Research Perspective Volume 5, Issue 8 pp 592—598

Rapamycin extends life- and health span because it slows aging

Mikhail V. Blagosklonny1, ,

  • 1 Department of Cell Stress Biology, Roswell Park Cancer Institute, BLSC, L3-312, Elm and Carlton Streets, Buffalo, NY, 14263, USA

Received: August 4, 2013       Accepted: August 10, 2013       Published: August 11, 2013      

https://doi.org/10.18632/aging.100591
How to Cite

Copyright: © 2013 Blagosklonny et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Making headlines, a thought-provocative paper by Neff, Ehninger and coworkers claims that rapamycin extends life span but has limited effects on aging. How is that possibly possible? And what is aging if not an increase of the probability of death with age. I discuss that the JCI paper actually shows that rapamycin slows aging and also extends lifespan regardless of its direct anti-cancer activities. Aging is, in part, MTOR-driven: a purposeless continuation of developmental growth. Rapamycin affects the same processes in young and old animals: young animals' traits and phenotypes, which continuations become hyperfunctional, harmful and lethal later in life.

Found by chance on the mystical Easter island [1], the anti-aging drug rapamycin gave birth to numerous myths. This time, it is claimed that rapamycin prolongs lifespan and prevents aging-associated changes by aging-independent mechanisms, not by affecting aging itself [2]. But what is then aging itself.

What is aging?

Aging is an exponential increase of the probability of death with age [3]. No one has died from health or without a cause. Most elderly humans die from age-related diseases [4-10], which are also called “natural causes”, if a precise diagnosis is unnecessary. In mammals, death from aging means death from age-related diseases. Not only humans and other mammals but also aging worms and flies die from pathologies [11-27].

Diseases are biomarkers of aging

Age-related diseases are biomarkers of aging [8]. The most common are cardiovascular diseases (associated with atherosclerosis, hypertension and cardiac hypertrophy), cancer, diabetes (and other complications of metabolic syndrome), Alzheimer and Parkinson diseases, macular degeneration and so on. Many manifestations of aging are not considered as diseases because they develop in everyone (e.g. female menopause).

The distinction is arbitrarily. For example, cancer-prone transgenic mice can exclusively die from cancer but still cancer is a disease. But many alterations, although associated with age, are not involved in aging. And these phenotypes are not affected by rapamycin.

Cellular aging

Unless we believe in vitalism, organismal aging should be linked to cellular aging. Cellular aging is, in part, MTOR-dependent process. The MTOR (mechanistic or mammalian target of rapamycin) pathway is activated by growth factors, hormones (such as insulin and testosterone), nutrients, oxygen and some conditions such as obesity [28-38]. Figuratively, MTOR is a “molecular hypothalamus”, a sensing pathway in every cell [39]. In turn, MTOR stimulates specific functions of differentiated cells and cellular mass growth. In proliferating cells, growth is balanced by division. In resting cells, active MTOR causes cellular hypertrophy, hyperfunctions (such as hypersecretion). MTOR-driven geroconversion is a conversion from quiescence to senescence [40-59]. And cellular senescence is characterized by increased cell-type-specific cellular functions (hyperfunctions), altering homeostasis and leading to age-related diseases [9].

Systemic hyperfunctions and aging

Except of terminal stages of age-related diseases, aging is associated with systemic hyperfunctions: increased blood pressure (hypertension), increased platelet aggregation (hyper-aggregation), hyper-contractility of arterial smooth muscle cells, hyper-coagulation, hyperlipidemia, hyperglycemia, hyperinsulinemia, increased resistance to hormones, pro-inflammatory conditions, organ hypertrophy, fibrosis and hyperplasia. These hyperfunctions are damaging to the organs and, when damage occurs, then some functions are lost. So only late stages of aging are decline and loss of functions. Terminal stages are MTOR-independent and will not be reversed by rapamycin. For example, hyperfunctional osteoclasts cause osteoporosis, leading to a broken bone and a sequence of events (immobilization, pneumonia, etc), which require standard medical interventions, not anti-aging drugs [60]. Not only in mammals, but also in C elegans and Drosophila, life-limiting pathologies are caused of exacerbated and intensified normal processes and functions [19,25,61,62].

Aging processes do not spring from nothing. They are continuations of normal cellular, tissue, organ and system functions in young animals. Unless miracle is possible, rapamycin must affect the same processes in old and young animals. And it does.

Aging is a quasi-program (not a program)

Why systemic hyperfunctions arise? Aging is an unintended continuation of organismal growth, like cellular senescence is a continuation of cellular growth [63]. In other words, aging is a quasi-program (not a program): an unintended and purposeless continuation of developmental programs, which are not switched off upon their completion [64-67], causing age-related diseases. For example, blood pressure is increased from birth to adulthood and continuation of this trend leads to hypertension. Menopause is a hyperfunctional continuation of reproductive program [68]. Aging-associated pathologies are continuation of normal functions of the young organism. Therefore, rapamycin must affect the same processes in young and old animals, because aging is a continuation of normal functions. Aging processes do not spring from nothing. They are continuations of normal cellular, tissue, organ and system functions in young animals.

Rapamycin extends life span independently of its anti-cancer effect and prevents cancer by slowing down aging.

