Skip to main content
  • Study protocol
  • Open access
  • Published:

Design and rationale of the ATHENA study – A 12-month, multicentre, prospective study evaluating the outcomes of a de novo everolimus-based regimen in combination with reduced cyclosporine or tacrolimus versus a standard regimen in kidney transplant patients: study protocol for a randomised controlled trial

Abstract

Background

Immunosuppression with calcineurin inhibitors remains the mainstay of treatment after kidney transplantation; however, long-term use of these drugs may be associated with nephrotoxicity. In this regard, the current approach is to optimise available immunosuppressive regimens to reduce the calcineurin inhibitor dose while protecting renal function without affecting the efficacy. The ATHENA study is designed to evaluate renal function in two regimens: an everolimus and reduced calcineurin inhibitor-based regimen versus a standard treatment protocol with mycophenolic acid and tacrolimus in de novo kidney transplant recipients.

Method/Design

ATHENA is a 12-month, multicentre, open-label, prospective, randomised, parallel-group study in de novo kidney transplant recipients (aged 18 years or older) receiving renal allografts from deceased or living donors. Eligible patients are randomised (1:1:1) prior to transplantation to one of the following three treatment arms: everolimus (starting dose 1.5 mg/day; C0 3–8 ng/mL) with cyclosporine or everolimus (starting dose 3 mg/day; C0 3–8 ng/mL) with tacrolimus or mycophenolic acid (enteric-coated mycophenolate sodium at 1.44 g/day or mycophenolate mofetil at 2 g/day) with tacrolimus; in combination with corticosteroids. All patients receive induction therapy with basiliximab. The primary objective is to demonstrate non-inferiority of renal function (eGFR by the Nankivell formula) in one of the everolimus arms compared with the standard group at month 12 post transplantation. The key secondary objective is to assess the incidence of treatment failure, defined as biopsy-proven acute rejection, graft loss, or death, among the treatment groups. Other objectives include assessment of the individual components of treatment failure, incidence and severity of viral infections, incidence and duration of delayed graft function, incidence of indication biopsies, slow graft function and wound healing complications, and overall safety and tolerability. Exploratory objectives include evaluation of left ventricular hypertrophy assessed by the left ventricular mass index, evolution of human leukocyte antigen and non-human leukocyte antigen antibodies, and a cytomegalovirus substudy.

Discussion

As one of the largest European multicentre kidney transplant studies, ATHENA will determine whether a de novo everolimus-based regimen can preserve renal function versus the standard of care. This study further assesses a number of clinical issues which impact long-term outcomes post transplantation; hence, its results will have a major clinical impact.

Trial registration

Clinicaltrials.gov: NCT01843348, date of registration – 18 April 2013; EUDRACT number: 2011-005238-21, date of registration – 20 March 2012

Peer Review reports

Background

The evolution of immunosuppressive regimens over the past few decades has led to considerable improvement in acute rejection rates and short-term graft survival [1]. Patient and graft survival at 1 year post transplantation now exceed 95 % in the case of living donation and 90 % after deceased donation. However, the long-term outcomes post kidney transplantation do not show a similar trend of improvement. At 10 years, graft survival remains about 50 % after deceased donation in the US and in Europe, with approximately 30 % of patients returning to dialysis and one of four patients dying with a functioning graft [1, 2]. The lack of improvement in long-term outcomes is further reflected by the fact that the number of re-transplants among adult kidney transplant patients has remained almost unchanged over the last decade [1].

