Review Volume 5, Issue 11 pp 793—801

Immunostimulatory activity of lifespan-extending agents

José Manuel Bravo-San Pedro1,2,3, , Laura Senovilla1,2,3,4, ,

  • 1 INSERM U848; Villejuif, France
  • 2 Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
  • 3 Gustave Roussy; Villejuif, France
  • 4 INSERM U1015; Villejuif, France

Received: November 26, 2013       Accepted: December 4, 2013       Published: December 5, 2013      

https://doi.org/10.18632/aging.100619
How to Cite

Copyright: © 2013 Bravo-San Pedro 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

During the past two decades, several interventions have been shown to increase the healthy lifespan of model organisms as evolutionarily distant from each other as yeast, worms, flies and mammals. These anti-aging maneuvers include (but are not limited to) cycles of caloric restriction, physical exercise as well as the administration of multiple, chemically unrelated agents, such as resveratrol, spermidine and various rapamycin-like compounds collectively known as rapalogs. Most, if not all, lifespan-extending agents promote macroautophagy (hereafter referred to as autophagy), an evolutionarily old mechanism that contributes to the maintenance of intracellular homeostasis and plays a critical role in the adaptive response of cells to stress. In line with this notion, the activation of autophagy appears to mediate significant anti-ageing effects in several organisms, including mice. Here, we focus on rapalogs to discuss the possibility that part of the beneficial activity of lifespan-extending agents stems from their ability to exert immunostimulatory effects. Accumulating evidence indicates indeed that the immune system can recognize and eliminate not only cells that are prone to undergo malignant transformation, but also senescent cells, thus playing a significant role in the control of organismal aging. In addition, it has recently become clear that rapamycin and other rapalogs, which for a long time have been viewed (and used in the clinic) as pure immunosuppressants, can mediate robust immunostimulatory functions, at least in some circumstances.

The hypothesis that organismal aging might be slowed down, and hence the appearance of aging-associated disorders delayed, has been the subject of intense investigation throughout the past two decades [1, 2]. In this context, several interventions have been demonstrated to significantly extend the healthy lifespan of model organisms as distant from each other on the evolution scale as yeast, worms, flies and mammals [3-6]. For illustrative purposes, such interventions can be classified into two large groups: (1) lifestyle modifications and (2) pharmacological/genetic maneuvers. The former include cycles of caloric restriction as well as a regular physical activity. These are actually known to extend the healthy lifespan of humans since a long time, although the molecular mechanisms underlying this phenomenon have only recently begun to emerge [7-10]. The latter encompass the administration of an increasingly wide panel of chemically unrelated molecules, including (but not limited to) resveratrol (a polyphenol found in grapes and red wine), spermidine (a polyamine that is abundant in grapefruits and soybeans), rapamycin (a macrolide originally isolated from the Easter Island micro-organism Streptomyces hygroscopicus) and multiple rapamycin-like compounds that are collectively known as rapalogs [11-15]. Most, if not all, these interventions share the ability to promote macroautophagy (hereafter referred to as autophagy), a mechanism for the lysosomal degradation of super-fluous, damaged or ectopic intracellular constituents [16, 17]. Moreover, the beneficial effects of both lifestyle modifications and pharmacological/genetic maneuvers have been shown to depend on an intact autophagic machinery, at least in some models [18-21]. In line with this notion, the moderate overexpression of one essential mediator of autophagy (i.e., ATG5) at the whole body level has recently been shown to extend the median lifespan of mice by approximately 17% [22]. Conversely, the genetic inhibition of autophagy-relevant proteins such as Beclin 1 (ATG6), ATG7 and ATG12 reportedly mediates a negative effect on the healthy lifespan of model organisms including Caenorhabditis elegans [23]. As a matter of fact, baseline levels of autophagy play a major role in the maintenance of intracellular (and hence organismal) homeostasis, hence mediating a robust oncosuppressive activity [24-26]. In addition, autophagy orchestrates the adaptive response of cells to multiple adverse conditions, including nutritional, physical and chemical cues [27]. It is therefore not surprising that autophagy might increase the organismal fitness and hence delay aging [28].

Nonetheless, the precise mechanisms whereby specific changes in lifestyle as well as selected chemicals or genetic manipulations delay aging (at least in model organisms) have not yet been fully elucidated. Thus, the efficacy of some anti-aging interventions may rely on mechanisms other than the upregulation of the autophagic flux. The immune system stands out as a good candidate for a part in this process, based on at least two lines of evidence: (1) autophagy plays a major role not only in the activation of innate responses against intracellular pathogens at the cell-autonomous level [29, 30], but also in the elicitation of adaptive immune responses based on the interaction between antigen-presenting cells and antigen-specific CD4+ and CD8+ T lymphocytes [31-33]; and (2) the immune system has been shown to recognize and eliminate not only cells that are prone to undergoing malignant transformation, but also senescent cells, thus contributing to the control of organismal aging [34, 35]. Interestingly, however, rapamycin and other rapalogs have long been known (and currently employed in the clinic) for their capacity to mediate robust immunosuppressive effects [36-38]. Indeed, rapamycin (which is also known as sirolimus) has first been approved by the US Food and Drug Administration (FDA) in 1999 for use in combination with ciclosporin and corticosteroids to prevent acute organ rejection in patients receiving kidney transplants [39, 40]. As it stands, however, the immunosuppressive potential of rapamycin and multiple rapalogs in humans has never been properly tested, as the clinical trials performed to date invariably employed as a control condition the gold-standard immunosuppressive regimens available [41]. In addition, accumulating preclinical and clinical evidence indicates that, at odds with immuno-suppressants that operate by inhibiting calcineurin, such as tacrolimus, rapamycin and other rapalogs might exert a significant immunostimulatory activity, at least under some circumstances.