Cancer and aging

Cancer is an aging-related disease and interventions that slow aging (e.g. calorie restriction) delay cancer [69-78]. Furthermore, compared with calorie restriction, rapamycin stronger inhibits MTOR. It is predictable that if rapamycin slows aging, it should delay cancer [79,80]. Studies support these predictions [81-84] and rapamycin extended lifespan and delayed cancer, even when calorie restriction did not [85]. Although rapamycin is a potent cancer-preventive agent, it is only modestly effective for cancer treatment. Rapalogs are most effective in drug combinations [86-93]. They also may decrease side effects by suppressing senescence of normal cells [51,58,59,94,95]. Also, senescence of normal cells creates cancer-promoting micro-environment [96-103]. If rapamycin indeed prevents cancer by slowing aging (not by killing cancer cells), the prevention must be started before cancer is initiated. In other words, if rapamycin treatment is started too late in life, then its anti-cancer effect will be blunted. This was shown in cancer-prone p53+/− mice [104]. The same was shown by Neff et al: rapamycin rapamycin did not prevent cancer when the treatment was started at middle and old age [2]. Thus, the JCI study confirms the notion that rapamycin delays cancer by slowing aging (see also discussion here in the last section). Anti-cancer effects simply cannot be responsible for life extension by rapamycin. First, effective anti-cancer drugs that are curative in lymphomas, testicular and ovarian cancers (methotrexate, cisplatin, paclitaxel) would greatly shorten murine lifespan, especially when started in young age. Even further, typical anti-cancer drugs accelerate cancer. For example, radiation (a classic anti-cancer intervention) dramatically accelerates cancer in p53+/− mice and shortens life span [105-109]. And anti-cancer drugs cause secondary cancers in patients. In contrast, not only rapamycin extends lifespan, it is the only known drug that extends life span consistently. Second, apart from cancer-prone strains of mice, cancer is not the main cause of death in most animals. MTOR is involved in most age-related diseases and rapamycin prevents them in mammals [64,110-123] and slows down aging [81,124-127]. Finally, yeast, worm and flies do not die from cancer and still inhibition of the MTOR pathway extends lifespan [128-137].

Inhibition of TOR slows aging: converging evidence [124]

  • Rapamycin suppresses geroconversion: conversion from cellular quiescence to senescence. Geroconversion is cellular basis of organismal aging

  • Genetic manipulations that inhibit the TOR pathway extend life-span in diverse species from yeast to mammals

  • Rapamycin extends lifespan in all species tested

  • Calorie restriction, which inhibits MTOR, extends lifespan

  • MTOR is involved in diseases of aging and rapamycin prevents these diseases in animal models

Rapamycin slows aging: the JCI paper [2]

How does the Neff et al study support the model of quasi-programmed aging?

  • As shown by Neff et al, chronic administration of rapamycin extends lifespan in male C57BL/6J mice, when started at both young and old age. Note: This extension is impressive given that (a) effects of rapamycin in male mice are blunted compared with female mice in previous studies, (b) C57BL/6J mice are intrinsically long-lived and (c) rapamycin was administrated in everyday schedule (chronic or immunosuppressive schedule) instead of intermittent or pulse administration (anti-aging schedule).

  • C57BL/6J mice are refractory to many tumors http://jaxmice.jax.org/strain/000664.html Therefore, life extension is difficult to explain by anti-cancer effects of rapamycin.

  • In fact, rapamycin did not prevent cancer when the treatment was started at middle and old age, but still extended life span. As stated by Neff et al [2]: “Rapamycin … had no measurable effect in the 25-month cohort (vehicle, 1 of 5; rapamycin, 2 of 8; P = 1.0, Fisher exact test) or the 34-month cohort (vehicle, 1 of 5; rapamycin, 3 of 10; P = 1.0, Fisher exact test).” As we discussed here, this indicates that effects of rapamycin are probably due to suppression of aging. Rapamycin treatment decreased cancer incidence only when it was started in young mice.

  • Rapamycin counteracted certain aging-related alterations in both young and old mice. This suggests that aging is a continuation of normal traits in young organisms. Aging is driven by intensified and exacerbated normal cellular functions.

  • Rapamycin did not affect many parameters that are not aging-specific such as alterations in plasma sodium, calcium and chloride concentrations. This is expectable. Aging is not associated with alterations of electrolyte homeostasis. These alterations are terminal phases of medical conditions due to organ (e.g. renal) failure.

  • Some age-related alterations actually counteract aging. For example, although RNA/protein synthesis is decreased with aging in model organisms, yet its further inhibition prolongs life span further [138-141]. As shown by Neff et al, rapamycin did not prevent anti-aging alterations such as a decrease in testosterone levels. Noteworthy, testosterone activates mTOR.

  • Some trends reported by Neff et al are not typical for aging. For example, while Neff reported a decrease in blood glucose and lipids with age, these parameters tend to increase with age, especially when age-related diseases develop. Perhaps mice with hyperglycemia and hyperlipidemia died during the study, while only surviving (the healthiest) mice were examined at the end of the study.

