, A.; Massanella, M.; Frick, A.; Rawlings, S.; Spina, C.; Vargas-Meneses, M.; Schrier, R.; Nakazawa, M.; Anderson, C.; Gianella, S. Subclinical Cytomegalovirus DNA Is Associated with CD4 T Cell Activation and Impaired CD8 T Cell CD107a Expression in People Living with HIV despite Early Antiretroviral Therapy. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
- Williams, B.; Landay, A.; Presti, R.M. Microbiome alterations in HIV infection a review. Cell. Microbiol. 2016, 18, 645–651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chomont, N.; El-Far, M.; Ancuta, P.; Trautmann, L.; Procopio, F.A.; Yassine-Diab, B.; Boucher, G.; Boulassel, M.-R.; Ghattas, G.; Brenchley, J.M.; et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat. Med. 2009, 15, 893–900. [Google Scholar] [CrossRef] [PubMed]
- Ho, D.D.; Rota, T.R.; Hirsch, M.S. Infection of monocyte/macrophages by human T lymphotropic virus type III. J. Clin. Investig. 1986, 77, 1712–1715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wong, M.E.; Jaworowski, A.; Hearps, A.C. The HIV Reservoir in Monocytes and Macrophages. Front. Immunol. 2019, 10, 1435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, N.C.; Partridge, A.T.; Tuzer, F.; Cohen, J.; Nacarelli, T.; Navas-Martín, S.; Sell, C.; Torres, C.; Martín-García, J. Induction of a Senescence-Like Phenotype in Cultured Human Fetal Microglia During HIV-1 Infection. J. Gerontol. Ser. A 2018, 73, 1187–1196. [Google Scholar] [CrossRef] [Green Version]
- Ribeiro, S.P.; Milush, J.M.; Cunha-Neto, E.; Kallas, E.; Kalil, J.; Passero, L.F.D.; Hunt, P.W.; Deeks, S.G.; Nixon, D.F.; Sengupta, D. Correction: p16INK4a Expression and Immunologic Aging in Chronic HIV Infection. PLoS ONE 2017, 12, e0176509. [Google Scholar] [CrossRef] [Green Version]
- Valdez, H.; Connick, E.; Smith, K.Y.; Lederman, M.M.; Bosch, R.J.; Kim, R.S.; Clair, M.S.; Kuritzkes, D.R.; Kessler, H.; Fox, L.; et al. Limited immune restoration after 3 years’ suppression of HIV-1 replication in patients with moderately advanced disease. AIDS 2002, 16, 1859–1866. [Google Scholar] [CrossRef]
- Jain, V.; Hartogensis, W.; Bacchetti, P.; Hunt, P.W.; Hatano, H.; Sinclair, E.; Epling, L.; Lee, T.-H.; Busch, M.P.; McCune, J.M.; et al. Antiretroviral Therapy Initiated Within 6 Months of HIV Infection Is Associated With Lower T-Cell Activation and Smaller HIV Reservoir Size. J. Infect. Dis. 2013, 208, 1202–1211. [Google Scholar] [CrossRef] [Green Version]
- Hocini, H.; Bonnabau, H.; Lacabaratz, C.; Lefebvre, C.; Tisserand, P.; Foucat, E.; Lelièvre, J.-D.; Lambotte, O.; Saez–Cirion, A.; Versmisse, P.; et al. HIV Controllers Have Low Inflammation Associated with a Strong HIV-Specific Immune Response in Blood. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
- Szaniawski, M.; Spivak, A.M. Senotherapeutics and HIV-1 Persistence. Curr. HIV/AIDS Rep. 2020, 17, 219–225. [Google Scholar] [CrossRef]
- Baker, D.J.; Wijshake, T.; Tchkonia, T.; Lebrasseur, N.K.; Childs, B.G.; Van De Sluis, B.; Kirkland, J.L.; Van Deursen, J.M. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef]
- Nelson, J.A.; Krishnamurthy, J.; Menezes, P.; Liu, Y.; Hudgens, M.G.; Sharpless, N.E.; Eron, J.J., Jr. Expression of p16(INK4a) as a biomarker of T-cell aging in HIV-infected patients prior to and during antiretroviral therapy. Aging Cell 2012, 11, 916–918. [Google Scholar] [CrossRef] [Green Version]
