220 results
Filters • 1
Sort By
Results Per Page
Filters
220 results
1
Download the following citations:
Email the following citations:
Print the following citations:
See all 16 Highlighted Expert Reviews articles matching your criteria
...
  • Stumpf J
  • Budde K
  • Witzke O
  • Sommerer C
  • Vogel T
  • et al.
E Clinical Medicine. 2023 Dec 22;67:102381 doi: 10.1016/j.eclinm.2023.102381.
BACKGROUND:

Optimal initial tacrolimus dosing and early exposure of tacrolimus after renal transplantation is not well studied.

METHODS:

In this open-label, 6 months, multicenter, randomized controlled, non-inferiority study, we randomly assigned 432 renal allograft recipients to receive basiliximab induction, mycophenolate and steroids and either standard prolonged-release tacrolimus (trough levels: 7-9 ng/ml; Standard Care arm), or an initial 7-day fixed 5 mg/day dose of prolonged-release tacrolimus followed by lower tacrolimus predose levels (trough levels: 5-7 ng/ml; Slow & Low arm). The primary end point was the combined incidence rate of biopsy-proven acute rejections (BPAR; including borderline), graft failure, or death at 6 months with a non-inferiority margin of 12.5%. (EudraCT-Nr: 2013-001770-19.

FINDINGS:

The combined primary endpoint in the Slow & Low arm was non-inferior compared to the Standard Care arm (22.1% versus 20.7%; difference: 1.4%, 90% CI -5.5% to 8.3%). The overall rate of BPAR including borderlines was similar (Slow & Low 17.4% versus Standard Care 16.6%). Safety parameters such as delayed graft function, kidney function, donor specific HLA-antibodies, infections, or post-transplantation diabetes mellitus did not differ.

INTERPRETATION:

This is the first study to show that an initial fixed dose of 5 mg per day followed by lower tacrolimus exposure is non-inferior compared to standard tacrolimus therapy and equally efficient and safe within 6 months after renal transplantation. These data suggest that therapeutic drug monitoring for prolonged release tacrolimus can be abandoned until start of the second week after transplantation.

FUNDING:

Investigator-initiated trial, financial support by Astellas Pharma GmbH.

  • Hirt-Minkowski P
  • Handschin J
  • Stampf S
  • Hopfer H
  • Menter T
  • et al.
J Am Soc Nephrol. 2023 Aug 1;34(8):1456-1469 doi: 10.1681/ASN.0000000000000160.
CET Conclusion
Reviewer: Mr Simon Knight, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: Whilst many studies have demonstrated associations between biomarkers and acute rejection in renal transplantation, few have prospectively assessed the value of routine monitoring. This study randomised de novo kidney recipients to monitoring with urine CXCL10, or to routine care. In the control arm, CXCL10 measurements were taken but concealed. In the study arm, response to CXCL10 measurements was protocolised, with a biopsy undertaken in patients with elevated levels to inform clinical management. The authors found no difference in one-year clinical outcomes in either intent-to-treat or per-protocol analysis with monitoring. This is an interesting study as it highlights the challenges of translating biomarker association into real-world clinical monitoring. It is possible that the timepoints selected for monitoring, or the frequency of monitoring, was not optimum. Successful monitoring also requires adequate response to an abnormal result – both by undertaking biopsy and treating abnormal findings. In this study, 23% indicated biopsies were not completed, and 24% detected rejection episodes were not treated. Successful monitoring also assumes that the biomarker detects an abnormality (e.g. rejection) at a stage where intervention can have an impact on clinical outcome.
Aims: This study aimed to examine whether renal allograft monitoring by urine CXCL10 led to an improvement in clinical outcomes posttransplantation.
Interventions: Participants were randomised into two groups: the intervention arm, where a renal allograft biopsy were performed if levels of urine CXCL10 were above a predefined cutoff without the presence of confounders; and the control arm, in which the urine CXCL10 analyses was also assessed but the results were concealed.
Participants: 241 adult kidney transplant recipients.
Outcomes: The primary outcome was a combined endpoint at 1 year following transplantation, including death-censored graft loss, clinical rejection, acute rejection, chronic active T-cell–mediated rejection, development of de novo donor-specific HLA antibodies, or an estimated glomerular filtration rate (eGFR) of 25 ml/min.
Follow Up: 1 year
SIGNIFICANCE STATEMENT:

