Skip to main content

Evaluating the efficacy, safety and evolution of renal function with early initiation of everolimus-facilitated tacrolimus reduction in de novo liver transplant recipients: Study protocol for a randomized controlled trial

Abstract

Background

Introduction of calcineurin inhibitors had led to improved survival rates in liver transplant recipients. However, long-term use of calcineurin inhibitors is associated with a higher risk of chronic renal failure, neurotoxicity, de novo malignancies, recurrence of hepatitis C viral (HCV) infection and hepatocellular carcinoma. Several studies have shown that everolimus has the potential to provide protection against viral replication, malignancy, and progression of fibrosis, as well as preventing nephrotoxicity by facilitating calcineurin inhibitor reduction without compromising efficacy. The Hephaistos study evaluates the beneficial effects of early initiation of everolimus in de novo liver transplant recipients.

Methods/Design

Hephaistos is an ongoing 12-month, multi-center, open-label, controlled study aiming to enroll 330 de novo liver transplant recipients from 15 centers across Germany. Patients are randomized in a 1:1 ratio (7–21 days post-transplantation) to receive everolimus (trough levels 3–8 ng/mL) with reduced tacrolimus (trough levels <5 ng/mL), or standard tacrolimus (trough levels 6–10 ng/mL) after entering a run-in period (3–5 days post-transplantation). In the run-in period, patients are treated with induction therapy, mycophenolate mofetil, tacrolimus, and corticosteroids according to local practice. Randomization is stratified by HCV status and model of end-stage liver disease scores at transplantation. The primary objective of the study is to exhibit superior renal function (estimated glomerular filtration rate assessed by the Modification of Diet in Renal Disease (MDRD)-4 formula) with everolimus plus reduced tacrolimus compared to standard tacrolimus at Month 12. Other objectives are: to assess the incidence of treated biopsy-proven acute rejection, graft loss, or death; the incidences of components of the composite efficacy endpoint; renal function via estimated glomerular filtration rate using various formulae (MDRD-4, Nankivell, Cockcroft-Gault, chronic kidney disease epidemiology collaboration and Hoek formulae); the incidence of proteinuria; the incidence of adverse events and serious adverse events; the incidence and severity of cytomegalovirus and HCV infections and HCV-related fibrosis.

Discussion

This study aims to demonstrate superior renal function, comparable efficacy, and safety in de novo liver transplant recipients receiving everolimus with reduced tacrolimus compared with standard tacrolimus. This study also evaluates the antiviral benefit by early initiation of everolimus.

Trial registration

NCT01551212.

Peer Review reports

Background

Liver transplantation: challenges for long-term outcomes

Liver transplantation is the most preferred and well-established mode of treatment in patients with end-stage liver diseases. From 1968 to 2009, approximately 93,634 liver transplantations were carried out in 83,816 patients in Europe. Of these, 14,116 liver transplantations were performed in Germany [1] from across 24 centers [2]. Around 1,097 liver transplantations were performed in 2012 and the 1-year patient survival rate is 76.6% (733 of the 957 patients with known survival status) [3]. Analysis of data across centers in Germany (2007 to 2010) reveals that better 1-year overall survival rates were significantly correlated with larger center size (R2 = 0.09, P = 0.009), whereas in-house mortality (R2 = 0.007, P = 0.52) and 3-year survival rates (R2 = 0.05, P = 0.09) showed non-significant correlation between center volume and outcome [4]. Model of end-stage liver disease (MELD) score was introduced in Germany in 2006 for thorough and transparent liver allograft allocation. Following the introduction of MELD, mortality on the waiting list decreased; however, 1-year survival rates also decreased from 90% to less than 80% [5,6]. The match MELD threshold increased from 25 to 34 [6,7]. A vast number (~40%) of patients are transplanted with a MELD score >30 [6,8].

Greater surgical expertise, better selection of patients, improved post-liver transplantation management of complications [1], and introduction of potent immunosuppressants, antibiotics and antiviral drugs are the key reasons for improvement in the survival rates [9]. Calcineurin inhibitors (CNIs) are the mainstay of immunosuppressive therapy in liver transplantation. However, post-transplant long-term use of CNIs is associated with a higher risk of chronic renal failure in liver transplant recipients [10,11] and in renal transplantation recipients [12]. Among the liver transplant recipients, approximately 20% of patients develop chronic renal failure (glomerular filtration rate of 29 mL/min/1.73 m2) by 5 years [10], leading to increased mortality [13,14]. Prolonged CNI exposure is also associated with neurotoxicity, de novo malignancies, recurrence of hepatitis C viral (HCV) infection and hepatocellular carcinoma (HCC) [15], and an increased risk of metabolic complications [11]. Therefore, it is important to identify alternate immunosuppressive regimens that: (1) maintain efficacy similar to CNI and optimize renal function while reducing CNI exposure and thus related nephrotoxicity; (2) minimize CNI-associated adverse events; and (3) reduce the post-transplant recurrence of HCV and HCC and occurrence of de novo malignancies [15].

