Advanced Search
Clear
40 results
Filters
Sort By
Results Per Page
Filters
Advanced Search
Clear
40 results
Download the following citations:
Email the following citations:
Print the following citations:
See all 1 Highlighted Expert Reviews articles matching your criteria
...
  • Hernández D
  • Vázquez-Sánchez T
  • Sola E
  • Lopez V
  • Ruiz-Esteban P
  • et al.
BMC Nephrol. 2022 Nov 7;23(1):357 doi: 10.1186/s12882-022-02989-z.
CET Conclusion
Reviewer: Mr John O'Callaghan, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: This is a clearly written protocol for a multicentre randomised controlled trial. The hypothesis is that treatment of early borderline lesions (within 3 months of transplant) will prevent or decrease progression of IFTA. The treatment used will be rabbit ATG, so a safety study will be as important as any improvement at a histological level. The study is based on the fact that early, subclinical lesions are an indicator of subsequent drop in graft function and reduced graft survival, and that treatment at this stage will have a positive impact. Length of follow up is 24 months, which should be long enough to assess efficacy and safety in general. Patients will require a 3-month protocol biopsy to then determine whether or not they enter randomisation. The sample size calculation has made reference to previous data and expected dropout rates.
Aims: This study aims to investigate the effects of treating early borderline lesions with polyclonal rabbit antithymocyte globulin (Grafalon®) in comparison to conventional therapy, in low immunological risk kidney transplant recipients.
Interventions: Participants will be randomly assigned to either the Grafalon® group or the standard treatment group.
Participants: The study will randomise 80 kidney transplant recipients with low immunological risk.
Outcomes: The primary outcomes are the presence of interstitial fibrosis/tubular atrophy (IFTA) and graft function. The main efficacy outcomes are function and histological lesions.
Follow Up: N/A
BACKGROUND:

Subclinical inflammation, including borderline lesions (BL), is very common (30-40%) after kidney transplantation (KT), even in low immunological risk patients, and can lead to interstitial fibrosis/tubular atrophy (IFTA) and worsening of renal function with graft loss. Few controlled studies have analyzed the therapeutic benefit of treating these BL on renal function and graft histology. Furthermore, these studies have only used bolus steroids, which may be insufficient to slow the progression of these lesions. Klotho, a transmembrane protein produced mainly in the kidney with antifibrotic properties, plays a crucial role in the senescence-inflammation binomial of kidney tissue. Systemic and local inflammation decrease renal tissue expression and soluble levels of α-klotho. It is therefore important to determine whether treatment of BL prevents a decrease in α-klotho levels, progression of IFTA, and loss of kidney function.

METHODS:

The TRAINING study will randomize 80 patients with low immunological risk who will receive their first KT. The aim of the study is to determine whether the treatment of early BL (3rd month post-KT) with polyclonal rabbit antithymocyte globulin (Grafalon®) (6 mg/kg/day) prevents or decreases the progression of IFTA and the worsening of graft function compared to conventional therapy after two years post-KT, as well as to analyze whether treatment of BL with Grafalon® can modify the expression and levels of klotho, as well as the pro-inflammatory cytokines that regulate its expression.

DISCUSSION:

This phase IV investigator-driven, randomized, placebo-controlled clinical trial will examine the efficacy and safety of Grafalon® treatment in low-immunological-risk KT patients with early BL.

TRIAL REGISTRATION:

clinicaltrials.gov : NCT04936282. Registered June 23, 2021, https://clinicaltrials.gov/ct2/show/NCT04936282?term=NCT04936282&draw=2&rank=1 . Protocol Version 2 of 21 January 2022.

SPONSOR:

Canary Isles Institute for Health Research Foundation, Canary Isles (FIISC). mgomez@fciisc.org .

  • Roufosse C
  • Becker JU
  • Rabant M
  • Seron D
  • Bellini MI
  • et al.
Transpl Int. 2022 May 20;35:10140 doi: 10.3389/ti.2022.10140.