This study is the first randomized controlled trial to investigate the clinical utility of a noninvasive monitoring biomarker in renal transplantation. Although urine CXCL10 monitoring could not demonstrate a beneficial effect on 1-year outcomes, the study is a rich source for future design of trials aiming to explore the clinical utility of noninvasive biomarkers. In addition, the study supports the use of urine CXCL10 to assess the inflammatory status of the renal allograft.

BACKGROUND:

Urine CXCL10 is a promising noninvasive biomarker for detection of renal allograft rejection. The aim of this study was to investigate the clinical utility of renal allograft monitoring by urine CXCL10 in a randomized trial.

METHODS:

We stratified 241 patients, 120 into an intervention and 121 into a control arm. In both arms, urine CXCL10 levels were monitored at three specific time points (1, 3, and 6 months post-transplant). In the intervention arm, elevated values triggered performance of an allograft biopsy with therapeutic adaptations according to the result. In the control arm, urine CXCL10 was measured, but the results concealed. The primary outcome was a combined end point at 1-year post-transplant (death-censored graft loss, clinical rejection between month 1 and 1-year, acute rejection in 1-year surveillance biopsy, chronic active T-cell-mediated rejection in 1-year surveillance biopsy, development of de novo donor-specific HLA antibodies, or eGFR <25 ml/min).

RESULTS:

The incidence of the primary outcome was not different between the intervention and the control arm (51% versus 49%; relative risk (RR), 1.04 [95% confidence interval, 0.81 to 1.34]; P = 0.80). When including 175 of 241 (73%) patients in a per-protocol analysis, the incidence of the primary outcome was also not different (55% versus 49%; RR, 1.11 [95% confidence interval, 0.84 to 1.47]; P = 0.54). The incidence of the individual end points was not different as well.

CONCLUSIONS:

This study could not demonstrate a beneficial effect of urine CXCL10 monitoring on 1-year outcomes (ClinicalTrials.gov_ NCT03140514 ).

  • Bezstarosti S
  • Meziyerh S
  • Reinders MEJ
  • Voogt-Bakker K
  • Groeneweg KE
  • et al.
Hla. 2023 Jul;102(1):3-12 doi: 10.1111/tan.15008.

Recently, the randomized phase-II Triton study demonstrated that mesenchymal stromal cell (MSC) therapy facilitated early tacrolimus withdrawal in living donor kidney transplant recipients. The current sub-study analyzed formation of de novo donor-specific HLA antibodies (dnDSA) in the context of the degree of HLA eplet mismatches. At the time of protocol biopsy at 6 months, 7/29 patients (24%) in the MSC group and 1/27 patient (3.7%) in the control group had developed dnDSA. In the MSC group, all dnDSA were anti-HLA-DQ; two patients had anti-DQ alone and five patients combined with anti-class I, HLA-DR or -DP. Despite excess dnDSA formation in the MSC-arm of the study, the evolution of eGFR (CKD-EPI) and proteinuria were comparable 2 years posttransplant. All dnDSA were complement-binding and three patients had antibody-mediated rejection in the protocol biopsy, but overall rejection episodes were not increased. Everolimus had to be discontinued in nine patients because of toxicity, and tacrolimus was reintroduced in six patients because of dnDSA formation. The HLA-DQ eplet mismatch load independently associated with dnDSA (adjusted hazard ratio = 1.07 per eplet mismatch, p = 0.008). A threshold of ≥11 HLA-DQ eplet mismatches predicted subsequent dnDSA in all 11 patients in the MSC group, but specificity was low (44%). Further research is warranted to explore HLA molecular mismatch load as a biomarker to guide personalized maintenance immunosuppression in kidney transplantation.