Eliminating/reducing calcineurin inhibitor exposure: mammalian target of rapamycin inhibitors

Mammalian target of rapamycin (mTOR) inhibitor (everolimus, sirolimus)-based CNI reduction or elimination is being practiced to overcome drug-induced adverse events. mTOR inhibitor-enabled reduced CNI exposure offers renal benefits without affecting efficacy in low-to-moderate risk de novo kidney transplant recipients [12]. Emerging data suggest that mTOR inhibitors offer antiviral benefits against BK virus, human papilloma virus, cytomegalovirus (CMV), human herpes virus 8 and several other herpes viruses [16]. Early initiation of mTOR inhibitor-based immunosuppression is more effective in reducing the risk of CMV infection and disease in solid organ transplant recipients [17]. Furthermore, a probable negative impact of mTOR inhibitors in post-operative surgical complications [15,18] was contradicted by findings from a single-center study in six liver transplant recipients, indicating that the rate of complications after major surgery is similar in patients receiving mTOR inhibitors to those not receiving mTOR inhibitors [19].

Everolimus in liver transplantation

Studies in de novo and maintenance liver transplant recipients demonstrated that everolimus facilitates CNI reduction/elimination without compromising efficacy (Table 1). Using an appropriate dose and switching to everolimus within 3 months of transplantation optimizes renal function and minimizes CNI-induced adverse events with comparable efficacy [20-32]. Other potential benefits of mTOR inhibitors related to HCV-related fibrosis, metabolic syndrome, and neurotoxicity have long-term implications for liver transplant recipients [15].

Table 1 Everolimus in liver transplantation

H2304, the registry study for everolimus use in liver transplantation, reported beneficial effects of everolimus [25]. Results from the H2304 study suggested that, despite the beneficial effects of everolimus initiation 30 ± 5 days post-transplantation, incidences of CMV and HCC recurrence were comparable (CMV: 4.9% versus 5.4%, P = 0.84; and HCC recurrence: 1.2% versus 1.2%, P = 1.0) between the everolimus plus reduced tacrolimus and the standard tacrolimus arms, respectively, at 24 months [25].

Initiation of everolimus earlier than 30 ± 5 days post-transplantation might provide antiviral benefits. Therefore, the Hephaistos study (NCT01551212; Efficacy of Everolimus in Combination With Tacrolimus in Liver Transplant Recipients) aims to establish the beneficial effects of early initiation of everolimus and the impact on the development or the rate of progression of fibrosis in HCV-positive recipients without affecting wound healing. A very low initial immunosuppressant dosage has been used, considering the relevant number of patients in Germany with a MELD score >30, who are in a poor health condition at the time of transplantation.

Methods/Design: Hephaistos study

Overview

Hephaistos (protocol version 3, 14 June 2013) is an ongoing, 12-month, multicenter, open-label, randomized, controlled study aimed to evaluate efficacy, safety, and evolution of renal function of everolimus with reduced tacrolimus in de novo liver transplant recipients. Patients undergoing a successful liver transplantation enter a run-in period between 3 and 5 days post-transplantation. During the run-in period, induction therapy, mycophenolate mofetil, tacrolimus and corticosteroids are initiated at the investigator’s discretion. Between 7 and 21 days post-transplantation, patients are randomized in a 1:1 ratio to receive either: (i) everolimus (trough level (C0) 3–8 ng/mL) with reduced tacrolimus (C0 <5 ng/mL), or (ii) standard tacrolimus (C0 6–10 ng/mL; Figure 1). Everolimus is initiated on the day of randomization and will be monitored throughout the study period (post 5 ± 2 days of everolimus/tacrolimus dose changes).

Figure 1
figure 1

Study design. *As per center practice. C0, trough levels; CS, corticosteroids; EVR, everolimus; HCV, hepatitis C virus; LTx, liver transplantation; M, month; MELD, model of end-stage liver disease; MMF, mycophenolate mofetil; RND, randomization; TAC, tacrolimus.