Antibody-mediated rejection (AMR) is caused by antibodies that recognize donor human leukocyte antigen (HLA) or other targets. As knowledge of AMR pathophysiology has increased, a combination of factors is necessary to confirm the diagnosis and phenotype. However, frequent modifications to the AMR definition have made it difficult to compare data and evaluate associations between AMR and graft outcome. The present paper was developed following a Broad Scientific Advice request from the European Society for Organ Transplantation (ESOT) to the European Medicines Agency (EMA), which explored whether updating guidelines on clinical trial endpoints would encourage innovations in kidney transplantation research. ESOT considers that an AMR diagnosis must be based on a combination of histopathological factors and presence of donor-specific HLA antibodies in the recipient. Evidence for associations between individual features of AMR and impaired graft outcome is noted for microvascular inflammation scores ≥2 and glomerular basement membrane splitting of >10% of the entire tuft in the most severely affected glomerulus. Together, these should form the basis for AMR-related endpoints in clinical trials of kidney transplantation, although modifications and restrictions to the Banff diagnostic definition of AMR are proposed for this purpose. The EMA provided recommendations based on this Broad Scientific Advice request in December 2020; further discussion, and consensus on the restricted definition of the AMR endpoint, is required.

  • Hernández D
  • Alonso-Titos J
  • Vázquez T
  • León M
  • Caballero A
  • et al.
J Clin Med. 2021 May 7;10(9) doi: 10.3390/jcm10092005.
CET Conclusion
Reviewer: Mr Simon Knight, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: This interesting multicentre study investigates the impact of late steroid withdrawal on graft histology following renal transplantation. Low risk patients 3 months post-transplant were randomized to withdrawal or continuation. Steroid withdrawal appeared safe for the majority of patients, with no increase in rejection or de-novo DSA and improved cardiovascular risk profile in keeping with previous studies. Of note, patients with subclinical inflammation at baseline (prior to withdrawal) demonstrated increased chronicity scores at 24 months if undergoing steroid withdrawal. This implies that baseline biopsy might be useful when considering steroid withdrawal, even in a low-risk cohort.
Aims: This study aimed to examine the impact of corticosteroid withdrawal (CSW) on inflammatory and chronic histological changes in kidney transplant recipients following transplantation.
Interventions: Participants were randomised to either the corticosteroid continuation group or the corticosteroid withdrawal group.
Participants: 105 low-immunological-risk kidney transplant recipients.
Outcomes: Acute rejection, inflammatory and chronicity scores, Banff scores, occurrence of de novo donor-specific antibodies (dnDSA), clinical and biochemical data during follow up, patient survival, graft survival and safety.
Follow Up: 24 months

The impact of corticosteroid withdrawal on medium-term graft histological changes in kidney transplant (KT) recipients under standard immunosuppression is uncertain. As part of an open-label, multicenter, prospective, phase IV, 24-month clinical trial (ClinicalTrials.gov, NCT02284464) in low-immunological-risk KT recipients, 105 patients were randomized, after a protocol-biopsy at 3 months, to corticosteroid continuation (CSC, n = 52) or corticosteroid withdrawal (CSW, n = 53). Both groups received tacrolimus and MMF and had another protocol-biopsy at 24 months. The acute rejection rate, including subclinical inflammation (SCI), was comparable between groups (21.2 vs. 24.5%). No patients developed dnDSA. Inflammatory and chronicity scores increased from 3 to 24 months in patients with, at baseline, no inflammation (NI) or SCI, regardless of treatment. CSW patients with SCI at 3 months had a significantly increased chronicity score at 24 months. HbA1c levels were lower in CSW patients (6.4 ± 1.2 vs. 5.7 ± 0.6%; p = 0.013) at 24 months, as was systolic blood pressure (134.2 ± 14.9 vs. 125.7 ± 15.3 mmHg; p = 0.016). Allograft function was comparable between groups and no patients died or lost their graft. An increase in chronicity scores at 2-years post-transplantation was observed in low-immunological-risk KT recipients with initial NI or SCI, but CSW may accelerate chronicity changes, especially in patients with early SCI. This strategy did, however, improve the cardiovascular profiles of patients.

