Minireviews Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Transplantation. Sep 10, 2018; 8(5): 150-155
Published online Sep 10, 2018. doi: 10.5500/wjt.v8.i5.150
Introduction of everolimus in kidney transplant recipients at a late posttransplant stage
Junji Uchida, Tomoaki Iwai, Tatsuya Nakatani, Department of Urology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
ORCID number: Junji Uchida (0000-0002-0113-8058); Tomoaki Iwai (0000-0003-2021-0673); Tatsuya Nakatani (0000-0002-0753-1571).
Author contributions: Uchida J, Iwai T and Nakatani T contributed equally to this work, generated the tables and wrote the manuscript.
Conflict-of-interest statement: We have no personal or financial interests to declare, and we have no financial support from an industry source for the current manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Junji Uchida, MD, PhD, Associate Professor, Department of Urology, Osaka City University Graduate School of Medicine, 1-4-3, Abeno-ku, Asahi-machi, Osaka 545-8585, Japan. m9492120@msic.med.osaka-cu.ac.jp
Telephone: +81-6-66453857 Fax: +81-6-66474426
Received: May 21, 2018
Peer-review started: May 21, 2018
First decision: June 6, 2018
Revised: June 23, 2018
Accepted: June 27, 2018
Article in press: June 28, 2018
Published online: September 10, 2018

Abstract

This minireview focuses on the current knowledge about the introduction of everolimus (EVL), a mammalian target of rapamycin inhibitor, with calcineurin inhibitor (CNI) elimination or minimization in kidney transplant recipients at a late posttransplant stage. Within, we have summarized two major clinical trials, ASCERTAIN and APOLLO, and seven other retrospective or nonrandomized studies. In the open-label multicenter ASCERTAIN study, the estimated glomerular filtration rate (eGFR) at 24 mo after conversion was not significantly different between three groups-EVL with CNI elimination, CNI minimization and continued CNI unchanged-at a mean of 5.4 years after transplantation. However, recipients with baseline creatinine clearance higher than 50 mL/min had a greater increase in measured GFR after CNI elimination. In the open-label multicenter APOLLO study, adjusted eGFR within the on-treatment population was significantly higher in the EVL continuation group than in the CNI continuation group at 12 mo after conversion at a mean of 7 years posttransplantation. Other studies on recipients without adverse events and already having satisfactory renal function showed favorable graft function by EVL late-induction with CNI elimination or reduction. These studies showed that chronic allograft nephropathy, CNI nephrotoxicity, CNI arteriolopathy, cancer and viral infection (especially cytomegalovirus infection) may be good indications for late conversion to EVL.

Key Words: Kidney transplantation, Everolimus, mTOR inhibitor, Late conversion, Calcineurine inhibitor

Core tip: Current immunosuppressive protocols consisting of calcineurin inhibitors (CNIs) and mycophenolate mofetil have improved short-term graft survival. However, improvements in long-term graft survival are restricted by nephrotoxicity associated with CNI. Everolimus is an exceedingly useful immunosuppressant for kidney transplant recipients when administered in combination with low-dose CNIs or with elimination of CNIs. Here, we summarize the current knowledge about the introduction of everolimus with CNI elimination or minimization in kidney transplant recipients at late posttransplant stage.



INTRODUCTION

Excellent short-to medium-term graft survival has been achieved in kidney transplantation owing to the low acute rejection rate of calcineurin inhibitor (CNI), cyclosporine (CsA) and tacrolimus (Tac)-based immunosuppressive therapies[1]. Therefore, the next step is to determine how to improve long-term graft and patient survival rates. CNIs are known to induce nephrotoxicity, malignancies and cardiovascular diseases and to promote interstitial fibrosis/tubular atrophy[2-5], strongly influencing long-term graft and patient survival. Thus, efforts to reduce CNI exposure have become extremely valuable.

