Abstract
The mechanistic/mammalian target of rapamycin (mTOR) is one of the systems that are necessary to maintain cell homeostasis, such as survival, proliferation, and differentiation. mTOR inhibitors (mTOR-Is) are utilized as immunosuppressants and anti-cancer drugs. In organ allotransplantation, current regimens infrequently include an mTOR-I, which are positioned more commonly as alternative immunosuppressants. In clinical allotransplantation, long-term efficacy has been established, but there is a significant incidence of adverse events, for example, inhibition of wound healing, buccal ulceration, anemia, hyperglycemia, dyslipidemia, and thrombocytopenia, some of which are dose-dependent. mTOR-Is have properties that may be especially beneficial in xenotransplantation. These include suppression of T cell proliferation, increases in the number of T regulatory cells, inhibition of pig graft growth, and anti-inflammatory, antiviral, and anti-cancer effects. We here review the potential benefits and risks of mTOR-Is in xenotransplantation and suggest that the benefits exceed the adverse effects.
Keywords: graft growth, immunosuppression, inflammation, mTOR inhibitor, neoplasia, Tregs, viral infection, xenotransplantation
1 |. INTRODUCTION
Initially, rapamycin was studied as a macrolide antibiotic.1 During these studies, the mechanistic/mammalian target of rapamycin (mTOR) was identified, and rapamycin was found to inhibit the cell cycle at the G1 phase.1 Although there are now a number of mTOR inhibitor (mTOR-I) compounds, some of which are used therapeutically, their basic mechanism of action is the same.2 However, pharmacokinetic parameters and sub-effects are different.3 Therefore, considering the limited information available, it may be necessary to compare them and consider switching to a different one depending on the situation. Today, mTOR-Is are well-known as immunosuppressants or anti-cancer drugs.
Although not administered to the patient who received the first pig heart transplant,4 there has been significant experience of mTOR-Is in pig-to-nonhuman primate (NHP) models5–10 (Table 1). The barriers to successful xenotransplantation include several that an mTOR-I might help overcome (Table 2). mTOR-Is have many qualities that make them advantageous in xenotransplantation (Table 3A).
Table 1:
Year (Reference) |
Pig organ (Gene-editing) |
Heterotopic/Life-supporting | Maximal graft survival (days) (Cause of termination of experiment) |
---|---|---|---|
2005 (McGregor et al)4 |
Heart (CD46) |
Heterotopic | 137 (Rejection) |
2005 (Brandl et al)5 |
Heart (CD55) |
Life-supporting | 25 (Rejection) |
2011 (Byrne et al)6 |
Heart (CD46/TPC) |
Life-supporting | 57 (Infection/PTLD) |
2015 (Iwase et al)7 |
Kidney (GTKO/CD46/CD55/TBM/EPCR/CD39) |
Life-supporting | 136 (Infection) |
2017 (Iwase et al)8 |
Kidney (GTKO/CD46/CD55/EPCR/ TFPI/CD47) |
Life-supporting | 260 (Infection) |
2020 (Reichart et al)9 |
Heart (GTKO/CD46/TBM) |
Life-supporting | 195 (Study end-point) |
EPCR = endothelial protein C receptor; PTLD = post-transplant lymphoproliferative disease; TBM = thrombomodulin; TFPI = tissue factor pathway inhibitor; TPC; polyethylene glycol α-Gal polymer.
Table 2:
Barriers | Effect of mTOR-I | Focus | Targets | References |
---|---|---|---|---|
Innate immune response | Some effect | Recipient | Dendritic cells | 24, 25, 26, 27, 28, 30 |
Adaptive immune response | Effective | Recipient | T cells, B cells Antibody producing cells |
11, 14, 15, 20, 21, 22 |
Inflammatory response | Effective | Recipient Donor |
Dendritic cells Endothelial cells |
27, 30, 31, 32, 33, 34 |
Coagulation dysfunction | - | - | - | - |
Viral infection | Effective | Recipient Donor |
CMV, EBV, BKV, COVID-19 | 23, 72, 77, 78, 79, 80, 81, 82, 83, 84, 86, 87 |
Rapid growth of the pig organ | Effective | Donor | Graft | 8, 9, 91, 92 |
De novo neoplasia | Effective | Recipient Donor |
Tumor cells | 83, 84, 99, 100, 101, 102 |
Table 3:
A. Advantages |
---|
Suppresses both cellular and antibody-mediated rejection (T cells, B cells, and antibody-producting cells) Associated with an increase in Tregs (especially when combined with CD40/CD154 co-stimulation pathway blockade) Inhibits the primate and pig IL-6/IL-6Rα/STAT3 pathway and suppresses inflammatory gene expression Decreases proinflammatory cytokines (e.g., IL-2, IFN-γ, and IL-6) Reduces pig organ growth Anti-viral/anti-cancer activity |
|
B. Disadvantages |
Increases IgM production (to specific antigens at low-concentration of mTOR-I) May inhibit wound healing (though this has never been a problem in our pig-to-NHP model) Not always tolerated by patients (but many side-effects are dose-dependent) Can be associated with proteinuria (though we have not seen this) May increase the risk of cardiovascular disease (e.g., dyslipidemia and hyperglycemia) May need supportive medications (e.g., erythropoietin, statin/fibrate, and insulin) |
1.1 |. Suppression of the immune and inflammatory responses
The main purpose of using an mTOR-I in organ transplantation is to suppress “signal III” in T cell activation. Signal III is the stimulation by interleukin (IL)-2 that triggers T cell proliferation.11 mTOR is downstream of the IL-2 and IL-15 receptors.11 IL-15 in tissues can promote CD4+T helper cell-mediated immunity and provide co-stimulatory signals to effector cytotoxic T cells.12 Inflammatory IL-15 has a variety of effector functions against memory CD8+T cells, such as proliferation, cell survival, enhanced cytotoxicity, and trafficking into non-lymphoid tissues.13 The immunosuppressive effect of mTOR-Is is by blockade of IL-2 and IL-15 induction of T cell proliferation14 (not by blocking all IL-2 signaling). Inhibition of mTOR also leads to suppression of B cell activation, proliferation, and differentiation into antibody-producing cells, and antibody production.14,15
The mTOR network consists of two main complexes, mTOR complex-1 (mTORC1) and −2 (mTORC2).16 mTORC1 is sensitive to an mTOR-I, and mTORC2 is inhibited by prolonged administration of an mTOR-I.17,18 mTORC2 is important for peripheral B lymphocyte maturation, homeostasis, function, and survival19 (low-dose rapamycin may increase IgM production against influenza vaccines, while maintaining inhibition of antibody class switch20). The mTOR-I, PP242 (not used clinically), inhibits mTORC1 and mTORC2 at low concentrations (without the need for prolonged administration) and increases the fraction of B cells undergoing antibody class switching.21 The use of PP242 at high concentrations inhibits B cell proliferation and differentiation.21
A recent study regarding SARS-CoV-2 mRNA vaccination showed that kidney transplant recipients who received an mTOR-I achieved higher IgG titers and specific T cell-derived interferon (IFN)-γ against SARS-CoV-2.22 This result is contrary to a report regarding influenza vaccination.20 There are some differences between the two reports such as the type of vaccine (e.g., mRNA or inactivated vaccine) and the timing of vaccination. The effect of mTOR-Is on antibody production after vaccination is influenced by the concentration of the mTOR-I, the type of antigen, and the amount and frequency of antigen exposure. In a clinical allotransplant study, de novo donor-specific antibody (DSA) tended to be lower when the immunosuppressive regimen included an mTOR-I.23
Collectively, these reports suggest that maintaining a sufficient concentration of mTOR-I for a prolonged period would help prevent AMR by sufficiently inhibiting both mTORC1 and mTORC2.
