1
|
Billany RE, Bishop NC, Castle EM, Graham-Brown MPM, Greenwood SA, Lightfoot CJ, Wilkinson TJ. Physical activity interventions in adult kidney transplant recipients: an updated systematic review and meta-analysis of randomized controlled trials. Ren Fail 2025; 47:2480246. [PMID: 40148080 PMCID: PMC11951324 DOI: 10.1080/0886022x.2025.2480246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/15/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Kidney transplant recipients (KTRs) exhibit a high prevalence of cardiovascular disease (CVD) and adverse changes in physical fitness and body composition. Post-transplant management recommends being physically active and evidence in this field is growing. The aim of this review was to update our previous systematic review and meta-analysis of randomized controlled trials (RCTs) assessing the effects of physical activity and exercise training interventions in KTRs. METHODS A comprehensive literature search between March 2021 and September 2024 identified seven additional RCTs. Therefore, this updated review and meta-analysis includes 23 RCTs. Outcomes included cardiorespiratory fitness (CRF), strength, blood pressure, body composition, heart rate, markers of dyslipidemia and kidney function, and health-related quality of life. RESULTS Twenty-three RCTs, including 1,139 KTRs, were included. The median intervention length was 12 weeks with participants exercising between 2 and 7×/week. Most studies used a mixture of aerobic and resistance training but reporting and intervention content was highly varied. Significant improvements were observed in CRF (V̇O2peak; +3.87 mL/kg/min, p = .0004), physical function (sit-to-stand-60; +7.72 repetitions, p = .0001), and high-density lipoprotein (HDL; +0.13 mmol/L, p = .02). Isolated studies reported improvements in strength, bone health, lean mass, and quality of life (QoL). All studies were found to have a high or moderate risk of bias. CONCLUSIONS Exercise training or increasing physical activity may confer several benefits in adult KTRs, especially through the improvements in CRF and HDL which have been linked to CVD risk. Despite new literature, there is still a need for long-term larger sampled RCTs and more detailed reporting of intervention details and program adherence.
Collapse
Affiliation(s)
- Roseanne E. Billany
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Nicolette C. Bishop
- School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, UK
| | - Ellen M. Castle
- Faculty of Health Sciences, Curtin School of Allied Health, Curtin University, Perth, Australia
- Physiotherapy Division, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | | | - Sharlene A. Greenwood
- Department of Renal Medicine, King’s College Hospital NHS Trust, London, UK
- Renal Sciences, Faculty of Life Sciences and Medicine, Kings College London, London, UK
| | - Courtney J. Lightfoot
- NIHR Leicester Biomedical Research Centre, Leicester, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Thomas J. Wilkinson
- NIHR Leicester Biomedical Research Centre, Leicester, UK
- Leicester Diabetes Centre, University of Leicester, Leicester, UK
| |
Collapse
|
2
|
Bellini MI, Angeletti S, Fresilli D, Di Segni M, Conte GML, Flavia R, Garofalo M, Pretagostini R, De Vito C, Pacini P, D'Andrea V, Barbato A, Drudi FM, Caratozzolo M, Vito C. Multiparametric ultrasound for non-invasive evaluation of kidney graft function. J Ultrasound 2025:10.1007/s40477-025-00989-x. [PMID: 40180766 DOI: 10.1007/s40477-025-00989-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/12/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Renal transplant (RT) remains the optimal treatment for end-stage renal disease and early complications might be detected in the postoperative period to improve long-term outcomes. To this regard, contrast enhanced ultrasound (CEUS) could be utilized to evaluate RT functional recovery and potentially detect acute rejection (AR) and/or renal ischemia signs. MATERIALS AND METHODS Observational study of 107 consecutive patients waitlisted for RT. Participants underwent conventional ultrasound (CUS) and color-doppler-ultrasound (CDUS) to evaluate resistive index of segmental and interlobar arteries and quantitative CEUS techniques recording the following perfusion parameters: peak intensity (PI-c), rising time (RT-c), area under (AUC-c) the time intensity curve (TIC), time to peak (TTP-c) and mean transit time (MTT-c). RESULTS CEUS parameters sensibility and specificity to predict AR in the early post-operative period resulted in: 90% and 69% for PI-c, 95% and 64% for RT-c, 85% and 65% for AUC-c. The overall diagnostic performance of these three CEUS parameters in comparison to the same in CUS and CDUS resulted in a sensitivity and specificity of 95% and 49%, versus 85% and 46%, respectively, therefore CEUS examination with the analysis of PI-c, RT-c and AUC-c values increases the diagnostic sensitivity in predicting AR by approximately 15-20% compared to CDUS and by 30-40% compared to CUS. CONCLUSION CEUS could be routinely included in RT follow-up, as it shows to be a non-invasive helpful diagnostic tool for early detection of renal graft complications, selecting patients eventually in need of further confirmation.