Everolimus (EVL) is an inhibitor of the mammalian target of rapamycin (mTOR), an evolutionarily conserved serine/threonine kinase playing an important role in the regulation of many cellular functions, which include metabolism, growth, proliferation, survival and memory[6]. EVL binds to the cytosolic FK-binding protein (FKBP)-12. The resulting complex then binds with high affinity to the FKBP12-rapamycin binding domain of mTOR, which inhibits mTOR activity, resulting in the inhibition of B cell and T cell proliferation, angiogenesis and cell metabolism[7,8]. EVL exhibits little nephrotoxicity and pleiotropic effects, such as antiproliferative[9], antineoplastic[10], antiviral[11] and antiatherosclerotic[12] properties. Therefore, it can be speculated that EVL is an exceedingly useful immunosuppressant for kidney transplant recipients in combination with low-dose or elimination of CNIs.

In the de novo use of EVL with low-dose CsA study (A2309) - a 24-mo randomized controlled study that compared EVL plus low-dose CsA against mycophenolate mofetil (MMF) plus standard-dose CsA in 833 kidney transplant recipients - the two treatment groups showed comparable graft function[13]. Meta-analysis of the CNI-sparing regimen in kidney transplantation showed an increase in graft failure rate associated with the combined use of mTOR inhibitors (mTORi) and mycophenolate, although improved graft function was noted among those surviving with functioning grafts[14].

In the early conversion of CNI to EVL study (ZEUS[15]), kidney transplant recipients were randomized at 4.5 mo for either conversion to EVL or continuance of CsA, and a higher estimated glomerular filtration rate (eGFR) was observed in the EVL group at year 3. However, the biopsy-proven acute rejection (BPAR) rate was 13.0% in the recipients who converted to EVL and 4.8% in the recipients who continued CsA (P = 0.015), although a statistically significant difference was not associated with long-term graft loss. In addition, the discontinuation rate of the EVL group was high (28.4%).

In a recent open-label, 24-mo study (the ELEVATE trial[16]), 715 kidney transplant recipients were randomized for either conversion to EVL or continuance of CNI at 10-14 wk after kidney transplantation. As a result, eGFR was comparable between the two groups, but the BPAR and discontinuation rates were higher in the EVL group (9.7% vs 4.8%, P = 0.014). Subsequently, some studies have been undertaken to explore the benefits of delayed introduction of EVL following initial CNI therapy in kidney transplantation (Tables 1 and 2). Possible pros and cons of late conversion to EVL with CNI elimination or minimization are shown in Table 3.