Study population

This study plans to enroll approximately 330 de novo liver transplant recipients, 165 in each study arm, from 15 transplant centers in Germany. The study population comprises adult patients aged between 18 and 65 years receiving a liver transplant from a deceased or living donor. At the time of randomization, only patients with an acceptable renal function defined as estimated glomerular filtration rate (eGFR) >30 mL/min/1.73 m2 assessed by the Modification of Diet in Renal Disease (MDRD)-4 formula [33] (obtained within 5 days before randomization) are eligible to enter the study. Inclusion and exclusion criteria at the time of study entry and randomization are summarized in Table 2. The study was approved by all competent Ethics Committees and regulatory authorities (Additional files 1 and 2). Informed consent is being obtained from all patients enrolled to the study by the investigators.

Table 2 Key inclusion and exclusion criteria for the Hephaistos study

Study objectives

This study is designed to evaluate the renal function, efficacy and safety of everolimus with reduced tacrolimus in liver transplant recipients. The primary objective of the study is to demonstrate superior eGFR (MDRD-4 formula) with the everolimus plus reduced tacrolimus regimen compared with standard tacrolimus at Month 12. The key secondary objective is to evaluate the incidence of treated biopsy-proven acute rejection, graft loss, or death at Month 12. Other secondary objectives are to evaluate: (i) incidences of components of the composite efficacy endpoint; (ii) renal function by eGFR using various formulae (MDRD-4, Nankivell, Cockcroft-Gault, chronic kidney disease epidemiology collaboration and Hoek formulae); (iii) incidence of proteinuria; (iv) incidence of adverse events and serious adverse events; and (v) incidence and severity of CMV and HCV infections and HCV-related fibrosis. Efficacy and safety objectives of the study are summarized in Table 3. All adverse events and serious adverse events occurring after administration of study treatment will be documented in case report forms and patient medical records for further monitoring.

Table 3 Objectives of the Hephaistos study

Randomization and immunosuppression

De novo liver transplant recipients are randomized 7–21 days post-transplantation to receive everolimus plus reduced tacrolimus or standard tacrolimus. All eligible patients are randomized using a validated system that automates the random assignment of treatment arms in the specified ratio. Randomization is stratified by HCV status (positive/negative) and the laboratory MELD score below/equal/above 30 at transplantation.

Immunosuppression up to the point of randomization comprises induction therapy, mycophenolate mofetil, tacrolimus, and corticosteroids administered according to local practice. Mycophenolic acid is discontinued at the time of randomization as per local practice. Patients in the everolimus plus reduced tacrolimus group are administered everolimus on the day of randomization at a dose of 1.0 mg b.i.d The target therapeutic range for everolimus is maintained at 3–8 ng/mL and dose adjustments will be monitored 5 ± 2 days post-randomization. Tacrolimus dose is adjusted to achieve a C0 <5 ng/mL after everolimus C0 is achieved. In the standard tacrolimus group, the target tacrolimus C0 is 6–10 ng/mL. Patients in both the groups receive corticosteroids at the investigator’s discretion. The treatment regimens will be continued until Month 12 post-randomization. Dose adjustments and interruptions are allowed only in patients who are unable to tolerate the protocol-specified dosing scheme. Everolimus dose will be: (i) interrupted in severe hematological adverse events until the condition is resolved; and (ii) permanently discontinued if a hemolytic uremic syndrome occurs. Tacrolimus dose will be adjusted if the whole blood levels are outside the target range and reduced in case of tacrolimus toxicity. Follow-up medical care is provided to all patients who prematurely discontinue from the study as per center practice.

Rejection episodes are treated as per local practice or at the investigator’s discretion. Other concomitant medications such as prophylactic treatment of CMV with valganciclovir, ganciclovir, CMV hyperimmune globulin, acyclovir or valacyclovir; Pneumocystis carinii with Bactrim® or equivalent (trimethoprim/sulfamethoxazole), oral candida with nystatin suspension and treatment of hepatitis B and C virus prophylaxis are permitted as per local practice.