  • Kim HW
  • Lee J
  • Heo SJ
  • Kim BS
  • Huh KH
  • et al.
Sci Rep. 2023 May 11;13(1):7682 doi: 10.1038/s41598-023-34804-6.
CET Conclusion
Reviewer: Mr John Fallon, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: The investigators found both groups IVIG+ and IVIG- were associated with dnDSA MFI, but that the addition of IVIG to rituximab had no added benefit for dnDSA reduction at either 3 or 12 months. This reduction is significant when they were compared with a matched group of retrospective controls. Between the two groups they also found no difference in their secondary outcome measures of eGFR at 3 or 12 months, with no episodes of anti-body mediated rejection (ABMR) in the study cohort. They also reported no difference in protein-creatinine ratio. However, then generalisability of the study is somewhat limited due to its small sample size and possibility of selection bias given that nearly all the participants were living-related kidney transplant recipients. No episodes of ABMR occurred, which due to the size of the trial isn’t entirely surprising but given a core part of pre-emptive dnDSA reduction is to hopefully reduce the incidence and severity of ABMR it is hard to assess the potential importance of the interventions. There is also no mention or inclusion of the ABMR rate in their retrospectively matched control cohort which may have been of interest. The study contributes to baseline data on the potential benefit of pre-emptive treatment of dnDSA, however, a larger randomised trial of rituximab vs placebo in an adult population would be of benefit.
Aims: This study aimed to evaluate the efficacy of pre-emptive treatment of De Novo DSA (dnDSA) using combination of high-dose intravenous immunoglobulin and rituximab (IVIG+) or rituximab alone (IVIG-) in reduction of dnDSA titre at 3 and 12 months after treatment compared to retrospective controls.
Interventions: Both groups received rituximab (375mg/m2) on day 0, and the IVIG+ group additionally received high-dose IVIG (2g/kg) after rituximab infusion.
Participants: 50 adult kidney recipients with functioning graft (eGFR >20 ml/min/1.73 m2) and subclinical class II dnDSA with mean fluorescent intensity (MFI) ≥1000 of the DR or DQ DSA.
Outcomes: The primary outcome measure was dnDSA titre at 3 and 12 months. Secondary outcomes were changes in eGFR and incidence of anti-body mediated rejection.
Follow Up: Participants were followed-up for 12 months.

De novo donor-specific antibody (dnDSA) is associated with a higher risk of kidney graft failure. However, it is unknown whether preemptive treatment of subclinical dnDSA is beneficial. Here, we assessed the efficacy of high-dose intravenous immunoglobulin (IVIG) and rituximab combination therapy for subclinical dnDSA. An open-label randomized controlled clinical trial was conducted at two Korean institutions. Adult (aged ≥ 19 years) kidney transplant patients with subclinical class II dnDSA (mean fluorescence intensity ≥ 1000) were enrolled. Eligible participants were randomly assigned to receive rituximab or rituximab with IVIG at a 1:1 ratio. The primary endpoint was the change in dnDSA titer at 3 and 12 months after treatment. A total of 46 patients (24 for rituximab and 22 for rituximab with IVIG) were included in the analysis. The mean baseline estimated glomerular filtration rate was 66.7 ± 16.3 mL/min/1.73 m2. The titer decline of immune-dominant dnDSA at 12 months in both the preemptive groups was significant. However, there was no difference between the two groups at 12 months. Either kidney allograft function or proteinuria did not differ between the two groups. No antibody-mediated rejection occurred in either group. Preemptive treatment with high-dose IVIG combined with rituximab did not show a better dnDSA reduction compared with rituximab alone.Trial registration: IVIG/Rituximab versus Rituximab in Kidney Transplant With de Novo Donor-specific Antibodies (ClinicalTrials.gov Identifier: NCT04033276, first trial registration (26/07/2019).