Table 1 Summary of late everolimus conversion clinical trials.
Ref.No. of subjects/follow-upEVLtreatmentGroupsOutcomes
ASCERTAIN[17] (2011)394/2 yrConversion to EVL with CNI elimination or minimization at mean of 5.6 yrGp 1: CNI elimination (EVL C0, 8-12 ng/mL), n = 127 Gp 2: CNI minimization (EVL C0, 3-8 ng/mL and CNI reduced to 80%-90% below baseline), n = 144 Gp 3: control (CsA C2, > 400 ng/mL; Tac C0, > 4 ng/mL), n = 123Graft survival: 96.9%, 94.6%, 95.1% (P = NS) Patient survival: 97.6%, 97.1%, 100% (P = NS) Comparable eGFR in 3 groups; recipients with baseline CrCl > 50 mL/min had greater increase in measured GFR after CNI elimination Adverse events resulted in discontinuation: 28.3%, 16.7%, 4.1% (Gp 1 vs GP 3, P < 0.001; Gp 2 vs Gp 3, P = 0.020)
APOLLO[18] (2015)93/1 yrConversion from CNI to EVL at mean of 7 yrGp 1: CNI elimination (EVL C0, 6-10 ng/mL), n = 46 Gp 2: control (CsA C0, 80-150 ng/mL; Tac C0, 5-10 ng/mL), n = 47Graft survival: 100%, 100% Patient survival: 97.8%, 97.9% (P = NS) Adjusted eGFR was significantly higher in Gp 1 within on-treatment population Adverse events resulted in discontinuation: 32.6%, 10.6% (P < 0.01)
Table 2 Summary of retrospective or nonrandomized studies for late everolimus conversion.
Ref.No. of subjects/follow-upEVL treatmentOutcomes
Morales et al[20] (2007)/ retrospective8/1-16 moConversion to EVL with CNI elimination or reduction at mean of 5 yrCrCl increased by 42% in recipients with CAN (grade 1 or 2) and CNI nephrotoxicity (P = 0.017)
Sanchez-Fructuoso et al[21] (2012)/ retrospective220/1 yrConversion from CNI to EVL at mean of 69.4 moCrCl increased in recipients with baseline CrCl ≥ 40 mL/min and baseline proteinuria < 550 mg/d (P = 0.005) Median proteinuria increased from 304 mg/d to 458 mg/d (P < 0.001) EVL discontinuation rate was 24%
Chow et al[22] (2015)/ open-label, single arm17/1 yrConversion to EVL with CNI minimization in recipients with CAN at mean of 4.2 yrMean slope of eGFR was - 4.31 mL/min/1.73 m2 per yr before conversion, as compared with 1.29 mL/min/1.73 m2 per yr at 12 mo after conversion (P = 0.036) Renal biopsy showed significant decrease of tubular atrophy (15.7% vs 7.1%, P = 0.005) and interstitial fibrosis (14.8% vs 7.2%, P = 0.013)
Miura et al[23] (2015)/ retrospective13/1 yrConversion to EVL with Tac reduction in recipients with CNIA at mean of 43 moaah scores improved in 5 recipients (38%); No improvement was observed in recipients with aah3; No deterioration was observed. eGFR improved from 44.3 mL/min/1.73 m2 to 49.8 mL/min/1.73 m2 (P < 0.01).
Uchida et al[24] (2016)/ retrospective (our report)26/1 yrConversion from antimetabolites (MMF or MZ) to EVL with CNI minimization at mean of 39.5 moeGFR significantly increased from 50.7 mL/min/1.73 m2 to 53.6 mL/min/1.73 m2 in the EVL continuation group EVL discontinuation rate was 42.3%
Nojima et al[25] (2017)/ retrospective56/1 yrConversion to EVL with CNI reduction in recipients with CNI nephrotoxicity or IF/TA at mean of 7.4 yreGFR increased by 7% (P < 0.005) EVL discontinuation rate was 11%
Nanmoku et al[26] (2017)/ nonrandomized86/ 1 yrConversion to EVL with Tac minimization, MMF reduction and steroid withdrawal in cases of complications such as diabetes, viral infection etcConventional group (n = 50); EVL group (n = 36) Biopsy-proven acute rejection rate exhibited no significant difference between these groups (12% vs 17%, P = 0.55) Serum creatinine significantly improved in the EVL group (P = 0.031) EVL discontinuation rate was 13.8%
Table 3 Pros and cons of late conversion to everolimus with calcineurin inhibitor elimination or minimization in kidney transplant recipients.
AdvantageDisadvantage
Due to EVL introduction Antitumoral effect (especially on nonmelanoma skin carcinoma) Antiviral effect (especially on CMV and BKV infection) Antiproliferative effect Antiatherosclerotic effectDue to EVL introduction Adverse events (gastrointestinal disorders, hyperlipidemia, interstitial pneumonitis, edema, mouth ulcers, proteinuria, impaired wound healing, hematotoxicity and so on)
Due to CNI elimination or minimization Favorable graft functionDue to CNI elimination or minimization Risk of de novo DSA

The aim of this minireview was to summarize the current knowledge on the introduction of EVL in kidney transplant recipients at a late posttransplant stage.

