Skip to main content
Nephrology Dialysis Transplantation logoLink to Nephrology Dialysis Transplantation
. 2024 Jan 3;39(3):531–549. doi: 10.1093/ndt/gfad258

International consensus on post-transplantation diabetes mellitus

Adnan Sharif 1,2,, Harini Chakkera 3, Aiko P J de Vries 4,5, Kathrin Eller 6, Martina Guthoff 7, Maria C Haller 8,9, Mads Hornum 10, Espen Nordheim 11,12, Alexandra Kautzky-Willer 13, Michael Krebs 14, Aleksandra Kukla 15,16, Amelie Kurnikowski 17, Elisabeth Schwaiger 18, Nuria Montero 19, Julio Pascual 20,21, Trond G Jenssen 22,23, Esteban Porrini 24, Manfred Hecking 25,26,27
PMCID: PMC11024828  PMID: 38171510

ABSTRACT

Post-transplantation diabetes mellitus (PTDM) remains a leading complication after solid organ transplantation. Previous international PTDM consensus meetings in 2003 and 2013 provided standardized frameworks to reduce heterogeneity in diagnosis, risk stratification and management. However, the last decade has seen significant advancements in our PTDM knowledge complemented by rapidly changing treatment algorithms for management of diabetes in the general population. In view of these developments, and to ensure reduced variation in clinical practice, a 3rd international PTDM Consensus Meeting was planned and held from 6–8 May 2022 in Vienna, Austria involving global delegates with PTDM expertise to update the previous reports. This update includes opinion statements concerning optimal diagnostic tools, recognition of prediabetes (impaired fasting glucose and/or impaired glucose tolerance), new mechanistic insights, immunosuppression modification, evidence-based strategies to prevent PTDM, treatment hierarchy for incorporating novel glucose-lowering agents and suggestions for the future direction of PTDM research to address unmet needs. Due to the paucity of good quality evidence, consensus meeting participants agreed that making GRADE (Grading of Recommendations, Assessment, Development, and Evaluations) recommendations would be flawed. Although kidney-allograft centric, we suggest that these opinion statements can be appraised by the transplantation community for implementation across different solid organ transplant cohorts. Acknowledging the paucity of published literature, this report reflects consensus expert opinion. Attaining evidence is desirable to ensure establishment of optimized care for any solid organ transplant recipient at risk of, or who develops, PTDM as we strive to improve long-term outcomes.

Keywords: GLP-1 analogues, metabolic syndrome, NODAT, post-transplant diabetes mellitus, SGLT2 inhibitors

INTRODUCTION

Post-transplantation diabetes mellitus (PTDM) significantly contributes to morbidity and mortality after solid organ transplantation (SOT). The last International PTDM Consensus Meeting in 2013 consolidated heterogenous clinical practice and suggested standards of care for the screening, diagnosis and management of PTDM [1]. However, the PTDM field has evolved dramatically since 2013, justifying an update. Research has enhanced our understanding, while expanded therapeutic options in the general population have dramatically shifted treatment algorithms. In this rapidly changing climate, ambitions to improve long-term SOT outcomes require optimized strategies to prevent/manage PTDM that are aligned with the latest scientific updates.

This Meeting Report summarizes proceedings from the 3rd International PTDM Consensus Meeting held in Vienna, Austria, from 6–8 May 2022. The meeting was endorsed by the European Renal Association (Diabesity Working Group) and the European Society for Organ Transplantation (EKITA Working Group). An international expert panel was convened by invitation, comprising 18 transplant clinicians, diabetologists and scientists with an active interest in the field, to deliberate updates to the previous consensus statement relevant for contemporary clinical practice. Invitations were based upon a meeting prerequisite to systematically review existing literature for presentation at open scientific sessions, encouraging debate and discussion. While targeting all SOT recipients, published data are kidney-centric and organ-specific considerations are required. After reviewing and reflecting upon the paucity of good quality evidence, consensus opinion agreed that making GRADE (Grading of Recommendations, Assessment, Development, and Evaluations) recommendations would be flawed [2]. Therefore, our terminological use of ‘Opinion Statement’ is deliberate to acknowledge this. This position statement reflects the consensus view of expert delegates. Ultimately, attaining this evidence is desirable to ensure establishment of optimized care for any solid organ transplant recipient at risk of, or who develops, PTDM as we strive to improve long-term outcomes.

OPINION STATEMENT 1: PERFORM AN ORAL GLUCOSE TOLERANCE TEST FOR DIAGNOSIS AND SCREENING; START ON THE WAITING LIST

Glucose thresholds for defining diabetes in the general population are based on the probability of developing retinopathy [3], but only one study explores this issue post-transplantation [4]. An oral glucose tolerance test (OGTT) is essential for diagnosis and screening (see Supplementary data, Table S1), as alternatives like haemoglobin A1c (HbA1c) lack diagnostic sensitivity [5–7] and association with adverse outcomes [1, 8, 9]. Patients with impaired glucose tolerance (IGT), exclusively diagnosed by OGTT, or PTDM are at risk for cardiovascular disease [9] and premature death [1, 8]. Importantly, OGTTs allow earlier identification of at-risk individuals on the waiting list [10]. When diagnosed early or by 2-h postprandial glucose only, PTDM may have greater chance of reversibility, although this may reflect low reproducibility [11]. Supplementary data, Table S2 summarizes the published evidence.

Long-term evolution of PTDM is characterized by metabolic variability [7, 11, 12]. Individuals with prediabetes (impaired fasting glucose and/or IGT) or PTDM risk factors will benefit from repeated (e.g. annual) OGTT testing. If diagnosed early (e.g. 3 months post-operatively), PTDM may need later confirmation. A diagnosis and screening algorithm is proposed (Fig. 1) but warrants validation for improvement of outcomes.

Figure 1:

Figure 1:

Five aspects of risk assessment for and diagnosis of PTDM and IGT.

OPINION STATEMENT 2: BE AWARE OF LONG-TERM CONSEQUENCES OF PREDIABETES AND PTDM

PTDM is associated with overall graft loss [13], cardiovascular events [8, 14] and all-cause mortality [8], while microvascular complications are less studied [4] and patient-reported outcomes are scarce. Some studies observe no association with patient/graft survival [15, 16], but this discrepancy might be influenced by heterogenous cohorts, diagnostic criteria or methodological differences. Importantly, the association of prediabetes with mortality and cardiovascular events should be appreciated [9]. Other long-term consequences of PTDM require evaluation. For example, diabetes is associated with several cancers in the general population but data with PTDM are limited. A recent cohort analysis has observed an association between PTDM and future renal cell cancer [17], consistent with observations from a Danish cohort describing increased risk for cardiovascular and cancer-related mortality in SOT recipients with pre-transplant diabetes or PTDM [18].

OPINION STATEMENT 3: PRIORITIZE CLINICAL ATTENTION TO ‘AT RISK’ GROUPS

SOT recipients are at risk for the development of prediabetes/PTDM, but certain patients have a disproportionately higher risk. Early identification of this high-risk group is crucial to ensure that resources are directed to the most vulnerable, who may be amenable to intervention.

This ‘at-risk’ group can be classified by clinical phenotypes or novel risk prediction methods like polygenic risk scores (PRS). The latter estimates an individual's genetic liability for a specific disease according to their genotypic profile and has been studied after liver and kidney transplantation [19]. PRS are associated with pre-transplant type 2 diabetes and post-surgery PTDM. PRS in liver donors, but not kidney donors, was an independent risk factor for PTDM development and a combined liver donor/recipient PRS improved PTDM prediction over-and-above a clinical variable model alone. Further research is recommended to identify the optimal way to identify at-risk groups.

OPINION STATEMENT 4: CONSIDER UNDERLYING PATHOMECHANISM OF PTDM DEVELOPMENT AND THE INTER-RELATIONSHIP BETWEEN β-CELL DYSFUNCTION AND METABOLIC STRESS

PTDM arises from an interaction between pre-transplant and post-transplant risk factors (Supplementary data, Fig. S1). Many pre-transplant risk factors are common to type 2 diabetes (i.e. obesity, metabolic syndrome), but immunosuppression is the most important post-transplant risk factor. Pre-transplant risk factors may identify individuals at risk from immunosuppression-induced β-cell toxicity amenable to intervention, supporting the use of waiting-list screening.

Mechanistically a combination of pancreatic β-cell dysfunction and insulin resistance are predisposing factors for PTDM, with superimposed immunosuppression accelerating pre-existing damage [20]. A mechanistic approach is depicted in Supplementary data, Fig. S2 according to an animal model of calcineurin inhibitor (CNI)-induced toxicity, potentiating similar cellular damage induced by obesity and insulin resistance, which indicates common pathways in β-cell dysfunction [20]. Importantly, this principle has been corroborated with slightly different pathways in human islets and pancreas transplant biopsies [21]. Tacrolimus induces β-cell damage provoked by the glucolipotoxicity state secondary to multi-factorial insults, pathogenic pathways [e.g. mammalian target of rapamycin (mTOR) pathway] [22] responsible for β-cell maintenance and function [20]. Furthermore, low-grade inflammatory stress is associated with early occurrence of PTDM [23] and early post-transplant mortality in general [24]. Thus, a ‘two-hit’ hypothesis combining transplantation-induced β-cell insult on a background of metabolic stress converging in a dysfunctional synergy is an attractive hypothesis for the development of prediabetes/PTDM. However, other confounders must not be overlooked. For example, Halden et al. demonstrated infusion of the incretin hormone glucagon-like peptide 1 (GLP-1) during fasting and hyperglycaemic conditions in patients with PTDM compared with normal glucose tolerance, rectified pathophysiological defects like hyperglucagonemia, and diminished first- and second-phase insulin secretion [25].

OPINION STATEMENT 5: CHOOSE AN IMMUNOSUPPRESSION REGIMEN FOR OPTIMIZATION OF PATIENT AND GRAFT SURVIVAL

Despite the association between immunosuppression and PTDM, de novo regimens should not be routinely modified to reduce PTDM risk or adjusted after PTDM development. However, for selected patients, tailored immunosuppression may be justified if development of diabetes outweighs other risks. Patient-specific factors, immunological considerations and competing risks must all be factored when choosing immunosuppression on a personalized basis.