  • Hernández D
  • Vázquez T
  • Alonso-Titos J
  • León M
  • Caballero A
  • et al.
J Clin Med. 2021 Apr 29;10(9) doi: 10.3390/jcm10091934.
CET Conclusion
Reviewer: Mr John O'Callaghan, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: This paper reports on sub-clinical inflammation in low-risk renal transplantation. It was performed within a trial of corticosteroid withdrawal, and hence the randomisation is not related directly to this paper. Protocol biopsies were performed at 3 months after transplant, during which time no patient had acute rejection or developed de novo DSA. The biopsies showed that 54% of recipients had sub-clinical inflammation (which is in agreement with previous studies). This inflammation was correlated significantly with the absolute number of HLA mismatches, and independently with Class II mismatches in multivariate analysis, but not Class I. At this level of breakdown, however, there is a question of adequate power to make this assessment. In the multivariate analysis, several other factors were assessed and found not to be independent predictors of sub-clinical inflammation: recipient age, DGF, transfusion prior to transplant, and tacrolimus. Subclinical inflammation is present in half of kidney transplants on protocol biopsies and there is evidence from other studies that these changes can lead to chronic damage and dysfunction. The study is limited by its small sample size and ethnic homogeneity (100% Caucasian). It also represents an analysis of results from an RCT, and therefore may show association but has not proven causation and does not have significant or novel implications for clinical practice.
Aims: This study is a part of a randomised controlled trial comparing corticosteroid withdrawal versus standard immunosuppression in low-immunological-risk renal transplantation. The aim of this study was to determine the effect of human leukocyte antigen (HLA)-mismatching on early subclinical inflammation (SCI) in low-immunological-risk renal transplant patients.
Interventions: Participants in the original trial were randomised to either the prednisone continuation group or the prednisone withdrawal group.
Participants: 105 Caucasian renal transplant patients.
Outcomes: Association between HLA mismatches and SCI risk.
Follow Up: 3 months

The impact of human leukocyte antigen (HLA)-mismatching on the early appearance of subclinical inflammation (SCI) in low-immunological-risk kidney transplant (KT) recipients is undetermined. We aimed to assess whether HLA-mismatching (A-B-C-DR-DQ) is a risk factor for early SCI. As part of a clinical trial (Clinicaltrials.gov, number NCT02284464), a total of 105 low-immunological-risk KT patients underwent a protocol biopsy on the third month post-KT. As a result, 54 presented SCI, showing a greater number of total HLA-mismatches (p = 0.008) and worse allograft function compared with the no inflammation group (48.5 ± 13.6 vs. 60 ± 23.4 mL/min; p = 0.003). Multiple logistic regression showed that the only risk factor associated with SCI was the total HLA-mismatch score (OR 1.32, 95%CI 1.06-1.64, p = 0.013) or class II HLA mismatching (OR 1.51; 95%CI 1.04-2.19, p = 0.032) after adjusting for confounder variables (recipient age, delayed graft function, transfusion prior KT, and tacrolimus levels). The ROC curve illustrated that the HLA mismatching of six antigens was the optimal value in terms of sensitivity and specificity for predicting the SCI. Finally, a significantly higher proportion of SCI was seen in patients with >6 vs. ≤6 HLA-mismatches (62.3 vs. 37.7%; p = 0.008). HLA compatibility is an independent risk factor associated with early SCI. Thus, transplant physicians should perhaps be more aware of HLA mismatching to reduce these early harmful lesions.

  • Jarque M
  • Crespo E
  • Melilli E
  • Gutiérrez A
  • Moreso F
  • et al.
Clin Infect Dis. 2020 Dec 3;71(9):2375-2385 doi: 10.1093/cid/ciz1209.
CET Conclusion
Reviewer: Mr Simon Knight, Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences University of Oxford
Conclusion: This somewhat complicated multicenter study investigated the use of pre-transplant testing for CMV-specific cell mediated immunity (using Elispot) to predict risk of post-transplant CMV infection. Patients were stratified into 2 groups according to pre-transplant immunity, and then each group was randomized to pre-emptive or prophylactic CMV prevention strategy post-transplant. The authors found that pre-transplant immunity predicts risk of post-transplant CMV infection, but this prediction is affected by the use of lymphocyte depleting induction agents. Measuring CMV CMI at day 15 post-transplant appears to offer better prediction of risk. It should be noted that the minority of patients received lymphocyte depleting agents so the power to detect differences in this subgroup is very small (the confidence intervals around the odd-ratios given are wide). Also, the randomized intervention was the CMV prevention strategy, rather than the monitoring strategy that is compared in the primary outcome, meaning that this is really an observational rather than randomized study.
Aims: The aim of this study was to investigate whether assessing pre-transplant Cytomegalovirus (CMV)-specific cell mediated immunity (CMI) could predict the risk of CMV infection in transplant patients
Interventions: Participants were first categorized as low risk or high risk based on CMV-specific CMI, and then subsequenty randomized to two groups: the 3-month antiviral prophylaxis group or the preemptive therapy group.
Participants: 160 CMV-seropositve renal transplant donors/recipients.
Outcomes: The primary outcome was the incidence of CMV infection. The secondary outcomes included rate of CMV infection in need of antiviral treatment, CMV disease rate and the rate of late-onset CMV infection following prophylaxis withdrawal; the influence of CMV-specific CMI based on the type of induction therapy; the effect of CMI on the pp65 CMV antigen; and the accuracy of prediction-risk of pre-transplant CMV-specific CMI after 15 days of transplantation.
Follow Up: 1 year
BACKGROUND:

Improving cytomegalovirus (CMV) immune-risk stratification in kidney transplantation is highly needed to establish guided preventive strategies.