- Egger, M.; May, M.; Chêne, G.; Phillips, A.N.; Ledergerber, B.; Dabis, F.; Costagliola, D.; Monforte, A.D.; de Wolf, F.; Reiss, P.; et al. Prognosis of HIV-1-infected patients starting highly active antiretroviral therapy: A collaborative analysis of prospective studies. Lancet 2002, 360, 119–129. [Google Scholar] [CrossRef]
- Palella, F.J., Jr.; Delaney, K.M.; Moorman, A.C.; Loveless, M.O.; Fuhrer, J.; Satten, G.A.; Aschman, D.J.; Holmberg, S.D.; The HIV Outpatient Study Investigators. Declining Morbidity and Mortality among Patients with Advanced Human Immunodeficiency Virus Infection. N. Engl. J. Med. 1998, 338, 853–860. [Google Scholar] [CrossRef]
- Lagoumtzi, S.M.; Chondrogianni, N. Senolytics and senomorphics: Natural and synthetic therapeutics in the treatment of aging and chronic diseases. Free Radic. Biol. Med. 2021, 171, 169–190. [Google Scholar] [CrossRef] [PubMed]
- Chandrasekar, A.P.; Cummins, N.W.; Badley, A.D. The Role of the BCL-2 Family of Proteins in HIV-1 Pathogenesis and Persistence. Clin. Microbiol. Rev. 2019, 33. [Google Scholar] [CrossRef] [PubMed]
- Zheng, L.; Yang, Y.; Guocai, L.; Pauza, C.D.; Salvato, M.S. HIV Tat Protein Increases Bcl-2 Expression in Monocytes Which Inhibits Monocyte Apoptosis Induced by Tumor Necrosis Factor-Alpha-Related Apoptosis-Induced Ligand. Intervirology 2007, 50, 224–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Szaniawski, M.; Spivak, A.M. Senotherapeutics for HIV and aging. Curr. Opin. HIV AIDS 2020, 15, 83–93. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Zhu, X.; Ji, H.; Deng, J.; Lu, P.; Jiang, Z.; Li, X.; Wang, Y.; Wang, C.; Zhao, J.; et al. Quercetin synergistically reactivates human immunodeficiency virus type 1 latency by activating nuclear factor-κB. Mol. Med. Rep. 2017, 17, 2501–2508. [Google Scholar] [CrossRef] [PubMed]
- Boidol, B.; Kornauth, C.; Van Der Kouwe, E.; Prutsch, N.; Kazianka, L.; Gültekin, S.; Hoermann, G.; Mayerhoefer, M.E.; Hopfinger, G.; Hauswirth, A.; et al. First-in-human response of BCL-2 inhibitor venetoclax in T-cell prolymphocytic leukemia. Blood 2017, 130, 2499–2503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cummins, N.W.; Sainski-Nguyen, A.M.; Natesampillai, S.; Aboulnasr, F.; Kaufmann, S.; Badley, A.D. Maintenance of the HIV Reservoir Is Antagonized by Selective BCL2 Inhibition. J. Virol. 2017, 91, e00012-17. [Google Scholar] [CrossRef] [Green Version]
- Stock, P.G.; Barin, B.; Hatano, H.; Rogers, R.L.; Roland, M.E.; Lee, T.-H.; Busch, M.; Deeks, S.G.; for Solid Organ Transplantation in HIV Study Investigators. Reduction of HIV persistence following transplantation in HIV-infected kidney transplant recipients. Am. J. Transplant. 2014, 14, 1136–1141. [Google Scholar] [CrossRef] [Green Version]
- Gavegnano, C.; Detorio, M.; Montero, C.; Bosque, A.; Planelles, V.; Schinazi, R.F. Ruxolitinib and Tofacitinib Are Potent and Selective Inhibitors of HIV-1 Replication and Virus ReactivationIn Vitro. Antimicrob. Agents Chemother. 2014, 58, 1977–1986. [Google Scholar] [CrossRef] [Green Version]