This hypothesis first originated from the observation that the recipients of solid organs maintained on rapamycin-based regimens manifested a reduced incidence of various tumors, notably lymphoma, as compared to patients subjected to organ transplantation and treated with conventional immunosuppressants such as corticosteroids, ciclosporin, azathioprine or tacrolimus [42-48]. Transplant recipients are indeed known to exhibit an increased incidence of multiple malignancies, encompassing lymphoma as well as hepatocellular carcinoma, Kaposi's sarcoma, and other cutaneous cancers, presumably owing to the state of systemic immunosuppression that is required to avoid rejection [49]. In transplanted patients, rapamycin was associated not only with robust oncosuppressive effects, but also with a bona fide anticancer activity against pre-existent tumors, in particular Kaposi's sarcomas [50-54]. Moreover, local or systemic inflammatory responses have been detected in a fraction of transplanted patients on rapamycin-based maintenance regimens [55, 56]. Often, such responses and the consequent toxicity (be it systemic or selectively affecting the transplant) could be promptly reversed by the reintroduction of calcineurin inhibitors [55, 56].

Recently, cancer-preventive and antineoplastic effects have also been attributed to everolimus (also known as RAD001), a rapalog approved by the US FDA for use in patients affected by various malignancies, including renal cell carcinoma (upon the failure of sunitinib- or sorafenib-based chemotherapeutic regimen) [57], subependymal giant cell astrocytoma [58], progressive neuroendocrine tumors of pancreatic origin [59], and advanced estrogen receptor (ER)+, v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2)+ breast carcinoma (in combination with the aromatase inhibitor exemestane [60]. Although such a beneficial (and completely unsuspected) activity of rapalogs was initially ascribed to their capacity to robustly inhibit the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) in cancer cells, accumulating preclinical evidence indicates that the therapeutic and oncopreventive effects of rapamycin-like compounds originates, at least in part, from cancer cell-extrinsic mechanisms that involve the immune system [61]. In further support of this notion, transplant recipients treated with rapamycin- or everolimus-based maintenance regimens appear to be significantly less predisposed to cytomegalovirus infections than their counterparts receiving conventional immuno-suppressants [62, 63].

Rapamycin and other rapalogs have been shown to exert robust antineoplastic and oncopreventive effects in both transplantable and oncogene-driven tumor models. In immunocompromised mice xenografted with human tumors, this activity obviously reflects cancer cell-intrinsic (or stromal) mechanisms. As a matter of fact, mTORC1 is hyperactivated (hence delivering critical pro-survival signals) in a large number of malignancies, most often due to genetic or epigenetic alterations that result in constitutive signaling via upstream tyrosine kinase receptors (e.g., the epidermal growth factor receptor, EGFR) [64-67]. Conversely, the anticancer effects of rapalogs in immunocompetent settings appear to rely, at least in part, on the elicitation of tumor-targeting immune responses. Rapamycin appears to enhance multiple facets of immune and inflammatory responses elicited in mice by stimuli encompassing replication-competent bacteria [68-73] and viruses [74-77], as well as purified components thereof [74, 78-82] and synthetic immunomodulatory agents [83, 84]. Ovalbumin-specific αβ T lymphocytes exposed to microspheres coated with an ovalbumin-derived peptide plus co-stimulatory CD80 molecules (or with anti-CD3/anti-CD28 antibodies) in the presence of rapamycin exhibited improved memory and antitumor functions in vivo than T cells of the same type activated in the presence of interleukin (IL)-12 [85-87]. Along similar lines, rapamycin has been shown to enhance tumor-targeting CD8+ T-cell memory responses elicited by a poxviral anticancer vaccine in mice [88, 89]. Such an immunostimulatory activity was observed only when rapamycin was administered in a high-dose short therapeutic course, as opposed to both a single, low-dose course as well as prolonged treatment schedules [88, 89]. Of note, rapamycin has also been shown to increase the yield and effector functions of human γδ T cells activated in vitro with isopentenyl pyrophosphate plus recombinant IL-2 [90]. In particular, γδ T cells subjected to antigen stimulation in the presence of rapamycin expressed increased levels of the activation marker CD69, the anti-apoptotic protein BCL-2 and IL-2 receptor α (IL2RA, best known as CD25) [90, 91]. These findings suggest that rapamycin may potentiate purely adaptive immune responses, such as those mediated by αβ T lymphocytes, as well as immune responses with mixed adaptive/innate features, such as those orchestrated by γδ T cells [92, 93]. Other rapalogs, including everolimus and temsirolimus (CCI-779, which has originally been approved by the US FDA for the treatment of advanced renal cell carcinoma in 2007) [94], have been demonstrated to exert immunostimulatory effects, in vitro and in vivo [77, 95, 96]. As a standalone example, temsirolimus was shown significantly improve the therapeutic potential of a peptide-based anticancer vaccine against established renal cell carcinomas and melanomas in mice [96, 97]. Thus, the potential immunostimulatory activity of rapalogs appears to stem from an on-target effect, i.e., the inhibition of mTORC1 [61, 98, 99].