Evidence is that the current target trough levels for mTOR-Is are in the optimal range. Caution should be exercised immediately after the initiation of administration of an mTOR-I or when switching to another type of immunosuppressant, as there may be a period of decreased blood concentration. In addition, rapamycin and everolimus have different strengths of interaction on mTORC2.3 Therefore, their effects may differ.
Dendritic cells (DCs) are also influenced by an mTOR-I.24 mTOR-Is impair endocytosis.25,26 Additionally, expression of the major histocompatibility complex (MHC)-I and -II, co-stimulatory molecules (including CD40), and antigen uptake receptors in DCs are decreased.26–28 It can be assumed that an immune response initiated by DC uptake of donor antigens by endocytosis (indirect recognition pathway) in allotransplantation is suppressed.29 Subsequently, DC-induced T cell stimulation and proliferation are reduced by an mTOR-I.26–28 T cell-primed mTOR-I-treated DCs decrease proinflammatory cytokines, for example, IL-2 and IFN-γ.27,30 Thus, mTOR-Is have the potential to suppress antigen presentation and the production of proinflammatory cytokines. They also block IL-6 and vascular endothelial growth factor (VEGF) production after reperfusion.31 Macrophages treated with an mTOR-I down-regulate the production of cytokines, including IL-6.32 In addition, mTOR-Is suppress the mTOR-signal transducer and activator of transcription (STAT) 3 pathway and reduce protein and mRNA levels of IFN-γ and IL-17 in splenocytes.33 In human vein endothelial cells, mTOR inhibition reduces IFN-γ-induced human leukocyte antigen (HLA)-class II expression.34
These results suggest that the effects of mTOR inhibition in suppressing the immune response and inflammation are exerted not only on the recipient’s immune system, but also on the donor graft. mTOR-Is have anti-inflammatory effects and subsequent suppression of the immune response.
In xenotransplantation, early upregulation of recipient IL-6 may result in the activation of coagulation.35 Additionally, increases in serum pig-IL-6 and baboon-IL-6 correlated with graft dysfunction.36 mTOR inhibition partially inhibited human, baboon, and pig IL-6/IL-6 receptor (IL-6R)α/STAT3 pathways and suppressed inflammatory gene expression.37
An mTOR-I is able to suppress pig endothelial cell-induced responder T cell proliferation.38 mTOR-I-preconditioned pig endothelial cells do not stimulate T cell proliferation.38
The prevention of systemic inflammation in xenograft recipients (SIXR) is beneficial in prolonging graft survival in xenotransplantation.35,39,40 Therefore, mTOR-Is have the potential to prevent SIXR and contribute to achieving long-term xenograft survival.
1.2 |. Induction of T regulatory cells (Tregs) and tolerance
mTOR-Is can suppress CD4+T helper cells and effector cytotoxic T cells, but may also have a beneficial effect on the induction of Tregs. Donor-specific tolerance is one of the goals of transplantation. CD4+CD25+Foxp3+ Tregs41 can suppress the immune response, and contribute to the induction of tolerance.41,42 Tregs help to maintain stable graft function and reduce the incidence of both acute and chronic rejection.42
Organ graft recipients who were treated with an mTOR-I had a higher frequency of Tregs compared with those treated with a calcineurin inhibitor (CNI).43 Low IL-2 receptor signaling acts to promote the maturation of immature Tregs (CD4+CD25lowFoxp3low) to CD4+CD25hiFoxp3hiTregs.44 IL-2 is required for Tregs, whereas CNIs inhibit the transcription of IL-2.45 Thus, it is suggested that mTOR inhibition maintains Tregs better than therapy with a CNI.
Some studies indicate that mTOR inhibition can expand the number of Tregs regardless of antigen levels.46–49 In a transforming growth factor-β (TGF-β)-dependent manner, an mTOR-I enhanced the expression of Foxp3.50 Tregs that are expanded by an mTOR-I suppress the proliferation of both syngeneic and allogeneic CD4+ and CD8+ T cells in vitro and prevent allograft rejection in vivo.46,51 Similarly in an in vitro study, purified Tregs enriched from CD4+ cells in the presence of an mTOR-I suppressed baboon anti-pig immune responses more strongly than freshly-isolated natural Tregs.52
The CD40/CD154 pathway is one of the co-stimulatory pathways between antigen-presenting cells (APCs) and T cells.53 Anti-CD154 (anti-CD40 ligand) monoclonal antibody (mAb) is attractive as an immunosuppressant, and may lead to tolerance.54 The mechanism by which anti-CD154 mAb induces tolerance is by inducing Tregs.55–57 In contrast, another report suggested that monotherapy with an anti-CD154mAb cannot induce tolerance and is not sufficient to prevent chronic graft vasculopathy.58 Thus, the combination of an mTOR-I and an anti-CD154 mAb may have additive or synergistic effects. As supportive information, in islet-kidney allotransplantation in NHPs, the combination of an mTOR-I and an anti-CD40mAb achieved long-term islet and kidney allograft survival.59 An mTOR-I combined with blockade of the CD40/CD154 pathway may therefore be an efficient regimen.
A regimen including an mTOR-I may therefore have advantages in the establishment of donor-specific tolerance.
1.3 |. Effect on viral infection
Cytomegalovirus (CMV) and Epstein-Barr virus (EBV) cause well-known infections after allotransplantation.60–67 After kidney allotransplantation, BK polyomavirus (BKV) is associated with rejection and kidney graft dysfunction.68 Furthermore, SARS-CoV-2 (COVID-19) has caused a pandemic since 2019.69–71 Although infections with CMV, EBV, BKV, and COVID-19 are not limited to immunosuppressed patients, the influence of immunosuppressants on viral infection should be considered.
CMV is one of the most frequent infectious agents seen in organ transplant recipients and is associated with higher risks of graft rejection/loss and patient morbidity and mortality.60–65 An mTOR-I reduced the incidence of human CMV viremia after allotransplantation.72 In xenotransplantation studies, pig CMV (pCMV) infection was associated with rejection and reduced survival.62,63,65,73–75 pCMV transmission was also associated with increased levels of IL-6 and TNFα.75 Anti-CMV agents, for example, ganciclovir or valganciclovir, are generally administered in clinical CMV infection,76 but these agents are much less effective in pCMV infection.64
There is increasing evidence that mTOR-Is have anti-CMV properties.23,77–79 They have indirect anti-viral activity and reduce the rate of CMV infection in CMV-seropositive organ transplant recipients.80 mTOR-Is inhibit proliferation of infected cells by activating specific signaling pathways.81 The mechanism of suppression of CMV infection by mTOR-Is has been reported as reinvigorating αβ and γδ T-cell function.82
EBV is a human oncovirus and is associated with the development of post-transplant lymphoproliferative disorders (PTLD).67 An mTOR-I decreased EBV+ B cell proliferation.83 After allotransplantation, the levels of EBV-DNA were significantly lower in patients receiving an mTOR-I than in those receiving a mycophenolate mofetil-based regimen.84 Thus, there is evidence that the risk of PTLD may be reduced in patients receiving an mTOR-I.7
Most adults have been exposed to BKV85 and this is particularly problematic after kidney transplantation.68 However, therapy with an mTOR-I reduced urinary BKV titers86 and the incidence of BK viremia.72
Although COVID-19 infection has been common recently,69–71 there are reports that the administration of an mTOR-I may reduce the incidence through a mechanism that may involve inhibition of viral replication.87
Therefore, although it is not definitively known whether mTOR inhibition reduces viral infections in xenotransplantation, we suggest that the anti-viral effect of mTOR-Is may be beneficial in xenotransplantation.