Collapse
Affiliation(s)
| | - Sergio Angeletti
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
- ASL Rieti, via del Terminillo 42, Rieti, Italy.
| | - Daniele Fresilli
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
- Department of Radiology, Ospedale S. Scolastica, Via S. Pasquale, 03043, Cassino, Frosinone, Italy
| | | | - Gian Marco Lo Conte
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Raponi Flavia
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Manuela Garofalo
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Renzo Pretagostini
- Department of General and Specialty Surgery, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Corrado De Vito
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185, Rome, Italy
| | - Patrizia Pacini
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Vito D'Andrea
- Department of Surgery, Sapienza University of Rome, 00161, Rome, Italy
| | | | - Francesco M Drudi
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Marcello Caratozzolo
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Cantisani Vito
- Department of Radiological, Oncological, and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| |
Collapse
|
3
|
Diebold M, Mayer KA, Hidalgo L, Kozakowski N, Budde K, Böhmig GA. Chronic Rejection After Kidney Transplantation. Transplantation 2025; 109:610-621. [PMID: 39192468 PMCID: PMC11927446 DOI: 10.1097/tp.0000000000005187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024]
Abstract
In kidney transplantation, ongoing alloimmune processes-commonly triggered by HLA incompatibilities-can trigger chronic transplant rejection, affecting the microcirculation and the tubulointerstitium. Continuous inflammation may lead to progressive, irreversible graft injury, culminating in graft dysfunction and accelerated transplant failure. Numerous experimental and translational studies have delineated a complex interplay of different immune mechanisms driving rejection, with antibody-mediated rejection (AMR) being an extensively studied rejection variant. In microvascular inflammation, a hallmark lesion of AMR, natural killer (NK) cells have emerged as pivotal effector cells. Their essential role is supported by immunohistologic evidence, bulk and spatial transcriptomics, and functional genetics. Despite significant research efforts, a substantial unmet need for approved rejection therapies persists, with many trials yielding negative outcomes. However, several promising therapies are currently under investigation, including felzartamab, a monoclonal antibody targeting the surface molecule CD38, which is highly expressed in NK cells and antibody-producing plasma cells. In an exploratory phase 2 trial in late AMR, this compound has demonstrated potential in resolving molecular and morphologic rejection activity and injury, predominantly by targeting NK cell effector function. These findings inspire hope for effective treatments and emphasize the necessity of further pivotal trials focusing on chronic transplant rejection.
Collapse
Affiliation(s)
- Matthias Diebold
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Katharina A. Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Luis Hidalgo
- HLA Laboratory, Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | | | - Klemens Budde
- Department of Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Georg A. Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Świątek Ł, Miedziaszczyk M, Lewandowski D, Robakowski F, Tyburski P, Jakubowska M, Karczewski M, Idasiak-Piechocka I. The Promising Effect of Tocilizumab on Chronic Antibody-Mediated Rejection (cAMR) of Kidney Transplant. Pharmaceutics 2025; 17:78. [PMID: 39861726 PMCID: PMC11768637 DOI: 10.3390/pharmaceutics17010078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Chronic antibody-mediated rejection (cAMR) constitutes a serious challenge in the long-term success of organ transplantation. It is associated with donor-specific antibodies (DSAs) which activate a complement pathway in response to the presence of human leukocyte antigens (HLAs) on the graft, which results in chronic inflammation and leads to graft dysfunction. One of the recent promising methods of cAMR treatment is a recombinant humanized anti-interleukin-6 receptor (IL-6R) monoclonal antibody referred to as Tocilizumab (TCZ). The aim of the presented systematic review is to explore the existing knowledge regarding the effect of tocilizumab treatment on cAMR and to perform a meta-analysis of the available data. Methods: A systematic review was performed using the PRISMA 2020 Checklist and Flow diagram. A systematic review protocol was registered in PROSPERO: CRD42024510996. The bias assessment was obtained with Methodical Index for Non-Randomized Studies (MINORS), whereas meta-analysis was performed using MedCalc. Results: Five clinical trials with a total number of 105 patients were included in our review. The mean loss of eGFR in time was -0.141 mL/min/1.73 m2 (95% CI: -0.409 to 0.126; p = 0.298) and was found to be statistically insignificant. The heterogeneity was low and was equal to I2 = 0.00%. The authors demonstrated a reduction in DSA titer by TCZ (-0.266 MFI (95% CI: -0.861 to 0.329; p = 0.377)). In the majority of studies, eGFR stabilization was associated with a reduction in DSAs. Conclusions: TCZ pharmacotherapy insignificantly reduced DSA titer and eGFR. Despite promising outcomes of potential eGFR stabilization, there is a need for large randomized controlled trials comparing standard management of cAMR and tocilizumab treatment.
Collapse
Affiliation(s)
- Łukasz Świątek
- Students Research Group of Transplantation and Kidney Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (Ł.Ś.)
| | - Miłosz Miedziaszczyk
- Department of General and Transplant Surgery, Poznan University of Medical Sciences, 60-355 Poznan, Poland
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Dominik Lewandowski
- Students Research Group of Transplantation and Kidney Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (Ł.Ś.)
| | - Filip Robakowski
- Students Research Group of Transplantation and Kidney Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (Ł.Ś.)
| | - Piotr Tyburski
- Students Research Group of Transplantation and Kidney Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (Ł.Ś.)
| | - Marta Jakubowska
- Students Research Group of Transplantation and Kidney Diseases, Poznan University of Medical Sciences, 60-355 Poznan, Poland; (Ł.Ś.)