References

  • 1. Stipp D. A new path to longevity. Sci Am. 2012; 306:32-39. [PubMed]
  • 2. Neff F, Flores-Dominguez D, Ryan DP, Horsch M, Schroder S, Adler T, Afonso LC, Aguilar-Pimentel JA, Becker L, Garrett L, Hans W, Hettich MM, Holtmeier R, Holter SM, Moreth K, Prehn C, et al. Rapamycin extends murine lifespan but has limited effects on aging. J Clin Invest. 2013;.
  • 3. Olshansky SJ, Passaro DJ, Hershow RC, Layden J, Carnes BA, Brody J, Hayflick L, Butler RN, Allison DB, Ludwig DS. A potential decline in life expectancy in the United States in the 21st century. N Engl J Med. 2005; 352:1138-1145. [PubMed]
  • 4. Horiuchi S and Wilmoth JR. Age patterns of the life table aging rate for major causes of death in Japan, 1951-1990. J Gerontol A Biol Sci Med Sci. 1997; 52:B67-77. [PubMed]
  • 5. Sonnenschein E and Brody JA. Effect of population aging on proportionate mortality from heart disease and cancer, U.S. 2000-2050. J Gerontol B Psychol Sci Soc Sci. 2005; 60:S110-112. [PubMed]
  • 6. Jemal A, Ward E, Hao Y, Thun M. Trends in the leading causes of death in the United States, 1970-2002. Jama. 2005; 294:1255-1259. [PubMed]
  • 7. Gorina Y and Lentzner H. Multiple causes of death in old age. Aging Trends. 2008; 1-9..
  • 8. Blagosklonny MV. Validation of anti-aging drugs by treating age-related diseases. Aging (Albany NY). 2009; 1:281-288. [PubMed]
  • 9. Blagosklonny MV. Prospective treatment of age-related diseases by slowing down aging. Am J Pathol. 2012; 181:1142-1146. [PubMed]
  • 10. Fontana L, Partridge L, Longo VD. Extending healthy life span--from yeast to humans. Science. 2010; 328:321-326. [PubMed]
  • 11. Partridge L and Gems D. Mechanisms of ageing: public or private? Nat Rev Genet. 2002; 3:165-175. [PubMed]
  • 12. Michno K, van de Hoef D, Wu H, Boulianne GL. Modeling age-related diseases in Drosophila: can this fly? Curr Top Dev Biol. 2005; 71:199-223. [PubMed]
  • 13. Birse RT, Choi J, Reardon K, Rodriguez J, Graham S, Diop S, Ocorr K, Bodmer R, Oldham S. High-fat-diet-induced obesity and heart dysfunction are regulated by the TOR pathway in Drosophila. Cell Metab. 2010; 12:533-544. [PubMed]
  • 14. Musselman LP, Fink JL, Narzinski K, Ramachandran PV, Hathiramani SS, Cagan RL, Baranski TJ. A high-sugar diet produces obesity and insulin resistance in wild-type Drosophila. Dis Model Mech. 2011; 4:842-849. [PubMed]
  • 15. Teleman AA, Ratzenbock I, Oldham S. Drosophila: a model for understanding obesity and diabetic complications. Exp Clin Endocrinol Diabetes. 2012; 120:184-185. [PubMed]
  • 16. Kumar S and Walter J. Phosphorylation of amyloid beta (Abeta) peptides - a trigger for formation of toxic aggregates in Alzheimer's disease. Aging (Albany NY). 2011; 3:803-812. [PubMed]
  • 17. Sarkar S, Ravikumar B, Floto RA, Rubinsztein DC. Rapamycin and mTOR-independent autophagy inducers ameliorate toxicity of polyglutamine-expanded huntingtin and related proteinopathies. Cell Death Differ. 2009; 16:46-56. [PubMed]
  • 18. Brignull HR, Morley JF, Garcia SM, Morimoto RI. Modeling polyglutamine pathogenesis in C. elegans. Methods Enzymol. 2006; 412:256-282. [PubMed]
  • 19. Rera M, Clark RI, Walker DW. Intestinal barrier dysfunction links metabolic and inflammatory markers of aging to death in Drosophila. Proc Natl Acad Sci U S A. 2012; 109:21528-21533. [PubMed]
  • 20. Wessells RJ, Fitzgerald E, Cypser JR, Tatar M, Bodmer R. Insulin regulation of heart function in aging fruit flies. Nat Genet. 2004; 36:1275-1281. [PubMed]
  • 21. Stefanatos R, Sriram A, Kiviranta E, Mohan A, Ayala V, Jacobs HT, Pamplona R, Sanz A. dj-1beta regulates oxidative stress, insulin-like signaling and development in Drosophila melanogaster. Cell Cycle. 2012; 11:3876-3886. [PubMed]
  • 22. Shmookler Reis RJ, Xu L, Lee H, Chae M, Thaden JJ, Bharill P, Tazearslan C, Siegel E, Alla R, Zimniak P, Ayyadevara S. Modulation of lipid biosynthesis contributes to stress resistance and longevity of C. elegans mutants. Aging (Albany NY). 2011; 3:125-147. [PubMed]
  • 23. Walker JA and Bernards A. Drosophila melanogaster neurofibromatosis-1: ROS, not Ras? Nat Genet. 2007; 39:443-445. [PubMed]
  • 24. Lee JH, Budanov AV, Park EJ, Birse R, Kim TE, Perkins GA, Ocorr K, Ellisman MH, Bodmer R, Bier E, Karin M. Sestrin as a feedback inhibitor of TOR that prevents age-related pathologies. Science. 2010; 327:1223-1228. [PubMed]