1.4 |. Effect on growth of a pig graft
mTOR coordinates eukaryotic cell growth,88 and mTOR inhibition inhibits the signal transduction pathways required for cell growth and proliferation.88,89 This inhibitory effect has been applied in various diseases.90 The observation that the growth of pig kidneys in NHPs receiving rapamycin8,9 appeared to be less than in those that did not91 suggested a beneficial effect on pig organ growth. In a cardiac xenotransplantation study, mTOR inhibition appeared to suppress growth,92 but its immunosuppressive effect may also have contributed to reduced cell infiltration, edema, and interstitial hemorrhage (thus reducing the apparent “growth” of the organ).
We suggest, therefore, that inclusion of an mTOR-I may inhibit graft growth and subsequent dysfunction.
1.5 |. Effect on de novo neoplasia
As a central regulator of cell growth, proliferation, differentiation, and survival, mTOR is also thought to play an important role in controlling tumor cell motility, invasion, and metastasis.93 There are increasing reports on the effects of mTOR inhibition on several types of cancer,94–97 indicating the efficacy of mTOR-Is against cancer. For example, immunosuppression with a CNI increases the risk of hepatocellular carcinoma,98,99 whereas an mTOR-I reduces it.99–101 In a human kidney transplantation study, mTOR inhibition protected from non-melanoma skin cancer and solid organ malignancies.102 Additionally, as mentioned above, an mTOR-I may contribute to preventing PTLD associated with EBV.83,84
1.6 |. Adverse effects
The potential beneficial effects of mTOR-Is are offset by potential adverse effects (Table 3B), which are related to the inhibition of mTOR-induced pathways.103
Impaired wound healing is related to several processes (hemostasis, inflammation, proliferation, and remodeling),104 and can be problematic as it is often accompanied by the development of a lymphocele.103,105,106 mTOR inhibition is associated with a higher frequency than is mycophenolate mofetil (MMF),107,108 but the absolute incidence trends lower with everolimus.108 A lower initial dosage of the mTOR-I and/or its delayed introduction may prevent this complication.103–106 Of note, in our pig-to-NHP kidney transplantation model, we have never experienced any wound healing problems. This may, of course, be associated with the fact that, in contrast to patients undergoing organ transplantation, the recipient animals are healthy, with no pre-transplant comorbidities.
There is a higher incidence of proteinuria than when a CNI is administered.109 In kidney xenotransplantation, proteinuria may be problematic even in the absence of an mTOR-I,110 but mTOR inhibition may exacerbate it. Podocyte cytotoxicity111 or reduced albumin endocytosis112 may be seen in mTOR-I-associated proteinuria. Angiotensin-converting enzyme inhibitors and angiotensin II type 1 receptor blockers may contribute to reversing it.112
Oral ulceration/stomatitis103,105 is more frequent than with a CNI.113 Combination therapy with an mTOR-I and mycophenolate mofetil tends to amplify the incidence.106 Although ulceration may be self-limiting,114 high-potency topical steroids (e.g., clobetasol), non-steroidal anti-inflammatory drugs (NSAIDs), and anesthetics (e.g., viscous lidocaine) can reduce pain and promote healing.105,106,115
Pneumonia/interstitial lung disease is a rare, but serious, adverse effect.103,105 Due to differences in dosing regimens and the frequency of radiation therapy, it is more common in the oncology setting than in the transplant setting.105 Early detection is desirable.106 Improvement in pneumonitis is expected with the reduction or discontinuation of the mTOR-I,116,117 or conversion from rapamycin to everolimus.118,119
Anemia is related to decreased iron availability, gastrointestinal absorption, and globin synthesis, and is dose-dependent,120,121 and generally not severe.121 The administration of iron and an agent that stimulates erythropoiesis, for example, erythropoietin, can be beneficial.105,120,121
Dyslipidemia, including hypercholesterolemia and hypertriglyceridemia, is a common side-effect of mTOR-Is,103,105,122 though the increases are rarely more than modest.106 Standard medications, for example, statins and fibrates, that prevent or reduce hypercholesterolemia and hypertriglyceridemia, are recommended to reduce cardiovascular events.103,105,106,122,123
Hyperglycemia or new-onset diabetes mellitus after transplantation (NODAT) has been reported to occur.103,105,124 mTOR inhibition is independently associated with NODAT.125 The mechanism of NODAT caused by an mTOR-I is multifactorial, but ultimately results in insulin resistance and/or defective insulin secretion.126–128 Hyperglycemia needs to be controlled to decrease the risk of cardiovascular disease.129,130 In islet allotransplantation, a regimen that included an mTOR-I without steroids131,132 resulted in prolonged graft function and controlled HbA1c.131,133,134 Thus, hyperglycemia may not result directly from the administration of an mTOR-I.
In transplantation, the trough levels of mTOR-Is are regularly monitored, and close monitoring can reduce the incidence and severity of adverse events,135 and yet ensure prolonged graft survival.
1.7 |. Target trough levels of rapamycin (sirolimus)
In clinical allotransplantation (when rapamycin is combined with cyclosporine and prednisolone), the target level of rapamycin is frequently 5–15 ng/mL.136 When rapamycin is combined with azathioprine and prednisolone, the trough levels may be maintained at 30 ng/mL for the first 2 months posttransplant, then reduced to 15 ng/mL.137,138 In xenotransplantation studies in NHPs, the target levels of rapamycin have generally been lower, for example, 8–20 ng/mL.5,7–9
In human whole blood, 95% of sirolimus is found in the RBCs, and the blood/plasma ratios are approximately 30. Furthermore, 97% of sirolimus is extensively bound to albumin. According to these data, it would be expected that the trough level of sirolimus depends on the RBC count (e.g., in anemia) and albumin concentration (e.g., proteinuria, or when there is suboptimal nutrition) in the recipient. However, in pharmacokinetics studies in kidney allotransplant recipients, the hematocrit, RBC count, and serum albumin levels were excluded from covariates that could affect blood concentrations.139,140 Therefore, factors affecting pharmacokinetics need to be further examined.
2 |. DISCUSSION AND CONCLUSIONS
An mTOR-I may exhibit a variety of beneficial effects, including (i) suppression of effector T cells, B cells, DCs, and antibody production, (ii) increases in Tregs, and (iii) anti-inflammatory, anti-viral, anti-cancer, and anti-growth effects. mTOR-Is have the potential to become valuable immunosuppressive agents in xenotransplantation, although it will be essential to ensure that appropriate blood levels are maintained and adverse effects minimized. It is possible that the adverse effects of mTOR-Is might not be observed in a healthy experimental animal but are more common in a debilitated patient.
There is one additional benefit associated with the administration of mTOR-Is that is gaining increasing attention, and that is their anti-aging effect.141,142 It is now accepted that the effect is not simply associated with a reduced incidence of viral infection and/or cancer, but is a direct effect that slows the aging process. Rapamycin extends lifespan in all tested models from yeast to mammals. It is more effective at slowing down the aging process than reversing it. This effect should be of benefit to the recipients of xenografts.
ACKNOWLEDGMENTS
The authors thank Elizier Katz, MD, for his valuable comments. Work on xenotransplantation in the authors’ laboratory is supported in part by NIH NIAID U19 grant AI090959, and by provision of genetically-modified pigs from Revivicor, Blacksburg, VA.
Funding information
NIH NIAID, Grant/Award Number: AI090959
Abbreviations:
- BKV
BK polyomavirus
- CMV
cytomegalovirus
- CNI
calcineurin inhibitor
- DCs
dendritic cells
- EBV
Epstein-Barr virus
- mTOR
the mechanistic/mammalian target of rapamycin
- mTOR-I
mTOR-inhibitor
- NHP
nonhuman primate
- NODAT
new-onset diabetes mellitus after transplantation
Footnotes
CONFLICT OF INTEREST STATEMENT
D.K.C.C. is a consultant to eGenesis Bio of Cambridge, MA, but the opinions expressed in this article are those of the authors, and do not necessarily reflect those of eGenesis. No other author reports a conflict of interest.