Currently, calcineurin inhibitors (CNIs), cyclosporine, and tacrolimus are the cornerstone of immunosuppressive therapy post kidney transplantation [1]. However, their long-term use may be associated with non-reversible nephrotoxicity, morphologically characterised by striped fibrosis, progressive arteriolar hyalinosis, and ischemic glomerulosclerosis, which is a well-recognised cause of morbidity in transplant patients [35]. Chronic allograft injury alone accounts for two thirds of kidney graft failures [6]. Clinical data has shown that lowering the dose of CNI can improve renal function [57]. In this regard, the focus should be on optimising the currently available immunosuppressive regimens with the aim of preserving long-term renal function while maintaining the efficacy [8]. Several studies with a reduced-dose CNI and everolimus regimen have shown that it maintained efficacy and preserved renal function (Table 1) [933]. In the large randomised A2309 trial, pre-emptive everolimus therapy was associated with a greater than 60 % reduction in cyclosporine exposure while preserving renal function with comparable efficacy to mycophenolic acid and standard-exposure cyclosporine in de novo kidney transplant patients [16, 17]. In the ASSET study, an everolimus-facilitated tacrolimus minimisation strategy achieved good renal function with an acceptable safety profile without compromising efficacy [23]. Moreover, everolimus exerts other non-immunosuppressive properties, including potential cardioprotective, anti-malignancy, and antiviral effects [3441]. These non-immunosuppressive benefits further suggest that everolimus-based regimens may be a preferred approach as cardiovascular disease, malignancy, and infections account for nearly four out of the five deaths occurring with functioning grafts [6].

Table 1 Everolimus in kidney transplantation

The ATHENA trial is designed to further increase our knowledge and seek answers relating to the use of everolimus in CNI minimisation protocols in de novo kidney transplant patients. The ATHENA study assesses the change in renal function at 12 months post transplant as the primary objective. The design of the trial is described here.

Methods/Design

Study design

ATHENA (Clinicaltrials.gov: NCT01843348; EUDRACT number: 2011-005238-21) is a 12-month, multicentre, randomised, international, prospective, controlled, open-label study with three parallel treatment groups in de novo kidney transplant recipients receiving renal allografts from deceased or living donors (protocol version 3, 29 July 2014). Eligible patients are randomised before transplantation using a validated system to ensure an unbiased treatment assignment in a 1:1:1 ratio to receive either everolimus with a reduced dose of cyclosporine, or everolimus with tacrolimus, or a standard regimen of mycophenolic acid with tacrolimus (Fig. 1). All patients receive induction therapy with basiliximab and maintenance steroids. At the time of randomisation, patients are stratified based on the donor type (living donor, deceased standard criteria donor, or deceased expanded criteria donor) and the participation of the recipient in the European Senior Program. The study protocol and the proposed informed consent form were reviewed and approved by the national institutional review boards or independent ethics committees at each centre and the federal institute for drugs and medical devices (Additional file 1). Written informed consent was obtained from all patients. The clinical study was designed and is conducted in accordance with the ethical principles laid down in the Declaration of Helsinki.

Fig. 1
figure 1

Study design. Steroid dose will be at least 5 mg prednisolone or equivalent, according to centre practice. EC-MPS enteric-coated mycophenolate sodium. M month, MMF mycophenolate mofetil, MPA mycophenolic acid, RND randomisation, Tx transplantation

Study population

The study population comprises de novo adult patients receiving a primary or secondary kidney transplant from a deceased or living donor. In the case of second kidney transplants, patients could be enrolled only if the first graft loss is due to non-immunological reasons. Patients are not eligible for the study if they are recipients of an ABO-incompatible transplants, have pre-existing donor-specific antibodies (DSA), or have an organ cold ischemia time longer than 30 h. Patients with pre-existing human leukocyte antigen (HLA)-antibodies not directed against the donor and less than 20 % panel reactivity at the time of transplant were included in the study. Detailed inclusion and exclusion criteria are shown in Table 2.