No robust data link induction therapy directly to PTDM risk. However, lymphocyte-depletion therapies (e.g. thymoglobulin, alemtuzumab) can facilitate lower exposure to maintenance CNIs and steroids which can reduce PTDM risk.

Regarding CNIs, Torres et al. randomized 128 de novo kidney transplant recipients (KTRs) at high-risk for PTDM but low immunological risk to: (i) tacrolimus and rapid steroid withdrawal, (ii) cyclosporine and steroid maintenance, or (iii) tacrolimus with steroid maintenance [26]. All arms received basiliximab and steroids. Patient/graft survival and graft function were similar between study arms, with tacrolimus and steroid maintenance providing the best balance between risk for PTDM versus acute rejection. There is limited evidence supporting conversion of CNI in established PTDM. In a randomized controlled trial (RCT) involving 87 KTRs, conversion from tacrolimus to cyclosporine significantly improved glycaemic control with no increased risk for acute rejection [27]. Late changes to immunosuppressive regimens may alleviate PTDM but this requires further evaluation to ensure glycaemic benefits outweigh long-term allograft risks. There is not enough evidence to support using different tacrolimus formulations, such as immediate versus prolonged release, but results from ongoing studies are awaited (see Supplementary data, Table S3).

Belatacept has a favourable metabolic risk profile, including less PTDM [28], in comparison with CNIs and different regimens have been explored in RCTs including KTRs [29]. Belatacept is an acceptable alternative to CNIs to reduce PTDM in low immunological–risk patients if logistical and cost implications are surmountable. Any studies to explore efficacy in non-renal SOT recipients should ensure data capture of PTDM as a secondary outcome.

Although mTOR inhibitors are diabetogenic, incidence of PTDM is not significantly increased by their use which may reflect reduced CNI exposure. A recent meta-analysis evaluating the combination of CNI plus mTOR inhibitors in de novo KTRs observed no increase of 1-year PTDM versus CNI plus antiproliferative agents in 13 studies [n = 4561 participants; relative risk 1.16, 95% confidence interval (CI) 0.97–1.38, = .10] [30]. These results were confirmed in the TRANSFORM (TRANSplant eFficacy and safety Outcomes with an eveRolimus-based regiMen) study, a 24-month, prospective, open-label trial in 2037 de novo KTRs randomized to receive everolimus with reduced-exposure CNI versus mycophenolate with standard-exposure CNI [31]. No difference in PTDM incidence was observed (risk ratio 1.09, 95% CI 0.87–1.37) with comparable efficacy and graft function.

There is no evidence to suggest any glycaemic risk from anti-proliferative agents such as mycophenolate mofetil or azathioprine.

Regarding steroids, a previous Cochrane analysis published in 2016 observed similar rates of mortality, graft loss and PTDM comparing regimens of steroid avoidance/withdrawal (stratified before or after 14 days, respectively) versus steroid maintenance, but higher rates of rejection [32]. In an updated analysis incorporating post-2016 RCTs of steroid avoidance [33, 34], lower rates of PTDM are now observed in steroid avoidance versus maintenance (risk ratio 0.70, 95% CI 0.56–0.88, = .002) but with similar mortality, graft loss and rejection observations to before (see Supplementary data, Fig. S3). However, the HARMONY study contributes a large effect size but is flawed by overreliance on HbA1c for PTDM diagnosis in the context of anemia rates between 27% and 39% across study arms [33]. Early steroid withdrawal may have differential impact stratified by age, with older SOT recipients in a population-cohort study demonstrating more favourable responses to steroid withdrawal (e.g. lower PTDM and mortality) but increased risk for rejection [35]. Balancing PTDM versus graft-related concerns with steroid avoidance/withdrawal is essential, although patient/graft survival should take priority. In a causal estimation effects registry analysis including 6070 KTRs, steroid withdrawal within 18 months post-transplantation was associated with increased risk of graft loss compared with steroid maintenance [36]. If a steroid avoidance regimen is desired then induction therapy with lymphocyte depletion should be considered.

OPINION STATEMENT 6: EMPHASIZE LIFESTYLE MODIFICATION TO ALL PATIENTS; CONSIDER MEDICAL OR SURGICAL INTERVENTION FOR TREATMENT OF OBESITY; USE INTERMITTENT EXOGENOUS INSULIN INTERVENTION EARLY POST-TRANSPLANTATION FOR POST-OPERATIVE HYPERGLYCAEMIA

Since the last meeting report [1], various groups have summarized suggestions on PTDM prevention [37–41]. These include: (i) dietary modification; (ii) physical exercise/training; (iii) pharmacological intervention; (iv) immunosuppression modification; (v) bariatric surgery; (vi) performing OGTTs pre-transplant for targeted intervention; and (vii) other measures including manipulation of microbiota. Meeting participants agreed any opinion regarding prevention would intuitively become stronger with increasing PTDM risk.

Regarding (i), uncertainty exists about the best dietary intervention [42], as observational evidence only supports Mediterranean diets [43] or increased vegetable intake [44]. With (ii), the CAVIAR (Comparing glycaemic benefits of Active Versus passive lifestyle Intervention in kidney Allograft Recipients) RCT implemented a graded exercise program with active dietician intervention (versus leaflet advice), which did not improve pathophysiological markers of glucose metabolism but reduced PTDM incidence [45]. An observational study demonstrated higher physical activity levels lowered risk of PTDM, and cardiovascular and all-cause mortality [46]. Although better evidence is desirable, meeting participants agreed that lifestyle modification, combining measures (i) and (ii), should be emphasized post-transplantation based upon evidence from the general population [47].

As for (iii), meeting participants agreed early exogenous insulin administration could be considered for PTDM prevention despite a recent RCT not reaching its primary endpoint (1-year PTDM incidence) [48]. This agreement acknowledged that the odds for overt PTDM at 1-year were significantly reduce in the adjusted per-protocol analysis only [48], and was also based on an earlier RCT (cited in previous meeting report) [1]. However, higher hypoglycaemia rates with this approach must be acknowledged [48] and enthusiasm may be influenced by inpatient length of stay post-operatively. An ongoing multicentre RCT testing early administration of vildagliptin for PTDM prevention is underway (Supplementary data, Table S3) [49], but another RCT was recently published demonstrating that post-operative sitagliptin was safe but did not lead to significant improvement in OGTT-derived 2-h glucose at 3 and 6 months post-transplantation [50].

The most controversial issue with PTDM prevention is immunosuppression tailoring for SOT patients at higher PTDM risk as per (iv), which is addressed under Opinion Statement 5. Meeting participants agreed further research is warranted to investigate immunosuppression modification strategies to prevent or treat PTDM.

Concerning (v), there is convincing evidence that bariatric surgery is beneficial for individuals with morbid obesity and chronic kidney disease (CKD), including those already waitlisted or seeking eligibility [51, 52]. In kidney transplant candidates with obesity (e.g. body mass index ≥35 kg/m2) refractory to lifestyle intervention, consider surgical or medical intervention which will enable successful transplantation and may aid PTDM prevention. A non-randomized study reported zero cases of PTDM in 12 non-diabetic KTRs transplanted after post-laparoscopic sleeve gastrectomy, in comparison with 3 of 18 patients from a matched non-laparoscopic sleeve gastrectomy control group (statistically not significant) [53]. As an alternative, GLP-1 receptor agonists might be a promising pharmacological option for individuals with advanced CKD and obesity who are transplant candidates. Studies are pending to determine feasibility (Supplementary data, Table S3).

Regarding measures (vi) and (vii), Hap et al. performed OGTTs among 80 waitlisted kidney transplant candidates and recommended a low carbohydrate diet, lifestyle modification and increased physical activity to 31 patients with dysglycaemia (with 28/31 showing attenuated glucose metabolism throughout the 12-month observational period post-transplant) [54]. These results align with several measures highlighted above showing that behavioural factors such as motivation are important to enable PTDM prevention.

OPINION STATEMENT 7: USE THE NOVEL AGENTS; PERSONALIZE GLUCOSE-LOWERING THERAPY BASED UPON A PATIENT-DEPENDENT HIERARCHY

Cardiovascular outcome trials using glucose-lowering treatment in KTRs are lacking. Novel agents, sodium-glucose co-transporter 2 (SGLT2) inhibitors and GLP-1 receptor agonists, now dominate diabetes treatment guidelines [55]. Meeting participants agreed that novel agents are under-utilized for PTDM management due to limitations of transplant-specific evidence (see Tables 1A/1B). However, prescribing is sub-optimal even in diabetic kidney disease patients in whom there are clear treatment benefits as per national/international recommendations [56]. This reflects a disconnect between clinical guidelines and real-world prescribing. Available transplant studies do not currently indicate a clear safety risk, which is why our personal view is more enthusiastic in comparison with recent KDIGO guidance on diabetes and CKD recommending more cautious adoption [57]. Meeting participants agreed targeted PTDM studies are desirable but adoption should not be delayed based on current evidence. Meeting participants also agreed that initiation of glucose-lowering agents will be reliant upon accessibility. However, if accessibility is not an issue, then a patient-dependent hierarchy (Fig. 2) is advisable.

Table 1A:

Prospective studies on glucose-lowering agents after kidney transplantation.