Taken together, these observations suggest that rapamycin and other rapalogs are capable of stimulating, rather than inhibiting, immune responses, at least under selected circumstances. Whether such an immunostimulatory function truly underlies the anti-aging effects of rapamycin remains to be formally demonstrated. Nonetheless, accumulating preclinical data (Table 1) as well as a large amount of circumstantial clinical evidence (Table 2) suggests that these lifespan-extending chemicals can be harnessed to promote therapeutically relevant antitumor immune responses. Properly designed trials that evaluate the actual immunotherapeutic potential of rapamycin-like compounds are urgently awaited. Alternatively, it will be interesting to see whether circulating or intratumoral biomarkers of pre-existing or therapy-elicited immune responses are capable of identifying a subset of cancer patients that obtain full-blown clinical benefits from the administration of rapalogs. The standardized immuno-monitoring procedures that are required in this context have just begun to be defined and implemented into clinical trials [100, 101]. Altogether, these studies will cast new light on whether rapamycin and other rapalogs should still be considered as immuno-suppressants or whether their immunomodulatory activity, similar that of other drugs like cyclophosphamide [102, 103], rather depends on a large panel of factors, including dose and administration schedule. In this latter scenario, rapalogs may turn out to constitute good candidates for the development of novel immunochemotherapeutic regimens [104].

Table 1. Preclinical evidence in support of the immunostimulatory activity of rapalogs

RapalogModelStimulusObservation(s)Ref.
Everolimus (RAD001)Breast cancer-bearing miceIL-15-coding plasmidImproved inhibition of tumor growth[95]
HTLV1-infected T cells and patient-derived ATLL cellsIKK inhibitorDecreased the secretion of IL-10[77]
Sprague-Dawley ratsRemnant kidney modelWorsened disease progression correlating with several markers of inflammation[105]
Sirolimus (Rapamycin)Human PBMCs and DCsLPS from Escherichia coliIncreased NF-κB activation and pro-inflammatory cytokine secretion; decreased STAT3 activation and IL-10 release[78]
Murine DCsLPS from Escherichia coliIncreased secretion of IL-12[79]
Monocytes, macrophages and primary DCsLPS from Escherichia coliIncreased NF-κB activation and pro-inflammatory cytokine secretion; decreased STAT3 activation and IL-10 release[80]
Murine DCs and C57Bl/10, C3H/HeJ, Il4ra−/− miceLPS from Escherichia coliIncreased cytokine secretion and improved T-cell co-stimulation[82]
Human whole bloodLPS, LTA or peptidoglycanInhibition of IL-10 secretion[81]
HEK293 cells stably expressing TLR2 of TLR4Mycobacterium tuberculosisIncreased IL-23 secretion at both the mRNA and protein level[69]
Murine macrophages, DCs and C57Bl/6 miceMycobacterium tuberculosisEnhanced TH1 responses in mice vaccinated with sirolimus-treated DCs[73]
THP1 cells, primary human PBMCs and DCsStaphylococcus aureusIncreased IL-12 secretion at both the mRNA and protein level[68]
Wild-type and transgenic C57Bl/6 miceListeria monocytogenesImproved antigen-specific T-cell responses in the course of infection[70]
Traf6−/− miceAttenuated Listeria monocytogenes strainImproved long-lived CD8+ memory T-cell responses[71]
DCs from wild-type and PI3K-deficient miceLeishmania majorImproved IL-12 secretion by DCs, robust TH1 responses in vivo[72]
Wild-type and transgenic Rag1−/− miceMyxoma virusIncreased anticancer activity of adoptively transferred T lymphocytes[75]
Old C57Bl/6 miceInfluenza virusImprove production of B lymphocytes and optimal responses to vaccination[76]
Wild-type and transgenic C57Bl/6 miceLCMV and engineered vaccinia virusIncreased amounts of antigen-specific T cells[74]
HUVECsThrombinIncreased NF-κB activation[106]
Tumor-bearing transgenic C57Bl/6 miceAnti-CD3/anti-CD8 antibodies Antigen-derived peptides plus CD80Generated OT-I cells that were more effective than IL-12-conditioned effector OT-I cells after adoptive transfer[85]
Human PBMCs and TU167 cellsIL-2 and isopentenyl pyrophosphateIncreased the yield and effector function of human γδ T cells in vitro[90]
Temsirolimus (CCI-779)RCC and melanoma-bearing miceHSP-based anticancer vaccineImproved CD8+ T-cell memory responses and effector functions[96]
Abbreviations: ATLL, adult T-cell leukemia-lymphoma; CAR, chimeric antigen receptor; DC, dendritic cell; HSP, heat-shock protein; HTLV-1, human T-cell lymphotropic virus type 1; HUVEC, human umbilical vein endothelial cell; IKK, IκB kinase; NF-κB, nuclear factor κ-light-chain-enhancer of activated B cells; IL, interleukin; LCMV, lymphocytic choriomeningitis virus; LPS, lipopolysaccharide; LTA, peptidoglycan; PBMC, peripheral blood mononuclear cell; PI3K, phosphoinositide-3-kinase; STAT3, signal transducer and activator of transcription 3; TLR, Toll-like receptor.