Table 2 Key inclusion and exclusion criteria

Study objectives

The primary objective at month 12 post transplantation is to demonstrate non-inferiority in renal function assessed by the glomerular filtration rate (Nankivell formula) [41] in at least one of the everolimus treatment regimens compared with the standard treatment group receiving mycophenolic acid and tacrolimus. The key secondary objective at month 12 is to evaluate the incidence of treatment failure defined as biopsy-proven acute rejection (BPAR), graft loss, or death among the treatment groups. Other objectives include assessment of individual components of treatment failure, incidence and severity of viral infections (cytomegalovirus (CMV), BK-virus (BKV)), incidence and duration of delayed graft function (DGF), incidence of indication biopsies, incidence of slow graft function, incidence of wound healing complications, and duration of healing. Incidence of viral infections (CMV and BKV) and changes to the viral load are closely monitored throughout the study. In addition, a patient subgroup analysis of CMV-specific T-cells and NK-cells is conducted in a central laboratory. Overall, the safety objectives include assessment of adverse events (AEs) and serious AEs (SAEs), infections and discontinuations due to AEs, and laboratory abnormalities. Exploratory objectives include evaluation of the incidence of HLA-antibodies and among those DSA and non-HLA antibodies (AT1R, ETAR) by treatment group and its association with acute rejection. HLA-antibodies including DSA were analysed using single antigen Luminex technology at baseline and month 12. In addition, left ventricular (LV) changes are measured by LV hypertrophy (LVH) assessment by echocardiography measurements. Detailed objectives of the study are outlined in Table 3.

Table 3 Objectives of the ATHENA study

Immunosuppression

All patients receive induction therapy with basiliximab dosed at 20 mg intravenously on the day of transplantation and on day 4 post transplantation, as per label recommendations. Patients are randomised to receive everolimus at an initial dose of 1.5 mg/day with a reduced dose of cyclosporine and 3 mg/day with tacrolimus within the first 24 h post transplantation. Thereafter, the dose of everolimus is adjusted to the target trough concentration of 3–8 ng/mL throughout the study period. Treatment with CNI, tacrolimus, or cyclosporine, is initiated within the first 24 h post transplantation, and the dose of CNI is adjusted to maintain the target trough levels, as shown in Fig. 1. In the control group, patients receive mycophenolic acid at a dose of 1.44 g/day enteric-coated mycophenolate sodium or at a dose of 2 g/day mycophenolate mofetil with a standard dose of tacrolimus. Dose adjustments and interruptions are allowed for tolerability reasons as defined in the protocol and are recorded. All patients receive corticosteroids at a minimum dose of 5 mg/day prednisolone or equivalent until month 12. Acute rejections are treated according to local practice and physicians’ discretion. All patients who prematurely withdraw from the study are provided with follow-up medical care/referred for appropriate ongoing care, as per the local practice.

Concomitant medication

Mandatory CMV prophylactic therapy with valganciclovir is recommended for at least 3 months in the case of high- to moderate-risk patients (CMV-positive donor/CMV-negative recipients or CMV-positive donor/CMV-positive recipients). All patients receive prophylactic treatment for Pneumocystis jirovecii pneumonia with trimethoprim/sulfamethoxazole for a period of 6 months. All medications and significant non-drug therapies administered after the initiation of the study drug are recorded.

Data collection

Patient visits are scheduled at baseline, and at months 1, 3, 6, 9, and 12 post transplantation. A detailed list of all study assessments and visits is shown in Table 4. Patients who discontinue the study drug and those who prematurely withdraw from the study are scheduled for a visit and all the assessments listed for visit 6 are performed.

Table 4 Assessment schedule

Renal function

Renal function is assessed by determining the glomerular filtrate rate using serum creatinine values according to the Nankivell formula [42] and used as the primary outcome measure in the study. In addition, as a secondary efficacy variable, the glomerular filtration rate is calculated using the Cockcroft-Gault method [43], the modification of diet in renal disease (MDRD) method [4446], and the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) method [47]. Serum creatinine levels are analysed using venous blood drawn and analysed in the local laboratory.

Outcome measures

BPAR is defined as rejections that are acute and proven by biopsy. The time to BPAR is the time from randomisation to the date of first documented BPAR. Graft loss is defined as a failure to discontinue dialysis or if the patient undergoes graft nephrectomy. Overall survival is defined as time from date of randomisation to death due to any cause. Delayed graft function is defined as the need for dialysis within the first 7 days post transplantation excluding the first day, and the duration is defined from the first dialysis day up to the last. Slow graft function is defined as serum creatinine >3.0 mg/dL at day 5.