Statistical analysis

This study tests the null hypothesis that there is zero treatment difference in the mean eGFR between the everolimus plus reduced tacrolimus and standard tacrolimus groups at Month 12 after baseline. The null hypothesis is tested with analysis of covariance with treatment, center, HCV class (positive/negative), and MELD (≤30 vs >30) as factors, and eGFR at Visit 1 (baseline) as a covariate. An alternative hypothesis assumes that the difference between the two arms is 7.0 mL/min/1.73 m2. A sample size of 105 in each group will have 80% power to detect a difference of 7.0 mL/min/1.73 m2 in the mean eGFR, assuming that the common standard deviation is 18.0 mL/min/1.73 m2 with a 5% two-sided significance level. Summary statistics of demographic and baseline characteristics, efficacy observations and measurements, safety observations and measurements, and pharmacokinetic measurements will be presented by treatment group. Categorical variables will be summarized by absolute and relative frequencies, and continuous variables will be summarized by descriptive statistics. Time-to-event data including rates of affected patients will be assessed by Kaplan-Meier statistics. Group comparisons will be performed using appropriate two-sided statistical tests. Physiological, laboratory and clinical assessments will be carried out as per plan (Additional file 3). Full analysis set will be used for the analysis of data.

Study conduct, analysis of study outcomes and documentation of the study results will be in accordance with the Declaration of Helsinki [34], Good Clinical Practice [35], and the local legal and regulatory requirements. Trial centers will be randomly audited by funding authorities. All data management activities will be carried out by a data monitoring committee according to the current standard operating procedures of Novartis Pharma GmbH, Nürnberg, Germany. The data monitoring committee has the right and duty to recommend study closure if necessary.

Sub-studies

The Hephaistos study will also include five sub-studies to evaluate:

  1. 1.

    Importance of non-human leukocyte antigen (HLA) antibodies targeting G-protein coupled receptor in liver transplant pathologies. This sub-study will determine: (i) the anti-AT1-receptor and anti-ETA-receptor antibodies status; (ii) correlation with immunologic and non-immunologic events; (iii) the correlation with histopathologic findings, if a biopsy is available; and (iv) functional outcome.

  2. 2.

    Cytochrome P450 (CYP450)-dependent vasoactive eicosanoids in serum and urine as a marker and mediator of nephrotoxicity after liver transplantation. This sub-study will determine: (i) CYP450-eicosanoid profile in plasma and urine and correlation with kidney and liver function as well as histopathologic findings if a biopsy is available; and (ii) genetic polymorphisms in CYP-isoforms (CYP3A4 and CYP3A5), drug transporters (ABCB1), and catechol-o-methyltransferase in donor and recipient, and correlation with renal and liver transplant outcomes.

  3. 3.

    Immunomodulatory effect of everolimus on natural killer cell subsets and plasma cytokine, chemokine and growth factor levels in patients after liver transplantation. The objectives of this sub-study are: (i) to determine alterations in the major natural killer cell subsets defined by CD56, CD16, and CD6 surface expression as well as in CD56 and CD6 expression on CD4+ or CD8+ T cells, respectively, in whole blood of patients before and after liver transplantation; (ii) to determine the individual course of the Th1, Th2, and Th17 response in combination with the chemokine and growth factor response in the plasma of liver transplant recipients; and (iii) to determine the correlation of these immune markers with the type of immunosuppression in both the treatment groups and with the clinical course after transplantation (rejection episodes).

  4. 4.

    Impact of everolimus on the development of alloimmunity and tolerance. The objectives of this sub-study are: (i) to determine the de novo HLA antibody development and its correlation with humoral rejection (for example, biliary alteration); (ii) to determine the changes in the T-cell activation marker sCD30, tolerance marker HLA-G, and regulatory antibody IgA-anti-Fab; and (iii) to determine the changes in regulatory T- and B-cell populations under everolimus and CNI minimization during the first year after transplantation. In addition, the study team plans to investigate the association between the immunological changes observed during the first year and outcome during the first 2 years after transplantation.

  5. 5.

    Immunomodulatory effect of everolimus on regulatory and innate lymphocyte populations and on CMV-specific T-cell immunity. The objective of this sub-study is to test if the immunosuppressive drug regimen differentially affects individual immunocompetence towards CMV, CMV-specific T-cell frequencies, phenotype, and functionality. The regulatory T-cells will be analyzed in parallel with CMV load analyses.

Blood samples will be drawn at screening, baseline, and Days 90 and 360 for the sub-studies and processed to obtain serum and/or plasma. For evaluating the genetic polymorphisms, blood samples from recipients and donors are collected in EDTA at screening. Liver biopsy is preserved if donor blood is not available. The protocol was amended in June 2013 to incorporate the details of the sub-studies.