REFERENCES
- 1.Heitman J, Movva NR, Hall MN. Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science (1979). 1991;253(5022):905–909. doi: 10.1126/science.1715094 [DOI] [PubMed] [Google Scholar]
- 2.Chen Y, Zhou X. Research progress of mTOR inhibitors. Eur J Med Chem 2020;208:112820. doi: 10.1016/j.ejmech.2020.112820 [DOI] [PubMed] [Google Scholar]
- 3.Klawitter J, Nashan B, Christians U. Everolimus and sirolimus in transplantation-related but different. Expert Opin Drug Saf 2015;14(7):1055–1070. doi: 10.1517/14740338.2015.1040388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Griffith BP, Goerlich CE, Singh AK, et al. Genetically modified porcine-to-human cardiac xenotransplantation. N Engl J Med 2022;387(1):35–44. doi: 10.1056/nejmoa2201422 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.McGregor CGA, Davies WR, Oi K, et al. Cardiac xenotransplantation: recent preclinical progress with 3-month median survival. J Thorac Cardiovasc Surg 2005;130(3):844.e1–844.e9. doi: 10.1016/j.jtcvs.2005.04.017 [DOI] [PubMed] [Google Scholar]
- 6.Brandl U, Michel S, Erhardt M, et al. Administration of GAS914 in an orthotopic pig-to-baboon heart transplantation model. Xenotransplantation. 2005;12(2):134–141. doi: 10.1111/j.1399-3089.2005.00208.x [DOI] [PubMed] [Google Scholar]
- 7.Byrne GW, Du Z, Sun Z, Asmann YW, McGregor CGA. Changes in cardiac gene expression after pig-to-primate orthotopic xenotransplantation. Xenotransplantation. 2011;18(1):14–27. doi: 10.1111/j.1399-3089.2010.00620.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Iwase H, Liu H, Wijkstrom M, et al. Pig kidney graft survival in a baboon for 136 days: longest life-supporting organ graft survival to date. Xenotransplantation. 2015;22(4):302–309. doi: 10.1111/xen.12174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Iwase H, Hara H, Ezzelarab M, et al. Immunological and physiological observations in baboons with life-supporting genetically engineered pig kidney grafts. Xenotransplantation. 2017;24(2). doi: 10.1111/xen.12293 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Reichart B, Längin M, Radan J, et al. Pig-to-non-human primate heart transplantation: the final step toward clinical xenotransplantation? J Heart Lung Transplant 2020;39(8):751–757. doi: 10.1016/j.healun.2020.05.004 [DOI] [PubMed] [Google Scholar]
- 11.Enderby C, Keller CA. An overview of immunosuppression in solid organ transplantation. Am J Manag Care. 2015;21(1 Suppl):s12–23. [PubMed] [Google Scholar]
- 12.Jabri B, Abadie V. IL-15 functions as a danger signal to regulate tissue-resident T cells and tissue destruction. Nat Rev Immunol 2015;15(12):771–783. doi: 10.1038/nri3919 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nolz JC, Richer MJ. Control of memory CD8+ T cell longevity and effector functions by IL-15. Mol Immunol 2020;117:180–188. doi: 10.1016/j.molimm.2019.11.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mukherjee S, Mukherjee U. A comprehensive review of immunosuppression used for liver transplantation. J Transplant 2009;2009:1–20. doi: 10.1155/2009/701464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Neuhaus P, Klupp J, Langrehr JM. mTOR inhibitors: an overview. Liver Transpl 2001;7(6):473–484. doi: 10.1053/jlts.2001.24645 [DOI] [PubMed] [Google Scholar]
- 16.Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124(3):471–484. doi: 10.1016/j.cell.2006.01. 016 [DOI] [PubMed] [Google Scholar]
- 17.Sarbassov DD, Ali SM, Sengupta S, et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell. 2006;22(2):159–168. doi: 10.1016/j.molcel.2006.03.029 [DOI] [PubMed] [Google Scholar]
- 18.Lamming DW, Ye L, Katajisto P, et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science (1979). 2012;335(6076):1638–1643. doi: 10.1126/science.1215135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lee K, Heffington L, Jellusova J, et al. Requirement for Rictor in homeostasis and function of mature B lymphoid cells Key Points. Blood. 2013;122(14):2369–2379. doi: 10.1182/blood-2013-01 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Keating R, Hertz T, Wehenkel M, et al. The kinase mTOR modulates the antibody response to provide cross-protective immunity to lethal infection with influenza virus. Nat Immunol 2013;14(12):1266–1276. doi: 10.1038/ni.2741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Limon JJ, So L, Jellbauer S, et al. mTOR kinase inhibitors promote antibody class switching via mTORC2 inhibition. Proc Natl Acad Sci U S A 2014;111(47):E5076–E5085. doi: 10.1073/pnas.1407104111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Netti GS, Infante B, Troise D, et al. mTOR inhibitors improve both humoral and cellular response to SARS-CoV-2 messenger RNA BNT16b2 vaccine in kidney transplant recipients. Am J Transplant 2022;22(5):1475–1482. doi: 10.1111/ajt.16958 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Berger SP, Sommerer C, Witzke O, et al. Two-year outcomes in de novo renal transplant recipients receiving everolimus-facilitated calcineurin inhibitor reduction regimen from the TRANSFORM study. Am J Transplant 2019;19(11):3018–3034. doi: 10.1111/ajt.15480 [DOI] [PubMed] [Google Scholar]
- 24.Stallone G, Infante B, Lorenzo A di, Rascio F, Zaza G, Grandaliano G. mTOR inhibitors effects on regulatory T cells and on dendritic cells. J Transl Med 2016;14(1):152. doi: 10.1186/s12967-016-0916-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hackstein H, Taner T, Logar AJ, Thomson AW. Rapamycin inhibits macropinocytosis and mannose receptor – mediated endocytosis by bone marrow – derived dendritic cells. Blood. 2002;100(3):1084–1087. doi: 10.1182/blood.V100.3.1084 [DOI] [PubMed] [Google Scholar]
- 26.Monti P, Mercalli A, Leone BE, Valerio DC, Allavena P, Piemonti L. Rapamycin impairs antigen uptake of human. Transplantation 2003;75:137–145. [DOI] [PubMed] [Google Scholar]
- 27.Hackstein H, Taner T, Zahorchak AF, et al. Rapamycin inhibits IL-4-induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo. Blood. 2003;101(11):4457–4463. doi: 10.1182/blood-2002-11-3370 [DOI] [PubMed] [Google Scholar]
- 28.Matsue H, Yang C, Matsue K, Edelbaum D, Mummert M, Takashima A. Contrasting impacts of immunosuppressive agents (rapamycin, FK506, cyclosporin A, and dexamethasone) on bidirectional dendritic cell-T cell interaction during antigen presentation. J Immunol 2002;169(7):3555–3564. doi: 10.4049/jimmunol.169.7.3555 [DOI] [PubMed] [Google Scholar]
- 29.Siu JHY, Surendrakumar V, Richards JA, Pettigrew GJ. T cell allorecognition pathways in solid organ transplantation. Front Immunol 2018;9(NOV). doi: 10.3389/fimmu.2018.02548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Taner T, Hackstein H, Wang Z, Morelli AE, Thomson AW. Rapamycin-treated, alloantigen-pulsed host dendritic cells induce Ag-specific T cell regulation and prolong graft survival. Am J Transplant 2005;5(2):228–236. doi: 10.1046/j.1600-6143.2004.00673.x [DOI] [PubMed] [Google Scholar]