Kidney allograft biopsy

Optional allograft biopsies are performed intra-operatively at the time of transplantation and at month 12. A control biopsy at month 6 may be performed according to centre practice. In all cases of suspected acute rejection, a graft biopsy is performed prior to, or within 24 h of initiation of anti-rejection therapy. All biopsies are read by the local pathologist according to the updated Banff 2009 criteria. Optional biopsies are assessed for the presence of interstitial fibrosis and tubular atrophy using the Banff 2007 criteria.

Non-HLA and HLA antibodies

The presence and evolution of non-HLA and HLA antibodies, and among those especially DSA antibodies in the serum, is evaluated at a central laboratory. Blood samples (5 mL) are collected for all patients at baseline, month 6, and month 12.

LVH and diastolic dysfunction

The echocardiographic analysis included assessments of the end-diastolic interventricular septum, LV end-diastolic posterior wall thickness, LV end-diastolic diameter, LV end-systolic diameter, LV end-diastolic volume, LV end-systolic volume, LV ejection fraction, relative wall thickness, and LV mass also expressed as LV mass index [48]. LVH was defined as an LV mass index exceeding 110 g/m2 in women and 125 g/m2 in men, and a value of 0.44 was taken as cut-off point for abnormal relative wall thickness [49]. Diastolic dysfunction was assessed and graded according to the guidelines of the American Society of Echocardiography [50].

CMV substudy

In this optional substudy, 4.7 mL of whole blood is drawn into a lithium heparin-containing tube at baseline and at month 12 or at the end of study/treatment, and shipped on the same day to a central laboratory at 4 °C. This substudy prospectively monitors the incidence of viraemia by analysis of viral load, CMV-specific T-cell frequency, phenotype and functionality, and regulatory T-cells, and T- and NK-cell subsets.

Data Monitoring Board

An external and independent Data Safety Monitoring Board was instituted before the start of the study. The board reviews safety-related issues on an ongoing basis and is entitled to make recommendations for changes in study conduct.

Statistical analysis

The primary efficacy variable, i.e., renal function at month 12 after randomisation between the treatment groups will be compared with the analysis of covariance (ANCOVA) model, using the treatment and centre as factors, and the estimated glomerular filtration rate at baseline as a covariate. Missing estimated glomerular filtration values will be handled within the ANCOVA analysis by multiple imputations by the last available post-baseline observation carried forward (LOCF) approach. Assuming a common dropout rate of 20 %, a sample size of 612 patients (204 patients in each treatment arm) is required in the study, so as to have at least 80 % power to demonstrate non-inferiority (2.5 % margin, one-sided t test) for the primary endpoint.

The primary analysis is based on the full analysis set that consists of all patients who receive at least one dose of the study drug. The per-protocol set includes all patients in the full analysis set who do not have any major deviations from the protocol procedures that may impact the study outcomes. The safety set consists of all patients who receive at least one dose of the study drug and had at least one post-baseline safety assessment.

Discussion

ATHENA is one of the largest European multicentre kidney transplant studies. It is the first study evaluating the non-inferiority of renal function as a primary objective in a de novo everolimus-based immunosuppressive protocol, and will determine whether an everolimus-based regimen can preserve renal function versus the current standard of care. The study will also provide insights into the evolution of HLA and non-HLA antibodies, occurrence of viral infections post transplantation, surveillance of cardiovascular comorbidities, and the incidence of wound healing complications [51]. In addition, the trial explores a regimen of everolimus with reduced-dose cyclosporine while the dose of tacrolimus in the everolimus arm and the standard arm is the same. These unique features will further enable the study to provide a direct comparison between the two arms, which will in turn help in optimising the immunosuppressive protocols. As the study addresses a wide range of issues that clinicians face today, its results are awaited with interest.

Trial status

The study is currently ongoing and actively recruiting patients across 27 sites, 15 centres in Germany and 12 centres in France. The study is expected to be complete by March 2016.