| | - Marek Karczewski
- Department of General and Transplant Surgery, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Ilona Idasiak-Piechocka
- Department of General and Transplant Surgery, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
5
|
Danković K, Stefanović N, Cvetković T, Vujić S, Jović M, Mitić B, Veličković-Radovanović R. Potential influence of interleukin-6 -174G/C gene polymorphism on kidney graft function and tacrolimus dose requirements: five-year follow-up. Xenobiotica 2024; 54:855-863. [PMID: 39535282 DOI: 10.1080/00498254.2024.2427032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
1. INTRODUCTION The study aimed to investigate the influence of interleukin (IL)-6 -174 G/C gene polymorphism on graft function (defined as estimated glomerular filtration rate, eGFR), as well as on the tacrolimus (Tac) pharmacokinetics during the five years after kidney transplantation. 2. METHODS The study included 115 Caucasian kidney transplant recipients on Tac-based immunosuppression. The patients were followed between 6 and 60 post-transplantation months. Interleukin-6 and CYP3A5 genotyping were performed. 3. RESULTS Patients carrying the IL-6 -174GG genotype had lower eGFR values compared to the patients with the IL-6 -174GC and -174CC genotypes at the 12th, 48th and 60th post-transplantation months. The linear regression analysis indicated that eGFR at the 6th post-transplantation month and IL-6 -174 G/C polymorphism are independent predictors of eGFR values in the late post-transplantation period. The IL-6 -174GG genotype carriers had lower dose-adjusted trough concentration (C0/D) of Tac compared to the IL-6 C allele carriers during the entire observation period (except at the 24th month), while this effect was independent of the CYP3A5 genotype within three years post-transplantation. 4. CONCLUSION Interleukin-6 genotyping could be an additional tool to categorise patients towards the risk of graft deterioration in the long-term post-transplantation period. The IL-6 genotyping could be supportive in genotype-guided dosing of Tac.
Collapse
Affiliation(s)
| | - Nikola Stefanović
- Department of Pharmacy, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Tatjana Cvetković
- Department of Biochemistry, Faculty of Medicine, University of Nis, Nis, Serbia
- Center for Clinical and Medical Biochemistry, University Clinical Center Nis, Nis, Serbia
| | - Stevan Vujić
- Faculty of Medicine, University of Nis, Nis, Serbia
| | - Maša Jović
- Faculty of Medicine, University of Nis, Nis, Serbia
| | - Branka Mitić
- Department of Internal Medicine, Faculty of Medicine, University of Nis, Nis, Serbia
- Clinic of Nephrology, University Clinical Center Nis, Nis, Serbia
| | - Radmila Veličković-Radovanović
- Clinic of Nephrology, University Clinical Center Nis, Nis, Serbia
- Department of Pharmacology with Toxicology, Faculty of Medicine, University of Nis, Nis, Serbia
| |
Collapse
|
6
|
Nađ Škegro S, Penezić L, Šimičević L, Hudolin T, Kaštelan Ž, Božina N, Trkulja V. The reduced function allele SLCO1B1 c.521T>C is of no practical relevance for the renal graft function over the first post-transplant year in patients treated with mycophenolic acid. Pharmacogenet Genomics 2024; 34:226-235. [PMID: 39101384 DOI: 10.1097/fpc.0000000000000539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
OBJECTIVE It is unclear whether renal transplant recipients treated with mycophenolic acid (MPA) who carry the reduced-function allele at polymorphism SLCO1B1 c.521T>C differ from their wild-type peers regarding renal outcomes and tolerability. We aimed to estimate the effect of this polymorphism on the graft function (estimated glomerular filtration rate, eGFR) over the first 12 post-transplant months in patients on MPA-based maintenance immunosuppression. METHODS In a 12-month observational cohort study, consecutive adult patients were repeatedly assessed for eGFR. The SLCO1B1 c.521C>T variant allele carriers (exposed) and wild-type subjects (controls) were balanced on a range of demographic, medical, and genetic variables at baseline, and eGFR trajectory was estimated with further adjustment for time-varying covariates. A subset of patients were assessed for exposure to MPA 5-7 days after the transplantation. RESULTS The adjusted eGFR slopes from day 1 to day 28 (daily), and from day 28 to day 365 (monthly) were practically identical in exposed (n = 86) and control (n = 168) patients [geometric means ratios (GMR) = 0.99, 95% confidence interval (CI) = 0.92-1.06 and GMR = 0.98, 0.94-1.01, respectively]. The rates of adverse renal outcomes and possible MPA-related adverse effects were low, and similar in exposed and controls [rate ratios (RR) = 0.94, 0.49-1.84 and RR = 1.08, 0.74-1.58, respectively]. The pharmacokinetic analysis did not signal meaningful differences regarding exposure to MPA, overall (exposed n = 23, control n = 45), if cotreated with cyclosporine (n = 17 vs. n = 26) or with tacrolimus (n = 8 vs. n = 17). CONCLUSIONS In patients treated with MPA, variant allele SLCO1B1 c.521T>C appears of no practical relevance regarding the 12-month renal graft function, MPA safety and exposure to MPA at early steady-state.