  • 25. Biteau B, Hochmuth CE, Jasper H. JNK activity in somatic stem cells causes loss of tissue homeostasis in the aging Drosophila gut. Cell Stem Cell. 2008; 3:442-455. [PubMed]
  • 26. McGee MD, Day N, Graham J, Melov S. cep-1/p53-dependent dysplastic pathology of the aging C. elegans gonad. Aging (Albany NY). 2012; 4:256-269. [PubMed]
  • 27. Banerjee KK, Ayyub C, Sengupta S, Kolthur-Seetharam U. dSir2 deficiency in the fatbody, but not muscles, affects systemic insulin signaling, fat mobilization and starvation survival in flies. Aging (Albany NY). 2012; 4:206-223. [PubMed]
  • 28. Loewith R and Hall MN. Target of rapamycin (TOR) in nutrient signaling and growth control. Genetics. 2011; 189:1177-1201. [PubMed]
  • 29. Sengupta S, Peterson TR, Sabatini DM. Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell. 2010; 40:310-322. [PubMed]
  • 30. Oh WJ and Jacinto E. mTOR complex 2 signaling and functions. Cell Cycle. 2011; 10:2305-2316. [PubMed]
  • 31. Duran RV and Hall MN. Glutaminolysis feeds mTORC1. Cell Cycle. 2012; 11:4107-4108. [PubMed]
  • 32. Conn CS and Qian SB. mTOR signaling in protein homeostasis: less is more? Cell Cycle. 2011; 10:1940-1947. [PubMed]
  • 33. Cornu M, Albert V, Hall MN. mTOR in aging, metabolism, and cancer. Curr Opin Genet Dev. 2013; 23:53-62. [PubMed]
  • 34. Kim S and Snyder SH. Nutrient amino acids signal to mTOR via inositol polyphosphate multikinase. Cell Cycle. 2011; 10:1708-1710. [PubMed]
  • 35. Feng Z and Levine AJ. The regulation of energy metabolism and the IGF-1/mTOR pathways by the p53 protein. Trends Cell Biol. 2010; 20:427-434. [PubMed]
  • 36. Leontieva OV and Blagosklonny MV. Hypoxia and gerosuppression: the mTOR saga continues. Cell Cycle. 2012; 11:3926-3931. [PubMed]
  • 37. Wang CY, Kim HH, Hiroi Y, Sawada N, Salomone S, Benjamin LE, Walsh K, Moskowitz MA, Liao JK. Obesity increases vascular senescence and susceptibility to ischemic injury through chronic activation of Akt and mTOR. Sci Signal. 2009; 2:ra11 [PubMed]
  • 38. Weber JD and Gutmann DH. Deconvoluting mTOR biology. Cell Cycle. 2012; 11:236-248. [PubMed]
  • 39. Blagosklonny MV. M(o)TOR of aging: MTOR as a universal molecular hypothalamus. Aging (Albany NY). 2013; 5:490-494. [PubMed]
  • 40. Blagosklonny MV. Cell senescence and hypermitogenic arrest. EMBO Rep. 2003; 4:358-362. [PubMed]
  • 41. Demidenko ZN and Blagosklonny MV. Growth stimulation leads to cellular senescence when the cell cycle is blocked. Cell Cycle. 2008; 7:3355-3361. [PubMed]
  • 42. Demidenko ZN, Zubova SG, Bukreeva EI, Pospelov VA, Pospelova TV, Blagosklonny MV. Rapamycin decelerates cellular senescence. Cell Cycle. 2009; 8:1888-1895. [PubMed]
  • 43. Demidenko ZN, Korotchkina LG, Gudkov AV, Blagosklonny MV. Paradoxical suppression of cellular senescence by p53. Proc Natl Acad Sci U S A. 2010; 107:9660-9664. [PubMed]
  • 44. Korotchkina LG, Leontieva OV, Bukreeva EI, Demidenko ZN, Gudkov AV, Blagosklonny MV. The choice between p53-induced senescence and quiescence is determined in part by the mTOR pathway. Aging (Albany NY). 2010; 2:344-352. [PubMed]
  • 45. Leontieva OV and Blagosklonny MV. DNA damaging agents and p53 do not cause senescence in quiescent cells, while consecutive re-activation of mTOR is associated with conversion to senescence. Aging (Albany NY). 2010; 2:924-935. [PubMed]
  • 46. Leontieva OV, Natarajan V, Demidenko ZN, Burdelya LG, Gudkov AV, Blagosklonny MV. Hypoxia suppresses conversion from proliferative arrest to cellular senescence. Proc Natl Acad Sci U S A. 2012; 109:13314-13318. [PubMed]
  • 47. Leontieva OV, Lenzo F, Demidenko ZN, Blagosklonny MV. Hyper-mitogenic drive coexists with mitotic incompetence in senescent cells. Cell Cycle. 2012; 11:4642-4649. [PubMed]
  • 48. Cho S and Hwang ES. Status of mTOR activity may phenotypically differentiate senescence and quiescence. Mol Cells. 2012; 33:597-604. [PubMed]
  • 49. Pani G. From growing to secreting: new roles for mTOR in aging cells. Cell Cycle. 2011; 10:2450-2453. [PubMed]
  • 50. Kolesnichenko M, Hong L, Liao R, Vogt PK, Sun P. Attenuation of TORC1 signaling delays replicative and oncogenic RAS-induced senescence. Cell Cycle. 2012; 11:2391-2401. [PubMed]
  • 51. Mercier I, Camacho J, Titchen K, Gonzales DM, Quann K, Bryant KG, Molchansky A, Milliman JN, Whitaker-Menezes D, Sotgia F, Jasmin JF, Schwarting R, Pestell RG, Blagosklonny MV, Lisanti MP. Caveolin-1 and accelerated host aging in the breast tumor microenvironment: chemoprevention with rapamycin, an mTOR inhibitor and anti-aging drug. Am J Pathol. 2012; 181:278-293. [PubMed]