GRAFT FUNCTION

Only two major clinical trials are available for the introduction of EVL in kidney transplant recipients at a late posttransplant stage, namely the ASCERTAIN[17] and APOLLO[18] trials (Table 1). In the open-label multicenter ASCERTAIN study, kidney transplant recipients receiving CNI were randomized to EVL with CNI elimination (n = 127), CNI minimization (n = 144) and continuation of CNI unchanged (controls, n = 123) at a mean of 5.4 years after transplantation. The eGFR at 24 mo was not significantly different among the three groups. However, recipients with baseline creatinine clearance higher than 50 mL/min had a greater increase in measured GFR after CNI elimination.

In the open-label multicenter APOLLO study, kidney transplant recipients were randomized to EVL with CNI elimination (n = 46) or for remaining on standard CNI-based immunosuppression (controls; n = 47) at a mean of 7 years after transplantation. Within the on-treatment population, adjusted eGFR was significantly higher in the EVL continuation group than in the CNI continuation group at 12 mo after conversion. In addition, the 5-year follow-up results showed that eGFR in the EVL continuation group was significantly higher, by 11 mL/min·1.73 m2 (P = 0.031), in recipients who remained on their randomized study regimen until 60 mo[19].

Other studies[20-26] have shown that favorable graft function was sustained by EVL late-induction with CNI elimination or reduction (Table 2). Our previous study[24] demonstrated that eGFR was significantly improved in stable kidney transplant recipients already having favorable renal function, after remaining on EVL treatment for 12 mo after conversion. As a histological assessment, Chow et al[22] demonstrated that EVL rescue therapy and CNI inhibitor minimization strategy slowed down the disease progression by reducing the tubular atrophy and interstitial fibrosis score in renal transplant recipients with biopsy-confirmed chronic allograft nephropathy. Miura et al[23] reported that Tac reduction with EVL addition histologically improved CNI arteriolopathy in 5 out of 9 selected recipients, whose alternate quantitative scoring for hyaline arteriolar thickening (aah scores) was under 3.

REJECTION

There was no significant difference in the number of BPAR episodes between the intervention group and the control group in both the ASCERTAIN and APOLLO studies. It was reported that EVL-based immunosuppression in early conversion from CNI was associated with an increased risk of developing donor-specific HLA antibodies (DSA) and antibody-mediated rejection[27]. In contrast, late conversion to CNI-free therapy with mTORi did not appear to affect the risk of de novo DSA[28], but there is concern about the development of DSA and antibody-mediated rejection because CNI level variability is a strong risk factor for de novo DSA development and death-censored graft loss[29].

ADVERSE EVENTS

Generally, mTORi administration has been associated with several adverse events, such as gastrointestinal disorders, hyperlipidemia, interstitial pneumonitis, edema, mouth ulcers, proteinuria, impaired wound healing, hematotoxicity and so on[7]. It was reported that adverse events of mTORi accounted for 20%-40% of the drop-out rate in a clinical phase III trial[30]. In the late conversion to EVL studies, the discontinuation of EVL treatment due to adverse events occurred at about the same rate (approximately 30%). In our report[24], the discontinuation rate of EVL treatment was relatively high, at 42.3%.

The common adverse events leading to discontinuation have been aphthous stomatitis, pneumonitis, progressive renal deterioration and proteinuria. Proteinuria is a well-known prognostic factor for graft and patient survival rates in kidney transplantation[31]. Sanchez-Fructuoso et al[21] reported that risk factors for the development of proteinuria ≥ 900 mg/d at 1 year after late conversion were creatinine clearance of < 60 mL/min, serum triglycerides of ≥ 150 mg/d, no treatment with steroid, baseline proteinuria of ≥ 550 mg/d and conversion at ≥ 3 years after transplantation. An interaction was observed between baseline proteinuria and time to conversion, and the authors concluded that the success of EVL conversion with CNI elimination depended on not making so late conversions and not converting recipients with high baseline proteinuria. On the other hand, Nojima et al[25] demonstrated that late immunosuppression conversion, at > 3 years after kidney transplantation, using EVL in addition to a reduction in CNI dose safely and significantly improved graft function.