Collapse
Affiliation(s)
| | - Luka Penezić
- Department of Urology, University Hospital Center Zagreb
| | - Livija Šimičević
- Divison of Pharmacogenomics and Therapy Individualization University Hospital Center Zagreb and Department of Biochemistry and Clinical Chemistry, Zagreb University School of Medicine
| | - Tvrtko Hudolin
- Department of Urology, University Hospital Center Zagreb
- Department of Urology, Zagreb University School of Medicine
| | - Željko Kaštelan
- Department of Urology, University Hospital Center Zagreb
- Department of Urology, Zagreb University School of Medicine
| | - Nada Božina
- Department of Pharmacology, Zagreb University School of Medicine, Zagreb Croatia
| | - Vladimir Trkulja
- Department of Pharmacology, Zagreb University School of Medicine, Zagreb Croatia
| |
Collapse
|
7
|
Canet E, Brule N, Pere M, Feuillet F, Blancho G, Martin-Lefevre L, Garandeau C, Asehnoune K, Rozec B, Duveau A, Dube L, Pierrot M, Humbert S, Tirot P, Boyer JM, Labadie F, Robert R, Benard T, Kerforne T, Thierry A, Lesieur O, Vincent JF, Lesouhaitier M, Larmet R, Vigneau C, Goepp A, Bouju P, Quentin C, Egreteau PY, Huet O, Renault A, Le Meur Y, Venhard JC, Buchler M, Voellmy MH, Herve F, Schnell D, Courte A, Glotz D, Amrouche L, Hazzan M, Kamar N, Moal V, Bourenne J, Le Quintrec M, Morelon E, Kamel T, Grimbert P, Heng AE, Merville P, Garin A, Hiesse C, Fermier B, Mousson C, Guyot-Colosio C, Bouvier N, Rerolle JP, Durrbach A, Drouin S, Caillard S, Frimat L, Girerd S, Albano L, Rostaing L, Bertrand D, Hertig A, Westeel PF, Montini F, Delpierre E, Dorez D, Alamartine E, Ouisse C, Sébille V, Reignier J. Hypothermia for expanded criteria organ donors in kidney transplantation in France (HYPOREME): a multicentre, randomised controlled trial. THE LANCET. RESPIRATORY MEDICINE 2024; 12:693-702. [PMID: 38876137 DOI: 10.1016/s2213-2600(24)00117-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND Expanded criteria donors help to increase graft availability, but provide organs with an increased risk of delayed graft function. We aimed to investigate whether donor hypothermia decreases the risk of delayed graft function compared with normothermia. METHODS We did this multicentre, randomised, controlled, parallel-arm trial at 53 intensive care units and transplant centres in France. We included expanded criteria donors in whom death was diagnosed based on neurological criteria, in compliance with French law, and the recipients of their kidney grafts. Eligible expanded criteria donors were older than 60 years or were aged 50-59 years and had at least two other risk factors (history of hypertension, creatinine >132 μmol/L, or cerebrovascular cause of death). Donors were randomly assigned to hypothermia (34-35°C) or normothermia (36·5-37·5°C). Machine perfusion was used routinely. Randomisation was done using a computer-generated, interactive, web-response system, in permuted blocks (block size six), stratified by centre. Outcome assessors were masked; investigator masking was not feasible. The primary outcome was the proportion of kidney recipients with delayed graft function, defined as renal replacement therapy within 7 days after transplantation, assessed in the modified intention-to-treat (mITT) population, which included all recipients who received at least one kidney from an expanded criteria donor, with the exception of those under guardianship. Secondary outcomes in expanded criteria donors were the number of organs recovered and transplanted, kidney function, body temperature, total volume of fluids administered, blood pressure and need for vasopressors and inotropes, and adverse events (cardiovascular events, metabolic disturbances, and coagulation disorders). Secondary outcomes in kidney recipients were duration of hospital stay, kidney graft function and vital status at day 7, day 28, 3 months, and 1 year after transplantation, and adverse events (infections, cardiovascular events, and surgical complications). Secondary outcomes were assessed in the mITT population. The trial was registered at ClinicalTrials.gov, NCT03098706. FINDINGS Between Nov 9, 2017, and March 3, 2021, 365 donors were randomly assigned, of whom 298 (151 [51%] male, 147 [49%] female) provided kidneys to 526 recipients (323 [61%] male, 203 [39%] female). 251 recipients in the hypothermia group and 275 recipients in the normothermia group were included in the analysis. Graft function was delayed in 40 (16%) of 251 recipients in the hypothermia group and 58 (21%) of 275 recipients in the normothermia group (odds ratio 0·71 [95% CI 0·44-1·13]; p=0·14; absolute difference -5·2% [95% CI 11·8-1·5]). Compared with donors in the normothermia group, donors in the hypothermia group had higher highest mean arterial pressure (115 mm Hg [SD 22] vs 108 mm Hg [20]; p=0·001). 1 year after transplantation, recipients in the hypothermia group had a lower mean creatinine concentration (152·4 μmol/L [SD 59·1] vs 169·7 μmol/L [51·4]; p=0·0351) and a higher mean creatinine clearance (42·3 mL/min/1·73 m2 [15·8] vs 40·5 mL/min/1·73 m2 [17·9]; p=0·0414) than those in the normothermia group. No significant differences between groups were identified for any other secondary outcomes. INTERPRETATION Hypothermia in expanded criteria donors whose organs were routinely stored using machine perfusion did not decrease the frequency of delayed kidney graft function. However, hypothermia was associated with a lower serum creatinine concentration and a higher creatinine clearance 1 year after transplantation. FUNDING French Ministry of Health and French Intensive Care Society.