Study Study size and design Duration Intervention/comparator Primary outcome/main outcome Secondary outcomes Primary outcome results/outcome results Strength Weakness
Insulin
 Hecking et al. 2012 [65] N = 56; RCT 12 months Basal (NPH) insulin ± short acting insulin/standard of care Difference in HbA1c at Month 3 Difference in HbA1c at Month 6 and 12, prevalence of NODAT and IGT, capillary blood glucose profile and the amount of insulin needed HbA1c at 3 months was significantly different and lower in the basal insulin group First study to prove that PTDM might be preventable. Pathophysiologically plausible Patients who dropped out were replaced. Small single-centre analysis. Results presented as unadjusted and adjusted results
 Schwaiger et al. 2021 [48] N = 263; RCT 24 months Basal (NPH) insulin ± short acting insulin/standard of care PTDM at 1 year PTDM at 2 years, glycaemic control, kidney function, patient and graft survival PTDM risk in unadjusted and adjusted ITT analyses: no statistically significant difference was observed between groups; PTDM risk in unadjusted PP analysis: no statistically significant difference was observed between groups; PTDM risk in adjusted PP analysis: a statistically significant difference with lower occurrence of PTDM was observed in the basal insulin group Relatively large multicentre RCT compared with other PTDM studies Significant baseline differences regarding ADPKD. Protocol deviations as described by the authors
Metformin
 Alnasrallah et al. 2019 [66] N = 19; pilot RCT 3–12 months Metformin/standard of care (lifestyle instruction) Feasibility of recruitment, tolerability of metformin, efficacy of metformin in IGT Lipid profile, change in body weight, cardiac events, adverse events, proportion of patients who revert to normal glucose metabolism, drug discontinuation, SAE 19 patients out of 78 with an OGTT recruited. Tolerability of metformin comparable between groups at 3 and 12 months. Efficacy of metformin on HbA1c and fasting plasma glucose not different at the tested time points First RCT with metformin in transplanted patients, focus on prevention (IGT patients), patient education taken seriously Sample size too small to prove absence of lactic acidosis
Thiazolidinediones
 Baldwin and Duffin 2004 [67] N = 18 (N = 11 with DM2, N = 7 with PTDM); prospective, observational (interventional) 133–718 days Rosiglitazone HbA1c improvement, avoidance of PTDM, avoidance of insulin dependency in PTDM Blood levels of cyclosporin A, tacrolimus, creatinine. Weight, peripheral oedema, pulmonary congestion, liver enzyme, lipids HbA1c improved significantly in DM2 and PTDM, PTDM patients did not depend on insulin Novelty at that time. Duration of follow-up Study design itself, potential selection bias, sample size
 Villanueva et al. 2005 [68] N = 40; prospective, observational (interventional) 12 months Rosiglitazone To evaluate the effect of rosiglitazone on insulin resistance in PTDM Physical examination, serum chemistry, weight, cyclosporin and tacrolimus levels 91% of patients initially treated with insulin were able to discontinue insulin. 30% were controlled with rosiglitazone monotherapy. Serum creatinine was stable during treatment with rosiglitazone. 13% treated with rosiglitazone developed oedema Real-world study Immunosuppressive regimen was modified
 Voytovich et al. 2005 [69] N = 10; prospective, observational (interventional) 4 weeks Rosiglitazone Impact on insulin sensitivity, plasma glucose and endothelial function in KTR with glucose intolerance Safety parameters Mean glucose disposal rate increased, the mean fasting plasma glucose and 2-h plasma glucose fell significantly, AUC glucose (from OGTT) was sig. reduced. Insulin secretion was not reduced. No sig. association between lowering plasma glucose and the improvement of endothelial function Mechanistically sophisticated (clamp-derived measurement of insulin sensitivity), pathophysiologically insightful Relatively short treatment time which limits clinical interpretation (also of safety parameters)
Han et al. 2010 [70] N = 83; RCT 12 months Pioglitazone/control (not receiving pioglitazone) Mean and max. carotid IMT Adiponectin levels, lipids, insulin secretory function and sensitivity Mean max IMT decreased only in the pioglitazone group. Association of adiponectin and IMT in pioglitazone group. Pioglitazone increased insulin sensitivity Study design, sample size, endpoints not limited to glucose metabolism Not placebo-controlled
 Werzowa et al. 2013 [71] N = 52; RCT 3 months Vildaglitpin/pioglitazone/placebo Difference in change in OGTT-derived 2-h plasma glucose Difference in 2hPG, FPG, HbA1c and fasting insulin within the groups before and after treatment, change in kidney and liver function, side effects The primary endpoint did not reach statistical significance Diabetologically comprehensive. The only study in prediabetes Weak effects. Limited pathophysiological information
Meglitinides
 Voytovich et al. 2007 [72] N = 14 (N = 6 with PTDM, N = 8 with IGT); prospective, observational (interventional) 2 weeks Nateglinide Insulin response and glucose excursions after a standardized liquid meal Carbohydrate and fat oxidation rates in indirect calorimetry, insulin, C-peptide, free fatty acids, triglycerides, lipids, liver enzymes, and creatinine, CsA and tacrolimus levels Significant decrease in 2hPG, decline of AUC glucose0–240 min, increase of AUC ins0–30 min, AUC ins30–120 min, and AUCC-peptide. Lower postprandial glucose in self-measurements Proof of mechanism of action Relatively short treatment time which limits clinical interpretation (also of safety parameters)
GLP-1 receptor agonists
 Pinelli et al. 2013 [73] N = 5; prospective, observational (interventional) (case series) 3 weeks Liraglutide Tacrolimus AUC0–12h Tacrolimus trough levels, allograft function, blood glucose Tac-AUC reduced, Tac trough levels unaltered, reduction of postprandial glucose and body weight The only study reporting an AUC for tacrolimus under GLP-1-RA treatment Small sample size, descriptive, difference in Tac-AUC not emphasized in the conclusion
 Halden et al. 2016 [25] N = 24; RCT 2–4 weeks GLP-1 infusion/0.9% saline. Hyperglycaemic clamp Fasting levels of plasma glucose, glucagon, and insulin, AUC concentrations Glucagon, proinsulin and insulin secretory response to arginine Patients with PTDM showed a reduced ability to suppress circulating glucagon levels during the hyperglycaemic clamp. First- and second-phase insulin secretion was lower compared with the control group Pathomechanistically sophisticated Relatively short treatment time which limits clinical interpretation (also of safety parameters)
DPP4 inhibitors
 Lane et al. 2011 [74] N = 15; prospective, observational (interventional) (pilot study) 3 months Sitagliptin Effect of sitagliptin on tacrolimus and sirolimus levels and changes in renal function Side effects and change in HbA1c Significant reduction in HbA1c, no significant change in tacrolimus or sirolimus levels, no significant change in eGFR First report on DPP4 inhibitors in transplanted patients None, apart from small sample size and descriptive design
 Werzowa et al. 2013 [71] N = 52; RCT 3 months Vildaglitpin/pioglitazone/placebo Difference in change in 2hPG Difference in 2hPG, FPG, HbA1c and fasting insulin within the groups before and after treatment, change in kidney and liver function, side effects The primary endpoint did not reach statistical significance Diabetologically comprehensive. The only study in prediabetes Weak effects. Limited pathophysiological information
 Soliman et al. 2013 [75] N = 62; RCT 12 weeks Sitagliptin/insulin glargine Change in HbA1c from baseline to Week 12 Change in body weight, fasting plasma glucose, lipid profile Significant reduction in HbA1c and fasting plasma glucose, comparable to insulin Study design clinically meaningful, answering a clinical need at that time Many drop-outs in the insulin group
 Haidinger et al. 2014 [76] N = 33; RCT 3 months (active), 4 months (including follow-up) Vildaglitpitn/placebo Difference in the intraindividual change in OGTT-derived 2hPG between Differences between the intraindividual change in OGTT-derived 2hPG from baseline to 4 months, FPG, HbA1c and fasting insulin, rate of side-effects, change in eGFR, albuminuria/proteinuria, change in liver function parameters from baseline, and immunosuppressant serum levels Intraindividual change in 2hPG between the vildagliptin, and placebo group was statistically significant at Month 3 OGTTs with insulin sensitivity and secretion during treatment and 1 month after drug discontinuation Short treatment duration
 Strøm Halden et al. 2014 [77] N = 19; cross-over RCT 8 weeks (4 weeks treatment) Sitagliptin/sitagliptin-free Effect of sitagliptin on insulin secretion Plasma glucose, insulin sensitivity, endothelial function, safety parameters (calcineurin inhibitor/everolimus levels and changes in renal function) Median (IQR) first- and second-phase insulin secretion responses increased following sitagliptin treatment as compared with control Study clinically well-intended to ensure treatment in all patients, OGTTs with insulin sensitivity and secretion, markers on cardiovascular risk Patients had different CNIs, temporal effects due to cross-over design and relatively short treatment duration
 Delos Santos et al. 2023 [50] N = 61; RCT 6 months Sitagliptin/placebo OGTT-derived 2-h glucose at 3 months PTDM prevention at 3 months (defined by normal OGTT) OGTT-derived 2-h glucose was 24 mg/dL lower and PTDM risk reduction was 18% in the sitagliptin group (not significantly different) Mechanistically compelling. The first study on PTDM prevention to date using a DPP4 inhibitor As lower 2-h glucose among patients on treatment was expectable, more information could have been derived and presented from the OGTTs
SGLT2is
 Schwaiger et al. 2019 [78] N = 14, N = 24 matched reference patients with PTDM; prospective, observational (interventional) 4 weeks run in, 4 weeks empagliflozin monotherapy, 12 months follow-up Empagliflozin monotherapy, followed by empagliflozin as add on Intra‐individual difference in the 2hPG between the baseline OGTT and the OGTT after 4 weeks: non-inferiority design Laboratory parameters, anthropometric measurements, blood pressure, and medications. Bioimpedance spectroscopy‐based assessment of fluid volume status and body composition, urinary tract infections compared with reference group OGTT-derived 2hPG increased during 4 weeks of empagliflozin treatment (P = ns), demonstrating clinically inferiority Many endpoints studied Inferiority of empagliflozin as substitute for insulin would have been expectable, small sample size
 Halden et al. 2019 [62] N = 44; double-blind RCT 24 weeks Empagliflozin/placebo Change in weighted mean glucose estimated with continuous glucose monitoring from iPro2 Change in HbA1c, FPG, 2hPG in OGTT, body weight, WHR, body composition including visceral fat, blood pressure, and eGFR Primary endpoint not evaluated (technical error), median change in HbA1c significantly reduced after 24 weeks of empagliflozin treatment compared with placebo Sophisticated study design, the authors confirmed that SGLT2is have no glucose-lowering effect at eGFR <45 mL/min/1.73 m2 in transplanted patients Prespecified primary endpoint not analysed, some uncertainty remains regarding urinary tract infections
 Mahling et al. 2019 [79] N = 10; prospective, observational (interventional) (case series) 12.0 (5.3–12.0) months Empagliflozin as add on therapy Changes in median eGFR, median HbA1c from baseline to end of follow-up Urinary tract infection, side effects Median eGFR remained stable, median HbA1c decreased Timely publication, real-world study Descriptive analysis, small sample size
 Shah et al. 2019 [80] N = 24; prospective, observational (interventional) 6 months Canagliflozin Not specified Body weight, blood pressure, HbA1c, serum creatinine, tacrolimus trough levels Reduction in weight, blood pressure and HbA1c, tacrolimus trough levels unchanged Only study with canagliflozin Descriptive, small sample size, only 1 woman
 Sánchez Fructuoso et al. 2023 [81] N = 338 (N = 204 with PTDM, N = 134 with T2DM); multicentre, prospective, observational (interventional) 6 months Canagliflozin, empagliflozin, dapagliflozin, ertugliflozin Assess adverse events, especially UTIs and/or mycoses in DKTRs placed on SGLT2i treatment Haemoglobin, eGFR, UACR and/or UPCR, glycaemia (FPG, HbA1c), lipid metabolism 26% patients had an adverse event over 6 months, the most frequent being a UTI (14% patients). In 10% patients, SGLT2i were suspended (mostly because of UTI). However, in a post hoc subgroup analysis, UTIs were similar between DKTRs treated with SGLT2i over 12 months, compared with non-DKTRs (17.9% versus 16.7%). Body weight, blood pressure, fasting glycaemia, HbA1c uric acid, UPCR lower after SGLT2i treatment; magnesium and haemoglobin levels higher The study provides comprehensive and useful clinical information, due in particular to its adequate sample size. Well designed (in the absence of funding for large RCTs). Meaningful way of researching UTI risk in this context 12-months’ follow-up not yet completed in N = 105 patients at the time of publication