Discussion

This study aims to demonstrate superior renal function, comparable efficacy, and safety in patients receiving everolimus plus reduced tacrolimus versus the standard tacrolimus regimens. This study also evaluates the antiviral benefit by initiation of everolimus as early as Day 7. This study allows very low initial immunosuppression because of a relevant number of high-MELD patients in Germany with a poor initial condition.

Trial status

Hephaistos is an ongoing study recruiting de novo liver transplant recipients at 15 centers across Germany. Currently 359 patients are screened and, of these, 156 patients are randomized (78 in each arm). At baseline, demographic characteristics such as mean age (54.04 versus 54.21 years), male patients (77.8% versus 62.5%), Caucasian patients (100% versus 95.8%) and mean body mass index (26.01 versus 26.81 kg/m2) were similar in the everolimus with reduced tacrolimus and standard tacrolimus regimens, respectively. All investigators are involved in final data interpretation and analysis. All trial results will be disclosed to the health authorities with subsequent publication in NIM and journals.

List of study sites

Universitaetsklinikum Hamburg-Eppendorf Hepatobiliare Chirurgie; Medizinische Hochschule Hannover; Charite Berlin Campus Virchow-Klinikum Allg.-,Visceral-,Transpl.-Chir; Universitatsklinikum Regensburg Chirurgie; Klinikum der Universitaet Heidelberg; Universitaetsklinikum Schleswig-Holstein Campus Kiel Allgemein- und Thoraxchirurgie; Universitatsklinikum Leipzig AoR Chirurgische Klinik II; Klinikum der Johannes-Gutenberg Universitaet; Universitatsklinikum Essen gGmbH Lebertransplantationsambulanz; Klinikum Grosshadern Muenchen Chirurgische Klinik Poliklinik; Universitaetsklinikum Frankfurt Chirurgie; Universitatsklinikum Tubingen Chirurgie; Universitatsklinikum Aachen Allgemein, Viszeral, Transpl.; Universitatsklinikum Erlangen Chirurgische Klinik; Universitatsklinikum Bonn Allg./Viszeral/Thorax/Gefass.

Abbreviations

ALT:

alanine aminotransferase

AP:

alkaline phosphatase

AST:

aspartate aminotransferase

BPAR:

biopsy-proven acute rejection

C0:

trough level

CG:

Cockcroft-Gault

CKD-EPI:

chronic kidney disease epidemiology collaboration

CMV:

cyto- megalovirus

CNI:

calcineurin inhibitor

CsA:

cyclosporine

CYP450:

cytochrome P450

eGFR:

estimated glomerular filtration rate

ESRD:

end stage renal disease

EVR:

everolimus

HCC:

hepatocellular carcinoma

HCV:

hepatitis C virus

HIV:

human immunodeficiency virus

HLA:

human leukocyte antigen

MDRD:

Modification of Diet in Renal Disease

MELD:

model of end-stage liver disease

MMF:

mycophenolate mofetil

mTOR:

mammalian target of rapamycin

NODM:

new onset diabetes mellitus

TAC:

tacrolimus

tBPAR:

treated biopsy-proven acute rejection

ULN:

upper limit of normal

References

  1. Adam R, Karam V, Delvart V, O'Grady J, Mirza D, Klempnauer J, et al. Evolution of indications and results of liver transplantation in Europe: a report from the European Liver Transplant Registry (ELTR). J Hepatol. 2012;57:675–88.

    Article  PubMed  Google Scholar 

  2. German foundation for organ transplantation. Assessed in December 2014, at [http://www.dso.de/servicecenter/krankenhaeuser/transplantationszentren.html]

  3. AQUA German Hospital Quality Report 2012. Assessed in December 2014, at [http://www.sqg.de/sqg/upload/CONTENT/EN/Quality-Report/AQUA-German-Hospital-Quality-Report-2012.pdf].

  4. Nijboer A, Ulrich F, Bechstein WO, Schnitzbauer AA. Volume and outcome relation in German liver transplant centers: what lessons can be learned? Transpl Res. 2014;3:5.

    Article  Google Scholar 

  5. Weismuller TJ, Negm A, Becker T, Barg-Hock H, Klempnauer J, Manns MP, et al. The introduction of MELD-based organ allocation impacts 3-month survival after liver transplantation by influencing pretransplant patient characteristics. Transpl Int. 2009;22:970–8.