Collapse
|
8
|
Penezić L, Nađ-Škegro S, Hadžavdić A, Ganoci L, Kaštelan Ž, Trkulja V, Božina N. Inosine monophosphate dehydrogenase type 2 polymorphism IMPDH2 3757T>C (rs11706052) and 12-month evolution of the graft function in renal transplant recipients on mycophenolate-based immunosuppression. THE PHARMACOGENOMICS JOURNAL 2024; 24:15. [PMID: 38769303 DOI: 10.1038/s41397-024-00335-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024]
Abstract
Variant allele at the inosine monophosphate dehydrogenase type 2 polymorphism IMPDH2 3757T>C has been associated with increased enzyme activity and reduced susceptibility to mycophenolic acid (MPA) in vitro. It has been suggested associated with an increased risk of acute rejection in renal transplant recipients on MPA-based immunosuppression, but not unambiguously. We assessed one-year evolution of the estimated glomerular filtration rate (eGFR) in transplanted variant allele carriers and wild-type subjects, while controlling for a number of demographic, pharmacogenetic, (co)morbidity, and treatment baseline and time-varying covariates. The eGFR slopes to day 28 (GMR = 1.01, 95% CI 0.93-1.09), and between days 28 and 365 (GMR = 1.01, 95% CI 0.99-1.02) were practically identical in 52 variant carriers and 202 wild-type controls. The estimates (95%CIs) remained within the limits of ±20% difference even after adjustment for a strong hypothetical effect of unmeasured confounders. Polymorphism IMPDH2 3757T>C does not affect the renal graft function over the 1st year after transplantation.
Collapse
Affiliation(s)
- Luka Penezić
- Department of Urology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Sandra Nađ-Škegro
- Department of Urology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Ayla Hadžavdić
- Teaching Institute for Emergency Medicine of Istria County, Zagreb, Croatia
| | - Lana Ganoci
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Center Zagreb, Zagreb, Croatia
| | - Željko Kaštelan
- Department of Urology, University Hospital Center Zagreb, Zagreb, Croatia
- Department of Urology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vladimir Trkulja
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Nada Božina
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
9
|
Streichart L, Felldin M, Ekberg J, Mjörnstedt L, Lindnér P, Lennerling A, Bröcker V, Mölne J, Holgersson J, Daenen K, Wennberg L, Lorant T, Baid-Agrawal S. Tocilizumab in chronic active antibody-mediated rejection: rationale and protocol of an in-progress randomized controlled open-label multi-center trial (INTERCEPT study). Trials 2024; 25:213. [PMID: 38519988 PMCID: PMC10958896 DOI: 10.1186/s13063-024-08020-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/26/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Chronic active antibody-mediated rejection (caAMR) in kidney transplants is associated with irreversible tissue damage and a leading cause of graft loss in the long-term. However, the treatment for caAMR remains a challenge to date. Recently, tocilizumab, a recombinant humanized monoclonal antibody directed against the human interleukin-6 (IL-6) receptor, has shown promise in the treatment of caAMR. However, it has not been systematically investigated so far underscoring the need for randomized controlled studies in this area. METHODS The INTERCEPT study is an investigator-driven randomized controlled open-label multi-center trial in kidney transplant recipients to assess the efficacy of tocilizumab in the treatment of biopsy-proven caAMR. A total of 50 recipients with biopsy-proven caAMR at least 12 months after transplantation will be randomized to receive either tocilizumab (n = 25) added to our standard of care (SOC) maintenance treatment or SOC alone (n = 25) for a period of 24 months. Patients will be followed for an additional 12 months after cessation of study medication. After the inclusion biopsies at baseline, protocol kidney graft biopsies will be performed at 12 and 24 months. The sample size calculation assumed a difference of 5 ml/year in slope of estimated glomerular filtration rate (eGFR) between the two groups for 80% power at an alpha of 0.05. The primary endpoint is the slope of eGFR at 24 months after start of treatment. The secondary endpoints include assessment of the following at 12, 24, and 36 months: composite risk score iBox, safety, evolution and characteristics of donor-specific antibodies (DSA), graft histology, proteinuria, kidney function assessed by measured GFR (mGFR), patient- and death-censored graft survival, and patient-reported outcomes that include transplant-specific well-being, adherence to immunosuppressive medications and perceived threat of the risk of graft rejection. DISCUSSION No effective treatment exists for caAMR at present. Based on the hypothesis that inhibition of IL-6 receptor by tocilizumab will reduce antibody production and reduce antibody-mediated damage, our randomized trial has a potential to provide evidence for a novel treatment strategy for caAMR, therewith slowing the decline in graft function in the long-term. TRIAL REGISTRATION ClinicalTrials.gov NCT04561986. Registered on September 24, 2020.