Abbreviations

AEs:

adverse events

ANCOVA:

analysis of covariance

BPAR:

biopsy-proven acute rejection

C0:

trough levels

cGFR:

calculated glomerular filtration rate

CKD-EPI:

Chronic Kidney Disease Epidemiology Collaboration

CMV:

cytomegalovirus

CNI:

calcineurin inhibitors

CrCl:

creatinine clearance

CsA:

cyclosporine

DGF:

delayed graft function

DSA:

donor-specific antibody

EC-MPS:

enteric-coated mycophenolate sodium

eGFR:

estimated glomerular filtration rate

EVR:

everolimus

HLA:

human leukocyte antigen

LV:

left ventricular

LVH:

left ventricular hypertrophy

MDRD:

modification of diet in renal disease

mGFR:

measured glomerular filtration rate

MMF:

mycophenolate mofetil

MPA:

mycophenolic acid

NODM:

new-onset diabetes mellitus

NS:

not significant

SAE:

serious adverse events

References

  1. Matas AJ, Smith JM, Skeans MA, Thompson B, Gustafson SK, Schnitzler MA, et al. OPTN/SRTR 2012 annual data report: kidney. Am J Transplant. 2014;14 Suppl 1:11–44.

    Article  PubMed  Google Scholar 

  2. Gondos A, Dohler B, Brenner H, Opelz G. Kidney graft survival in Europe and the United States: strikingly different long-term outcomes. Transplantation. 2013;95:267–74.

    Article  PubMed  Google Scholar 

  3. Chapman JR. Chronic calcineurin inhibitor nephrotoxicity – lest we forget. Am J Transplant. 2011;11:693–7.

    Article  CAS  PubMed  Google Scholar 

  4. Ojo AO, Held PJ, Port FK, Wolfe RA, Leichtman AB, Young EW, et al. Chronic renal failure after transplantation of a nonrenal organ. N Engl J Med. 2003;349:931–40.

    Article  CAS  PubMed  Google Scholar 

  5. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Chapman JR, Allen RD. Calcineurin inhibitor nephrotoxicity: longitudinal assessment by protocol histology. Transplantation. 2004;78:557–65.

    Article  CAS  PubMed  Google Scholar 

  6. Australia and New Zealand Dialysis and Transplant Registry 35th annual report 2012. http://www.anzdata.org.au/anzdata/AnzdataReport/35thReport/2012_annual_report.pdf. Accessed 31 May 2015.

  7. 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;22:2107–18.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. Chadban S, Morris R, Hirsch HH, Bunnapradist S, Arns W, Budde K. Immunosuppression in renal transplantation: some aspects for the modern era. Transplant Rev (Orlando). 2008;22:241–51.

    Article  Google Scholar 

  9. Nashan B, Curtis J, Ponticelli C, Mourad G, Jaffe J, Haas T. Everolimus and reduced-exposure cyclosporine in de novo renal-transplant recipients: a three-year phase II, randomized, multicenter, open-label study. Transplantation. 2004;78:1332–40.

    Article  CAS  PubMed  Google Scholar 

  10. Vitko S, Margreiter R, Weimar W, Dantal J, Viljoen HG, Li Y, et al. Everolimus (Certican) 12-month safety and efficacy versus mycophenolate mofetil in de novo renal transplant recipients. Transplantation. 2004;78:1532–40.

    Article  PubMed  Google Scholar 

  11. Vitko S, Margreiter R, Weimar W, Dantal J, Kuypers D, Winkler M, et al. Three-year efficacy and safety results from a study of everolimus versus mycophenolate mofetil in de novo renal transplant patients. Am J Transplant. 2005;5:2521–30.

    Article  CAS  PubMed  Google Scholar 

  12. Lorber MI, Mulgaonkar S, Butt KM, Elkhammas E, Mendez R, Rajagopalan PR, et al. Everolimus versus mycophenolate mofetil in the prevention of rejection in de novo renal transplant recipients: a 3-year randomized, multicenter, phase III study. Transplantation. 2005;80:244–52.

    Article  CAS  PubMed  Google Scholar 

  13. Vitko S, Tedesco H, Eris J, Pascual J, Whelchel J, Magee JC, et al. Everolimus with optimized cyclosporine dosing in renal transplant recipients: 6-month safety and efficacy results of two randomized studies. Am J Transplant. 2004;4:626–35.