MALIGNANCIES

Kidney transplant recipients late-converted to sirolimus-based, CNI-free immunotherapy had a lower risk of malignancies at 2 years postconversion, with a high degree of heterogeneity attributed in the CONVERT trial[32]. The reduction was driven by a significant reduction in nonmelanoma skin carcinoma rate (P < 0.001), while the rate of all other malignancies was numerically lower, although without statistical significance (P = 0.058). It has been reported that switching from CNIs to sirolimus had an antitumoral effect among kidney transplant recipients with previous nonmelanoma skin carcinoma[33]. In the cases of late EVL conversion, however, the ASCERTAIN study[17] showed that the incidence rates of malignancies were 7.1%, 7.6% and 5.7%, respectively in the CNI elimination, CNI minimization and control groups at 2 years after EVL conversion.

CAUSE OF LATE CONVERSION TO EVL

Chronic allograft nephropathy, CNI nephrotoxicity and CNI arteriolopathy may be good indications for late conversion to EVL[20-23,25]. Furthermore, cancer is one of the main indications for late conversion to EVL[20,21]. As mentioned in the above section on “malignancies”, there is no evidence to date for the superiority of EVL in suppressing malignancies at late conversion. However, Lim et al[34] published that de novo use of EVL with reduced exposure to CNIs may enable a reduction in malignancy burden after transplantation.

Viral infection is also an indication for late conversion to EVL. It is well known that kidney transplant recipients receiving mTORi have a lower risk of developing cytomegalovirus (CMV) infection[35]. Furthermore, cases with ganciclovir-resistant cytomegalovirus infection have been reported to be cured after switching to mTORi[36]. Kidney transplant recipients who have BK virus infection may benefit from conversion to mTORi[35]. Polanco et al[37] reported a recent prospective study of 15 recipients with BK virus-associated nephropathy. As a result, MMF elimination and conversion from Tac to EVL occurred in 9 recipients (60%), and 6 (67%) of the 9 recipients had improvement and 3 maintained stable renal function. In addition, BK viremia cleared in 5 (56%) of the recipients and decreased more than 95% in the remaining 4. With respect to Epstein-Barr virus infection, there is lack of evidence on whether the use of mTORi reduces the risk of infection in solid organ transplant recipients[35].

ABO-INCOMPATIBLE KIDNEY TRANSPLANTATION

Only two short-term pilot studies have been published about the introduction of EVL in ABO-incompatible kidney transplant recipients at a late posttransplant stage[38,39]. In our study, 16 stable ABO-incompatible kidney transplant recipients were switched from MMF to EVL with CNI minimization. Our results showed that conversion to EVL with CNI minimization for 3 mo did not induce acute rejection and C4d deposition in all recipients, and the mean eGFR value significantly increased at 3 mo after conversion compared to baseline[38]. In another study, 7 stable ABO-incompatible kidney transplant recipients were converted from mycophenolate acid to EVL at a late posttransplant phase because of active BK virus replication, and then compared with a reference group of 14 ABO-incompatible patients receiving standard Tac and mycophenolate acid[39]. Conversion from mycophenolate acid to EVL decreased the BK viral load in 5 patients. Thus, this study demonstrated that ABO-incompatible kidney transplant recipients with an active BK virus infection may benefit from conversion to EVL[39].