Green coloured boxes: randomized controlled trials; yellow-coloured boxes: prospective observational/(interventional) studies.

ns: not statistically significant; SAE: serious adverse events; IMT: intima media thickness; AUC: area under the curve; BMI: body mass index; NODAT: new-onset diabetes after transplantation; SGLT2i: SGLT2 inhibitor; ITT: intention-to-treat; ADPKD: autosomal dominant polycystic kidney disease; DM2: type 2 diabetes mellitus; 2hPG: 2-h plasma glucose; FPG: fasting plasma glucose; sig.: significant; IQR: interquartile range; WHR: waist-to-hip ratio; UTI: urinary tract infection; DKTR: diabetic KTR; UACR: urine albumin:creatinine ratio; UPCR: urine protein:creatinine ratio; PP: per protocol.

Table 1B:

Retrospective studies on glucose-lowering agents after kidney transplantation or SOT including kidney.

Study Study size Organ Primary results Secondary results Strength Weakness
Insulin
 Chandra et al. 2023 [82] N = 23 (N = 10 treated with insulin isophane, N = 13 treated with insulin glargine) Kidney 12 episodes of hypoglycaemia in glargine-treated PTDM patients compared with 3 in isophane-treated PTDM patients (= .056) Significantly lower blood glucose and HbA1c in the glargine vs. isophane group. In the glargine group, 8 out of 12 hypoglycaemic episodes were nocturnal (1 out of 3 hypoglycaemic episodes were nocturnal in the isophane group) First report on hypoglycaemia risk with various basal insulin regimens Patient population predominantly male (87% males) and of relatively young age (average age <40 years in both groups)
Sulfonylureas
 Tuerk et al. 2008 [83] N = 47 gliquidone + 28 rosiglitazone (N = 75) Kidney Mean fasting blood glucose improved, success rate was similar in both groups In 4 patients the dose of gliquidone therapy had to be reduced due to hypoglycaemia. Pretreatment with other antidiabetics was identified as a negative prognostic factor First report on SUs in PTDM Comparison against TZDs (rosiglitazone) with non-standard treatment goals may be somewhat unusual
Metformin
 Kurian et al. 2008 [84] N = 32 in the metformin and N = 46 in the thiazolidinedione group (pioglitazone, rosiglitazone) Kidney No significant difference in HbA1c before and after metformin therapy or thiazolidinedione therapy No case of lactic acidosis in the metformin group. A slight decrease in eGFR was only significant in the preexisting DM group Long observational period, first data on safety of metformin The fact that no treatment effect was observed may not be meaningful in view of sample size and study design
 Stephen et al. 2014 [60] N = 46 914 (4609 with metformin, 42 305 non-metformin glucose-lowering agent Kidney Metformin claims were filled later and were associated with higher eGFR before the first claim Metformin was associated with lower adjusted hazard for living and deceased donor allograft survival at 3 years. Metformin was associated with lower mortality Sample size, outcome data No clear distinction between DM and PTDM, bias by indication
 Kwon et al. 2023 [59] N = 1193 with metformin, N = 802 without Kidney Metformin reduced death-censored graft failure, no association with all-cause mortality No association with BPAR, no confirmed case of lactic acidosis Sample size, outcome data Bias by indication
Thiazolidinediones
 Pietruck et al. 2005 [85] N = 22 (rosiglitazone) Kidney 73% had sufficient glycaemic control Diabetologically comprehensive. Novelty at that time. Duration of follow-up Sample size
 Luther and Baldwin 2004 [86] N = 10 with DM2 and PTDM in KTR and LTR (pioglitazone) Kidney Mean HbA1c and mean total daily insulin dose was significantly lower after pioglitazone initiation. Mean serum creatinine levels did not change. Mean blood tacrolimus levels were lower in the pioglitazone group (no difference in dose-normalized tacrolimus blood levels) Mean BMI increased after pioglitazone. Mean daily prednisolone dose decreased non- significantly. No significant fluid retention and no differences in mean serum lipid values after pioglitazone initiation Emphasis on safety. Duration of follow-up Study design itself, potential selection bias, sample size, similarity to study by Baldwin and Duffin
 Kurian et al. 2008 [84] N = 32 in the metformin and N = 46 in the thiazolidinedione group (pioglitazone, rosiglitazone) Kidney No significant difference in HbA1c before and after metformin therapy or thiazolidinedione therapy No case of lactic acidosis in the metformin group. A slight decrease in eGFR was only significant in the preexisting DM group Long observational period, first data on safety of metformin The fact that no treatment effect was observed may not be meaningful in view of sample size and study design
Meglitinides
 Türk et al. 2006 [87] N = 44 (N = 23 repaglinide, N = 21 rosiglitazone) Kidney After 6 months, 14/23 patients showed successful repaglinide treatment (significant improvement of blood glucose concentrations and HbA1c <7%, no other medication needed) No significant change in creatinine, cyclosporine A and tacrolimus levels. Similar success rate and HbA1c as in rosiglitazone group First report on glinides in PTDM Comparison of various subgroups with non-standard treatment goals
GLP-1 receptor agonists
 Liou et al. 2018 [88] N = 7 (liraglutide) Kidney Glycaemia improved incl. HbA1c eGFR improved Long treatment duration Small sample size
 Singh et al. 2019 [89] N = 63 (dulaglutide) Kidney, liver, heart Weight loss Reduction in insulin requirements Relatively large cohort Inhomogeneous cohort (multiple organs)
 Thangavelu et al. 2020 [90] N = 19 Kidney, liver, heart Stability of the tacrolimus level Reduction in body weight, BMI and HbA1c Relatively early study Inhomogeneous cohort (multiple organs), small sample size
 Singh et al. 2020 [91] N = 63 (dulaglutide) N = 25 (liraglutide Kidney, liver, heart Weight loss Reduction in insulin requirement Relatively large cohort Similar data as previous study
 Vigara et al. 2022 [92] N = 50 (semaglutide, liraglutide, duaglutide) Kidney Improvement in eGFR and reduction in proteinuria Body weight reduction, improvement in HbA1c Relatively large cohort Exclusion criteria not clear
 Sweiss et al. 2022 [93] N = 118, 70% KTRs, 32% PTDM (liraglutide, dulaglutide, semaglutide, exenatide) Kidney, lung, liver Significant difference fasting blood glucose and HbA1c at baseline to 3- to 12-month nadir, weight loss 7% nausea, 4% pancreatitis, 7% hypo- glycaemic events Large cohort of SOT with GLP-1-RA treatment Various transplanted organs and various GLP-1-RA
DPP4 inhibitors
 Sanyal et al. 2013 [94] N = 21 (linagliptin) Kidney Linaglitpin monotherapy was effective for glycaemic control in patients with NODAT Insulin requirement in 2 patients, 1 hypoglycaemic episode Early real-world data Entirely descriptive
 Boerner et al. 2014 [95] N = 22 (sitagliptin) Kidney Diabetes control (defined by HbA1c) improved at 6 months and persisted at 12 months Graft function (serum creatinine and eGFR) did not differ at month 12. No effect on liver transaminase levels and rare occurrence of transplant associated adverse events Systematic follow-up Entirely descriptive
 Bae et al. 2016 [96] N = 65 (vildagliptin, sitagliptin, linagliptin) Kidney HbA1c at 3 months significantly decreased from baseline in the linagliptin group compared with other DPP4i Cyclosporin trough levels were increased in the sitagliptin group compared with the vildagliptin group Various DPP4 inhibitors analysed Superiority of one gliptine versus others is clinically implausible and not known in DM2, may have been dose-dependent
 Guardado-Mednoza et al. 2019 [97] N = 14 (linagliptin + basal (NPH) and lispro insulin) N = 14 basal (NPH) and lispro insulin Kidney Significant lower fasting plasma glucose levels in the linagliptin plus insulin group after 5 days and at 1 year Lower insulin doses in the insulin plus linagliptin group and less severe hypoglycaemic events Data from the early post-transplant period Treatment duration unclear, therefore, follow-up data not meaningful
 Sanyal et al. 2021 [98] N = 95 any agent [all received linagliptin (alone or in combination)] Kidney NODAT patients achieved long-term glycaemic control and improved renal function Most patients needed a combination therapy. Linagliptin was effective without producing hypoglycaemia Manuscript describes a real-world outpatient scenario Bias by indication
SGLT2is
 Rajasekeran et al. 2017 [99] N = 10 (6 KTRs, 4 SPKTs, PTDM and T2DM) (canagliflozin) Kidney Meaningful changes in various parameters (incl. HbA1c, weight, and blood pressure), but none of them significant First study of SGLT2is in transplanted patients Small sample size
 Attallah and Yassine 2019 [100] N = 8 (empagliflozin) Kidney Increase in creatinine, decrease in HbA1c, body weight and urinary protein excretion Meaningful HbA1c reduction shown for patients with excellent allograft function Descriptive, small sample size
 AlKindi et al. 2020 [101] N = 8 (empagliflozin, dapagliflozin) Kidney Decrease in HbA1c and body mass index, kidney function remained stable Meaningful HbA1c reduction shown for patients with excellent allograft function Descriptive, small sample size
 Song et al. 2021 [102] N = 50 (empagliflozin, canagliflozin, dapagliflozin) Kidney Weight reduction Improvement in hypomagnesemia, reduction in insulin requirement Relatively large cohort Low incidence of UTIs is difficult to interpret (more clarity would have been helpful)
 Lim et al. 2022 [103] N = 226 (empagliflozin, dapagliflozin) among N = 2083 (propensity score matching 1:3) Kidney Improvement in a composite outcome, consisting of all-cause mortality, death-censored graft failure, and serum creatinine doubling Graft failure reduced (this item was also part of the composite outcome) First study to describe hard outcome data in KTRs Written like an RCT (misleading)
 Lemke et al. 2022 [104] N = 39 (canagliflozin, dapagliflozin) Kidney Decrease in HbA1c Kidney function and tacrolimus levels not meaningfully altered Honest discussion of therapy pros and cons UTIs not clarified further