Table 2. Clinical evidence in support of the immunostimulatory activity of rapalogs

RapalogSettingObservation(s)Ref.
Everolimus (RAD001)Cardiac transplantationDecreased incidence of CMV infection among everolimus-treated patients[62]
Sirolimus (Rapamycin)Liver transplantationLimited rate of HCV progression and associated hepatic fibrosis[107]
Renal transplantationAnemia correlating with biochemical evidence of a chronic inflammatory state[56]
Renal transplantationDevelopment of glomerulonephritis upon conversion from a calcineurin inhibitor-based immunosuppression to rapamycin[55]
Solid organ transplantationDecreased incidence of CMV infection among sirolimus-treated patients[63]
Solid organ transplantationDecreased incidence of multiple tumors among sirolimus-treated patients[42-48]
Solid organ transplantationConsistent antitumor responses in patients with post-transplantation neoplasms treated with sirolimus[50-54]
Temsirolimus (CCI-779)Advanced cancerNo signs of immunosuppression among everolimus-treated patients[108]
Abbreviations: CMV, cytomegalovirus; HCV, hepatitis C virus.

Conflicts of Interest

The authors have no conflict of interests to declare.

References

  • 1. Campisi J and d'Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nature reviews Molecular cell biology. 2007; 8:729-740..
  • 2. Fossel M. Cell senescence in human aging and disease. Annals of the New York Academy of Sciences. 2002; 959:14-23. [PubMed]
  • 3. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, Nadon NL, Wilkinson JE, Frenkel K, Carter CS, Pahor M, Javors MA, Fernandez E, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009; 460:392-395. [PubMed]
  • 4. 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 metabolism. 2010; 11:35-46. [PubMed]
  • 5. Robida-Stubbs S, Glover-Cutter K, Lamming DW, Mizunuma M, Narasimhan SD, Neumann-Haefelin E, Sabatini DM, Blackwell TK. TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO. Cell metabolism. 2012; 15:713-724. [PubMed]
  • 6. Powers RW 3rd, Kaeberlein M, Caldwell SD, Kennedy BK, Fields S. Extension of chronological life span in yeast by decreased TOR pathway signaling. Genes & development. 2006; 20:174-184. [PubMed]
  • 7. He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, An Z, Loh J, Fisher J, Sun Q, Korsmeyer S, Packer M, May HI, et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature. 2012; 481:511-515. [PubMed]
  • 8. Galluzzi L and Kroemer G. Autophagy mediates the metabolic benefits of endurance training. Circulation research. 2012; 110:1276-1278. [PubMed]
  • 9. Blagosklonny MV. Linking calorie restriction to longevity through sirtuins and autophagy: any role for TOR. Cell death & disease. 2010; 1:e12 [PubMed]
  • 10. Warr MR, Binnewies M, Flach J, Reynaud D, Garg T, Malhotra R, Debnath J, Passegue E. FOXO3A directs a protective autophagy program in haematopoietic stem cells. Nature. 2013; 494:323-327. [PubMed]
  • 11. Eisenberg T, Knauer H, Schauer A, Buttner S, Ruckenstuhl C, Carmona-Gutierrez D, Ring J, Schroeder S, Magnes C, Antonacci L, Fussi H, Deszcz L, Hartl R, et al. Induction of autophagy by spermidine promotes longevity. Nature cell biology. 2009; 11:1305-1314..
  • 12. Morselli E, Maiuri MC, Markaki M, Megalou E, Pasparaki A, Palikaras K, Criollo A, Galluzzi L, Malik SA, Vitale I, Michaud M, Madeo F, Tavernarakis N, et al. Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell death & disease. 2010; 1:e10 [PubMed]
  • 13. Morselli E, Galluzzi L, Kepp O, Criollo A, Maiuri MC, Tavernarakis N, Madeo F, Kroemer G. Autophagy mediates pharmacological lifespan extension by spermidine and resveratrol. Aging. 2009; 1:961-970. [PubMed]
  • 14. Benjamin D, Colombi M, Moroni C, Hall MN. Rapamycin passes the torch: a new generation of mTOR inhibitors. Nature reviews Drug discovery. 2011; 10:868-880..
  • 15. Lamming DW, Ye L, Sabatini DM, Baur JA. Rapalogs and mTOR inhibitors as anti-aging therapeutics. The Journal of clinical investigation. 2013; 123:980-989. [PubMed]
  • 16. Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. The New England journal of medicine. 2013; 368:1845-1846. [PubMed]
  • 17. Klionsky DJ. Autophagy: from phenomenology to molecular understanding in less than a decade. Nature reviews Molecular cell biology. 2007; 8:931-937..
  • 18. Jia K and Levine B. Autophagy is required for dietary restriction-mediated life span extension in C. elegans. Autophagy. 2007; 3:597-599. [PubMed]
  • 19. Hansen M, Chandra A, Mitic LL, Onken B, Driscoll M, Kenyon C. A role for autophagy in the extension of lifespan by dietary restriction in C. elegans. PLoS genetics. 2008; 4:e24.
  • 20. Dwivedi M, Song HO, Ahnn J. Autophagy genes mediate the effect of calcineurin on life span in C. elegans. Autophagy. 2009; 5:604-607. [PubMed]