    Article  CAS  PubMed  Google Scholar 

  14. Tedesco-Silva Jr H, Vitko S, Pascual J, Eris J, Magee JC, Whelchel J, et al. 12-month safety and efficacy of everolimus with reduced exposure cyclosporine in de novo renal transplant recipients. Transpl Int. 2007;20:27–36.

    Article  CAS  PubMed  Google Scholar 

  15. Chan L, Greenstein S, Hardy MA, Hartmann E, Bunnapradist S, Cibrik D, et al. Multicenter, randomized study of the use of everolimus with tacrolimus after renal transplantation demonstrates its effectiveness. Transplantation. 2008;85:821–6.

    Article  CAS  PubMed  Google Scholar 

  16. Tedesco Jr SH, Cibrik D, Johnston T, Lackova E, Mange K, Panis C, et al. Everolimus plus reduced-exposure CsA versus mycophenolic acid plus standard-exposure CsA in renal-transplant recipients. Am J Transplant. 2010;10:1401–13.

    Article  Google Scholar 

  17. Cibrik D, Silva Jr HT, Vathsala A, Lackova E, Cornu-Artis C, Walker RG, et al. Randomized trial of everolimus-facilitated calcineurin inhibitor minimization over 24 months in renal transplantation. Transplantation. 2013;95:933–42.

    Article  CAS  PubMed  Google Scholar 

  18. Budde K, Becker T, Arns W, Sommerer C, Reinke P, Eisenberger U, et al. Everolimus-based, calcineurin-inhibitor-free regimen in recipients of de-novo kidney transplants: an open-label, randomised, controlled trial. Lancet. 2011;377:837–47.

    Article  CAS  PubMed  Google Scholar 

  19. Budde K, Lehner F, Sommerer C, Reinke P, Arns W, Eisenberger U, et al. Five-year outcomes in kidney transplant patients converted from cyclosporine to everolimus: the randomized ZEUS study. Am J Transplant. 2015;15:119–28.

    Article  CAS  PubMed  Google Scholar 

  20. Albano L, Berthoux F, Moal MC, Rostaing L, Legendre C, Genin R, et al. Incidence of delayed graft function and wound healing complications after deceased-donor kidney transplantation is not affected by de novo everolimus. Transplantation. 2009;88:69–76.

    Article  CAS  PubMed  Google Scholar 

  21. Dantal J, Berthoux F, Moal MC, Rostaing L, Legendre C, Genin R, et al. Efficacy and safety of de novo or early everolimus with low cyclosporine in deceased-donor kidney transplant recipients at specified risk of delayed graft function: 12-month results of a randomized, multicenter trial. Transpl Int. 2010;23:1084–93.

    Article  CAS  PubMed  Google Scholar 

  22. Novoa PA, Grinyo JM, Ramos FJ, Errasti P, Franco A, Aldana G, et al. De novo use of everolimus with elimination or minimization of cyclosporine in renal transplant recipients. Transplant Proc. 2011;43:3331–9.

    Article  CAS  PubMed  Google Scholar 

  23. Langer RM, Hene R, Vitko S, Christiaans M, Tedesco-Silva Jr H, Ciechanowski K, et al. Everolimus plus early tacrolimus minimization: a phase III, randomized, open-label, multicentre trial in renal transplantation. Transpl Int. 2012;25:592–602.

    Article  CAS  PubMed  Google Scholar 

  24. Budde K, Rath T, Sommerer C, Haller H, Reinke P, Witzke O, et al. 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.

    Article  CAS  PubMed  Google Scholar 

  25. Budde K, Sommerer C, Rath T, Reinke P, Haller H, Witzke O, et al. Renal function to 5 years after late conversion of kidney transplant patients to everolimus: a randomized trial. J Nephrol. 2015;28:115–23.