Both tables contain studies from patients with disorders of the glucose metabolism that became known after transplantation (hyperglycaemia/PTDM/IGT). If studies were entirely conducted with patients who had type 2 diabetes before transplantation, they were not listed.

DM: diabetes mellitus; DM2: type 2 diabetes mellitus; NODAT: new-onset diabetes after transplantation; BMI: body mass index; GLP-1-RA: GLP-1 receptor agonist; DPP4i: DPP4 inhibitor; SU: sulfonylurea; TZDs: thiazolidinediones; BPAR: biopsy proved acute rejection.

Figure 2:

Figure 2:

Glucose-lowering treatment in KTRs: suggested algorithm.

Metformin is cheap and easily available. While advised for use only with estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2, renal restrictions are not an absolute requirement [58]. Observational studies show an association with lower risk for death-censored graft failure [59] and post-transplant mortality [60, 61] but not cardiovascular-related mortality. Metformin may be an appropriate choice for solid organ transplant recipients at low risk for adverse cardio-renal outcomes or if access to novel anti-diabetics is an issue. However, for solid organ transplant recipients at moderate to high risk for adverse cardio-renal outcomes with no accessibility issues, the consensus opinion was novel anti-diabetic therapies should be strongly considered before metformin.

SGLT2 inhibitors can be used for the treatment of PTDM once stable graft function is achieved [62]. Initiation should be influenced by comorbidities like heart failure (supporting use) and significant urosepsis or severe mycotic genital infection risk (discouraging use), although current studies have not shown increased urinary tract infection risk with SGLT2 inhibitors (see Tables 1A and 1B). Enthusiasm for early post-operative commencement will be influenced by local urological practices (e.g. length of post-operative urinary catheter placement, ureteric stent removal). Improvement of glycaemic control may vary based on kidney function (less effective at lower eGFR) [62]. Awareness of the risk for euglycaemic diabetic ketoacidosis is critical, especially in patients with insulin deficiency. SGLT2 inhibitors should be suspended if fasting is required or during an acute illness.

GLP-1 receptor agonists are preferable in patients with obesity. Several non-randomized published reports indicate an acceptable safety profile with no increased rejection or graft failure risk, although gastrointestinal side effects are common. Appropriate education is required for patients who are initiated on incretin mimetics with emphasis on slow dose up-titration to improve tolerance, and suspension of treatment with acute illness [25].

Insulin should be used for treatment of post-operative hyperglycaemia. For stable patients, oral or non-insulin injectable agents (and their combination) are preferable unless diabetes control cannot be achieved. Of note, data on the glucose-lowering effect of basal insulin in KTRs exist for basal neutral protamine Hagedorn (NPH)-insulin alone [48], the peak effect of which can be matched to the glucose peak exhibited by KTRs in the afternoon.

Dipeptidylpeptidase 4 (DPP4) inhibitors are safe but demonstrate no cardio-renal benefit. Thiazolidinediones are better options than sulfonylureas and meglitinides (both have risk of hypoglycaemia), and no evidence exists for alpha-glucosidase inhibitors. Meeting participants agree these drug classes have the lowest priority for clinical use.

In summary, and in view of the pros and cons for each pharmacological therapy, meeting participants agreed that any decision to initiate one glucose-lowering agent versus another is best guided by a patient-dependent hierarchy (shown in Fig. 2) if accessibility is not an issue. Personalization of glucose-lowering therapy is essential, with treatment goals depending on comorbidities, awareness of hypoglycaemia risk and allograft function.

OPINION STATEMENT 8: INCREASE COLLABORATIVE RESEARCH BETWEEN ACADEMIC MEDICINE, MULTI-DISCIPLINARY CLINICAL TEAMS, INDUSTRY PARTNERS AND PATIENTS

Exclusion of SOT recipients from pioneering cardiovascular and renal outcome trials of new glucose-lowering agents has resulted in sub-optimal uptake post-transplantation. Observational studies and RCTs relating to PTDM are in progress (see Supplementary data, Table S3), but more are required and should target at-risk groups for maximum benefit. Patient-reported outcomes, health economic analyses and cost effectiveness models are lacking and require dedicated studies and incorporation as secondary outcomes into RCTs where feasible (suggested PTDM clinical trial endpoints in Supplementary data, Table S4). Lack of robust PTDM data capture by national transplant registries limits the ability to ascertain PTDM-associated outcomes [63]. Acquiring these data should be encouraged to improve our understanding of long-term outcomes with record linkage. Collaboration between healthcare professionals, academic groups, industry and patient groups is essential.

Finally, most published research is after kidney transplantation, but PTDM is a complication affecting all SOT recipients with prevalence rates between 20% and 40% in heart, lung and liver transplant recipients [64]. In a Danish SOT cohort (n = 959), the highest incidence of PTDM is seen 46–365 days post-transplantation. SOT recipients with PTDM had higher risk for all-cause mortality (1.89, 95% CI 1.17–3.06), with cardiovascular and cancer-related causality more common than in non-diabetic SOT recipients [18]. More studies are warranted in non-renal transplant cohorts. While most of this report is valid across SOT cohorts, bespoke differences may be apparent between different solid organ settings to justify organ-specific versus organ-generic recommendations.

CONCLUSION

PTDM is a complex and multi-factorial post-transplant complication, spanning a continuum of disease that may begin prior to transplantation in many cases. This Meeting Report summarizes proceedings from the 3rd International PTDM Consensus meeting, reflecting expert opinion. Optimizing long-term outcomes after SOT, with attenuation of both premature mortality and/or graft loss, is a clinical priority. Therefore, improving our diagnosis, prevention and management of PTDM should be considered an integral component of long-term post-transplant care.

Supplementary Material

gfad258_Supplemental_Files

ACKNOWLEDGEMENTS

The 3rd International PTDM Consensus meeting was endorsed by the European Kidney Transplant Association, an organ expert section of ESOT on kidney transplantation, and Diabesity, a working group of the European Renal Association.

Contributor Information

Adnan Sharif, Department of Nephrology and Transplantation, University Hospitals Birmingham, Birmingham, United Kingdom; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.

Harini Chakkera, Division of Nephrology and Hypertension, Mayo Clinic, Scottsdale, AZ, United States of America.

Aiko P J de Vries, Leiden Transplant Center, Leiden University Medical Center, Leiden, The Netherlands; Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.

Kathrin Eller, Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz Austria.

Martina Guthoff, Department of Diabetology, Endocrinology, Nephrology, University of Tübingen, Tübingen, Germany.

Maria C Haller, Ordensklinikum Linz, Elisabethinen Hospital, Department of Medicine III, Nephrology, Hypertension, Transplantation, Rheumatology, Geriatrics, Linz, Austria; Medical University of Vienna, CeMSIIS, Section for Clinical Biometrics, Vienna, Austria.

Mads Hornum, Department of Nephrology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.

Espen Nordheim, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Nydalen, Norway; Department of Nephrology, Oslo University Hospital-Ullevål, Oslo, Nydalen, Norway.

Alexandra Kautzky-Willer, Department of Internal Medicine III, Clinical Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria.

Michael Krebs, Department of Internal Medicine III, Clinical Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria.

Aleksandra Kukla, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America; William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States of America.

Amelie Kurnikowski, Department of Internal Medicine III, Clinical Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria.

Elisabeth Schwaiger, Department of Internal Medicine, Brothers of Saint John of God Eisenstadt, Eisenstadt, Austria.

Nuria Montero, Nephrology Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, University of Barcelona, Barcelona Spain.

Julio Pascual, Institute Mar for Medical Research-IMIM, Barcelona, Spain; Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain.

Trond G Jenssen, Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Nydalen, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway.

Esteban Porrini, Instituto de Tecnologías Biomédicas (ITB), University of La Laguna, Research Unit Department, Hospital Universitario de Canarias, Tenerife, Spain.

Manfred Hecking, Department of Internal Medicine III, Clinical Division of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria; Center for Public Health, Department of Epidemiology, Medical University of Vienna, Vienna, Austria; Kuratorium for Dialysis and Kidney Transplantation (KfH), Neu-Isenburg, Germany.

FUNDING

No funding was sought for convening this meeting.

DATA AVAILABILITY STATEMENT

No empirical data collected for this manuscript.