    Article  PubMed  Google Scholar 

  6. Schlitt HJ, Loss M, Scherer MN, Becker T, Jauch KW, Nashan B, et al. Current developments in liver transplantation in Germany: MELD-based organ allocation and incentives for transplant centres. Z Gastroenterol. 2011;49:30–8.

    Article  CAS  PubMed  Google Scholar 

  7. Bahra M, Neuhaus P. Liver transplantation in the high MELD era: a fair chance for everyone? Langenbecks Arch Surg. 2011;396:461–5.

    Article  PubMed  Google Scholar 

  8. Rahmel A. Eurotransplant annual report 2012. Published annually by annual report/Eurotransplant International Foundation. Leiden: Eurotransplant Foundation. -III., graf., tab.

  9. Braun F, Teren K, Wilms P, Gunther R, Allmann J, Broering DC, et al. Quality of life after liver transplantation. Transplant Proc. 2009;41:2564–6.

    Article  CAS  PubMed  Google Scholar 

  10. 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 

  11. Farkas SA, Schnitzbauer AA, Kirchner G, Obed A, Banas B, Schlitt HJ. Calcineurin inhibitor minimization protocols in liver transplantation. Transpl Int. 2009;22:49–60.

    Article  PubMed  Google Scholar 

  12. Nashan B. Induction therapy and mTOR inhibition: minimizing calcineurin inhibitor exposure in de novo renal transplant patients. Clin Transplant. 2013;27 Suppl 25:16–29.

    Article  CAS  PubMed  Google Scholar 

  13. Pawarode A, Fine DM, Thuluvath PJ. Independent risk factors and natural history of renal dysfunction in liver transplant recipients. Liver Transpl. 2003;9:741–7.

    Article  PubMed  Google Scholar 

  14. Moreno JM, Cuervas-Mons V, Rubio E, Pons F, Herreros AT, Turrion VS, et al. Chronic renal dysfunction after liver transplantation in adult patients: prevalence, risk factors, and impact on mortality. Transpl Proc. 2003;35:1907–8.

    Article  CAS  Google Scholar 

  15. Klintmalm GB, Nashan B. The role of mTOR inhibitors in liver transplantation: reviewing the evidence. J Transplant. 2014;845438.

  16. Brennan DC, Aguado JM, Potena L, Jardine AG, Legendre C, Saemann MD, et al. Effect of maintenance immunosuppressive drugs on virus pathobiology: evidence and potential mechanisms. Rev Med Virol. 2013;23:97–125.

    Article  CAS  PubMed  Google Scholar 

  17. 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 

  18. Nashan B, Citterio F. 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 

  19. Nashan B. mTOR inhibitors and their role in modern concepts of immunosuppression. World J Surg. 2014;38:3199–201.

    Article  PubMed  Google Scholar 

  20. Levy G, Schmidli H, Punch J, Tuttle-Newhall E, Mayer D, Neuhaus P, et al. Safety, tolerability, and efficacy of everolimus in de novo liver transplant recipients: 12- and 36-month results. Liver Transpl. 2006;12:1640–8.

    Article  PubMed  Google Scholar 

  21. Masetti M, Montalti R, Rompianesi G, Codeluppi M, Gerring R, Romano A, et al. Early withdrawal of calcineurin inhibitors and everolimus monotherapy in de novo liver transplant recipients preserves renal function. Am J Transplant. 2010;10:2252–62.

    Article  CAS  PubMed  Google Scholar 

  22. Fischer L, Klempnauer J, Beckebaum S, Metselaar HJ, Neuhaus P, Schemmer P, et al. A randomized, controlled study to assess the conversion from calcineurin-inhibitors to everolimus after liver transplantation-PROTECT. Am J Transplant. 2012;12:1855–65.

    Article  CAS  PubMed  Google Scholar 

  23. Sterneck M, Kaiser GM, Heyne N, Richter N, Rauchfuss F, Pascher A, et al. Everolimus and early calcineurin inhibitor withdrawal: 3-year results from a randomized trial in liver transplantation. Am J Transplant. 2014;14:701–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. De Simone P, Nevens F, De Carlis L, Metselaar HJ, Beckebaum S, Saliba F, et al. Everolimus with reduced tacrolimus improves renal function in de novo liver transplant recipients: a randomized controlled trial. Am J Transplant. 2012;12:3008–20.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Saliba F, De Simone P, Nevens F, De Carlis L, Metselaar HJ, Beckebaum S, Jonas S, et al. Renal function at two years in liver transplant patients receiving everolimus: results of a randomized, multicenter study. Am J Transplant. 2013;13:1734–45.