  • 52. Halicka HD, Zhao H, Li J, Lee YS, Hsieh TC, Wu JM, Darzynkiewicz Z. Potential anti-aging agents suppress the level of constitutive mTOR- and DNA damage- signaling. Aging (Albany NY). 2012; 4:952-965. [PubMed]
  • 53. Denchi EL. A critical role for TORC1 in cellular senescence. Cell Cycle. 2012; 11:2976 [PubMed]
  • 54. Menendez JA, Vellon L, Oliveras-Ferraros C, Cufi S, Vazquez-Martin A. mTOR-regulated senescence and autophagy during reprogramming of somatic cells to pluripotency: a roadmap from energy metabolism to stem cell renewal and aging. Cell Cycle. 2011; 10:3658-3677. [PubMed]
  • 55. Serrano M. Dissecting the role of mTOR complexes in cellular senescence. Cell Cycle. 2012; 11:2231-2232. [PubMed]
  • 56. Gan B and DePinho RA. mTORC1 signaling governs hematopoietic stem cell quiescence. Cell Cycle. 2009; 8:1003-1006. [PubMed]
  • 57. Chen C, Liu Y, Zheng P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Sci Signal. 2009; 2:ra75 [PubMed]
  • 58. Iglesias-Bartolome R, Patel V, Cotrim A, Leelahavanichkul K, Molinolo AA, Mitchell JB, Gutkind JS. mTOR inhibition prevents epithelial stem cell senescence and protects from radiation-induced mucositis. Cell Stem Cell. 2012; 11:401-414. [PubMed]
  • 59. Iglesias-Bartolome R and Gutkind SJ. Exploiting the mTOR paradox for disease prevention. Oncotarget. 2012; 3:1061-1063. [PubMed]
  • 60. Blagosklonny MV. How to save Medicare: the anti-aging remedy. Aging (Albany NY). 2012; 4:547-552. [PubMed]
  • 61. Gems D and Partridge L. Genetics of Longevity in Model Organisms: Debates and Paradigm Shifts. Annu Rev Physiol. 2013; 75:621-644. [PubMed]
  • 62. Gems DH and de la Guardia YI. Alternative Perspectives on Aging in C. elegans: Reactive Oxygen Species or Hyperfunction? Antioxid Redox Signal. 2013; 19:321-329. [PubMed]
  • 63. Blagosklonny MV and Hall MN. Growth and aging: a common molecular mechanism. Aging (Albany NY). 2009; 1:357-362. [PubMed]
  • 64. Blagosklonny MV. Aging and immortality: quasi-programmed senescence and its pharmacologic inhibition. Cell Cycle. 2006; 5:2087-2102. [PubMed]
  • 65. Blagosklonny MV. mTOR-driven aging: speeding car without brakes. Cell Cycle. 2009; 8:4055-4059. [PubMed]
  • 66. Blagosklonny MV. Revisiting the antagonistic pleiotropy theory of aging: TOR-driven program and quasi-program. Cell Cycle. 2010; 9:3151-3156. [PubMed]
  • 67. Blagosklonny M. Answering the ultimate question “What is the Proximal Cause of Aging?”. Aging (Albany NY). 2012; 4:861-877. [PubMed]
  • 68. Blagosklonny MV. Why men age faster but reproduce longer than women: mTOR and evolutionary perspectives. Aging (Albany NY). 2010; 2:265-273. [PubMed]
  • 69. Yancik R. Population aging and cancer: a cross-national concern. Cancer J. 2005; 11:437-441. [PubMed]
  • 70. Ikeno Y, Bronson RT, Hubbard GB, Lee S, Bartke A. Delayed occurrence of fatal neoplastic diseases in ames dwarf mice: correlation to extended longevity. J Gerontol A Biol Sci Med Sci. 2003; 58:291-296. [PubMed]
  • 71. Berrigan D, Perkins SN, Haines DC, Hursting SD. Adult-onset calorie restriction and fasting delay spontaneous tumorigenesis in p53-deficient mice. Carcinogenesis. 2002; 23:817-822. [PubMed]
  • 72. Ikeno Y, Hubbard GB, Lee S, Cortez LA, Lew CM, Webb CR, Berryman DE, List EO, Kopchick JJ, Bartke A. Reduced incidence and delayed occurrence of fatal neoplastic diseases in growth hormone receptor/binding protein knockout mice. J Gerontol A Biol Sci Med Sci. 2009; 64:522-529. [PubMed]
  • 73. Anisimov VN, Berstein LM, Popovich IG, Zabezhinski MA, Egormin PA, Piskunova TS, Semenchenko AV, Tyndyk ML, Yurova MN, Kovalenko IG, Poroshina TE. If started early in life, metformin treatment increases life span and postpones tumors in female SHR mice. Aging (Albany NY). 2011; 3:148-157. [PubMed]
  • 74. Pinkston JM, Garigan D, Hansen M, Kenyon C. Mutations that increase the life span of C. elegans inhibit tumor growth. Science. 2006; 313:971-975. [PubMed]
  • 75. Buffenstein R. The naked mole-rat: a new long-living model for human aging research. J Gerontol A Biol Sci Med Sci. 2005; 60:1369-1377. [PubMed]