- 31.Wenzel M, Haffer H, Wang P, et al. Influence of an early application of mammalian target of rapamycin inhibitors everolimus and sirolimus on acute vascular inflammatory responses after ischemia-reperfusion injury. Exp Clin Transplant 2021;19(1):50–57. doi: 10.6002/ect.2020.0111 [DOI] [PubMed] [Google Scholar]
- 32.Ko JH, Yoon SO, Lee HJ, Oh JY. Rapamycin regulates macrophage activation by inhibiting NLRP3 inflammasome-p38 MAPK-NFκB pathways in autophagy- and p62-dependent manners. Oncotarget 2017;8(25):40817–40831. doi: 10.18632/oncotarget.17256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hou H, Miao J, Cao R, et al. Rapamycin ameliorates experimental autoimmune encephalomyelitis by suppressing the mTOR-STAT3 pathway. Neurochem Res 2017;42(10):2831–2840. doi: 10.1007/s11064-017-2296-7 [DOI] [PubMed] [Google Scholar]
- 34.Maenaka A, Kenta I, Ota A, et al. Interferon-γ-induced HLA Class II expression on endothelial cells is decreased by inhibition of mTOR and HMG-CoA reductase. FEBS Open Bio 2020;10(5):927–936. doi: 10.1002/2211-5463.12854 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ezzelarab MB, Ekser B, Azimzadeh A, et al. Systemic inflammation in xenograft recipients precedes activation of coagulation. Xenotransplantation 2015;22(1):32–47. doi: 10.1111/xen.12133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhang G, Iwase H, Li Q, et al. The role of interleukin-6 (IL-6) in the systemic inflammatory response in xenograft recipients and in pig kidney xenograft failure. Front Immunol 2021;12. doi: 10.3389/fimmu.2021.788949 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhang G, Iwase H, Wang L, et al. Is interleukin-6 receptor blockade (tocilizumab) beneficial or detrimental to pig-to-baboon organ xenotransplantation? Am J Transplant 2020;20(4):999–1013. doi: 10.1111/ajt.15712 [DOI] [PubMed] [Google Scholar]
- 38.Li S, Xu H, Kirk AD. Modulation of xenogeneic T-cell proliferation by B7 and mTOR blockade of T cells and porcine endothelial cells. Transplantation. 2022;106(5):950–962. doi: 10.1097/TP.0000000000003920 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ezzelarab MB, Cooper DKC. Systemic inflammation in xenograft recipients (SIXR): a new paradigm in pig-to-primate xenotransplantation? Int J Surg 2015;23:301–305. doi: 10.1016/j.ijsu.2015.07.643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li J, Hara H, Wang Y, Esmon C, Cooper DKC, Iwase H. Evidence for the important role of inflammation in xenotransplantation. J Inflamm 2019;16(1). doi: 10.1186/s12950-019-0213-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775–787. doi: 10.1016/j.cell.2008.05.009 [DOI] [PubMed] [Google Scholar]
- 42.Shan J, Guo Y, Luo L, et al. Do CD4+Foxp3+ Treg cells correlate with transplant outcomes: a systematic review on recipients of solid organ transplantation. Cell Immunol 2011;270(1):5–12. doi: 10.1016/j.cellimm.2011.05.006 [DOI] [PubMed] [Google Scholar]
- 43.Shan J, Feng L, Li Y, Sun G, Chen X, Chen P. The effects of rapamycin on regulatory T cells: its potential time-dependent role in inducing transplant tolerance. Immunol Lett 2015;162(1):74–86. doi: 10.1016/j.imlet.2014.07.006 [DOI] [PubMed] [Google Scholar]
- 44.Malek TR, Castro I. Interleukin-2 receptor signaling: at the interface between tolerance and immunity. Immunity 2010;33(2):153–165. doi: 10.1016/j.immuni.2010.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Fruman DA, Klee CB, Bierer BE, Burakoff SJ. Calcineurin phosphatase activity in T lymphocytes is inhibited by FK 506 and cyclosporin A. Proc Natl Acad Sci U S A 1992;89(9):3686–3690. doi: 10.1073/pnas.89.9.3686 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Battaglia M, Stabilini A, Roncarolo MG. Rapamycin selectively expands CD4+CD25+FoxP3 + regulatory T cells. Blood. 2005;105(12):4743–4748. doi: 10.1182/blood-2004-10-3932 [DOI] [PubMed] [Google Scholar]
- 47.Kang J, Huddleston SJ, Fraser JM, Khoruts A. De novo induction of antigen-specific CD4+CD25+Foxp3+ regulatory T cells in vivo following systemic antigen administration accompanied by blockade of mTOR. J Leukoc Biol 2008;83(5):1230–1239. doi: 10.1189/jlb.1207851 [DOI] [PubMed] [Google Scholar]
- 48.Battaglia M, Stabilini A, Tresoldi E. Expanding human T regulatory cells with the mTOR-inhibitor rapamycin. Methods Mol Biol 2012;821:279–293. doi: 10.1007/978-1-61779-430-8_17 [DOI] [PubMed] [Google Scholar]
- 49.Janyst M, Kaleta B, Janyst K, Zagożdżon R, Kozlowska E, Lasek W. Comparative study of immunomodulatory agents to induce human T regulatory (Treg) cells: preferential Treg-stimulatory effect of prednisolone and rapamycin. Arch Immunol Ther Exp (Warsz). 2020;68(4). doi: 10.1007/s00005-020-00582-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gabryšová L, Christensen JR, Wu X, Kissenpfennig A, Malissen B, O’Garra A. Integrated T-cell receptor and costimulatory signals determine TGF-β-dependent differentiation and maintenance of Foxp3+ regulatory T cells. Eur J Immunol 2011;41(5):1242–1248. doi: 10.1002/eji.201041073 [DOI] [PubMed] [Google Scholar]
- 51.Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J, Kaupper T, Roncarolo MG. Rapamycin promotes expansion of functional CD4 + CD25 + FOXP3 + regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol 2006;177(12):8338–8347. doi: 10.4049/jimmunol.177.12.8338 [DOI] [PubMed] [Google Scholar]
- 52.Singh AK, Horvath KA, Mohiuddin MM. Rapamycin promotes the enrichment of CD4+CD25hiFoxP3+ T regulatory cells from naïve CD4+ T cells of baboon that suppress antiporcine xenogenic response in vitro. Transplant Proc 2009;41(1):418–421. doi: 10.1016/j.transproceed.2008.10.079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang T, Pierson RN, Azimzadeh AM. Update on CD40 and CD154 blockade in transplant models. Immunotherapy 2015;7(8):899–911. doi: 10.2217/IMT.15.54 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pinelli DF, Ford ML. Novel insights into anti-CD40/CD154 immunotherapy in transplant tolerance. Immunotherapy 2015;7(4):399–410. doi: 10.2217/imt.15.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhao Y, Hu W, Chen P, et al. Immunosuppressive and metabolic agents that influence allo- and xenograft survival by in vivo expansion of T regulatory cells. Xenotransplantation. 2020;27(6). doi: 10.1111/xen.12640 [DOI] [PubMed] [Google Scholar]
- 56.Ferrer IR, Wagener ME, Song M, Kirk AD, Larsen CP, Ford ML. Antigen-specific induced Foxp3 + regulatory T cells are generated following CD40/CD154 blockade. Proc Natl Acad Sci U S A 2011;108(51):20701–20706. doi: 10.1073/pnas.1105500108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Pinelli DF, Wagener ME, Liu D, et al. An anti-CD154 domain antibody prolongs graft survival and induces Foxp3+ iTreg in the absence and presence of CTLA-4 Ig. Am J Transplant 2013;13(11):3021–3030. doi: 10.1111/ajt.12417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yamada A, Sayegh MH. The CD154-CD40 costimulatory pathway in transplantation. Transplantation. 2002;73(1 Suppl):S36–S39. doi: 10.1097/00007890-200201151-00012 [DOI] [PubMed] [Google Scholar]