    Article  CAS  PubMed  Google Scholar 

  26. Salvadori M, Scolari MP, Bertoni E, Citterio F, Rigotti P, Cossu M, et al. Everolimus with very low-exposure cyclosporine a in de novo kidney transplantation: a multicenter, randomized, controlled trial. Transplantation. 2009;88:1194–202.

    Article  CAS  PubMed  Google Scholar 

  27. Ponticelli C, Salvadori M, Scolari MP, Citterio F, Rigotti P, Veneziano A, et al. Everolimus and minimization of cyclosporine in renal transplantation: 24-month follow-up of the EVEREST study. Transplantation. 2011;91:e72–e3.

    Article  PubMed  Google Scholar 

  28. Takahashi K, Uchida K, Yoshimura N, Takahara S, Teraoka S, Teshima R, et al. Efficacy and safety of concentration-controlled everolimus with reduced-dose cyclosporine in Japanese de novo renal transplant patients: 12-month results. Transplant Res. 2013;2:14.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  29. Holdaas H, Rostaing L, Seron D, Cole E, Chapman J, Fellstrom B, et al. Conversion of long-term kidney transplant recipients from calcineurin inhibitor therapy to everolimus: a randomized, multicenter, 24-month study. Transplantation. 2011;92:410–8.

    Article  CAS  PubMed  Google Scholar 

  30. Chadban SJ, Eris JM, Kanellis J, Pilmore H, Lee PC, Lim SK, et al. A randomized, controlled trial of everolimus-based dual immunosuppression versus standard of care in de novo kidney transplant recipients. Transpl Int. 2014;27:302–11.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. Bemelman FJ. de Maar EF, Press RR, van Kan HJ, ten Berge IJ, Homan van der Heide JJ, et al. Minimization of maintenance immunosuppression early after renal transplantation: an interim analysis. Transplantation. 2009;88:421–8.

    Article  PubMed  Google Scholar 

  32. Mjörnstedt L, Sorensen SS, von Zur MB, Jespersen B, Hansen JM, Bistrup C, et al. Improved renal function after early conversion from a calcineurin inhibitor to everolimus: a randomized trial in kidney transplantation. Am J Transplant. 2012;12:2744–53.

    Article  PubMed  Google Scholar 

  33. Nashan B. Induction therapy and mTOR inhibition: minimizing calcineurin inhibitor exposure in de novo renal transplant patients. Clin Transplant. 2013;27:16–29.

    Article  CAS  PubMed  Google Scholar 

  34. Paoletti E, Marsano L, Bellino D, Cassottana P, Cannella G. Effect of everolimus on left ventricular hypertrophy of de novo kidney transplant recipients: a 1 year, randomized, controlled trial. Transplantation. 2012;93:503–8.

    Article  CAS  PubMed  Google Scholar 

  35. Eisen H. Long-term cardiovascular risk in transplantation – insights from the use of everolimus in heart transplantation. Nephrol Dial Transplant. 2006;21 Suppl 3:iii9–13.

    PubMed  Google Scholar 

  36. Eisen HJ, Kobashigawa J, Starling RC, Pauly DF, Kfoury A, Ross H, et al. Everolimus versus mycophenolate mofetil in heart transplantation: a randomized, multicenter trial. Am J Transplant. 2013;13:1203–16.

    Article  CAS  PubMed  Google Scholar 

  37. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008;372:449–56.

    Article  CAS  PubMed  Google Scholar 

  38. Euvrard S, Boissonnat P, Roussoulieres A, Kanitakis J, Decullier E, Claudy A, et al. Effect of everolimus on skin cancers in calcineurin inhibitor-treated heart transplant recipients. Transpl Int. 2010;23:855–7.

    Article  CAS  PubMed  Google Scholar 

  39. Nashan B. Maximizing the clinical outcome with mTOR inhibitors in the renal transplant recipient: defining the role of calcineurin inhibitors. Transpl Int. 2004;17:279–85.

    Article  CAS  PubMed  Google Scholar 

  40. Nashan B, Gaston R, Emery V, Saemann MD, Mueller NJ, Couzi L, et al. Review of cytomegalovirus infection findings with mammalian target of rapamycin inhibitor-based immunosuppressive therapy in de novo renal transplant recipients. Transplantation. 2012;93:1075–85.