  • 21. Minina EA, Sanchez-Vera V, Moschou PN, Suarez MF, Sundberg E, Weih M, Bozhkov PV. Autophagy mediates caloric restriction-induced lifespan extension in Arabidopsis. Aging cell. 2013; 12:327-329. [PubMed]
  • 22. Pyo JO, Yoo SM, Ahn HH, Nah J, Hong SH, Kam TI, Jung S, Jung YK. Overexpression of Atg5 in mice activates autophagy and extends lifespan. Nature communications. 2013; 4:2300.
  • 23. Hars ES, Qi H, Ryazanov AG, Jin S, Cai L, Hu C, Liu LF. Autophagy regulates ageing in C. elegans. Autophagy. 2007; 3:93-95. [PubMed]
  • 24. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science. 2011; 333:1109-1112. [PubMed]
  • 25. Morselli E, Galluzzi L, Kepp O, Vicencio JM, Criollo A, Maiuri MC, Kroemer G. Anti- and pro-tumor functions of autophagy. Biochimica et biophysica acta. 2009; 1793:9 1524-1532. [PubMed]
  • 26. White E. Deconvoluting the context-dependent role for autophagy in cancer. Nature reviews Cancer. 2012; 12:401-410..
  • 27. Kroemer G, Marino G, Levine B. Autophagy and the integrated stress response. Molecular cell. 2010; 40:280-293. [PubMed]
  • 28. Martins I, Galluzzi L, Kroemer G. Hormesis, cell death and aging. Aging. 2011; 3:821-828. [PubMed]
  • 29. Levine B, Mizushima N, Virgin HW. Autophagy in immunity and inflammation. Nature. 2011; 469:323-335. [PubMed]
  • 30. Benjamin JL, Sumpter R Jr, Levine B, Hooper LV. Intestinal epithelial autophagy is essential for host defense against invasive bacteria. Cell host & microbe. 2013; 13:723-734. [PubMed]
  • 31. Yi Y, Zhou Z, Shu S, Fang Y, Twitty C, Hilton TL, Aung S, Urba WJ, Fox BA, Hu HM, Li Y. Autophagy-assisted antigen cross-presentation: Autophagosome as the argo of shared tumor-specific antigens and DAMPs. Oncoimmunology. 2012; 1:976-978. [PubMed]
  • 32. Ma Y, Galluzzi L, Zitvogel L, Kroemer G. Autophagy and cellular immune responses. Immunity. 2013; 39:211-227. [PubMed]
  • 33. Michaud M, Sukkurwala AQ, Martins I, Shen S, Zitvogel L, Kroemer G. Subversion of the chemotherapy-induced anticancer immune response by the ecto-ATPase CD39. Oncoimmunology. 2012; 1:393-395. [PubMed]
  • 34. Senovilla L, Galluzzi L, Zitvogel L, Kroemer G. Immunosurveillance as a regulator of tissue homeostasis. Trends in immunology. 2013; 34:471-481. [PubMed]
  • 35. Prendergast GC and Metz R. A perspective on new immune adjuvant principles: Reprogramming inflammatory states to permit clearance of cancer cells and other age-associated cellular pathologies. Oncoimmunology. 2012; 1:924-929. [PubMed]
  • 36. Law BK. Rapamycin: an anti-cancer immunosuppressant? Critical reviews in oncology/hematology. 2005; 56:47-60. [PubMed]
  • 37. Morelon E, Mamzer-Bruneel MF, Peraldi MN, Kreis H. Sirolimus: a new promising immunosuppressive drug. Towards a rationale for its use in renal transplantation. Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association - European Renal Association. 2001; 16:18-20..
  • 38. Sehgal SN, Molnar-Kimber K, Ocain TD, Weichman BM. Rapamycin: a novel immunosuppressive macrolide. Medicinal research reviews. 1994; 14:1-22. [PubMed]
  • 39. Kahan BD, Podbielski J, Napoli KL, Katz SM, Meier-Kriesche HU, Van Buren CT. Immunosuppressive effects and safety of a sirolimus/cyclosporine combination regimen for renal transplantation. Transplantation. 1998; 66:1040-1046. [PubMed]
  • 40. Brattstrom C, Sawe J, Tyden G, Herlenius G, Claesson K, Zimmerman J, Groth CG. Kinetics and dynamics of single oral doses of sirolimus in sixteen renal transplant recipients. Therapeutic drug monitoring. 1997; 19:397-406. [PubMed]
  • 41. Blagosklonny MV. Immunosuppressants in cancer prevention and therapy Oncoimmunology. 2013; 2:e26961.
  • 42. Mathew T, Kreis H, Friend P. Two-year incidence of malignancy in sirolimus-treated renal transplant recipients: results from five multicenter studies. Clinical transplantation. 2004; 18:446-449. [PubMed]
  • 43. Yakupoglu YK, Buell JF, Woodle S, Kahan BD. Individualization of immunosuppressive therapy. III. Sirolimus associated with a reduced incidence of malignancy. Transplantation proceedings. 2006; 38:358-361. [PubMed]
  • 44. Campistol JM, Eris J, Oberbauer R, Friend P, Hutchison B, Morales JM, Claesson K, Stallone G, Russ G, Rostaing L, Kreis H, Burke JT, Brault Y, et al. Sirolimus therapy after early cyclosporine withdrawal reduces the risk for cancer in adult renal transplantation. Journal of the American Society of Nephrology: JASN. 2006; 17:581-589. [PubMed]
  • 45. Alberu J, Pascoe MD, Campistol JM, Schena FP, Rial Mdel C, Polinsky M, Neylan JF, Korth-Bradley J, Goldberg-Alberts R, Maller ES, Sirolimus CTSG. Lower malignancy rates in renal allograft recipients converted to sirolimus-based, calcineurin inhibitor-free immunotherapy: 24-month results from the CONVERT trial. Transplantation. 2011; 92:303-310. [PubMed]