    Article  CAS  PubMed  Google Scholar 

  26. Bilbao I, Sapisochin G, Dopazo C, Lazaro JL, Pou L, Castells L, et al. Indications and management of everolimus after liver transplantation. Transplant Proc. 2009;41:2172–6.

    Article  CAS  PubMed  Google Scholar 

  27. Casanovas T, Argudo A, Pena-Cala MC. Everolimus in clinical practice in long-term liver transplantation: an observational study. Transplant Proc. 2011;43:2216–9.

    Article  CAS  PubMed  Google Scholar 

  28. Castroagudin JF, Molina E, Romero R, Otero E, Tome S, Varo E. Improvement of renal function after the switch from a calcineurin inhibitor to everolimus in liver transplant recipients with chronic renal dysfunction. Liver Transpl. 2009;15:1792–7.

    Article  PubMed  Google Scholar 

  29. De Simone P, Carrai P, Precisi A, Petruccelli S, Baldoni L, Balzano E, et al. Conversion to everolimus monotherapy in maintenance liver transplantation: feasibility, safety, and impact on renal function. Transpl Int. 2009;22:279–86.

    Article  PubMed  Google Scholar 

  30. De Simone P, Metselaar HJ, Fischer L, Dumortier J, Boudjema K, Hardwigsen J, et al. Conversion from a calcineurin inhibitor to everolimus therapy in maintenance liver transplant recipients: a prospective, randomized, multicenter trial. Liver Transpl. 2009;15:1262–9.

    Article  PubMed  Google Scholar 

  31. Saliba F, Dharancy S, Lorho R, Conti F, Radenne S, Neau-Cransac M, et al. Conversion to everolimus in maintenance liver transplant patients: a multicenter, retrospective analysis. Liver Transpl. 2011;17:905–13.

    Article  PubMed  Google Scholar 

  32. Vallin M, Guillaud O, Morard I, Gagnieu MC, Mentha G, Adham M, et al. Tolerability of everolimus-based immunosuppression in maintenance liver transplant recipients. Clin Transpl. 2011;25:660–9.

    Article  CAS  Google Scholar 

  33. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D, et al. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Ann Intern Med. 1999;130:461–70.

    Article  CAS  PubMed  Google Scholar 

  34. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH). Guideline E6. Note for Guidance on good clinical practice (GCP). Published by the European Medicines Agency, 7 Westferry Circus, London, UK. Assessed in December 2014, at. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500002874.pdf.

  35. World Medical Association Declaration of Helsinki. Recommendations guiding physicians in biomedical research involving human subjects. Adopted by the 18th World Medical Assembly Helsinki, Finland, June 1964, amended by 48th General Assembly, Somerset West, South Africa 1996. JAMA. 1997;277:925–6.

Download references

Acknowledgements

The study was funded by Novartis Pharma GmbH, Nürnberg, Germany. We thank Aruna Meka, Novartis Healthcare Pvt. Ltd. India, for providing medical writing assistance.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bjorn Nashan.

Additional information

Competing interests

All authors are lead investigators in the Hephaistos study and received respective research funds/honorary fees. BN received honoraria from Astellas, Chiesi, Novartis and is a member of the speaker bureau of Astellas, Chiesi, and Novartis. PS received fees from Astellas, Novartis and Sanofi. FB received speaker fees and travel grants from Novartis. HS received speaker fees from Astellas, BMS, Novartis, and Roche. MD and PW are full-time employees at Novartis Pharmaceuticals Germany.

Authors’ contributions

BN is the representing investigator of this trial. All authors participated in the scientific design/implementation/conduct of the Hephaistos study as well as manuscript writing. All authors read and approved the final manuscript.

Additional files

Additional file 1:

List of ethics committees [in German].

Additional file 2:

List of ethics committees [in English].

Additional file 3:

Assessment schedule.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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

Nashan, B., Schemmer, P., Braun, F. et al. Evaluating the efficacy, safety and evolution of renal function with early initiation of everolimus-facilitated tacrolimus reduction in de novo liver transplant recipients: Study protocol for a randomized controlled trial. Trials 16, 118 (2015). https://0-doi-org.brum.beds.ac.uk/10.1186/s13063-015-0626-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s13063-015-0626-0

Keywords