Collapse
Affiliation(s)
- Lillian Streichart
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Marie Felldin
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Jana Ekberg
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Lars Mjörnstedt
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Per Lindnér
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Annette Lennerling
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Verena Bröcker
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Mölne
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Holgersson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg and Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kristien Daenen
- Department of Nephrology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lars Wennberg
- Department of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Lorant
- Section of Transplantation Surgery, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Seema Baid-Agrawal
- Transplant Institute, Sahlgrenska University Hospital, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, 413 45, Gothenburg, Sweden.
| |
Collapse
|
10
|
López Del Moral C, Wu K, Naik M, Osmanodja B, Akifova A, Lachmann N, Stauch D, Hergovits S, Choi M, Bachmann F, Halleck F, Schrezenmeier E, Schmidt D, Budde K. Predictors of graft failure after first detection of de novo donor-specific HLA antibodies in kidney transplant recipients. Nephrol Dial Transplant 2023; 39:84-94. [PMID: 37410616 DOI: 10.1093/ndt/gfad149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND De novo donor-specific antibodies (dnDSAs) may cause antibody-mediated rejection and graft dysfunction. Little is known about the clinical course after first detection of dnDSAs during screening in asymptomatic patients. We aimed to assess the value of estimated glomerular filtration rate (eGFR) and proteinuria to predict graft failure in patients with dnDSAs and their potential utility as surrogate endpoints. METHODS All 400 kidney transplant recipients with dnDSAs at our centre (1 March 2000-31 May 2021) were included in this retrospective study. The dates of graft loss, rejection, doubling of creatinine, ≥30% eGFR decline, proteinuria ≥500 mg/g and ≥1000 mg/g were registered from the first dnDSA appearance. RESULTS During 8.3 years of follow-up, graft failure occurred in 33.3% of patients. Baseline eGFR and proteinuria correlated with 5-year graft loss (area under the receiver operating characteristics curve 0.75 and 0.80, P < .001). Creatinine doubled after a median of 2.8 years [interquartile range (IQR) 1.5-5.0] from dnDSA and the time from doubling creatinine to graft failure was 1.0 year (IQR 0.4-2.9). Analysing eGFR reduction ≥30% as a surrogate endpoint (148/400), the time from dnDSA to this event was 2.0 years (IQR 0.6-4.2), with a positive predictive value (PPV) of 45.9% to predict graft loss, which occurred after 2.0 years (IQR 0.8-3.2). The median time from proteinuria ≥500 mg/g and ≥1000 mg/g to graft failure was identical, 1.8 years, with a PPV of 43.8% and 49.0%, respectively. Composite endpoints did not improve PPV. Multivariable analysis showed that rejection was the most important independent risk factor for all renal endpoints and graft loss. CONCLUSIONS Renal function, proteinuria and rejection are strongly associated with graft failure in patients with dnDSA and may serve as surrogate endpoints.
Collapse
Affiliation(s)
- Covadonga López Del Moral
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Nephrology, Marqués de Valdecilla University Hospital-IDIVAL, Santander, Spain
| | - Kaiyin Wu
- Department of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel Naik
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Aylin Akifova
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Lachmann
- Institute for Transfusion Medicine, HLA-Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Diana Stauch
- Institute for Transfusion Medicine, HLA-Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sabine Hergovits
- Institute for Transfusion Medicine, HLA-Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health Charité - Universitätsmedizin Berlin, BIH Academy, Berlin, Germany
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
11
|
Barbosa ACS, Mauroner LG, Kumar J, Sims-Lucas S. Delayed graft function post renal transplantation: a review on animal models and therapeutics. Am J Physiol Renal Physiol 2023; 325:F817-F825. [PMID: 37855040 PMCID: PMC10878700 DOI: 10.1152/ajprenal.00146.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023] Open
Abstract
The incidence of end-stage renal disease (ESRD) has been increasing worldwide. Its treatment involves renal replacement therapy, either by dialyses or renal transplantation from a living or deceased donor. Although the initial mortality rates for patients on dialysis are comparable with kidney transplant recipients, the quality of life and long-term prognosis are greatly improved in transplanted patients. However, there is a large gap between availability and need for donor kidneys. This has led to the increase in the use of expanded kidney donor criteria. Allograft dysfunction immediately after transplant sets it up for many complications, such as acute rejection and shorter allograft survival. Delayed graft function (DGF) is one of the immediate posttransplant insults to the kidney allograft, which is increasing in prevalence due to efforts to maximize the available donor pool for kidneys and use of expanded kidney donor criteria. In this review, we discuss the risk factors for DGF, its implications for long-term allograft survival, animal models of DGF, and the therapeutic options currently under evaluation for prevention and management of DGF.