CONCLUSION

In this minireview, we summarized reports published on the introduction of EVL in kidney transplant recipients at a late posttransplant stage. Selected recipients, who can continue EVL treatment without adverse events and who already have satisfactory renal function, may profit by late conversion to EVL with CNI elimination or minimization. In addition, chronic allograft nephropathy, CNI nephrotoxicity, CNI arteriolopathy, cancer and viral infection (especially cytomegalovirus infection) may be good indications for late conversion to EVL.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Transplantation

Country of origin: Japan

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Sureshkumar K, Yildiz B S- Editor: Ma YJ L- Editor: A E- Editor: Yin SY

References
1.  Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B. Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era. Am J Transplant. 2004;4:378-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 945]  [Cited by in F6Publishing: 913]  [Article Influence: 45.7]  [Reference Citation Analysis (0)]
2.  Fletcher JT, Nankivell BJ, Alexander SI. Chronic allograft nephropathy. Pediatr Nephrol. 2009;24:1465-1471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
3.  Naesens M, Kuypers DR, Sarwal M. Calcineurin inhibitor nephrotoxicity. Clin J Am Soc Nephrol. 2009;4:481-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 950]  [Cited by in F6Publishing: 998]  [Article Influence: 66.5]  [Reference Citation Analysis (0)]
4.  Marcén R. Immunosuppressive drugs in kidney transplantation: impact on patient survival, and incidence of cardiovascular disease, malignancy and infection. Drugs. 2009;69:2227-2243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 215]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
5.  Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Chapman JR, Allen RD. Calcineurin inhibitor nephrotoxicity: longitudinal assessment by protocol histology. Transplantation. 2004;78:557-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 392]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
6.  Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124:471-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4260]  [Cited by in F6Publishing: 4276]  [Article Influence: 237.6]  [Reference Citation Analysis (0)]
7.  Shipkova M, Hesselink DA, Holt DW, Billaud EM, van Gelder T, Kunicki PK, Brunet M, Budde K, Barten MJ, De Simone P. Therapeutic Drug Monitoring of Everolimus: A Consensus Report. Ther Drug Monit. 2016;38:143-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 83]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
8.  Baroja-Mazo A, Revilla-Nuin B, Ramírez P, Pons JA. Immunosuppressive potency of mechanistic target of rapamycin inhibitors in solid-organ transplantation. World J Transplant. 2016;6:183-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 46]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
9.  Schuler W, Sedrani R, Cottens S, Häberlin B, Schulz M, Schuurman HJ, Zenke G, Zerwes HG, Schreier MH. SDZ RAD, a new rapamycin derivative: pharmacological properties in vitro and in vivo. Transplantation. 1997;64:36-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 454]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
10.  Campistol JM, Eris J, Oberbauer R, Friend P, Hutchison B, Morales JM, Claesson K, Stallone G, Russ G, Rostaing L. Sirolimus therapy after early cyclosporine withdrawal reduces the risk for cancer in adult renal transplantation. J Am Soc Nephrol. 2006;17:581-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 379]  [Cited by in F6Publishing: 399]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
11.  Brennan DC, Legendre C, Patel D, Mange K, Wiland A, McCague K, Shihab FS. Cytomegalovirus incidence between everolimus versus mycophenolate in de novo renal transplants: pooled analysis of three clinical trials. Am J Transplant. 2011;11:2453-2462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 115]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
12.  Mueller MA, Beutner F, Teupser D, Ceglarek U, Thiery J. Prevention of atherosclerosis by the mTOR inhibitor everolimus in LDLR-/- mice despite severe hypercholesterolemia. Atherosclerosis. 2008;198:39-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 122]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