METHODS:

This prospective, interventional, multicenter clinical trial assessed the value of monitoring pretransplant CMV-specific cell-mediated immunity (CMI) using an interferon-γ release assay to predict CMV infection in kidney transplantation. One hundred sixty donor/recipient CMV-seropositive (D+/R+) patients, stratified by their baseline CMV (immediate-early protein 1)-specific CMI risk, were randomized to receive either preemptive or 3-month antiviral prophylaxis. Also, 15-day posttransplant CMI risk stratification and CMI specific to the 65 kDa phosphoprotein (pp65) CMV antigen were investigated. Immunosuppression consisted of basiliximab, tacrolimus, mycophenolate mofetil, and corticosteroids in 80% of patients, whereas 20% received thymoglobulin induction therapy.

RESULTS:

Patients at high risk for CMV based on pretransplant CMI developed significantly higher CMV infection rates than those deemed to be at low risk with both preemptive (73.3% vs 44.4%; odds ratio [OR], 3.44 [95% confidence interval {CI}, 1.30-9.08]) and prophylaxis (33.3% vs 4.1%; OR, 11.75 [95% CI, 2.31-59.71]) approaches. The predictive capacity for CMV-specific CMI was only found in basiliximab-treated patients for both preemptive and prophylaxis therapy. Fifteen-day CMI risk stratification better predicted CMV infection (81.3% vs 9.1%; OR, 43.33 [95% CI, 7.89-237.96]).

CONCLUSIONS:

Pretransplant CMV-specific CMI identifies D+/R+ kidney recipients at high risk of developing CMV infection if not receiving T-cell-depleting antibodies. Monitoring CMV-specific CMI soon after transplantation further defines the CMV infection prediction risk. Monitoring CMV-specific CMI may guide decision making regarding the type of CMV preventive strategy in kidney transplantation.

CLINICAL TRIALS REGISTRATION:

NCT02550639.

  • Favà A
  • Cucchiari D
  • Montero N
  • Toapanta N
  • Centellas FJ
  • et al.
Am J Transplant. 2020 Nov;20(11):3030-3041 doi: 10.1111/ajt.16246.

Kidney transplant recipients might be at higher risk for severe coronavirus disease 2019 (COVID-19). However, risk factors for relevant outcomes remain uncertain in this population. This is a multicentric kidney transplant cohort including 104 hospitalized patients between March 4 and April 17, 2020. Risk factors for death and acute respiratory distress syndrome (ARDS) were investigated, and clinical and laboratory data were analyzed. The mean age was 60 years. Forty-seven patients (54.8%) developed ARDS. Obesity was associated to ARDS development (OR 2.63; P = .04). Significant age differences were not found among patients developing and not developing ARDS (61.3 vs 57.8 years, P = .16). Seventy-six (73%) patients were discharged, and 28 (27%) died. Death was more common among the elderly (55 and 70.8 years, P < .001) and those with preexisting pulmonary disease (OR 2.89, P = .009). At admission, higher baseline lactate dehydrogenase (257 vs 358 IU/mL, P = .001) or ARDS conferred higher risk of death (HR 2.09, P = .044). In our cohort, ARDS was equally present among young and old kidney recipients. However, the elderly might be at higher risk of death, along with those showing higher baseline LDH at admission.

  • Schinstock CA
  • Mannon RB
  • Budde K
  • Chong AS
  • Haas M
  • et al.
Transplantation. 2020 May;104(5):911-922 doi: 10.1097/TP.0000000000003095.