Collapse
Affiliation(s)
- Anne C S Barbosa
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, UPMC Children's Hospital, Pittsburgh, Pennsylvania, United States
| | - Lillian G Mauroner
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, UPMC Children's Hospital, Pittsburgh, Pennsylvania, United States
| | - Juhi Kumar
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, UPMC Children's Hospital, Pittsburgh, Pennsylvania, United States
| | - Sunder Sims-Lucas
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh, UPMC Children's Hospital, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
12
|
Kosinski L, Frey E, Klein A, O'Doherty I, Romero K, Stegall M, Helanterä I, Gaber AO, Fitzsimmons WE, Aggarwal V. Longitudinal estimated glomerular filtration rate (eGFR) modeling in long-term renal function to inform clinical trial design in kidney transplantation. Clin Transl Sci 2023; 16:1680-1690. [PMID: 37350196 PMCID: PMC10499426 DOI: 10.1111/cts.13579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/10/2023] [Indexed: 06/24/2023] Open
Abstract
Kidney transplantation is the preferred treatment for individuals with end-stage kidney disease. From a modeling perspective, our understanding of kidney function trajectories after transplantation remains limited. Current modeling of kidney function post-transplantation is focused on linear slopes or percent decline and often excludes the highly variable early timepoints post-transplantation, where kidney function recovers and then stabilizes. Using estimated glomerular filtration rate (eGFR), a well-known biomarker of kidney function, from an aggregated dataset of 4904 kidney transplant patients including both observational studies and clinical trials, we developed a longitudinal model of kidney function trajectories from time of transplant to 6 years post-transplant. Our model is a nonlinear, mixed-effects model built in NONMEM that captured both the recovery phase after kidney transplantation, where the graft recovers function, and the long-term phase of stabilization and slow decline. Model fit was assessed using diagnostic plots and individual fits. Model performance, assessed via visual predictive checks, suggests accurate model predictions of eGFR at the median and lower 95% quantiles of eGFR, ranges which are of critical clinical importance for assessing loss of kidney function. Various clinically relevant covariates were also explored and found to improve the model. For example, transplant recipients of deceased donors recover function more slowly after transplantation and calcineurin inhibitor use promotes faster long-term decay. Our work provides a generalizable, nonlinear model of kidney allograft function that will be useful for estimating eGFR up to 6 years post-transplant in various clinically relevant populations.
Collapse
Affiliation(s)
| | - Eric Frey
- Critical Path InstituteTucsonArizonaUSA
| | | | | | | | - Mark Stegall
- Department of SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Ilkka Helanterä
- Department of Transplantation and Liver SurgeryHelsinki University HospitalHelsinkiFinland
| | - Ahmed Osama Gaber
- Department of Surgery, Houston Methodist HospitalHoustonTexasUSA
- Weill Cornell MedicineNew YorkNew YorkUSA
| | | | | | | |
Collapse
|
13
|
Guo J, Qin C, Li X, Zhuang X. The Flow Cytometric Analysis of Peripheral Blood Lymphocytes and Expression of HLA II Molecules in Lymphocyte During Acute Rejection After Renal Transplantation. J Inflamm Res 2023; 16:2607-2613. [PMID: 37360623 PMCID: PMC10289173 DOI: 10.2147/jir.s410341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Objective To investigate the changes in the proportion of peripheral blood lymphocytes and the expression of HLA II molecules in lymphocytes during acute rejection after renal transplantation. Methods Thirty-five patients who had undergone renal transplantation were selected. Eighteen patients with clinical and pathological confirmed acute rejection were selected as the test group, and twelve patients without clinical acute rejection symptoms were selected as the control group. Flow cytometry analysis was used to determine the proportion of peripheral blood lymphocytes. The mRNA and protein expression of HLA II molecules on peripheral blood lymphocytes were detected using real-time fluorescence quantification and immunoblotting, respectively. Results The proportion of T lymphocytes, B lymphocytes, and CD4CD8 double positive T cells in the Control Group were 67.48% ± 5.35%, 10.82% ± 1.26%, and 0.88% ± 0.06%, respectively, and in the Test Group were 87.52% ± 6.28%, 3.36% ± 0.26%, and 0.34% ± 0.03%, with a significant difference respectively. The mRNA and protein expressions of HLA II molecules of peripheral blood B lymphocytes in the control group were significantly higher that these in the test group. Conclusion The proportion of peripheral blood T lymphocytes, B lymphocytes, CD4CD8 double positive T cells, and the expression of HLA II molecules of peripheral blood lymphocytes can all indicate the occurrence of acute renal transplantation rejection, which were exceedingly useful to clinicians in judging the acute rejection of renal transplantation in the early stages.
Collapse
Affiliation(s)
- Jianzhuang Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, Shandong, People’s Republic of China
| | - Chengwei Qin
- Department of Anesthesiology, Shandong Second Provincial General Hospital, Jinan, Shandong, People’s Republic of China
| | - Xiangdong Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, Shandong, People’s Republic of China
| | - Xiaoxuan Zhuang
- International Medical College of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
14
|
van Vugt LK, Schagen MR, de Weerd A, Reinders ME, de Winter BC, Hesselink DA. Investigational drugs for the treatment of kidney transplant rejection. Expert Opin Investig Drugs 2022; 31:1087-1100. [PMID: 36175360 DOI: 10.1080/13543784.2022.2130751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Kidney transplant rejection remains an important clinical problem despite the development of effective immunosuppressive drug combination therapy. Two major types of rejection are recognized, namely T-cell-mediated rejection (TCMR) and antibody-mediated rejection (ABMR), which have a different pathophysiology and are treated differently. Unfortunately, long-term outcomes of both TCMR and ABMR remain unsatisfactory despite current therapy. Hence, alternative therapeutic drugs are urgently needed. AREAS COVERED This review covers novel and investigational drugs for the pharmacological treatment of kidney transplant rejection. Potential therapeutic strategies and future directions are discussed. EXPERT OPINION The development of alternative pharmacologic treatment of rejection has focused mostly on ABMR, since this is the leading cause of kidney allograft loss and currently lacks an effective, evidence-based therapy. At present, there is insufficient high-quality evidence for any of the covered investigational drugs to support their use in ABMR. However, with the emergence of targeted therapies, this potential arises for individualized treatment strategies. In order to generate more high-quality evidence for such strategies and overcome the obstacles of classic, randomized, controlled trials, we advocate the implementation of adaptive trial designs and surrogate clinical endpoints. We believe such adaptive trial designs could help to understand the risks and benefits of promising drugs such as tocilizumab, clazakizumab, belimumab, and imlifidase.