    Article  CAS  PubMed  Google Scholar 

  41. Brennan DC, Legendre C, Patel D, Mange K, Wiland A, McCague K, et al. Cytomegalovirus incidence between everolimus versus mycophenolate in de novo renal transplants: pooled analysis of three clinical trials. Am J Transplant. 2011;11:2453–62.

    Article  CAS  PubMed  Google Scholar 

  42. Nankivell BJ, Gruenewald SM, Allen RD, Chapman JR. Predicting glomerular filtration rate after kidney transplantation. Transplantation. 1995;59:1683–9.

    Article  CAS  PubMed  Google Scholar 

  43. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16:31–41.

    Article  CAS  PubMed  Google Scholar 

  44. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of Diet in Renal Disease Study Group. Ann Intern Med. 1999;130:461–70.

    Article  CAS  PubMed  Google Scholar 

  45. Rodrigo E, Fernandez-Fresnedo G, Ruiz JC, Pinera C, Heras M, de Francisco AL, et al. Assessment of glomerular filtration rate in transplant recipients with severe renal insufficiency by Nankivell, Modification of Diet in Renal Disease (MDRD), and Cockcroft-Gault equations. Transplant Proc. 2003;35:1671–2.

    Article  CAS  PubMed  Google Scholar 

  46. Pierrat A, Gravier E, Saunders C, Caira MV, Ait-Djafer Z, Legras B, et al. Predicting GFR in children and adults: a comparison of the Cockcroft-Gault, Schwartz, and modification of diet in renal disease formulas. Kidney Int. 2003;64:1425–36.

    Article  PubMed  Google Scholar 

  47. Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro III AF, Feldman HI, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150:604–12.

    Article  PubMed Central  PubMed  Google Scholar 

  48. Devereux RB, Alonso DR, Lutas EM, Gottlieb GJ, Campo E, Sachs I, et al. Echocardiographic assessment of left ventricular hypertrophy: comparison to necropsy findings. Am J Cardiol. 1986;57:450–8.

    Article  CAS  PubMed  Google Scholar 

  49. Savage DD, Garrison RJ, Kannel WB, Levy D, Anderson SJ, Stokes 3rd J, et al. The spectrum of left ventricular hypertrophy in a general population sample: the Framingham Study. Circulation. 1987;75:I26–33.

    CAS  PubMed  Google Scholar 

  50. Nagueh SF, Appleton CP, Gillebert TC, Marino PN, Oh JK, Smiseth OA, et al. Recommendations for the evaluation of left ventricular diastolic function by echocardiography. J Am Soc Echocardiogr. 2009;22:107–33.

    Article  PubMed  Google Scholar 

  51. Nashan B, Citterio F, et al. Wound healing complications and the use of mammalian target of rapamycin inhibitors in kidney transplantation: a critical review of the literature. Transplantation. 2012;94:547–61.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The study was funded by Novartis Pharma GmbH, Nürnberg, Germany. We thank Dhaval Gupta and Daniel Baeumer of Novartis for providing medical writing assistance.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudia Sommerer.

Additional information

Competing interests

CS, BS, DD, PS, IH, FT and BN received investigator fees from Novartis.

Authors’ contributions

FT is the principal investigator of the trial. CS, BS, DD, PS, IH, BN, and FT participated in the study design/implementation/conduct of the ATHENA study. All authors contributed to the review of the protocol and approved the final manuscript.

Additional file

Additional file 1:

List of ethics committees. (PDF 184 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sommerer, C., Suwelack, B., Dragun, D. et al. Design and rationale of the ATHENA study – A 12-month, multicentre, prospective study evaluating the outcomes of a de novo everolimus-based regimen in combination with reduced cyclosporine or tacrolimus versus a standard regimen in kidney transplant patients: study protocol for a randomised controlled trial. Trials 17, 92 (2016). https://doi.org/10.1186/s13063-016-1220-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13063-016-1220-9

Keywords