CONFLICT OF INTEREST STATEMENT

A.S. has received lecture fees from Chiesi and Napp Pharamceuticals, travel support from Sandoz, grant money from Chiesi and advisory board fees from Novartis. J.P. received lecture fees from Novartis, Chiesi and Sanofi. A.Kukla has received product support from Dexcom and is on the NovoNordisk Advisory Board. M.G. has received research support from Chiesi, lecture fees from Alexion, Astellas, AstraZeneca, Baxter, Bayer, Lilly and Novartis, advisory board fees from Alexion, Boehringer/Lilly and Chiesi, and travel support from Alexion, Astellas and Boehringer/Lilly. K.E. has received grant support from Chiesi, lecture fees from AstraZeneca, Alexion, Chiesi and Novartis, and advisory board fees from AstraZeneca, Alexion and Chiesi. M.Hecking served as a speaker and/or consultant for Astellas Pharma, AstraZeneca, Bayer, Eli Lilly, Fresenius Medical Care, Janssen-Cilag, Siemens Healthcare and Vifor, and received academic study support from Astellas Pharma, Boehringer Ingelheim, Eli Lilly, Nikkiso and Siemens Healthcare. M.Hornum received advisory board fee from AstraZeneca and Bayer, and travel support from AstraZeneca, and also served as speaker and moderator for AstraZeneca. N.M. has received travel support from NovoNordisk and Nordic Pharma and lecture fees from Sanofi. M.K. has received research support from AstraZeneca and Fit for Me, speaker and consulting fees from Lilly, Takeda, Ipsen and Sanofi, and travel support from Pfizer, Novo Nordisk, Merck, Ipsen, HRA Pharma and Boehringer-Ingelheim. A.P.J.V. has received speaker and advisory board fees from Novartis, Sandoz, Chiesi, Astellas, AstraZeneca and CSL Behring (all fees to employer). E.S. has received speaker fees from Amgen and Novartis, and travel support from Takeda and Astellas. M.C.H. has received speaker fees from AstraZeneca and Vifor. T.G.J. has received lecture fees from Boehring Ingelheim, AstraZeneca, NovoNordisk and Takeda, and advisory board fees from Bayer and Abbot Diagnostics. E.N. has received lecture fees from AstraZeneca. A.Kurnikowski, H.C., A.K.-W. and E.P. have no relevant disclosures to report