  • Stringer D
  • Gardner L
  • Shaw O
  • Clarke B
  • Briggs D
  • et al.
E Clinical Medicine. 2023 Jan 12;56:101819 doi: 10.1016/j.eclinm.2022.101819.
CET Conclusion
Reviewer: Mr John O'Callaghan, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: This is a very interesting study conducted across 13 transplant centres in the UK with 3-year graft survival as the primary outcome. The study tested the hypothesis that screening for the development of Donor Specific Antibodies (DSA), combined with an intervention to improve adherence and optimised oral treatment would prevent transplant failure. Patients developing DSA in the “biomarker-led care” group were changed to maximum tolerated doses of MMF/mycophenolic acid (MPA) and tacrolimus, and were encouraged to take prednisolone 20 mg daily for two weeks, reducing by 5 mg down to their previous maintenance dose. 2037 patients were randomised. The study again confirmed that the development of DSA after transplantation was strongly predictive of a poorer long term graft survival. The key output from the study however is the indication that measures taken in this at-risk group did not improve the outcome and prevent the inevitable negative impact on graft survival.
Aims: This study aimed to determine whether allograft failure could be prevented through routine surveillance for human leukocyte antigens antibodies (HLA Ab), in combination with an intervention to improve adherence and optimization of immunosuppression.
Interventions: Participants were randomised to either the blinded standard care (SC) arm or the unblinded biomarker led care (BLC) arm.
Participants: 2037 renal transplant patients.
Outcomes: The primary endpoint was graft failure. The secondary endpoints included time to graft failure, patient survival, levels of graft dysfunction or change in eGFR, presence of biopsy-proven acute rejection, adverse effect profiles, the cost effectiveness of the screening/treatment protocol, and the impact of HLA Ab screening and treatment on the patients’ drug therapy adherence and their perceptions of risk to the health of the transplant.
Follow Up: 64 months
BACKGROUND:

3% of kidney transplant recipients return to dialysis annually upon allograft failure. Development of antibodies (Ab) against human leukocyte antigens (HLA) is a validated prognostic biomarker of allograft failure. We tested whether screening for HLA Ab, combined with an intervention to improve adherence and optimization of immunosuppression could prevent allograft failure.

METHODS:

Prospective, open-labelled randomised biomarker-based strategy (hybrid) trial in 13 UK transplant centres [EudraCT (2012-004308-36) and ISRCTN (46157828)]. Patients were randomly allocated (1:1) to unblinded or double-blinded arms and screened every 8 months. Unblinded HLA Ab+ patients were interviewed to encourage medication adherence and had tailored optimisation of Tacrolimus, Mycophenolate mofetil and Prednisolone. The primary outcome was time to graft failure in an intention to treat analysis. The trial had 80% power to detect a hazard ratio of 0.49 in donor specific antibody (DSA)+ patients.

FINDINGS:

From 11/9/13 to 27/10/16, 5519 were screened for eligibility and 2037 randomised (1028 to unblinded care and 1009 to double blinded care). We identified 198 with DSA and 818 with non-DSA. Development of DSA, but not non-DSA was predictive of graft failure. HRs for graft failure in unblinded DSA+ and non-DSA+ groups were 1.54 (95% CI: 0.72 to 3.30) and 0.97 (0.54-1.74) respectively, providing no evidence of an intervention effect. Non-inferiority for the overall unblinded versus blinded comparison was not demonstrated as the upper confidence limit of the HR for graft failure exceeded 1.4 (1.02, 95% CI: 0.72 to 1.44). The only secondary endpoint reduced in the unblinded arm was biopsy-proven rejection.

INTERPRETATION:

Intervention to improve adherence and optimize immunosuppression does not delay failure of renal transplants after development of DSA. Whilst DSA predicts increased risk of allograft failure, novel interventions are needed before screening can be used to direct therapy.

FUNDING:

The National Institute for Health Research Efficacy and Mechanism Evaluation programme grant (ref 11/100/34).

  • Nickerson PW
  • Böhmig GA
  • Chadban S
  • Kumar D
  • Mannon RB
  • et al.
Trials. 2022 Dec 22;23(1):1042 doi: 10.1186/s13063-022-06897-3.
BACKGROUND:

Chronic active antibody-mediated rejection (AMR) is a major cause of graft loss with no approved drugs for its treatment. Currently, off-label regimens are used, reflecting the high unmet need for effective therapies based on well-controlled trials. Clazakizumab is a high-affinity, humanized monoclonal antibody that binds interleukin-6 and decreases donor-specific antibody (DSA) production and inflammation. Phase 2 pilot studies of clazakizumab in kidney transplant recipients with chronic active AMR suggest modulation of DSA, stabilization of glomerular filtration rate (GFR), and a manageable safety profile. We report the design of the Phase 3 IMAGINE study (NCT03744910) to evaluate the safety and efficacy of clazakizumab for the treatment of chronic active AMR.