  • 76. Kim EB, Fang X, Fushan AA, Huang Z, Lobanov AV, Han L, Marino SM, Sun X, Turanov AA, Yang P, Yim SH, Zhao X, Kasaikina MV, Stoletzki N, Peng C, Polak P, et al. Genome sequencing reveals insights into physiology and longevity of the naked mole rat. Nature. 2011; 479:223-227. [PubMed]
  • 77. Berman AE, Leontieva OV, Natarajan V, McCubrey JA, Demidenko ZN, Nikiforov MA. Recent progress in genetics of aging, senescence and longevity: focusing on cancer-related genes. Oncotarget. 2012; 3:1522-1532. [PubMed]
  • 78. Borras C, Monleon D, Lopez-Grueso R, Gambini J, Orlando L, Pallardo FV, Santos E, Vina J, Font de Mora J. RasGrf1 deficiency delays aging in mice. Aging (Albany NY). 2011; 3:262-276. [PubMed]
  • 79. Blagosklonny MV. Prevention of cancer by inhibiting aging. Cancer Biol Ther. 2008; 7:1520-1524. [PubMed]
  • 80. Blagosklonny MV. Rapalogs in cancer prevention: Anti-aging or anticancer? Cancer Biol Ther. 2012; 13:1349-1354. [PubMed]
  • 81. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandezr E, Miller RA. Rapamycin fed late in life extends lifespan in genetically heterogenous mice. Nature. 2009; 460:392-396. [PubMed]
  • 82. Anisimov VN, Zabezhinski MA, Popovich IG, Piskunova TS, Semenchenko AV, Tyndyk ML, Yurova MN, Rosenfeld SV, Blagosklonny MV. Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice. Cell Cycle. 2011; 10:4230-4236. [PubMed]
  • 83. Anisimov VN, Zabezhinski MA, Popovich IG, Piskunova TS, Semenchenko AV, Tyndyk ML, Yurova MN, Antoch MP, Blagosklonny MV. Rapamycin extends maximal lifespan in cancer-prone mice. Am J Pathol. 2010; 176:2092-2097. [PubMed]
  • 84. Selman C and Partridge L. A double whammy for aging? Rapamycin extends lifespan and inhibits cancer in inbred female mice. Cell Cycle. 2012; 11:17-18. [PubMed]
  • 85. Livi CB, Hardman RL, Christy BA, Dodds SG, Jones D, Williams C, Strong R, Bokov A, Javors MA, Ikeno Y, Hubbard G, Hasty P, Sharp ZD. Rapamycin extends life span of Rb1+/− mice by inhibiting neuroendocrine tumors. Aging (Albany NY). 2013; 5:100-110. [PubMed]
  • 86. Chiarini F, Lonetti A, Teti G, Orsini E, Bressanin D, Cappellini A, Ricci F, Tazzari PL, Ognibene A, Falconi M, Pagliaro P, Iacobucci I, Martinelli G, Amadori S, McCubrey JA, Martelli AM. A combination of temsirolimus, an allosteric mTOR inhibitor, with clofarabine as a new therapeutic option for patients with acute myeloid leukemia. Oncotarget. 2012; 3:1615-1628. [PubMed]
  • 87. Garrett JT, Chakrabarty A, Arteaga CL. Will PI3K pathway inhibitors be effective as single agents in patients with cancer? Oncotarget. 2011; 2:1314-1321. [PubMed]
  • 88. Zhang Y and Zheng XF. mTOR-independent 4E-BP1 phosphorylation is associated with cancer resistance to mTOR kinase inhibitors. Cell Cycle. 2012; 11:594-603. [PubMed]
  • 89. Kurmasheva RT, Dudkin L, Billups C, Debelenko LV, Morton CL, Houghton PJ. The insulin-like growth factor-1 receptor-targeting antibody, CP-751,871, suppresses tumor-derived VEGF and synergizes with rapamycin in models of childhood sarcoma. Cancer Res. 2009; 69:7662-7671. [PubMed]
  • 90. Martelli AM, Chiarini F, Evangelisti C, Cappellini A, Buontempo F, Bressanin D, Fini M, McCubrey JA. Two hits are better than one: targeting both phosphatidylinositol 3-kinase and mammalian target of rapamycin as a therapeutic strategy for acute leukemia treatment. Oncotarget. 2012; 3:371-394. [PubMed]
  • 91. McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Franklin RA, Montalto G, Cervello M, Libra M, Candido S, Malaponte G, Mazzarino MC, Fagone P, Nicoletti F, Basecke J, Mijatovic S, Maksimovic-Ivanic D, et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascade inhibitors: how mutations can result in therapy resistance and how to overcome resistance. Oncotarget. 2012; 3:1068-1111. [PubMed]
  • 92. Steelman LS, Navolanic P, Chappell WH, Abrams SL, Wong EW, Martelli AM, Cocco L, Stivala F, Libra M, Nicoletti F, Drobot LB, Franklin RA, McCubrey JA. Involvement of Akt and mTOR in chemotherapeutic- and hormonal-based drug resistance and response to radiation in breast cancer cells. Cell Cycle. 2011; 10:3003-3015. [PubMed]
  • 93. Bressanin D, Evangelisti C, Ricci F, Tabellini G, Chiarini F, Tazzari PL, Melchionda F, Buontempo F, Pagliaro P, Pession A, McCubrey JA, Martelli AM. Harnessing the PI3K/Akt/mTOR pathway in T-cell acute lymphoblastic leukemia: eliminating activity by targeting at different levels. Oncotarget. 2012; 3:811-823. [PubMed]