With the development of modern solid-phase assays to detect anti-HLA antibodies and a more precise histological classification, the diagnosis of antibody-mediated rejection (AMR) has become more common and is a major cause of kidney graft loss. Currently, there are no approved therapies and treatment guidelines are based on low-level evidence. The number of prospective randomized trials for the treatment of AMR is small, and the lack of an accepted common standard for care has been an impediment to the development of new therapies. To help alleviate this, The Transplantation Society convened a meeting of international experts to develop a consensus as to what is appropriate treatment for active and chronic active AMR. The aim was to reach a consensus for standard of care treatment against which new therapies could be evaluated. At the meeting, the underlying biology of AMR, the criteria for diagnosis, the clinical phenotypes, and outcomes were discussed. The evidence for different treatments was reviewed, and a consensus for what is acceptable standard of care for the treatment of active and chronic active AMR was presented. While it was agreed that the aims of treatment are to preserve renal function, reduce histological injury, and reduce the titer of donor-specific antibody, there was no conclusive evidence to support any specific therapy. As a result, the treatment recommendations are largely based on expert opinion. It is acknowledged that properly conducted and powered clinical trials of biologically plausible agents are urgently needed to improve patient outcomes.

  • Torres A
  • Hernández D
  • Moreso F
  • Serón D
  • Burgos MD
  • et al.
Kidney Int Rep. 2018 Jul 11;3(6):1304-1315 doi: 10.1016/j.ekir.2018.07.009.
INTRODUCTION:

Despite the high incidence of posttransplant diabetes mellitus (PTDM) among high-risk recipients, no studies have investigated its prevention by immunosuppression optimization.

METHODS:

We conducted an open-label, multicenter, randomized trial testing whether a tacrolimus-based immunosuppression and rapid steroid withdrawal (SW) within 1 week (Tac-SW) or cyclosporine A (CsA) with steroid minimization (SM) (CsA-SM), decreased the incidence of PTDM compared with tacrolimus with SM (Tac-SM). All arms received basiliximab and mycophenolate mofetil. High risk was defined by age >60 or >45 years plus metabolic criteria based on body mass index, triglycerides, and high-density lipoprotein-cholesterol levels. The primary endpoint was the incidence of PTDM after 12 months.

RESULTS:

The study comprised 128 de novo renal transplant recipients without pretransplant diabetes (Tac-SW: 44, Tac-SM: 42, CsA-SM: 42). The 1-year incidence of PTDM in each arm was 37.8% for Tac-SW, 25.7% for Tac-SM, and 9.7% for CsA-SM (relative risk [RR] Tac-SW vs. CsA-SM 3.9 [1.2-12.4; P = 0.01]; RR Tac-SM vs. CsA-SM 2.7 [0.8-8.9; P = 0.1]). Antidiabetic therapy was required less commonly in the CsA-SM arm (P = 0.06); however, acute rejection rate was higher in CsA-SM arm (Tac-SW 11.4%, Tac-SM 4.8%, and CsA-SM 21.4% of patients; cumulative incidence P = 0.04). Graft and patient survival, and graft function were similar among arms.

CONCLUSION:

In high-risk patients, tacrolimus-based immunosuppression with SM provides the best balance between PTDM and acute rejection incidence.

  • Moreso F
  • Crespo M
  • Ruiz JC
  • Torres A
  • Gutierrez-Dalmau A
  • et al.
Am J Transplant. 2018 Apr;18(4):927-935 doi: 10.1111/ajt.14520.
CET Conclusion
Reviewer: Centre for Evidence in Transplantation
Conclusion: This small study investigates the role of rituximab, combined with IvIG, for renal transplant recipients with chronic antibody-mediated rejection (CAMR). The study is placebo-controlled. Despite being multicentre and spanning a 3-year period, only 25 of the planned 50 patients were recruited. The authors conclude that this regimen is unlikely to be useful in the management of CAMR, as no differences were seen in renal function, proteinuria, biopsy scores or MFI of donor-specific antibodies. The major limitation here is the small number of patients recruited, leading to a significant difference in baseline renal function between the groups. 95% confidence intervals for the outcomes are not presented, but it is likely that there is insufficient power to draw any firm conclusions on the basis of these data.
Expert Review
Reviewer: Professor John Kanellis, Department of Nephrology, Department of Medicine, Monash University, Clayton, Melbourne.
Conflicts of Interest: No
Clinical Impact Rating 3
Review: This double-blind, placebo controlled study examined the use of IVIG and rituximab to treat chronic antibody-mediated rejection (cAMR). Participants had donor specific antibody (DSA), transplant glomerulopathy and an eGFR of >20ml/min. No benefit was seen after analysis of change in eGFR and proteinuria, biopsy scores and DSA MFI. This study is of significant interest as it suggests no benefit from currently used treatments for cAMR. The use of IVIG and rituximab is onerous and has significant side effects, therefore it is important to have studies that reveal benefit. The results need to be interpreted with caution however, as the study was small with only half the planned participants being recruited. It is therefore significantly underpowered. Furthermore, as the authors also acknowledge, subjects already had significant injury present and the baseline eGFR differed in the two arms (placebo 45±18 ml/min vs treatment arm 35±17 ml/min). These issues may also have influenced the results. Further well designed and adequately powered studies in cAMR are desperately needed as there is a significant clinical challenge and few proven therapeutic options. It is hoped that new therapies being currently trialled (IL-6 blockade, IdeS therapy) will show significant benefit.
Aims: To evaluate the efficacy and safety of intravenous immunoglobulins (IVIG) combined with rituximab (RTX) in renal transplant patients with chronic antibody mediated rejection (ABMR), displaying transplant glomerulopathy and donor-specific antibodies (DSA).
Interventions: Participants were randomised to receive either four consecutive doses of IVIG (0.5 g/kg) every 3 weeks and one single dose of RTX (375 mg/m2), 1 week after the last IVIG dose (treatment group), versus an isovolumetric saline solution following the same schedule (control group).
Participants: 25 stable renal transplant recipients aged ≥18 years with biopsy proven chronic ABMR diagnosed < 6 months prior to randomisation.
Outcomes: The primary outcome measured was the delicine in estimated glomerular filtration rate. Secondary measured outcomes included proteinuria renal lesions, and DSA at 1 year. Adverse events, opportunistic infections and hospitalisations were also measured.
Follow Up: 12 months