13.  Cibrik D, Silva HT Jr, Vathsala A, Lackova E, Cornu-Artis C, Walker RG, Wang Z, Zibari GB, Shihab F, Kim YS. Randomized trial of everolimus-facilitated calcineurin inhibitor minimization over 24 months in renal transplantation. Transplantation. 2013;95:933-942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 83]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
14.  Sharif A, Shabir S, Chand S, Cockwell P, Ball S, Borrows R. Meta-analysis of calcineurin-inhibitor-sparing regimens in kidney transplantation. J Am Soc Nephrol. 2011;22:2107-2118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 102]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
15.  Budde K, Lehner F, Sommerer C, Arns W, Reinke P, Eisenberger U, Wüthrich RP, Scheidl S, May C, Paulus EM. Conversion from cyclosporine to everolimus at 4.5 months posttransplant: 3-year results from the randomized ZEUS study. Am J Transplant. 2012;12:1528-1540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 70]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
16.  de Fijter JW, Holdaas H, Øyen O, Sanders JS, Sundar S, Bemelman FJ, Sommerer C, Pascual J, Avihingsanon Y, Pongskul C. Early Conversion From Calcineurin Inhibitor- to Everolimus-Based Therapy Following Kidney Transplantation: Results of the Randomized ELEVATE Trial. Am J Transplant. 2017;17:1853-1867.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 57]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
17.  Holdaas H, Rostaing L, Serón D, Cole E, Chapman J, Fellstrøm B, Strom EH, Jardine A, Midtvedt K, Machein U. Conversion of long-term kidney transplant recipients from calcineurin inhibitor therapy to everolimus: a randomized, multicenter, 24-month study. Transplantation. 2011;92:410-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 113]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
18.  Budde K, Rath T, Sommerer C, Haller H, Reinke P, Witzke O, Suwelack B, Baeumer D, May C, Porstner M. Renal, efficacy and safety outcomes following late conversion of kidney transplant patients from calcineurin inhibitor therapy to everolimus: the randomized APOLLO study. Clin Nephrol. 2015;83:11-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
19.  Budde K, Sommerer C, Rath T, Reinke P, Haller H, Witzke O, Suwelack B, Baeumer D, Sieder C, Porstner M. Renal function to 5 years after late conversion of kidney transplant patients to everolimus: a randomized trial. J Nephrol. 2015;28:115-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
20.  Morales J, Fierro A, Benavente D, Zehnder C, Ferrario M, Contreras L, Herzog C, Buckel E. Conversion from a calcineurin inhibitor-based immunosuppressive regimen to everolimus in renal transplant recipients: effect on renal function and proteinuria. Transplant Proc. 2007;39:591-593.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
21.  Sánchez-Fructuoso AI, Ruiz JC, Calvo N, Rodrigo E, Perez-Flores I, Gómez-Alamillo C, Fernández-Pérez C, Arias M, Barrientos A. Everolimus as primary immunosuppression in kidney transplantation: experience in conversion from calcineurin inhibitors. Transplantation. 2012;93:398-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
22.  Chow KM, Szeto CC, Lai FM, Luk CC, Kwan BC, Leung CB, Li PK. Functional and histological improvement after everolimus rescue of chronic allograft dysfunction in renal transplant recipients. Ther Clin Risk Manag. 2015;11:829-835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
23.  Miura M, Higashiyama H, Fukasawa Y, Itoh Y, Tamaki T. Tacrolimus reduction with everolimus addition for calcineurin inhibitor-induced arteriolopathy in kidney allografts. Nephrology (Carlton). 2015;20 Suppl 2:58-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
24.  Uchida J, Iwai T, Kuwabara N, Kabei K, Nishide S, Yamasaki T, Naganuma T, Kumada N, Takemoto Y, Nakatanti T. Clinical Experience of Late Conversion From Antimetabolites With Standard Exposure Calcineurin Inhibitors to Everolimus With Calcineurin Inhibitor Minimization in Stable Kidney Transplant Recipients With Good Renal Function. Transplant Proc. 2016;48:775-780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
25.  Nojima M, Yamada Y, Higuchi Y, Shimatani K, Kanematsu A, Yamamoto S. Immunosuppression Modification by Everolimus With Minimization of Calcineurin Inhibitors Recovers Kidney Graft Function Even in Patients With Very Late Conversion and Also With Poor Graft Function. Transplant Proc. 2017;49:41-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