METHODS:

IMAGINE is a multicenter, double-blind trial of approximately 350 kidney transplant recipients with chronic active AMR (Banff chronic glomerulopathy [cg] >0 with concurrent positive human leukocyte antigen DSA) randomized 1:1 to receive clazakizumab or placebo (12.5 mg subcutaneous once every 4 weeks). The event-driven trial design will follow patients until 221 occurrences of all-cause graft loss are observed, defined as return to dialysis, graft nephrectomy, re-transplantation, estimated GFR (eGFR) <15 mL/min/1.73m2, or death from any cause. A surrogate for graft loss (eGFR slope) will be assessed at 1 year based on prior modeling validation. Secondary endpoints will include measures of pharmacokinetics/pharmacodynamics. Recruitment is ongoing across North America, Europe, Asia, and Australia.

DISCUSSION:

IMAGINE represents the first Phase 3 clinical trial investigating the safety and efficacy of clazakizumab in kidney transplant recipients with chronic active AMR, and the largest placebo-controlled trial in this patient population. This trial includes prognostic biomarker enrichment and uniquely utilizes the eGFR slope at 1 year as a surrogate endpoint for graft loss, which may accelerate the approval of a novel therapy for patients at risk of graft loss. The findings of this study will be fundamental in helping to address the unmet need for novel therapies for chronic active AMR.

TRIAL REGISTRATION:

ClinicalTrials.gov NCT03744910 . Registered on November 19, 2018.

  • Masset C
  • Dantal J
  • Soulillou JP
  • Walencik A
  • Delbos F
  • et al.
Front Immunol. 2022 Nov 28;13:1021481 doi: 10.3389/fimmu.2022.1021481.

Whilst calcineurin inhibitors (CNI) are the cornerstone of immunosuppressive maintenance therapy in kidney transplantation, several studies have investigated the safety of CNI withdrawal in order to avoid their numerous side effects. In this context, we performed several years ago a clinical randomized trial evaluating CNI weaning in stable kidney transplant recipients without anti-HLA immunization. The trial was interrupted prematurely due to a high number of de novo DSA (dnDSA) and biopsy proven acute rejection (BPAR) in patients who underwent tacrolimus weaning, resulting in treatment for rejection and resumption of tacrolimus. We report here the long-term outcomes of patients included in this clinical trial. Ten years after randomization, all patients are alive with a functional allograft. They all receive tacrolimus therapy except one with recurrent cutaneous neoplasia issues. Long-term eGFR was comparable between patients of the two randomized groups (46.4 ml/min vs 42.8 ml/min). All dnDSA that occurred during the study period became non-detectable and all rejections episodes were reversed. The retrospective assessment of HLA DQ single molecule epitope mismatching determined that a majority of patients who developed dnDSA after tacrolimus withdrawal would have been considered at high immunological risk. Minimization of immunosuppression remains a challenging objective, mainly because of the issues to properly select very low immunological risk patients. Valuable improvements have been made the last decade regarding evaluation of the allograft rejection notably through the determination of numerous at-risk biomarkers. However, even if the impact of such tools still need to be clarify in clinical routine, they may permit an improvement in patients' selection for immunosuppression minimization without increasing the risk of allograft rejection.