- 59.Oura T, Hotta K, Lei J, et al. Immunosuppression with CD40 costimulatory blockade plus rapamycin for simultaneous islet – kidney transplantation in nonhuman primates. Am J Transplant 2017;17(3):646–656. doi: 10.1111/ajt.13999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kotton CN, Kumar D, Caliendo AM, et al. The third international consensus guidelines on the management of cytomegalovirus in solid-organ transplantation. Transplantation. 2018;102(6):900–931. doi: 10.1097/TP.0000000000002191 [DOI] [PubMed] [Google Scholar]
- 61.Stern M, Hirsch H, Cusini A, et al. Cytomegalovirus serology and replication remain associated with solid organ graft rejection and graft loss in the era of prophylactic treatment. Transplantation. 2014;98(9):1013–1018. doi: 10.1097/TP.0000000000000160 [DOI] [PubMed] [Google Scholar]
- 62.Mueller NJ, Kuwaki K, Dor FJMF, et al. Reduction of consumptive coagulopathy using porcine cytomegalovirus-free cardiac porcine grafts in pig-to-primate xenotransplantation. Transplantation. 2004;78(10):1449–1453. doi: 10.1097/01.TP.0000141361.68446.1F [DOI] [PubMed] [Google Scholar]
- 63.Mueller NJ, Barth RN, Yamamoto S, et al. Activation of cytomegalovirus in pig-to-primate organ xenotransplantation. J Virol 2002;76(10):4734–4740. doi: 10.1128/jvi.76.10.4734-4740.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mueller NJ, Sulling K, Gollackner B, et al. Reduced efficacy of ganciclovir against porcine and baboon cytomegalovirus in pig-to-baboon xenotransplantation. Am J Transplant 2003;3(9):1057–1064. doi: 10.1034/j.1600-6143.2003.00192.x [DOI] [PubMed] [Google Scholar]
- 65.Gollackner B, Mueller NJ, Houser S, et al. Porcine cytomegalovirus and coagulopathy in pig-to-primate xenotransplantation. Transplantation. 2003;75(11):1841–1847. doi: 10.1097/01.TP.0000065806.90840.C1 [DOI] [PubMed] [Google Scholar]
- 66.Silva JT, Fernández-Ruiz M, Aguado JM. Prevention and therapy of viral infections in patients with solid organ transplantation. Enferm Infecc Microbiol Clin 2021;39(2):87–97. doi: 10.1016/j.eimc.2020.01.021 [DOI] [PubMed] [Google Scholar]
- 67.Prockop SE, Vatsayan A. Epstein-Barr virus lymphoproliferative disease after solid organ transplantation. Cytotherapy. 2017;19(11):1270–1283. doi: 10.1016/j.jcyt.2017.08.010 [DOI] [PubMed] [Google Scholar]
- 68.Shen CL, Wu BS, Lien TJ, Yang AH, Yang CY. BK polyomavirus nephropathy in kidney transplantation: balancing rejection and infection. Viruses. 2021;13(3). doi: 10.3390/v13030487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hui DS, E I Azhar, Madani TA, et al. The continuing 2019-nCoV epidemic threat of novel coronaviruses to global health − The latest 2019 novel coronavirus outbreak in Wuhan, China. Int J Infect Dis 2020;91:264–266. doi: 10.1016/j.ijid.2020.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lu H, Stratton CW, Tang YW. Outbreak of pneumonia of unknown etiology in Wuhan, China: the mystery and the miracle. J Med Virol 2020;92(4):401–402. doi: 10.1002/jmv.25678 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhu N, Zhang D, Wang W, et al. A novel coronavirus from patients with pneumonia in China, 2019. N EnglJ Med 2020;382(8):727–733. doi: 10.1056/nejmoa2001017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Knight RJ, Graviss EA, Nguyen DT, et al. Conversion from tacrolimus-mycophenolate mofetil to tacrolimus-mTOR immunosuppression after kidney-pancreas transplantation reduces the incidence of both BK and CMV viremia. Clin Transplant 2018;32(6). doi: 10.1111/ctr.13265 [DOI] [PubMed] [Google Scholar]
- 73.Yamada K, Tasaki M, Sekijima M, et al. Porcine cytomegalovirus infection is associated with early rejection of kidney grafts in a pig to baboon xenotransplantation model. Transplantation 2014;98(4):411–418. doi: 10.1097/TP.0000000000000232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Denner J Reduction of the survival time of pig xenotransplants by porcine cytomegalovirus. Virol J 2018;15(1). doi: 10.1186/s12985-018-1088-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Denner J, Längin M, Reichart B, et al. Impact of porcine cytomegalovirus on long-term orthotopic cardiac xenotransplant survival. Sci Rep 2020;10(1). doi: 10.1038/s41598-020-73150-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kotton CN. CMV: prevention, diagnosis and therapy. Am J Transplant 2013;13(Suppl 3):24–40. doi: 10.1111/ajt.12006 [DOI] [PubMed] [Google Scholar]
- 77.Kaminski H, Kamar N, Thaunat O, et al. Incidence of cytomegalovirus infection in seropositive kidney transplant recipients treated with everolimus: a randomized, open-label, multicenter phase 4 trial. Am J Transplant 2022;22(5):1430–1441. doi: 10.1111/ajt.16946 [DOI] [PubMed] [Google Scholar]
- 78.Nashan B, Gaston R, Emery V, et al. Review of cytomegalovirus infection findings with mammalian target of rapamycin inhibitor-based immunosuppressive therapy in de novo renal transplant recipients. Transplantation. 2012;93(11):1075–1085. doi: 10.1097/TP.0b013e31824810e6 [DOI] [PubMed] [Google Scholar]
- 79.Tedesco-Silva H, Pascual J, Viklicky O, et al. Safety of everolimus with reduced calcineurin inhibitor exposure in de novo kidney transplants: an analysis from the randomized transform study. Transplantation. 2019;103(9):1953–1963. doi: 10.1097/TP.0000000000002626 [DOI] [PubMed] [Google Scholar]
- 80.Radtke J, Dietze N, Spetzler VN, et al. Fewer cytomegalovirus complications after kidney transplantation by de novo use of mTOR inhibitors in comparison to mycophenolic acid. Transpl Infect Dis 2016;18(1):79–88. doi: 10.1111/tid.12494 [DOI] [PubMed] [Google Scholar]
- 81.Lurain NS, Chou S. Antiviral drug resistance of human cytomegalovirus. Clin Microbiol Rev 2010;23(4):689–712. doi: 10.1128/CMR.00009-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kaminski H, Marseres G, Yared N, et al. mTOR inhibitors prevent CMV infection through the restoration of functional ab and gd T cells in kidney transplantation. J Am Soc Nephrol 2022;33(1):121–137. doi: 10.1681/ASN.2020121753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wowro SJ, Schmitt KRL, Tong G, Berger F, Schubert S. Effects of mTOR and calcineurin inhibitors combined therapy in Epstein-Barr virus positive and negative Burkitt lymphoma cells. Int Immunopharmacol 2016;30:9–17. doi: 10.1016/j.intimp.2015.11.020 [DOI] [PubMed] [Google Scholar]
- 84.Petrara MR, Serraino D, di Bella C, et al. Immune activation, immune senescence and levels of Epstein Barr Virus in kidney transplant patients: impact of mTOR inhibitors. Cancer Lett 2020;469:323–331. doi: 10.1016/j.canlet.2019.10.045 [DOI] [PubMed] [Google Scholar]