Collapse
Affiliation(s)
- Lukas K van Vugt
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maaike R Schagen
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Annelies de Weerd
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marlies Ej Reinders
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brenda Cm de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Erasmus MC Transplant Institute, Rotterdam, the Netherlands.,Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
15
|
Naesens M, Loupy A, Hilbrands L, Oberbauer R, Bellini MI, Glotz D, Grinyó J, Heemann U, Jochmans I, Pengel L, Reinders M, Schneeberger S, Budde K. Rationale for Surrogate Endpoints and Conditional Marketing Authorization of New Therapies for Kidney Transplantation. Transpl Int 2022; 35:10137. [PMID: 35669977 PMCID: PMC9163307 DOI: 10.3389/ti.2022.10137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022]
Abstract
Conditional marketing authorization (CMA) facilitates timely access to new drugs for illnesses with unmet clinical needs, such as late graft failure after kidney transplantation. Late graft failure remains a serious, burdensome, and life-threatening condition for recipients. This article has been developed from content prepared by members of a working group within the European Society for Organ Transplantation (ESOT) for a Broad Scientific Advice request, submitted by ESOT to the European Medicines Agency (EMA), and reviewed by the EMA in 2020. The article presents the rationale for using surrogate endpoints in clinical trials aiming at improving late graft failure rates, to enable novel kidney transplantation therapies to be considered for CMA and improve access to medicines. The paper also provides background data to illustrate the relationship between primary and surrogate endpoints. Developing surrogate endpoints and a CMA strategy could be particularly beneficial for studies where the use of primary endpoints would yield insufficient statistical power or insufficient indication of long-term benefit following transplantation.
Collapse
Affiliation(s)
- Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- *Correspondence: Maarten Naesens,
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Hôpital Necker, Paris, France
| | - Luuk Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | | | - Denis Glotz
- Paris Translational Research Center for Organ Transplantation, Hôpital Saint Louis, Paris, France
| | | | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, Munich, Germany
| | - Ina Jochmans
- Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Liset Pengel
- Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Marlies Reinders
- Erasmus MC Transplant Institute, Department of Internal Medicine, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Stefan Schneeberger
- Department of General, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
16
|
Naesens M, Budde K, Hilbrands L, Oberbauer R, Bellini MI, Glotz D, Grinyó J, Heemann U, Jochmans I, Pengel L, Reinders M, Schneeberger S, Loupy A. Surrogate Endpoints for Late Kidney Transplantation Failure. Transpl Int 2022; 35:10136. [PMID: 35669974 PMCID: PMC9163814 DOI: 10.3389/ti.2022.10136] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022]
Abstract
In kidney transplant recipients, late graft failure is often multifactorial. In addition, primary endpoints in kidney transplantation studies seek to demonstrate the short-term efficacy and safety of clinical interventions. Although such endpoints might demonstrate short-term improvement in specific aspects of graft function or incidence of rejection, such findings do not automatically translate into meaningful long-term graft survival benefits. Combining many factors into a well-validated model is therefore more likely to predict long-term outcome and better reflect the complexity of late graft failure than using single endpoints. If conditional marketing authorization could be considered for therapies that aim to improve long-term outcomes following kidney transplantation, then the surrogate endpoint for graft failure in clinical trial settings needs clearer definition. This Consensus Report considers the potential benefits and drawbacks of several candidate surrogate endpoints (including estimated glomerular filtration rate, proteinuria, histological lesions, and donor-specific anti-human leukocyte antigen antibodies) and composite scoring systems. The content was created from information prepared by a working group within the European Society for Organ Transplantation (ESOT). The group submitted a Broad Scientific Advice request to the European Medicines Agency (EMA), June 2020: the request focused on clinical trial design and endpoints in kidney transplantation. Following discussion and refinement, the EMA made final recommendations to ESOT in December 2020 regarding the potential to use surrogate endpoints in clinical studies that aim to improving late graft failure.
Collapse
Affiliation(s)
- Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Luuk Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria
| | | | - Denis Glotz
- Paris Translational Research Center for Organ Transplantation, Hôpital Saint Louis, Paris, France
| | | | - Uwe Heemann
- Department of Nephrology, Technical University of Munich, Munich, Germany
| | - Ina Jochmans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Liset Pengel
- Centre for Evidence in Transplantation, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Marlies Reinders
- Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Stefan Schneeberger
- Department of General, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Hôpital Necker, Paris, France
| |
Collapse
|