  • 46. Kauffman HM, Cherikh WS, Cheng Y, Hanto DW, Kahan BD. Maintenance immunosuppression with target-of-rapamycin inhibitors is associated with a reduced incidence of de novo malignancies. Transplantation. 2005; 80:883-889. [PubMed]
  • 47. Salgo R, Gossmann J, Schofer H, Kachel HG, Kuck J, Geiger H, Kaufmann R, Scheuermann EH. Switch to a sirolimus-based immunosuppression in long-term renal transplant recipients: reduced rate of (pre-)malignancies and nonmelanoma skin cancer in a prospective, randomized, assessor-blinded, controlled clinical trial. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2010; 10:1385-1393. [PubMed]
  • 48. Campbell SB, Walker R, Tai SS, Jiang Q, Russ GR. Randomized controlled trial of sirolimus for renal transplant recipients at high risk for nonmelanoma skin cancer. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2012; 12:1146-1156. [PubMed]
  • 49. London NJ, Farmery SM, Will EJ, Davison AM, Lodge JP. Risk of neoplasia in renal transplant patients. Lancet. 1995; 346:403-406. [PubMed]
  • 50. Mohsin N, Budruddin M, Pakkyara A, Darweesh A, Nayyer M, Amitabh J, Daar AS. Complete regression of visceral Kaposi's sarcoma after conversion to sirolimus. Experimental and clinical transplantation: official journal of the Middle East Society for Organ Transplantation. 2005; 3:366-369. [PubMed]
  • 51. Zmonarski SC, Boratynska M, Puziewicz-Zmonarska A, Kazimierczak K, Klinger M. Kaposi's sarcoma in renal transplant recipients. Annals of transplantation: quarterly of the Polish Transplantation Society. 2005; 10:59-65. [PubMed]
  • 52. Zmonarski SC, Boratynska M, Rabczynski J, Kazimierczak K, Klinger M. Regression of Kaposi's sarcoma in renal graft recipients after conversion to sirolimus treatment. Transplantation proceedings. 2005; 37:964-966. [PubMed]
  • 53. Cullis B, D'Souza R, McCullagh P, Harries S, Nicholls A, Lee R, Bingham C. Sirolimus-induced remission of posttransplantation lymphoproliferative disorder. American journal of kidney diseases: the official journal of the National Kidney Foundation. 2006; 47:e67-72. [PubMed]
  • 54. Stallone G, Schena A, Infante B, Di Paolo S, Loverre A, Maggio G, Ranieri E, Gesualdo L, Schena FP, Grandaliano G. Sirolimus for Kaposi's sarcoma in renal-transplant recipients. The New England journal of medicine. 2005; 352:1317-1323. [PubMed]
  • 55. Dittrich E, Schmaldienst S, Soleiman A, Horl WH, Pohanka E. Rapamycin-associated post-transplantation glomerulonephritis and its remission after reintroduction of calcineurin-inhibitor therapy. Transplant international: official journal of the European Society for Organ Transplantation. 2004; 17:215-220. [PubMed]
  • 56. Thaunat O, Beaumont C, Chatenoud L, Lechaton S, Mamzer-Bruneel MF, Varet B, Kreis H, Morelon E. Anemia after late introduction of sirolimus may correlate with biochemical evidence of a chronic inflammatory state. Transplantation. 2005; 80:1212-1219. [PubMed]
  • 57. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grunwald V, Thompson JA, Figlin RA, Hollaender N, Urbanowitz G, Berg WJ, Kay A, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008; 372:449-456. [PubMed]
  • 58. Krueger DA, Care MM, Holland K, Agricola K, Tudor C, Mangeshkar P, Wilson KA, Byars A, Sahmoud T, Franz DN. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. The New England journal of medicine. 2010; 363:1801-1811. [PubMed]
  • 59. Yao JC, Shah MH, Ito T, Bohas CL, Wolin EM, Van Cutsem E, Hobday TJ, Okusaka T, Capdevila J, de Vries EG, Tomassetti P, Pavel ME, Hoosen S, et al. Everolimus for advanced pancreatic neuroendocrine tumors. The New England journal of medicine. 2011; 364:514-523. [PubMed]
  • 60. Baselga J, Campone M, Piccart M, Burris HA 3rd, Rugo HS, Sahmoud T, Noguchi S, Gnant M, Pritchard KI, Lebrun F, Beck JT, Ito Y, Yardley D, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. The New England journal of medicine. 2012; 366:520-529. [PubMed]
  • 61. Araki K, Ellebedy AH, Ahmed R. TOR in the immune system. Current opinion in cell biology. 2011; 23:707-715. [PubMed]
  • 62. Hill JA, Hummel M, Starling RC, Kobashigawa JA, Perrone SV, Arizon JM, Simonsen S, Abeywickrama KH, Bara C. A lower incidence of cytomegalovirus infection in de novo heart transplant recipients randomized to everolimus. Transplantation. 2007; 84:1436-1442. [PubMed]
  • 63. Demopoulos L, Polinsky M, Steele G, Mines D, Blum M, Caulfield M, Adamkovic A, Liu Q, Harler MB, Hahn C, Singh A. Reduced risk of cytomegalovirus infection in solid organ transplant recipients treated with sirolimus: a pooled analysis of clinical trials. Transplantation proceedings. 2008; 40:1407-1410. [PubMed]