There are no approved treatments for chronic antibody mediated rejection (ABMR). We conducted a multicenter, prospective, randomized, placebo-controlled, double-blind clinical trial to evaluate efficacy and safety of intravenous immunoglobulins (IVIG) combined with rituximab (RTX) (EudraCT 2010-023746-67). Patients with transplant glomerulopathy and anti-HLA donor-specific antibodies (DSA) were eligible. Patients with estimated glomerular filtration rate (eGFR) <20 mL/min per 1.73m2 and/or severe interstitial fibrosis/tubular atrophy were excluded. Patients were randomized to receive IVIG (4 doses of 0.5 g/kg) and RTX (375 mg/m2 ) or a wrapped isovolumetric saline infusion. Primary efficacy variable was the decline of eGFR at one year. Secondary efficacy variables included evolution of proteinuria, renal lesions, and DSA at 1 year. The planned sample size was 25 patients per group. During 2012-2015, 25 patients were randomized (13 to the treatment and 12 to the placebo group). The planned patient enrollment was not achieved because of budgetary constraints and slow patient recruitment. There were no differences between the treatment and placebo groups in eGFR decline (-4.2 ± 14.4 vs. -6.6 ± 12.0 mL/min per 1.73 m2 , P-value = .475), increase of proteinuria (+0.9 ± 2.1 vs. +0.9 ± 2.1 g/day, P-value = .378), Banff scores at one year and MFI of the immunodominant DSA. Safety was similar between groups. These data suggest that the combination of IVIG and RTX is not useful in patients displaying transplant glomerulopathy and DSA.

  • Neuberger JM
  • Bechstein WO
  • Kuypers DR
  • Burra P
  • Citterio F
  • et al.
Transplantation. 2017 Apr;101(4S Suppl 2):S1-S56 doi: 10.1097/TP.0000000000001651.

Short-term patient and graft outcomes continue to improve after kidney and liver transplantation, with 1-year survival rates over 80%; however, improving longer-term outcomes remains a challenge. Improving the function of grafts and health of recipients would not only enhance quality and length of life, but would also reduce the need for retransplantation, and thus increase the number of organs available for transplant. The clinical transplant community needs to identify and manage those patient modifiable factors, to decrease the risk of graft failure, and improve longer-term outcomes.COMMIT was formed in 2015 and is composed of 20 leading kidney and liver transplant specialists from 9 countries across Europe. The group's remit is to provide expert guidance for the long-term management of kidney and liver transplant patients, with the aim of improving outcomes by minimizing modifiable risks associated with poor graft and patient survival posttransplant.The objective of this supplement is to provide specific, practical recommendations, through the discussion of current evidence and best practice, for the management of modifiable risks in those kidney and liver transplant patients who have survived the first postoperative year. In addition, the provision of a checklist increases the clinical utility and accessibility of these recommendations, by offering a systematic and efficient way to implement screening and monitoring of modifiable risks in the clinical setting.