REFERENCES

  • 1.Sharif A, Hecking M, de Vries APet al. Proceedings from an international consensus meeting on posttransplantation diabetes mellitus: recommendations and future directions. Am J Transplant 2014;14:1992–2000. 10.1111/ajt.12850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lv M, Luo X, Chen Y. GRADE misuse in systematic reviews. Lancet Infect Dis 2022;22:590–1. 10.1016/S1473-3099(22)00218-3 [DOI] [PubMed] [Google Scholar]
  • 3.American Diabetes Association. 2. Classification and diagnosis of diabetes: standards of medical care in diabetes—2021. Diabetes Care 2021;44:S15–33. [DOI] [PubMed] [Google Scholar]
  • 4.Londero TM, Giaretta LS, Farenzena LPet al. Microvascular complications of posttransplant diabetes mellitus in kidney transplant recipients: a longitudinal study. J Clin Endocrinol Metab 2019;104:557–67. 10.1210/jc.2018-01521 [DOI] [PubMed] [Google Scholar]
  • 5.Eide IA, Halden TA, Hartmann Aet al. Limitations of hemoglobin A1c for the diagnosis of posttransplant diabetes mellitus. Transplantation 2015;99:629–35. 10.1097/TP.0000000000000376 [DOI] [PubMed] [Google Scholar]
  • 6.Ussif AM, Åsberg A, Halden TASet al. Validation of diagnostic utility of fasting plasma glucose and HbA1c in stable renal transplant recipients one year after transplantation. BMC Nephrol 2019;20:12. 10.1186/s12882-018-1171-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kurnikowski A, Nordheim E, Schwaiger Eet al. Criteria for prediabetes and posttransplant diabetes mellitus after kidney transplantation: a 2-year diagnostic accuracy study of participants from a randomized controlled trial. Am J Transplant 2022;22:2880–91. 10.1111/ajt.17187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Eide IA, Halden TA, Hartmann Aet al. Mortality risk in post-transplantation diabetes mellitus based on glucose and HbA1c diagnostic criteria. Transpl Int 2016;29:568–78. 10.1111/tri.12757 [DOI] [PubMed] [Google Scholar]
  • 9.Porrini E, Diaz JM, Moreso Fet al. Prediabetes is a risk factor for cardiovascular disease following renal transplantation. Kidney Int 2019;96:1374–80. 10.1016/j.kint.2019.06.026 [DOI] [PubMed] [Google Scholar]
  • 10.Singer J, Aouad LJ, Wyburn Ket al. The utility of pre- and post-transplant oral glucose tolerance tests: identifying kidney transplant recipients with or at risk of new onset diabetes after transplant. Transpl Int 2022;35:10078. 10.3389/ti.2022.10078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Porrini EL, Diaz JM, Moreso Fet al. Clinical evolution of post-transplant diabetes mellitus. Nephrol Dial Transplant 2016;31:495–505. 10.1093/ndt/gfv368 [DOI] [PubMed] [Google Scholar]
  • 12.Guthoff M, Wagner R, Weichbrodt Ket al. Dynamics of glucose metabolism after kidney transplantation. Kidney Blood Press Res 2017;42:598–607. 10.1159/000481375 [DOI] [PubMed] [Google Scholar]
  • 13.Eide IA, Halden TAS, Hartmann Aet al. Associations between posttransplantation diabetes mellitus and renal graft survival. Transplantation 2017;101:1282–9. 10.1097/TP.0000000000001259 [DOI] [PubMed] [Google Scholar]
  • 14.Topitz D, Schwaiger E, Frommlet Fet al. Cardiovascular events associate with diabetes status rather than with early basal insulin treatment for the prevention of post-transplantation diabetes mellitus. Nephrol Dial Transplant 2020;35:544–6. 10.1093/ndt/gfz244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gaynor JJ, Ciancio G, Guerra Get al. Single-centre study of 628 adult, primary kidney transplant recipients showing no unfavourable effect of new-onset diabetes after transplant. Diabetologia 2015;58:334–45. 10.1007/s00125-014-3428-0 [DOI] [PubMed] [Google Scholar]
  • 16.Hussain A, Culliford A, Phagura Net al. Comparing survival outcomes for kidney transplant recipients with pre-existing diabetes versus those who develop post-transplantation diabetes. Diabet Med 2022;39:e14707. [DOI] [PubMed] [Google Scholar]
  • 17.Porrini E, Monetro N, Díaz Jet al. Post-transplant diabetes mellitus and renal cell cancer after renal transplantation. Nephrol Dial Transplant 2023;38:1552–9. [DOI] [PubMed] [Google Scholar]
  • 18.Dos Santos Q, Hornum M, Terrones-Campos Cet al. Posttransplantation diabetes mellitus among solid organ recipients in a Danish cohort. Transpl Int 2022;35:10352. 10.3389/ti.2022.10352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Shaked A, Loza BL, Van Loon Eet al. Donor and recipient polygenic risk scores influence the risk of post-transplant diabetes. Nat Med 2022;28:999–1005. 10.1038/s41591-022-01758-7 [DOI] [PubMed] [Google Scholar]
  • 20.Rodriguez-Rodriguez AE, Porrini E, Torres A.. Beta-cell dysfunction induced by tacrolimus: a way to explain type 2 diabetes? Int J Mol Sci 2021;22:10311. 10.3390/ijms221910311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Trinanes J, Ten Dijke P, Groen Net al. Tacrolimus-induced BMP/SMAD signaling associates with metabolic stress-activated FOXO1 to trigger beta-cell failure. Diabetes 2020;69:193–204. 10.2337/db19-0828 [DOI] [PubMed] [Google Scholar]
  • 22.Rodriguez-Rodriguez AE, Donate-Correa J, Rovira Jet al. Inhibition of the mTOR pathway: a new mechanism of beta cell toxicity induced by tacrolimus. Am J Transplant 2019;19:3240–9. 10.1111/ajt.15483 [DOI] [PubMed] [Google Scholar]
  • 23.Heldal TF, Ueland T, Jenssen Tet al. Inflammatory and related biomarkers are associated with post-transplant diabetes mellitus in kidney recipients: a retrospective study. Transpl Int 2018;31:510–9. 10.1111/tri.13116 [DOI] [PubMed] [Google Scholar]
  • 24.Heldal TF, Asberg A, Ueland Tet al. Inflammation in the early phase after kidney transplantation is associated with increased long-term all-cause mortality. Am J Transplant 2022;22:2016–27. 10.1111/ajt.17047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Halden TA, Egeland EJ, Asberg Aet al. GLP-1 restores altered insulin and glucagon secretion in posttransplantation diabetes. Diabetes Care 2016;39:617–24. 10.2337/dc15-2383 [DOI] [PubMed] [Google Scholar]
  • 26.Torres A, Hernandez D, Moreso Fet al. Randomized controlled trial assessing the impact of tacrolimus versus cyclosporine on the incidence of posttransplant diabetes mellitus. Kidney Int Rep 2018;3:1304–15. 10.1016/j.ekir.2018.07.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wissing KM, Abramowicz D, Weekers Let al. Prospective randomized study of conversion from tacrolimus to cyclosporine A to improve glucose metabolism in patients with posttransplant diabetes mellitus after renal transplantation. Am J Transplant 2018;18:1726–34. 10.1111/ajt.14665 [DOI] [PubMed] [Google Scholar]
  • 28.Muller MM, Schwaiger E, Kurnikowski Aet al. Glucose metabolism after kidney transplantation: insulin release and sensitivity with tacrolimus- versus belatacept-based immunosuppression. Am J Kidney Dis 2021;77:462–4. 10.1053/j.ajkd.2020.07.016 [DOI] [PubMed] [Google Scholar]
  • 29.Kirk AD, Adams AB, Durrbach Aet al. Optimization of de novo belatacept-based immunosuppression administered to renal transplant recipients. Am J Transplant 2021;21:1691–8. 10.1111/ajt.16386 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Montero N, Quero M, Melilli Eet al. Mammalian target of rapamycin inhibitors combined with calcineurin inhibitors as initial immunosuppression in renal transplantation: a meta-analysis. Transplantation 2019;103:2031–56. 10.1097/TP.0000000000002769 [DOI] [PubMed] [Google Scholar]
  • 31.Berger SP, Sommerer C, Witzke Oet al. Two-year outcomes in de novo renal transplant recipients receiving everolimus-facilitated calcineurin inhibitor reduction regimen from the TRANSFORM study. Am J Transplant 2019;19:3018–34. 10.1111/ajt.15480 [DOI] [PubMed] [Google Scholar]
  • 32.Haller MC, Royuela A, Nagler EVet al. Steroid avoidance or withdrawal for kidney transplant recipients. Cochrane Database Syst Rev 2016;2016:CD005632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Thomusch O, Wiesener M, Opgenoorth Met al. Rabbit-ATG or basiliximab induction for rapid steroid withdrawal after renal transplantation (Harmony): an open-label, multicentre, randomised controlled trial. Lancet 2016;388:3006–16. 10.1016/S0140-6736(16)32187-0 [DOI] [PubMed] [Google Scholar]
  • 34.van Sandwijk MS, de Vries APJ, Bakker SJLet al. Early steroid withdrawal compared with standard immunosuppression in kidney transplantation - interim analysis of the Amsterdam-Leiden-Groningen randomized controlled trial. Transplant Direct 2018;4:e354. 10.1097/TXD.0000000000000794 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ahn JB, Bae S, Schnitzler Met al. Effect of early steroid withdrawal on posttransplant diabetes among kidney transplant recipients differs by recipient age. Transplant Direct 2022;8:e1260. 10.1097/TXD.0000000000001260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Haller MC, Kammer M, Kainz Aet al. Steroid withdrawal after renal transplantation: a retrospective cohort study. BMC Med 2017;15:8. 10.1186/s12916-016-0772-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ducloux D, Courivaud C.. Prevention of post-transplant diabetes mellitus: towards a personalized approach. J Pers Med 2022;12:116. 10.3390/jpm12010116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Iqbal A, Zhou K, Kashyap SRet al. Early post-renal transplant hyperglycemia. J Clin Endocrinol Metab 2022;107:549–62. 10.1210/clinem/dgab697 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sridhar VS, Ambinathan JPN, Gillard Pet al. Cardiometabolic and kidney protection in kidney transplant recipients with diabetes: mechanisms, clinical applications, and summary of clinical trials. Transplantation 2022;106:734–48. 10.1097/TP.0000000000003919 [DOI] [PubMed] [Google Scholar]
  • 40.Hecking M, Sharif A, Eller Ket al. Management of post-transplant diabetes: immunosuppression, early prevention, and novel antidiabetics. Transpl Int 2021;34:27–48. 10.1111/tri.13783 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Langsford D, Steinberg A, Dwyer KM.. Diabetes mellitus following renal transplantation: clinical and pharmacological considerations for the elderly patient. Drugs Aging 2017;34:589–601. 10.1007/s40266-017-0478-2 [DOI] [PubMed] [Google Scholar]
  • 42.Li J, Chong A, Carey S.. Dietary interventions on the prevention and management of diabetes in post-kidney transplantation - A systematic review. Nephrology (Carlton) 2022;27:269–80. 10.1111/nep.13982 [DOI] [PubMed] [Google Scholar]
  • 43.Oste MC, Corpeleijn E, Navis GJet al. Mediterranean style diet is associated with low risk of new-onset diabetes after renal transplantation. BMJ Open Diabetes Res Care 2017;5:e000283. 10.1136/bmjdrc-2016-000283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gomes-Neto AW, Oste MCJ, Sotomayor CGet al. Fruit and vegetable intake and risk of posttransplantation diabetes in renal transplant recipients. Diabetes Care 2019;42:1645–52. 10.2337/dc19-0224 [DOI] [PubMed] [Google Scholar]
  • 45.Kuningas K, Driscoll J, Mair Ret al. Comparing glycaemic benefits of active versus passive lifestyle intervention in kidney allograft recipients: a randomized controlled trial. Transplantation 2020;104:1491–9. 10.1097/TP.0000000000002969 [DOI] [PubMed] [Google Scholar]
  • 46.Byambasukh O, Oste MCJ, Gomes-Neto AWet al. Physical activity and the development of post-transplant diabetes mellitus, and cardiovascular- and all-cause mortality in renal transplant recipients. J Clin Med 2020;9:415. 10.3390/jcm9020415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Haw JS, Galaviz KI, Straus ANet al. Long-term sustainability of diabetes prevention approaches: a systematic review and meta-analysis of randomized clinical trials. JAMA Intern Med 2017;177:1808–17. 10.1001/jamainternmed.2017.6040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Schwaiger E, Krenn S, Kurnikowski Aet al. Early postoperative basal insulin therapy versus standard of care for the prevention of diabetes mellitus after kidney transplantation: a multicenter randomized trial. J Am Soc Nephrol 2021;32:2083–98. 10.1681/ASN.2021010127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gaiffe E, Crepin T, Bamoulid Jet al. PRODIG (Prevention of new onset diabetes after transplantation by a short term treatment of vildagliptin in the early renal post-transplant period) study: study protocol for a randomized controlled study. Trials 2019;20:375. 10.1186/s13063-019-3392-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Delos Santos RB, Hagopian JC, Chen Let al. Sitagliptin versus placebo to reduce the incidence and severity of posttransplant diabetes mellitus after kidney transplantation-A single-center, randomized, double-blind controlled trial. Transplantation 2023;107:1180–7. 10.1097/TP.0000000000004373 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kassam AF, Mirza A, Kim Yet al. Long-term outcomes in patients with obesity and renal disease after sleeve gastrectomy. Am J Transplant 2020;20:422–9. 10.1111/ajt.15650 [DOI] [PubMed] [Google Scholar]
  • 52.Yemini R, Nesher E, Carmeli Iet al. Bariatric surgery is efficacious and improves access to transplantation for morbidly obese renal transplant candidates. Obes Surg 2019;29:2373–80. 10.1007/s11695-019-03925-1 [DOI] [PubMed] [Google Scholar]