26.  Nanmoku K, Kurosawa A, Kubo T, Shinzato T, Shimizu T, Kimura T, Yagisawa T. Effective and Safe Reduction of Conventional Immunosuppressants Using Everolimus in Maintenance Kidney Transplant Recipients. Transplant Proc. 2017;49:1724-1728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
27.  Liefeldt L, Brakemeier S, Glander P, Waiser J, Lachmann N, Schönemann C, Zukunft B, Illigens P, Schmidt D, Wu K. Donor-specific HLA antibodies in a cohort comparing everolimus with cyclosporine after kidney transplantation. Am J Transplant. 2012;12:1192-1198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 197]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
28.  Grimbert P, Thaunat O. mTOR inhibitors and risk of chronic antibody-mediated rejection after kidney transplantation: where are we now? Transpl Int. 2017;30:647-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
29.  Rodrigo E, Segundo DS, Fernández-Fresnedo G, López-Hoyos M, Benito A, Ruiz JC, de Cos MA, Arias M. Within-Patient Variability in Tacrolimus Blood Levels Predicts Kidney Graft Loss and Donor-Specific Antibody Development. Transplantation. 2016;100:2479-2485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 146]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
30.  Rostaing L, Kamar N. mTOR inhibitor/proliferation signal inhibitors: entering or leaving the field? J Nephrol. 2010;23:133-142.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Roodnat JI, Mulder PG, Rischen-Vos J, van Riemsdijk IC, van Gelder T, Zietse R, IJzermans JN, Weimar W. Proteinuria after renal transplantation affects not only graft survival but also patient survival. Transplantation. 2001;72:438-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 146]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
32.  Alberú J, Pascoe MD, Campistol JM, Schena FP, Rial Mdel C, Polinsky M, Neylan JF, Korth-Bradley J, Goldberg-Alberts R, Maller ES; Sirolimus CONVERT Trial Study Group. 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]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 164]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
33.  Euvrard S, Morelon E, Rostaing L, Goffin E, Brocard A, Tromme I, Broeders N, del Marmol V, Chatelet V, Dompmartin A. Sirolimus and secondary skin-cancer prevention in kidney transplantation. N Engl J Med. 2012;367:329-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 437]  [Cited by in F6Publishing: 385]  [Article Influence: 32.1]  [Reference Citation Analysis (0)]
34.  Lim WH, Russ GR, Wong G, Pilmore H, Kanellis J, Chadban SJ. The risk of cancer in kidney transplant recipients may be reduced in those maintained on everolimus and reduced cyclosporine. Kidney Int. 2017;91:954-963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
35.  Pascual J, Royuela A, Fernández AM, Herrero I, Delgado JF, Solé A, Guirado L, Serrano T, de la Torre-Cisneros J, Moreno A. Role of mTOR inhibitors for the control of viral infection in solid organ transplant recipients. Transpl Infect Dis. 2016;18:819-831.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 48]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
36.  Sabé N, González-Costello J, Rama I, Niubó J, Bodro M, Roca J, Cruzado JM, Manito N, Carratalà J. Successful outcome of ganciclovir-resistant cytomegalovirus infection in organ transplant recipients after conversion to mTOR inhibitors. Transpl Int. 2012;25:e78-e82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
37.  Polanco N, González Monte E, Folgueira MD, Morales E, Gutiérrez Martínez E, Bengoa I, Hernández A, Morales JM, Praga M, Andrés A. Everolimus-based immunosuppression therapy for BK virus nephropathy. Transplant Proc. 2015;47:57-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
38.  Uchida J, Machida Y, Iwai T, Kuwabara N, Kabei K, Naganuma T, Kumada N, Kawashima H, Nakatani T. Conversion of stable ABO-incompatible kidney transplant recipients from mycophenolate mofetil with standard exposure calcineurin inhibitors (CNIs) to everolimus with very low exposure CNIs-a short-term pilot study. Clin Transplant. 2014;28:80-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
39.  Belliere J, Kamar N, Mengelle C, Allal A, Sallusto F, Doumerc N, Game X, Congy-Jolivet N, Esposito L, Debiol B. Pilot conversion trial from mycophenolic acid to everolimus in ABO-incompatible kidney-transplant recipients with BK viruria and/or viremia. Transpl Int. 2016;29:315-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]