  • 64. Pollack BP. EGFR inhibitors, MHC expression and immune responses: Can EGFR inhibitors be used as immune response modifiers? Oncoimmunology. 2012; 1:71-74. [PubMed]
  • 65. Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nature reviews Molecular cell biology. 2011; 12:21-35..
  • 66. Wouters BG and Koritzinsky M. Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nature reviews Cancer. 2008; 8:851-864..
  • 67. Shackelford DB and Shaw RJ. The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nature reviews Cancer. 2009; 9:563-575..
  • 68. Uthaisangsook S, Day NK, Hitchcock R, Lerner A, James-Yarish M, Good RA, Haraguchi S. Negative regulation of interleukin-12 production by a rapamycin-sensitive signaling pathway: a brief communication. Experimental biology and medicine. 2003; 228:1023-1027. [PubMed]
  • 69. Yang CS, Song CH, Lee JS, Jung SB, Oh JH, Park J, Kim HJ, Park JK, Paik TH, Jo EK. Intracellular network of phosphatidylinositol 3-kinase, mammalian target of the rapamycin/70 kDa ribosomal S6 kinase 1, and mitogen-activated protein kinases pathways for regulating mycobacteria-induced IL-23 expression in human macrophages. Cellular microbiology. 2006; 8:1158-1171. [PubMed]
  • 70. Ferrer IR, Wagener ME, Robertson JM, Turner AP, Araki K, Ahmed R, Kirk AD, Larsen CP, Ford ML. Cutting edge: Rapamycin augments pathogen-specific but not graft-reactive CD8+ T cell responses. Journal of immunology. 2010; 185:2004-2008..
  • 71. Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, Jones RG, Choi Y. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009; 460:103-107. [PubMed]
  • 72. Fukao T, Tanabe M, Terauchi Y, Ota T, Matsuda S, Asano T, Kadowaki T, Takeuchi T, Koyasu S. PI3K-mediated negative feedback regulation of IL-12 production in DCs. Nature immunology. 2002; 3:875-881. [PubMed]
  • 73. Jagannath C, Lindsey DR, Dhandayuthapani S, Xu Y, Hunter RL Jr, Eissa NT. Autophagy enhances the efficacy of BCG vaccine by increasing peptide presentation in mouse dendritic cells. Nature medicine. 2009; 15:267-276..
  • 74. Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP, Ahmed R. mTOR regulates memory CD8 T-cell differentiation. Nature. 2009; 460:108-112. [PubMed]
  • 75. Thomas DL, Doty R, Tosic V, Liu J, Kranz DM, McFadden G, Macneill AL, Roy EJ. Myxoma virus combined with rapamycin treatment enhances adoptive T cell therapy for murine melanoma brain tumors. Cancer immunology, immunotherapy: CII. 2011; 60:1461-1472..
  • 76. Chen C, Liu Y, Liu Y, Zheng P. mTOR regulation and therapeutic rejuvenation of aging hematopoietic stem cells. Science signaling. 2009; 2:ra75 [PubMed]
  • 77. Nishioka C, Ikezoe T, Yang J, Udaka K, Yokoyama A. The combination of IkappaB kinase beta inhibitor and everolimus modulates expression of interleukin-10 in human T-cell lymphotropic virus type-1-infected T cells. Immunology. 2013; 138:216-227. [PubMed]
  • 78. Haidinger M, Poglitsch M, Geyeregger R, Kasturi S, Zeyda M, Zlabinger GJ, Pulendran B, Horl WH, Saemann MD, Weichhart T. A versatile role of mammalian target of rapamycin in human dendritic cell function and differentiation. Journal of immunology. 2010; 185:3919-3931..
  • 79. Ohtani M, Nagai S, Kondo S, Mizuno S, Nakamura K, Tanabe M, Takeuchi T, Matsuda S, Koyasu S. Mammalian target of rapamycin and glycogen synthase kinase 3 differentially regulate lipopolysaccharide-induced interleukin-12 production in dendritic cells. Blood. 2008; 112:635-643. [PubMed]
  • 80. Weichhart T, Costantino G, Poglitsch M, Rosner M, Zeyda M, Stuhlmeier KM, Kolbe T, Stulnig TM, Horl WH, Hengstschlager M, Muller M, Saemann MD. The TSC-mTOR signaling pathway regulates the innate inflammatory response. Immunity. 2008; 29:565-577. [PubMed]
  • 81. Jorgensen PF, Wang JE, Almlof M, Solberg R, Okkenhaug C, Scholz T, Thiemermann C, Foster SJ, Aasen AO. Sirolimus interferes with the innate response to bacterial products in human whole blood by attenuation of IL-10 production. Scandinavian journal of immunology. 2001; 53:184-191. [PubMed]
  • 82. Hackstein H, Taner T, Zahorchak AF, Morelli AE, Logar AJ, Gessner A, Thomson AW. Rapamycin inhibits IL-4--induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo. Blood. 2003; 101:4457-4463. [PubMed]
  • 83. Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012; 1:894-907. [PubMed]
  • 84. Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012; 1:699-716. [PubMed]
  • 85. Rao RR, Li Q, Odunsi K, Shrikant PA. The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin. Immunity. 2010; 32:67-78. [PubMed]
  • 86. You CX, Shi M, Liu Y, Cao M, Luo R, Hermonat PL. AAV2/IL-12 gene delivery into dendritic cells (DC) enhances CTL stimulation above other IL-12 applications: Evidence for IL-12 intracrine activity in DC. Oncoimmunology. 2012; 1:847-855. [PubMed]