- Bermejo, M.; López-Huertas, M.R.; García-Pérez, J.; Climent, N.; Descours, B.; Ambrosioni, J.; Mateos, E.; Rodríguez-Mora, S.; Rus-Bercial, L.; Benkirane, M.; et al. Dasatinib inhibits HIV-1 replication through the interference of SAMHD1 phosphorylation in CD4+ T cells. Biochem. Pharmacol. 2016, 106, 30–45. [Google Scholar] [CrossRef]
- Kjær, A.S.H.K.; Brinkmann, C.R.; Dinarello, C.A.; Olesen, R.; Østergaard, L.; Søgaard, O.S.; Tolstrup, M.; Rasmussen, T.A. The histone deacetylase inhibitor panobinostat lowers biomarkers of cardiovascular risk and inflammation in HIV patients. AIDS 2015, 29, 1195–1200. [Google Scholar] [CrossRef]
- De Armas, L.R.; Gavegnano, C.; Pallikkuth, S.; Rinaldi, S.; Pan, L.; Battivelli, E.; Verdin, E.; Younis, R.T.; Pahwa, R.; Williams, S.L. The Effect of JAK1/2 Inhibitors on HIV Reservoir Using Primary Lymphoid Cell Model of HIV Latency. Front. Immunol. 2021, 12, 720697. [Google Scholar] [CrossRef]
- Gavegnano, C.; Haile, W.B.; Hurwitz, S.; Tao, S.; Jiang, Y.; Schinazi, R.F.; Tyor, W.R. Baricitinib reverses HIV-associated neurocognitive disorders in a SCID mouse model and reservoir seeding in vitro. J. Neuroinflamm. 2019, 16, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Planas, D.; Pagliuzza, A.; Ponte, R.; Fert, A.; Marchand, L.R.; Massanella, M.; Gosselin, A.; Mehraj, V.; Dupuy, F.P.; Isnard, S.; et al. LILAC pilot study: Effects of metformin on mTOR activation and HIV reservoir persistence during antiretroviral therapy. EBioMedicine 2021, 65, 103270. [Google Scholar] [CrossRef]
SASP Factors | |
---|---|
Soluble factors | |
Interleukins (IL): IL-6, IL-7, IL-8, IL-10, IL-1a, -1b, IL-13, IL-15 | Soluble or shed receptors or ligands ICAM-1, -3, OPG, sTNFRI, TRAIL-R3, Fas, sTNFRII, Fas, uPAR, SGP130, EGF-R |
Chemokines (CXCL, CCL): CCL2, CCL20, GRO-a,-b,-g, MCP-2, MCP-4, MIP-1ª, MIP-3ª, HCC-4, Eotaxin, Eotaxin-3, TECK, ENA-78, I-309, I-TAC, CXCL1, -2, -5, -11, -12. | Nonprotein soluble factors PGE2, Nitric oxide, Reactive oxygen species |
Other inflammatory factors: GM-CSE, G-CSE, IFN-γ, BLC, MIF | Insoluble factors (ECM) Fibronectin, Collagens, Laminin |
Growth factors and regulators: Amphiregulin, Epiregulin, Heregulin, EGF, bFGF, HGF, KGF (FGF7), VEGF, Angiogenin, SCF, SDF-1, PIGF, NGF, IGFBP-2, -3, -4, -6, -7, GDNF, PDGF, | |
Proteases and regulators: MMP-1, -3, -9, -10, -12, -13, -14, TIMP-1, TIMP-2, PAI-1, -2; tPA; uPA, Cathepsin B |
Agent | |
---|---|
Quercetin |
|
Venetoclax (ABT-199) | |
Rapamycin | |
Ruxolitinib |
|
Dasatinib | |
Tanespimycin (17-AAG) |
|
Panobinostat |
|
Everolimus |
|
Baricitinib | |
Metformin |
|
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sánchez-Díaz, L.; Espinosa-Sánchez, A.; Blanco, J.-R.; Carnero, A. Senotherapeutics in Cancer and HIV. Cells 2022, 11, 1222. https://doi.org/10.3390/cells11071222
Sánchez-Díaz L, Espinosa-Sánchez A, Blanco J-R, Carnero A. Senotherapeutics in Cancer and HIV. Cells. 2022; 11(7):1222. https://doi.org/10.3390/cells11071222
Chicago/Turabian StyleSánchez-Díaz, Laura, Asunción Espinosa-Sánchez, José-Ramón Blanco, and Amancio Carnero. 2022. "Senotherapeutics in Cancer and HIV" Cells 11, no. 7: 1222. https://doi.org/10.3390/cells11071222
APA StyleSánchez-Díaz, L., Espinosa-Sánchez, A., Blanco, J. -R., & Carnero, A. (2022). Senotherapeutics in Cancer and HIV. Cells, 11(7), 1222. https://doi.org/10.3390/cells11071222