- 85.BK virus. Am J Transplant 2004;4(10 Suppl.):89–91. doi: 10.1111/j.1600-6135.2004.00730.x [DOI] [PubMed] [Google Scholar]
- 86.Yen CL, Tian YC, Wu HH, et al. Conversion to mTOR-inhibitors with calcineurin inhibitor elimination or minimization reduces urinary polyomavirus BK load in kidney transplant recipients. J Formos Med Assoc 2016;115(7):539–546. doi: 10.1016/j.jfma.2016.01.008 [DOI] [PubMed] [Google Scholar]
- 87.Mashayekhi-Sardoo H, Hosseinjani H. A new application of mTOR inhibitor drugs as potential therapeutic agents for COVID-19. J Basic Clin Physiol Pharmacol 2022;33(1):17–25. doi: 10.1515/jbcpp-2020-0495 [DOI] [PubMed] [Google Scholar]
- 88.Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168(6):960–976. doi: 10.1016/j.cell.2017.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chung J, Kuo CJ, Crabtree GR, Blenis J. Rapamycin-FKBP specifically blocks growth-dependent activation of and signaling by the 70 kd S6 protein kinases. Cell. 1992;69(7):1227–1236. doi: 10.1016/0092-8674(92)90643-Q [DOI] [PubMed] [Google Scholar]
- 90.Manning BD. Game of TOR – The target of rapamycin rules four kingdoms. N Engl J Med 2017;377(13):1297–1299. doi: 10.1056/nejmcibr1709384 [DOI] [PubMed] [Google Scholar]
- 91.Tanabe T, Watanabe H, Shah JA, et al. Role of intrinsic (graft) versus extrinsic (host) factors in the growth of transplanted organs following allogeneic and xenogeneic transplantation. Am J Transplant 2017;17(7):1778–1790. doi: 10.1111/ajt.14210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Längin M, Mayr T, Reichart B, et al. Consistent success in life-supporting porcine cardiac xenotransplantation. Nature 2018;564(7736):430–433. doi: 10.1038/s41586-018-0765-z [DOI] [PubMed] [Google Scholar]
- 93.Huang S, Zhou H. Role of mTOR signaling in tumor cell motility, invasion and metastasis. Curr Protein Pept Sci 2011;12(1):30–42. doi: 10.2174/138920311795659407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Watanabe R, Wei L, Huang J. mTOR signaling, function, novel inhibitors, and therapeutic targets. J Nucl Med 2011;52(4):497–500. doi: 10.2967/jnumed.111.089623 [DOI] [PubMed] [Google Scholar]
- 95.Ciołczyk-Wierzbicka D, Gil D, Zarzycka M, Laidler P. mTOR inhibitor everolimus reduces invasiveness of melanoma cells. Hum Cell 2020;33(1):88–97. doi: 10.1007/s13577-019-00270-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kim JO, Kim KH, Song IS, et al. Potentiation of the anticancer effects of everolimus using a dual mTORC1/2 inhibitor in hepatocellular carcinoma cells. Oncotarget 2017;8(2):2936–2948. doi: 10.18632/oncotarget.13808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Pal SK, Quinn DI. Differentiating mTOR inhibitors in renal cell carcinoma. Cancer Treat Rev 2013;39(7):709–719. doi: 10.1016/j.ctrv.2012.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Vivarelli M, Cucchetti A, Barba G la, et al. Liver transplantation for hepatocellular carcinoma under calcineurin inhibitors: reassessment of risk factors for tumor recurrence. Ann Surg 2008;248(5):857–862. doi: 10.1097/SLA.0b013e3181896278 [DOI] [PubMed] [Google Scholar]
- 99.Vivarelli M, Dazzi A, Zanello M, et al. Effect of different immunosuppressive schedules on recurrence-free survival after liver transplantation for hepatocellular carcinoma. Transplantation 2010;89(2):227–231. doi: 10.1097/TP.0b013e3181c3c540 [DOI] [PubMed] [Google Scholar]
- 100.Duvoux C, Toso C. mTOR inhibitor therapy: does it prevent HCC recurrence after liver transplantation? Transplant Rev 2015;29(3):168–174. doi: 10.1016/j.trre.2015.02.003 [DOI] [PubMed] [Google Scholar]
- 101.Schnitzbauer AA, Filmann N, Adam R, et al. mTOR inhibition is most beneficial after liver transplantation for hepatocellular carcinoma in patients with active tumors. Ann Surg 2020;272(5):855–862. doi: 10.1097/SLA.0000000000004280 [DOI] [PubMed] [Google Scholar]
- 102.Santos AH, Chen C, Leghrouz MA, Bueno EP, Lee JJ, Wen X. Association of HLA mismatch and mTOR inhibitor regimens with malignancy and mortality after kidney transplantation. Transpl Immunol 2021;66. doi: 10.1016/j.trim.2021.101391 [DOI] [PubMed] [Google Scholar]
- 103.Nguyen LS, Vautier M, Allenbach Y, et al. Sirolimus and mTOR inhibitors: a review of side effects and specific management in solid organ transplantation. Drug Saf 2019;42(7):813–825. doi: 10.1007/s40264-019-00810-9 [DOI] [PubMed] [Google Scholar]
- 104.Bootun R Effects of immunosuppressive therapy on wound healing. Int Wound J 2013;10(1):98–104. doi: 10.1111/j.1742-481X.2012.00950.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kaplan B, Qazi Y, Wellen JR. Strategies for the management of adverse events associated with mTOR inhibitors. Transplant Rev 2014;28(3):126–133. doi: 10.1016/j.trre.2014.03.002 [DOI] [PubMed] [Google Scholar]
- 106.Pallet N, Legendre C. Adverse events associated with mTOR inhibitors. Expert Opin Drug Saf. 2013;12(2):177–186. doi: 10.1517/14740338.2013.752814 [DOI] [PubMed] [Google Scholar]
- 107.Ueno P, Felipe C, Ferreira A, et al. Wound healing complications in kidney transplant recipients receiving everolimus. Transplantation 2017;101(4):844–850. doi: 10.1097/TP.0000000000001392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Citterio F, Henry M, Kim DY, et al. Wound healing adverse events in kidney transplant recipients receiving everolimus with reduced calcineurin inhibitor exposure or current standard-of-care: insights from the 24-month TRANSFORM study. Expert Opin Drug Saf 2020;19(10):1339–1348. doi: 10.1080/14740338.2020.1792441 [DOI] [PubMed] [Google Scholar]
- 109.Diekmann F, Andrés A, Oppenheimer F. mTOR inhibitor-associated proteinuria in kidney transplant recipients. Transplant Rev 2012;26(1):27–29. doi: 10.1016/j.trre.2011.10.003 [DOI] [PubMed] [Google Scholar]
- 110.Shah JA, Lanaspa MA, Tanabe T, Watanabe H, Johnson RJ, Yamada K. Remaining physiological barriers in porcine kidney xenotransplantation: potential pathways behind proteinuria as well as factors related to growth discrepancies following pig-to-kidney xenotransplantation. J Immunol Res 2018;2018. doi: 10.1155/2018/6413012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Letavernier E, Bruneval P, Vandermeersch S, et al. Sirolimus interacts with pathways essential for podocyte integrity. Nephrol Dial Transplant 2009;24(2):630–638. doi: 10.1093/ndt/gfn574 [DOI] [PubMed] [Google Scholar]
- 112.Oroszlán M, Bieri M, Ligeti N, et al. Sirolimus and everolimus reduce albumin endocytosis in proximal tubule cells via an angiotensin II-dependent pathway. Transpl Immunol 2010;23(3):125–132. doi: 10.1016/j.trim.2010.05.003 [DOI] [PubMed] [Google Scholar]