  • 87. Song DG and Powell DJ. Pro-survival signaling via CD27 costimulation drives effective CAR T-cell therapy. Oncoimmunology. 2012; 1:547-549. [PubMed]
  • 88. Li Q, Rao R, Vazzana J, Goedegebuure P, Odunsi K, Gillanders W, Shrikant PA. Regulating mammalian target of rapamycin to tune vaccination-induced CD8(+) T cell responses for tumor immunity. Journal of immunology. 2012; 188:3080-3087..
  • 89. Srivastava RK, Utley A, Shrikant PA. Rapamycin: A rheostat for CD8(+) T-cell-mediated tumor therapy. Oncoimmunology. 2012; 1:1189-1190. [PubMed]
  • 90. Li H and Pauza CD. Rapamycin increases the yield and effector function of human gammadelta T cells stimulated in vitro. Cancer immunology, immunotherapy: CII. 2011; 60:361-370..
  • 91. Putz EM, Schuster C, Sexl V. Bcl-2: Live and let die. Oncoimmunology. 2012; 1:749-750. [PubMed]
  • 92. Anderson J, Gustafsson K, Himoudi N, Yan M, Heuijerjans J. Licensing of gammadeltaT cells for professional antigen presentation: A new role for antibodies in regulation of antitumor immune responses. Oncoimmunology. 2012; 1:1652-1654. [PubMed]
  • 93. Gertner-Dardenne J, Fauriat C, Vey N, Olive D. Immunotherapy of acute myeloid leukemia based on gammadelta T cells. Oncoimmunology. 2012; 1:1614-1616. [PubMed]
  • 94. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, Kovacevic Z, Lesovoy V, Schmidt-Wolf IG, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. The New England journal of medicine. 2007; 356:2271-2281. [PubMed]
  • 95. Zhao N, Li X, He X, Qiu Y, Zhu L, Qi F. Interleukin-15 gene therapy and the mammalian target of rapamycin inhibitor everolimus inhibit the growth of metastatic breast cancer. The journal of gene medicine. 2013; 15:366-374. [PubMed]
  • 96. Wang Y, Wang XY, Subjeck JR, Shrikant PA, Kim HL. Temsirolimus, an mTOR inhibitor, enhances anti-tumour effects of heat shock protein cancer vaccines. British journal of cancer. 2011; 104:643-652. [PubMed]
  • 97. Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology. 2012; 1:1557-1576. [PubMed]
  • 98. Metz R, Rust S, Duhadaway JB, Mautino MR, Munn DH, Vahanian NN, Link CJ, Prendergast GC. IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: A novel IDO effector pathway targeted by D-1-methyl-tryptophan. Oncoimmunology. 2012; 1:1460-1468. [PubMed]
  • 99. Araki K, Youngblood B, Ahmed R. The role of mTOR in memory CD8 T-cell differentiation. Immunological reviews. 2010; 235:234-243. [PubMed]
  • 100. Hoos A, Janetzki S, Britten CM. Advancing the field of cancer immunotherapy: MIATA consensus guidelines become available to improve data reporting and interpretation for T-cell immune monitoring. Oncoimmunology. 2012; 1:1457-1459. [PubMed]
  • 101. Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautes-Fridman C, Ma Y, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology. 2012; 1:1323-1343. [PubMed]
  • 102. Malvicini M, Alaniz L, Bayo J, Garcia M, Piccioni F, Fiore E, Atorrasagasti C, Aquino JB, Matar P, Mazzolini G. Single low-dose cyclophosphamide combined with interleukin-12 gene therapy is superior to a metronomic schedule in inducing immunity against colorectal carcinoma in mice. Oncoimmunology. 2012; 1:1038-1047. [PubMed]
  • 103. Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nature reviews Drug discovery. 2012; 11:215-233..
  • 104. Egilmez NK, Harden JL, Rowswell-Turner RB. Chemoimmunotherapy as long-term maintenance therapy for cancer. Oncoimmunology. 2012; 1:563-565. [PubMed]
  • 105. Vogelbacher R, Wittmann S, Braun A, Daniel C, Hugo C. The mTOR inhibitor everolimus induces proteinuria and renal deterioration in the remnant kidney model in the rat. Transplantation. 2007; 84:1492-1499. [PubMed]
  • 106. Minhajuddin M, Fazal F, Bijli KM, Amin MR, Rahman A. Inhibition of mammalian target of rapamycin potentiates thrombin-induced intercellular adhesion molecule-1 expression by accelerating and stabilizing NF-kappa B activation in endothelial cells. Journal of immunology. 2005; 174:5823-5829..
  • 107. McKenna GJ, Trotter JF, Klintmalm E, Onaca N, Ruiz R, Jennings LW, Neri M, O'Leary JG, Davis GL, Levy MF, Goldstein RM, Klintmalm GB. Limiting hepatitis C virus progression in liver transplant recipients using sirolimus-based immunosuppression. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2011; 11:2379-2387. [PubMed]
  • 108. Raymond E, Alexandre J, Faivre S, Vera K, Materman E, Boni J, Leister C, Korth-Bradley J, Hanauske A, Armand JP. Safety and pharmacokinetics of escalated doses of weekly intravenous infusion of CCI-779, a novel mTOR inhibitor, in patients with cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2004; 22:2336-2347. [PubMed]
Download PDF