  • 94. Blagosklonny MV. The power of chemotherapeutic engineering: arresting cell cycle and suppressing senescence to protect from mitotic inhibitors. Cell Cycle. 2011; 10:2295-2298. [PubMed]
  • 95. Apontes P, Leontieva OV, Demidenko ZN, Li F, Blagosklonny MV. Exploring long-term protection of normal human fibroblasts and epithelial cells from chemotherapy in cell culture. Oncotarget. 2011; 2:222-233. [PubMed]
  • 96. Liu D and Hornsby PJ. Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res. 2007; 67:3117-3126. [PubMed]
  • 97. Pasciu D, Montisci S, Greco M, Doratiotto S, Pitzalis S, Pani P, Laconi S, Laconi E. Aging is associated with increased clonogenic potential in rat liver in vivo. Aging Cell. 2006; 5:373-377. [PubMed]
  • 98. Caruso C, Lio D, Cavallone L, Franceschi C. Aging, longevity, inflammation, and cancer. Ann N Y Acad Sci. 2004; 1028:1-13. [PubMed]
  • 99. Bavik C, Coleman I, Dean JP, Knudsen B, Plymate S, Nelson PS. The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Res. 2006; 66:794-802. [PubMed]
  • 100. Campisi J. Senescent cells, tumor suppression, and organismal aging: good citizens, bad neighbors. Cell. 2005; 120:513-522. [PubMed]
  • 101. Krtolica A, Parrinello S, Lockett S, Desprez PY, Campisi J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc Natl Acad Sci U S A. 2001; 98:12072-12077. [PubMed]
  • 102. Parrinello S, Coppe JP, Krtolica A, Campisi J. Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J Cell Sci. 2005; 118:485-496. [PubMed]
  • 103. Henry CJ, Marusyk A, DeGregori J. Aging-associated changes in hematopoiesis and leukemogenesis: what's the connection? Aging (Albany NY). 2011; 3:643-656. [PubMed]
  • 104. Komarova EA, Antoch MP, Novototskaya LR, Chernova OB, Paszkiewicz G, Leontieva OV, Blagosklonny MV, Gudkov AV. Rapamycin extends lifespan and delays tumorigenesis in heterozygous p53+/− mice. Aging (Albany NY). 2012; 4:709-714. [PubMed]
  • 105. Kemp CJ, Wheldon T, Balmain A. p53-deficient mice are extremely susceptible to radiation-induced tumorigenesis. Nat Genet. 1994; 8:66-69. [PubMed]
  • 106. Kony SJ, de Vathaire F, Chompret A, Shamsaldim A, Grimaud E, Raquin MA, Oberlin O, Brugieres L, Feunteun J, Eschwege F, Chavaudra J, Lemerle J, Bonaiti-Pellie C. Radiation and genetic factors in the risk of second malignant neoplasms after a first cancer in childhood. Lancet. 1997; 350:91-95. [PubMed]
  • 107. Venkatachalam S, Shi YP, Jones SN, Vogel H, Bradley A, Pinkel D, Donehower LA. Retention of wild-type p53 in tumors from p53 heterozygous mice: reduction of p53 dosage can promote cancer formation. Embo J. 1998; 17:4657-4667. [PubMed]
  • 108. Lee JM, Abrahamson JL, Kandel R, Donehower LA, Bernstein A. Susceptibility to radiation-carcinogenesis and accumulation of chromosomal breakage in p53 deficient mice. Oncogene. 1994; 9:3731-3736. [PubMed]
  • 109. Leonova KI, Shneyder J, Antoch MP, Toshkov IA, Novototskaya LR, Komarov PG, Komarova EA, Gudkov AV. A small molecule inhibitor of p53 stimulates amplification of hematopoietic stem cells but does not promote tumor development in mice. Cell Cycle. 2010; 9:1434-1443. [PubMed]
  • 110. Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, Oroz LG, Scaravilli F, Easton DF, Duden R, O'Kane CJ, Rubinsztein DC. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet. 2004; 36:585-595. [PubMed]
  • 111. Mueller MA, Beutner F, Teupser D, Ceglarek U, Thiery J. Prevention of atherosclerosis by the mTOR inhibitor everolimus in LDLR−/− mice despite severe hypercholesterolemia. Atherosclerosis. 2008; 198:39-48. [PubMed]
  • 112. Tsang CK, Qi H, Liu LF, Zheng XFS. Targeting mammalian target of rapamycin (mTOR) for health and diseases. Drug Disc Today. 2007; 12:112-124..
  • 113. Blagosklonny MV. An anti-aging drug today: from senescence-promoting genes to anti-aging pill. Drug Disc Today. 2007; 12:218-224..
  • 114. Neef M, Ledermann M, Saegesser H, Schneider V, Reichen J. Low-dose oral rapamycin treatment reduces fibrogenesis, improves liver function, and prolongs survival in rats with established liver cirrhosis. J Hepatol. 2006; 45:786-796. [PubMed]
  • 115. Nistala R, Sowers JR, Whaley-Connell A. Over-nutrition contributes to tubulointerstitial fibrosis by targeting nutrient-sensing kinases: role for the mTOR/S6K pathway. Cell Cycle. 2012; 11:831-832. [PubMed]
  • 116. Dazert E and Hall MN. mTOR signaling in disease. Curr Opin Cell Biol. 2011;.