  • de Weerd AE
  • Fatly ZA
  • Boer-Verschragen M
  • Kal-van Gestel JA
  • Roelen DL
  • et al.
Transpl Int. 2022 Oct 24;35:10839 doi: 10.3389/ti.2022.10839.
CET Conclusion
Reviewer: Mr Keno Mentor, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: This pilot study explores the possibility of reducing immunosuppression therapy in patients undergoing kidney transplantation. With previous studies showing that weaning of tacrolimus results in significantly poorer graft and patient survival, this study focused on withdrawal of MMF in those patients with low immunological risk. It was intended as a feasibility study for a larger trial, but the findings suggest that with careful patient selection, long-term graft survival can be achieved with tacrolimus monotherapy while significantly reducing the incidence of infectious complications. Being a pilot study, the main weakness is the small sample size. The older patient cohort (average age 59) means the conclusions may also not be generalisable to younger patients. However, the findings did demonstrate the feasibility of investigating this study question in a larger non-inferiority trial.
Aims: This study aimed to examine the feasibility and safety of tacrolimus (TAC) monotherapy in renal transplant patients with low immunological risk.
Interventions: Participants were randomly assigned to either TAC/mycophenolate mofetil (MMF) or to taper and discontinue MMF at month 9.
Participants: 79 kidney transplant recipients.
Outcomes: The main outcomes of interest were biopsy-proven acute rejection (BPAR), kidney function, hospital admission, infectious burden, patient survival, death-censored graft survival and proteinuria.
Follow Up: 5 years

In this randomized-controlled pilot study, the feasibility and safety of tacrolimus monotherapy in immunologically low-risk kidney transplant recipients was evaluated [NTR4824, www.trialregister.nl]. Low immunological risk was defined as maximal 3 HLA mismatches and the absence of panel reactive antibodies. Six months after transplantation, recipients were randomized if eGFR >30 ml/min, proteinuria <50 mg protein/mmol creatinine, no biopsy-proven rejection after 3 months, and no lymphocyte depleting therapy given. Recipients were randomized to tacrolimus/mycophenolate mofetil (TAC/MMF) or to taper and discontinue MMF at month 9 (TACmono). 79 of the 121 recipients were randomized to either TACmono (n = 38) or TAC/MMF (n = 41). Mean recipient age was 59 years and 59% received a living donor transplant. The median follow-up was 62 months. After randomization, 3 TACmono and 4 TAC/MMF recipients experienced a biopsy-proven rejection. At 5 years follow-up, patient survival was 84% in TACmono versus 76% in TAC/MMF with death-censored graft survival of 97% for both groups and no differences in eGFR and proteinuria. Eleven TACmono recipients had an infectious episode versus 22 TAC/MMF recipients (p < 0.03). Donor-specific anti-HLA antibodies were not detected during follow-up in both groups. Tacrolimus monotherapy in selected immunologically low-risk kidney transplant recipients appears safe and reduces the number of infections.

  • Mayer KA
  • Budde K
  • Halloran PF
  • Doberer K
  • Rostaing L
  • et al.
Trials. 2022 Apr 8;23(1):270 doi: 10.1186/s13063-022-06198-9.
BACKGROUND:

Antibody-mediated rejection (ABMR) is a cardinal cause of renal allograft loss. This rejection type, which may occur at any time after transplantation, commonly presents as a continuum of microvascular inflammation (MVI) culminating in chronic tissue injury. While the clinical relevance of ABMR is well recognized, its treatment, particularly a long time after transplantation, has remained a big challenge. A promising strategy to counteract ABMR may be the use of CD38-directed treatment to deplete alloantibody-producing plasma cells (PC) and natural killer (NK) cells.

METHODS:

This investigator-initiated trial is planned as a randomized, placebo-controlled, double-blind, parallel-group, multi-center phase 2 trial designed to assess the safety and tolerability (primary endpoint), pharmacokinetics, immunogenicity, and efficacy of the fully human CD38 monoclonal antibody felzartamab (MOR202) in late ABMR. The trial will include 20 anti-HLA donor-specific antibody (DSA)-positive renal allograft recipients diagnosed with active or chronic active ABMR ≥ 180 days post-transplantation. Subjects will be randomized 1:1 to receive felzartamab (16 mg/kg per infusion) or placebo for a period of 6 months (intravenous administration on day 0, and after 1, 2, 3, 4, 8, 12, 16, and 20 weeks). Two follow-up allograft biopsies will be performed at weeks 24 and 52. Secondary endpoints (preliminary assessment) will include morphologic and molecular rejection activity in renal biopsies, immunologic biomarkers in the blood and urine, and surrogate parameters predicting the progression to allograft failure (slope of renal function; iBOX prediction score).