- 113.Liu J, Liu D, Li J, et al. Efficacy and safety of everolimus for maintenance immunosuppression of kidney transplantation: a meta-analysis of randomized controlled trials. PLoS One 2017;12(1). doi: 10.1371/journal.pone.0170246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Campistol JM, de Fijter JW, Flechner SM, Langone A, Morelon E, Stockfleth E. mTOR inhibitor-associated dermatologic and mucosal problems. Clin Transplant 2010;24(2):149–156. doi: 10.1111/j.1399-0012.2010.01232.x [DOI] [PubMed] [Google Scholar]
- 115.Chuang P, Langone AJ. Clobetasol ameliorates aphthous ulceration in renal transplant patients on sirolimus. Am J Transplant 2007;7(3):714–717. doi: 10.1111/j.1600-6143.2006.01678.x [DOI] [PubMed] [Google Scholar]
- 116.Pham PTT, Pham PCT, Danovitch GM, et al. Sirolimus-associated pulmonary toxicity. Transplantation 2004;77(8):1215–1220. doi: 10.1097/01.TP.0000118413.92211.B6 [DOI] [PubMed] [Google Scholar]
- 117.Alexandru S, Ortiz A, Baldovi S, et al. Severe everolimus-associated pneumonitis in a renal transplant recipient. Nephrol Dial Transplant 2008;23(10):3353–3355. doi: 10.1093/ndt/gfn401 [DOI] [PubMed] [Google Scholar]
- 118.Rehm B, Keller F, Mayer J, Stracke S. Resolution of sirolimus-induced pneumonitis after conversion to everolimus. Transplant Proc. 2006;38(3):711–713. doi: 10.1016/j.transproceed.2006.01.052 [DOI] [PubMed] [Google Scholar]
- 119.Alkhunaizi AM, Al-Khouzaie TH, Alsagheir AI. Sirolimus-induced interstitial lung disease and resolution after conversion to everolimus. Respir Med Case Rep 2020;30. doi: 10.1016/j.rmcr.2020.101109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Fishbane S, Cohen DJ, Coyne DW, Djamali A, Singh AK, Wish JB. Post-transplant anemia: the role of sirolimus. Kidney Int 2009;76(4):376–382. doi: 10.1038/ki.2009.231 [DOI] [PubMed] [Google Scholar]
- 121.Sofroniadou S, Kassimatis T, Goldsmith D. Anaemia, microcytosis and sirolimus-is iron the missing link? Nephrol Dial Transplant 2010;25(5):1667–1675. doi: 10.1093/ndt/gfp674 [DOI] [PubMed] [Google Scholar]
- 122.Kurdi A, Martinet W, de Meyer GRY. mTOR inhibition and cardiovascular diseases: dyslipidemia and atherosclerosis. Transplantation 2018;102(2S):S44–S46. doi: 10.1097/TP.0000000000001693 [DOI] [PubMed] [Google Scholar]
- 123.Agarwal A, Prasad GVR. Post-transplant dyslipidemia: mechanisms, diagnosis and management. World J Transplant 2016;6(1):125. doi: 10.5500/wjt.v6.i1.125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Pham PTT, Pham PCT, Lipshutz GS, Wilkinson AH. New onset diabetes mellitus after solid organ transplantation. Endocrinol Metab Clin North Am 2007;36(4):873–890. doi: 10.1016/j.ecl.2007.07.007 [DOI] [PubMed] [Google Scholar]
- 125.Johnston O, Rose CL, Webster AC, Gill JS. Sirolimus is associated with new-onset diabetes in kidney transplant recipients. J Am Soc Nephrol 2008;19(7):1411–1418. doi: 10.1681/ASN.2007111202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dong M, Parsaik AK, Eberhardt NL, Basu A, Cosio FG, Kudva YC. Cellular and physiological mechanisms of new-onset diabetes mellitus after solid organ transplantation. Diabet Med 2012;29(7). doi: 10.1111/j.1464-5491.2012.03617.x [DOI] [PubMed] [Google Scholar]
- 127.Vergès B, Cariou B. mTOR inhibitors and diabetes. Diabetes Res Clin Pract 2015;110(2):101–108. doi: 10.1016/j.diabres.2015.09.014 [DOI] [PubMed] [Google Scholar]
- 128.di Paolo S, Teutonico A, Leogrande D, Capobianco C, Schena PF. Chronic inhibition of mammalian target of rapamycin signaling downregulates insulin receptor substrates 1 and 2 and AKT activation: a crossroad between cancer and diabetes? J Am Soc Nephrol 2006;17(8):2236–2244. doi: 10.1681/ASN.2006030196 [DOI] [PubMed] [Google Scholar]
- 129.Cosio FG, Hickson LJ, Griffin MD, Stegall MD, Kudva Y. Patient survival and cardiovascular risk after kidney transplantation: the challenge of diabetes. Am J Transplant 2008;8(3):593–599. doi: 10.1111/j.1600-6143.2007.02101.x [DOI] [PubMed] [Google Scholar]
- 130.Maskey R New-Onset Diabetes after Transplant (NODAT). Kathmandu Univ Med J (KUMJ) 2014;12(48):301–305. doi: 10.3126/kumj.v12i4.13740 [DOI] [PubMed] [Google Scholar]
- 131.Berney T, Andres A, Toso C, Majno P, Squifflet JP. mTOR inhibition and clinical transplantation: pancreas and islet. Transplantation 2018;102(2S Suppl 1):S30–S31. doi: 10.1097/TP.0000000000001700 [DOI] [PubMed] [Google Scholar]
- 132.Shapiro AM, Lakey JR, Ryan EA, et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med 2000;343(4):230–238. doi: 10.1056/NEJM200007273430401 [DOI] [PubMed] [Google Scholar]
- 133.Lablanche S, Borot S, Wojtusciszyn A, et al. Five-year metabolic, functional, and safety results of patients with type 1 diabetes transplanted with allogenic islets within the Swiss-French GRAGIL network. Diabetes Care 2015;38(9):1714–1722. doi: 10.2337/dc15-0094 [DOI] [PubMed] [Google Scholar]
- 134.Hering BJ, Clarke WR, Bridges ND, et al. Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia. Diabetes Care 2016;39(7):1230–1240. doi: 10.2337/dc15-1988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Ventura-Aguiar P, Campistol JM, Diekmann F. Safety of mTOR inhibitors in adult solid organ transplantation. Expert Opin Drug Saf 2016;15(3):303–319. doi: 10.1517/14740338.2016.1132698 [DOI] [PubMed] [Google Scholar]
- 136.Kahan BD, Napoli KL, Kelly PA, et al. Therapeutic drug monitoring of sirolimus: correlations with efficacy and toxicity. Clin Transplant 2000;14(2):97–109. doi: 10.1034/j.1399-0012.2000.140201.x [DOI] [PubMed] [Google Scholar]
- 137.Groth CG, Bäckman L, Morales JM, et al. Sirolimus (rapamycin)-based therapy in human renal transplantation: similar efficacy and different toxicity compared with cyclosporine. Sirolimus European Renal Transplant Study Group. Transplantation 1999;67(7):1036–1042. doi: 10.1097/00007890-199904150-00017 [DOI] [PubMed] [Google Scholar]
- 138.Kreis H, Cisterne JM, Land W, et al. Sirolimus in association with mycophenolate mofetil induction for the prevention of acute graft rejection in renal allograft recipients. Transplantation 2000;69(7):1252–1260. doi: 10.1097/00007890-200004150-00009 [DOI] [PubMed] [Google Scholar]
- 139.Djebli N, Rousseau A, Hoizey G, et al. Sirolimus population pharmacokinetic/pharmacogenetic analysis and bayesian modelling in kidney transplant recipients. Clin Pharmacokinet 2006;45(11):1135–1148. doi: 10.2165/00003088-200645110-00007 [DOI] [PubMed] [Google Scholar]
- 140.Dansirikul C, Morris RG, Tett SE, Duffull SB. A Bayesian approach for population pharmacokinetic modelling of sirolimus. Br J Clin Pharmacol 2006;62(4):420–434. doi: 10.1111/j.1365-2125.2005.02533.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Harrison DE, Strong R, Sharp ZD, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 2009;460(7253):392–395. doi: 10.1038/nature08221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Blagosklonny M v. Rapamycin for longevity: opinion article. Aging 2019;11(19):8048–8067. doi: 10.18632/aging.102355 [DOI] [PMC free article] [PubMed] [Google Scholar]