  • 117. Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011; 12:21-35. [PubMed]
  • 118. Bove J, Martinez-Vicente M, Vila M. Fighting neurodegeneration with rapamycin: mechanistic insights. Nat Rev Neurosci. 2011; 12:437-452. [PubMed]
  • 119. Flynn JM, O'Leary MN, Zambataro CA, Academia EC, Presley MP, Garrett BJ, Zykovich A, Mooney SD, Strong R, Rosen CJ, Kapahi P, Nelson MD, Kennedy BK, Melov S. Late life rapamycin treatment reverses age-related heart dysfunction. Aging Cell. 2013;.
  • 120. Kolosova NG, Muraleva NA, Zhdankina AA, Stefanova NA, Fursova AZ, Blagosklonny MV. Prevention of age-related macular degeneration-like retinopathy by rapamycin in rats. Am J Pathol. 2012; 181:472-477. [PubMed]
  • 121. Blagosklonny MV. Once again on rapamycin-induced insulin resistance and longevity: despite of or owing to. Aging (Albany NY). 2012; 4:350-358. [PubMed]
  • 122. Ramos FJ, Chen SC, Garelick MG, Dai DF, Liao CY, Schreiber KH, MacKay VL, An EH, Strong R, Ladiges WC, Rabinovitch PS, Kaeberlein M, Kennedy BK. Rapamycin reverses elevated mTORC1 signaling in lamin A/C-deficient mice, rescues cardiac and skeletal muscle function, and extends survival. Sci Transl Med. 4:144ra103.
  • 123. Passtoors WM, Beekman M, Deelen J, van der Breggen R, Maier AB, Guigas B, Derhovanessian E, van Heemst D, de Craen AJ, Gunn DA, Pawelec G, Slagboom PE. Gene expression analysis of mTOR pathway: association with human longevity. Aging Cell. 2013; 12:24-31. [PubMed]
  • 124. Blagosklonny MV. Rapamycin and quasi-programmed aging: Four years later. Cell Cycle. 2010; 9:1859-1862. [PubMed]
  • 125. Spong A and Bartke A. Rapamycin slows aging in mice. Cell Cycle. 2012; 11:845 [PubMed]
  • 126. Wilkinson JE, Burmeister L, Brooks SV, Chan CC, Friedline S, Harrison DE, Hejtmancik JF, Nadon N, Strong R, Wood LK, Woodward MA, Miller RA. Rapamycin slows aging in mice. Aging Cell. 2012; 11:675-682. [PubMed]
  • 127. Zhang Y, Bokov A, Gelfond J, Soto V, Ikeno Y, Hubbard G, Diaz V, Sloane L, Maslin K, Treaster S, Rendon S, van Remmen H, Ward W, Javors M, Richardson A, Austad SN, et al. Rapamycin Extends Life and Health in C57BL/6 Mice. J Gerontol A Biol Sci Med Sci. 2013;.
  • 128. Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S. Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr Biol. 2004; 14:885-890. [PubMed]
  • 129. Kapahi P, Chen D, Rogers AN, Katewa SD, Li PW, Thomas EL, Kockel L. With TOR, less is more: a key role for the conserved nutrient-sensing TOR pathway in aging. Cell Metab. 2010; 11:453-465. [PubMed]
  • 130. Katewa SD and Kapahi P. Role of TOR signaling in aging and related biological processes in Drosophila melanogaster. Exp Gerontol. 2011; 46:382-390. [PubMed]
  • 131. Moskalev AA and Shaposhnikov MV. Pharmacological Inhibition of Phosphoinositide 3 and TOR Kinases Improves Survival of Drosophila melanogaster. Rejuvenation Res. 2010; 13:246-247. [PubMed]
  • 132. Partridge L, Alic N, Bjedov I, Piper MD. Ageing in Drosophila: the role of the insulin/Igf and TOR signalling network. Exp Gerontol. 2011; 46:376-381. [PubMed]
  • 133. Bjedov I, Toivonen JM, Kerr F, Slack C, Jacobson J, Foley A, Partridge L. Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Cell Metab. 2010; 11:35-46. [PubMed]
  • 134. Bjedov I and Partridge L. A longer and healthier life with TOR down-regulation: genetics and drugs. Biochem Soc Trans. 2011; 39:460-465. [PubMed]
  • 135. Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and age-related disease. Nature. 2013; 493:338-345. [PubMed]
  • 136. Kaeberlein M. Longevity and aging. F1000Prime Rep. 2013; 5:5 [PubMed]
  • 137. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 153:1194-1217. [PubMed]
  • 138. Pan KZ, Palter JE, Rogers AN, Olsen A, Chen D, Lithgow GJ, Kapahi P. Inhibition of mRNA translation extends lifespan in Caenorhabditis elegans. Aging Cell. 2007; 6:111-119. [PubMed]
  • 139. Syntichaki P, Troulinaki K, Tavernarakis N. eIF4E function in somatic cells modulates ageing in Caenorhabditis elegans. Nature. 2007; 445:922-926. [PubMed]
  • 140. Hansen M, Taubert S, Crawford D, Libina N, Lee SJ, Kenyon C. Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans. Aging Cell. 2007; 6:95-110. [PubMed]
  • 141. Mehta R, Chandler-Brown D, Ramos FJ, Shamieh LS, Kaeberlein M. Regulation of mRNA translation as a conserved mechanism of longevity control. Adv Exp Med Biol. 2011; 694:14-29. [PubMed]