DISCUSSION:

Based on the hypothesis that felzartamab is able to halt the progression of ABMR via targeting antibody-producing PC and NK cells, we believe that our trial could potentially provide the first proof of concept of a new treatment in ABMR based on a prospective randomized clinical trial.

TRIAL REGISTRATION:

EU Clinical Trials Register (EudraCT) 2021-000545-40 . Registered on 23 June 2021.

CLINICALTRIALS:

gov NCT05021484 . Registered on 25 August 2021.

  • Nunes Ficher K
  • Dreige Y
  • Gessolo Lins PR
  • Nicolau Ferreira A
  • Toniato de Rezende Freschi J
  • et al.
Transplantation. 2022 Feb 1;106(2):381-390 doi: 10.1097/TP.0000000000003714.
CET Conclusion
Reviewer: Mr John O'Callaghan, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: This paper reports the 5-year outcomes from a previously published trial comparing 3 different immune suppression regimens. The number lost to follow up was small and similar between the study arms. The primary outcome was a composite “treatment failure” defined as biopsy proven rejection >1A, graft loss, death or loss to follow up. There was no significant difference in the rate of this primary outcome. There was no significant difference GFR. There was however a higher risk of post-transplant diabetes with the groups receiving everolimus (30-33% versus 15%). In this group of low/moderate risk renal transplant recipients, all three immune suppression regimens were associated with acceptable outcomes. Similar proportions of each group were still receiving their randomised therapy at 5 years.
Aims: This post hoc analysis aimed to report the 5-year follow-up outcomes of a randomised controlled trial investigating the efficacy and safety of everolimus compared to mycophenolate in kidney transplant recipients receiving tacrolimus-based immunosuppressive regimen.
Interventions: Participants in the original trial were randomised to one of three groups: rabbit antithymocyte globulin ( r-ATG) plus everolimus, basiliximab plus everolimus, or basiliximab plus mycophenolate.
Participants: 300 de novo kidney transplant recipients.
Outcomes: The main outcomes included assessment of treatment failure, treated clinical acute rejection episodes, serum HLA antibodies, estimated glomerular filtration rate (eGFR), proteinuria, blood pressure, body mass index (BMI), high-density lipoprotein cholesterol (HDL)/ low-density lipoprotein cholesterol (LDL), posttransplant diabetes mellitus (PTDM), use of lipidlowering agents, major adverse cardiovascular events (MACEs), and malignancies.
Follow Up: 5 years
BACKGROUND:

The short-term efficacy and safety of everolimus in combination with tacrolimus have been described in several clinical trials. Yet, detailed long-term data comparing the use of everolimus or mycophenolate in kidney transplant recipients receiving tacrolimus are lacking.

METHODS:

This is a 5-y follow-up post hoc analysis of a prospective trial including 288 patients who were randomized to receive a single 3-mg/kg dose of rabbit antithymocyte globulin, tacrolimus, everolimus (EVR), and prednisone (rabbit antithymocyte globulin/EVR, n = 85); basiliximab, tacrolimus, everolimus, and prednisone (basiliximab/EVR, n = 102); or basiliximab, tacrolimus, mycophenolate, and prednisone (basiliximab/mycophenolate, n = 101).

RESULTS:

There were no differences in the incidence of treatment failure (31.8% versus 40.2% versus 34.7%, P = 0.468), de novo donor-specific HLA antibodies (6.5% versus 11.7% versus 4.0%, P = 0.185), patient (92.9% versus 94.1% versus 92.1%, P = 0.854), and death-censored graft (87.1% versus 90.2% versus 85.1%, P = 0.498) survivals. Using a sensitive analysis, the trajectories of estimated glomerular filtration rate were comparable in the intention-to-treat (P = 0.145) and per protocol (P = 0.354) populations. There were no differences in study drug discontinuation rate (22.4% versus 30.4% versus 17.8%, P = 0.103).

CONCLUSIONS:

In summary, this analysis in a cohort of de novo low/moderate immunologic risk kidney transplant recipients suggests that the use of a single 3 mg/kg rabbit antithymocyte globulin dose followed by EVR combined with reduced tacrolimus concentrations was associated with similar efficacy and renal function compared with the standard of care immunosuppressive regimen.