  • 53.Kim Y, Jung AD, Dhar VKet al. Laparoscopic sleeve gastrectomy improves renal transplant candidacy and posttransplant outcomes in morbidly obese patients. Am J Transplant 2018;18:410–6. 10.1111/ajt.14463 [DOI] [PubMed] [Google Scholar]
  • 54.Hap K, Madziarska K, Zmonarski Set al. Pretransplantation oral glucose tolerance test can prevent posttransplant diabetes mellitus after renal transplantation: preliminary study. Transplant Proc 2018;50:1776–80. 10.1016/j.transproceed.2018.03.125 [DOI] [PubMed] [Google Scholar]
  • 55.Sarafidis P, Ferro CJ, Morales Eet al. SGLT-2 inhibitors and GLP-1 receptor agonists for nephroprotection and cardioprotection in patients with diabetes mellitus and chronic kidney disease. A consensus statement by the EURECA-m and the DIABESITY working groups of the ERA-EDTA. Nephrol Dial Transplant 2019;34:208–30. 10.1093/ndt/gfy407 [DOI] [PubMed] [Google Scholar]
  • 56.Lamprea-Montealegre JA, Madden E, Tummalapalli SLet al. Prescription patterns of cardiovascular- and kidney-protective therapies among patients with type 2 diabetes and chronic kidney disease. Diabetes Care 2022;45:2900–6. 10.2337/dc22-0614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.de Boer IH, Khunti K, Sadusky Tet al. Diabetes management in chronic kidney disease: a consensus report by the American Diabetes Association (ADA) and Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 2022;45:3075–90. 10.1016/j.kint.2022.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lalau JD, Arnouts P, Sharif Aet al. Metformin and other antidiabetic agents in renal failure patients. Kidney Int 2015;87:308–22. 10.1038/ki.2014.19 [DOI] [PubMed] [Google Scholar]
  • 59.Kwon S, Kim YC, Kwon Het al. Metformin use and long-term clinical outcomes in kidney transplant recipients. Am J Kidney Dis 2023;82:290–9.e1. 10.1053/j.ajkd.2023.01.446 [DOI] [PubMed] [Google Scholar]
  • 60.Stephen J, Anderson-Haag TL, Gustafson Set al. Metformin use in kidney transplant recipients in the United States: an observational study. Am J Nephrol 2014;40:546–53. 10.1159/000370034 [DOI] [PubMed] [Google Scholar]
  • 61.Vest LS, Koraishy FM, Zhang Zet al. Metformin use in the first year after kidney transplant, correlates, and associated outcomes in diabetic transplant recipients: a retrospective analysis of integrated registry and pharmacy claims data. Clin Transplant 2018;32:e13302. 10.1111/ctr.13302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Halden TAS, Kvitne KE, Midtvedt Ket al. Efficacy and safety of empagliflozin in renal transplant recipients with posttransplant diabetes mellitus. Diabetes Care 2019;42:1067–74. 10.2337/dc19-0093 [DOI] [PubMed] [Google Scholar]
  • 63.Sharif A. Prevention of post-transplantation diabetes: small steps, big opportunities. J Am Soc Nephrol 2021;32:1833–4. 10.1681/ASN.2021060777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jenssen T, Hartmann A.. Post-transplant diabetes mellitus in patients with solid organ transplants. Nat Rev Endocrinol 2019;15:172–88. 10.1038/s41574-018-0137-7 [DOI] [PubMed] [Google Scholar]
  • 65.Hecking M, Haidinger M, Döller Det al. Early basal insulin therapy decreases new-onset diabetes after renal transplantation. J Am Soc Nephrol 2012;23:739–49. 10.1681/ASN.2011080835 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Alnasrallah B, Goh TL, Chan LWet al. Transplantation and diabetes (Transdiab): a pilot randomised controlled trial of metformin in impaired glucose tolerance after kidney transplantation. BMC Nephrol 2019;20:147. 10.1186/s12882-019-1321-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Baldwin D, Duffin KE.. Rosiglitazone treatment of diabetes mellitus after solid organ transplantation. Transplantation 2004;77:1009–14. 10.1097/01.TP.0000116393.98934.6F [DOI] [PubMed] [Google Scholar]
  • 68.Villanueva G, Baldwin D.. Rosiglitazone therapy of posttransplant diabetes mellitus. Transplantation 2005;80:1402–5. 10.1097/01.tp.0000181165.19788.95 [DOI] [PubMed] [Google Scholar]
  • 69.Voytovich MH, Simonsen C, Jenssen Tet al. Short-term treatment with rosiglitazone improves glucose tolerance, insulin sensitivity and endothelial function in renal transplant recipients. Nephrol Dial Transplant 2005;20:413–8. 10.1093/ndt/gfh641 [DOI] [PubMed] [Google Scholar]
  • 70.Han SJ, Hur KY, Kim YSet al. Effects of pioglitazone on subclinical atherosclerosis and insulin resistance in nondiabetic renal allograft recipients. Nephrol Dial Transplant 2010;25:976–84. 10.1093/ndt/gfp567 [DOI] [PubMed] [Google Scholar]
  • 71.Werzowa J, Hecking M, Haidinger Met al. Vildagliptin and pioglitazone in patients with impaired glucose tolerance after kidney transplantation: a randomized, placebo-controlled clinical trial. Transplantation 2013;95:456–62. 10.1097/TP.0b013e318276a20e [DOI] [PubMed] [Google Scholar]
  • 72.Voytovich MH, Haukereid C, Hjelmesaeth Jet al. Nateglinide improves postprandial hyperglycemia and insulin secretion in renal transplant recipients. Clin Transplant 2007;21:246–51. 10.1111/j.1399-0012.2006.00634.x [DOI] [PubMed] [Google Scholar]
  • 73.Pinelli NR, Patel A, Salinitri FD.. Coadministration of liraglutide with tacrolimus in kidney transplant recipients: a case series. Diabetes Care 2013;36:e171–2. 10.2337/dc13-1066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lane JT, Odegaard DE, Haire CEet al. Sitagliptin therapy in kidney transplant recipients with new-onset diabetes after transplantation. Transplantation 2011;92:e56–7. 10.1097/TP.0b013e3182347ea4 [DOI] [PubMed] [Google Scholar]
  • 75.Soliman AR, Fathy A, Khashab Set al. Sitagliptin might be a favorable antiobesity drug for new onset diabetes after a renal transplant. Exp Clin Transplant 2013;11:494–8. 10.6002/ect.2013.0018 [DOI] [PubMed] [Google Scholar]
  • 76.Haidinger M, Werzowa J, Hecking Met al. Efficacy and safety of vildagliptin in new-onset diabetes after kidney transplantation—A randomized, double-blind, placebo-controlled trial. Am J Transplant 2014;14:115–23. 10.1111/ajt.12518 [DOI] [PubMed] [Google Scholar]
  • 77.Strøm Halden TA, Åsberg A, Vik Ket al. Short-term efficacy and safety of sitagliptin treatment in long-term stable renal recipients with new-onset diabetes after transplantation. Nephrol Dial Transplant 2014;29:926–33. 10.1093/ndt/gft536 [DOI] [PubMed] [Google Scholar]
  • 78.Schwaiger E, Burghart L, Signorini Let al. Empagliflozin in posttransplantation diabetes mellitus: a prospective, interventional pilot study on glucose metabolism, fluid volume, and patient safety. Am J Transplant 2019;19:907–19. 10.1111/ajt.15223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Mahling M, Schork A, Nadalin Set al. Sodium-glucose cotransporter 2 (SGLT2) inhibition in kidney transplant recipients with diabetes mellitus. Kidney Blood Press Res 2019;44:984–92. 10.1159/000501854 [DOI] [PubMed] [Google Scholar]
  • 80.Shah M, Virani Z, Rajput Pet al. Efficacy and safety of canagliflozin in kidney transplant patients. Indian J Nephrol 2019;29:278–81. 10.4103/ijn.IJN_2_18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sánchez Fructuoso AI, Bedia Raba A, Banegas Deras Eet al. Sodium-glucose cotransporter-2 inhibitor therapy in kidney transplant patients with type 2 or post-transplant diabetes: an observational multicentre study. Clin Kidney J 2023;16:1022–34. 10.1093/ckj/sfad007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Chandra A, Rao N, Pooniya Vet al. Hypoglycemia with insulin in post-transplant diabetes mellitus. Transpl Immunol 2023;78:101833. 10.1016/j.trim.2023.101833 [DOI] [PubMed] [Google Scholar]
  • 83.Tuerk TR, Bandur S, Nuernberger Jet al. Gliquidone therapy of new-onset diabetes mellitus after kidney transplantation. Clin Nephrol 2008;70:26–32. 10.5414/CNP70026 [DOI] [PubMed] [Google Scholar]
  • 84.Kurian B, Joshi R, Helmuth A.. Effectiveness and long-term safety of thiazolidinediones and metformin in renal transplant recipients. Endocr Pract 2008;14:979–84. 10.4158/EP.14.8.979 [DOI] [PubMed] [Google Scholar]
  • 85.Pietruck F, Kribben A, Van TNet al. Rosiglitazone is a safe and effective treatment option of new-onset diabetes mellitus after renal transplantation. Transpl Int 2005;18:483–6. 10.1111/j.1432-2277.2004.00076.x [DOI] [PubMed] [Google Scholar]
  • 86.Luther P, Baldwin D.. Pioglitazone in the management of diabetes mellitus after transplantation. Am J Transplant 2004;4:2135–8. 10.1111/j.1600-6143.2004.00613.x [DOI] [PubMed] [Google Scholar]
  • 87.Türk T, Pietruck F, Dolff Set al. Repaglinide in the management of new-onset diabetes mellitus after renal transplantation. Am J Transplant 2006;6:842–6. 10.1111/j.1600-6143.2006.01250.x [DOI] [PubMed] [Google Scholar]
  • 88.Liou JH, Liu YM, Chen CH.. Management of diabetes mellitus with glucagonlike peptide-1 agonist liraglutide in renal transplant recipients: a retrospective study. Transplant Proc 2018;50:2502–5. 10.1016/j.transproceed.2018.03.087 [DOI] [PubMed] [Google Scholar]
  • 89.Singh P, Pesavento TE, Washburn Ket al. Largest single-centre experience of dulaglutide for management of diabetes mellitus in solid organ transplant recipients. Diabetes Obes Metab 2019;21:1061–5. 10.1111/dom.13619 [DOI] [PubMed] [Google Scholar]
  • 90.Thangavelu T, Lyden E, Shivaswamy V.. A retrospective study of glucagon-like peptide 1 receptor agonists for the management of diabetes after transplantation. Diabetes Ther 2020;11:987–94. 10.1007/s13300-020-00786-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Singh P, Taufeeq M, Pesavento TEet al. Comparison of the glucagon-like-peptide-1 receptor agonists dulaglutide and liraglutide for the management of diabetes in solid organ transplant: a retrospective study. Diabetes Obes Metab 2020;22:879–84. 10.1111/dom.13964 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Vigara LA, Villanego F, Orellana Cet al. Effectiveness and safety of glucagon-like peptide-1 receptor agonist in a cohort of kidney transplant recipients. Clin Transplant 2022;36:e14633. 10.1111/ctr.14633 [DOI] [PubMed] [Google Scholar]
  • 93.Sweiss H, Hall R, Zeilmann Det al. Single-center evaluation of safety & efficacy of glucagon-like peptide-1 receptor agonists in solid organ transplantation. Prog Transplant 2022;32:357–62. [DOI] [PubMed] [Google Scholar]
  • 94.Sanyal D, Gupta S, Das P.. A retrospective study evaluating efficacy and safety of linagliptin in treatment of NODAT (in renal transplant recipients) in a real world setting. Indian J Endocrinol Metab 2013;17:S203–5. 10.4103/2230-8210.119572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Boerner BP, Miles CD, Shivaswamy V.. Efficacy and safety of sitagliptin for the treatment of new-onset diabetes after renal transplantation. Int J Endocrinol 2014;2014:617638. 10.1155/2014/617638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bae J, Lee MJ, Choe EYet al. Effects of dipeptidyl peptidase-4 inhibitors on hyperglycemia and blood cyclosporine levels in renal transplant patients with diabetes: a pilot study. Endocrinol Metab (Seoul) 2016;31:161–7. 10.3803/EnM.2016.31.1.161 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Guardado-Mendoza R, Cázares-Sánchez D, Evia-Viscarra MLet al. Linagliptin plus insulin for hyperglycemia immediately after renal transplantation: a comparative study. Diabetes Res Clin Pract 2019;156:107864. 10.1016/j.diabres.2019.107864 [DOI] [PubMed] [Google Scholar]
  • 98.Sanyal D, Biswas M, Chaudhari N.. Long-term efficacy and safety of anti-hyperglycaemic agents in new-onset diabetes after transplant: results from outpatient-based 1-year follow-up and a brief review of treatment options. Diabetes Metab Syndr 2021;15:13–9. 10.1016/j.dsx.2020.11.019 [DOI] [PubMed] [Google Scholar]
  • 99.Rajasekeran H, Kim SJ, Cardella CJet al. Use of canagliflozin in kidney transplant recipients for the treatment of type 2 diabetes: a case series. Diabetes Care 2017;40:e75–6. 10.2337/dc17-0237 [DOI] [PubMed] [Google Scholar]
  • 100.Attallah N, Yassine L.. Use of empagliflozin in recipients of kidney transplant: a report of 8 cases. Transplant Proc 2019;51:3275–80. 10.1016/j.transproceed.2019.05.023 [DOI] [PubMed] [Google Scholar]
  • 101.AlKindi F, Al-Omary HL, Hussain Qet al. Outcomes of SGLT2 inhibitors use in diabetic renal transplant patients. Transplant Proc 2020;52:175–8. 10.1016/j.transproceed.2019.11.007 [DOI] [PubMed] [Google Scholar]
  • 102.Song CC, Brown A, Winstead Ret al. Early initiation of sodium-glucose linked transporter inhibitors (SGLT-2i) and associated metabolic and electrolyte outcomes in diabetic kidney transplant recipients. Endocrinol Diabetes Metab 2021;4:e00185. 10.1002/edm2.185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lim JH, Kwon S, Jeon Yet al. The efficacy and safety of SGLT2 inhibitor in diabetic kidney transplant recipients. Transplantation 2022;106:e404–12. 10.1097/TP.0000000000004228 [DOI] [PubMed] [Google Scholar]
  • 104.Lemke A, Brokmeier HM, Leung SBet al. Sodium-glucose cotransporter 2 inhibitors for treatment of diabetes mellitus after kidney transplantation. Clin Transplant 2022;36:e14718. 10.1111/ctr.14718 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

gfad258_Supplemental_Files

Data Availability Statement

No empirical data collected for this manuscript.


Articles from Nephrology Dialysis Transplantation are provided here courtesy of Oxford University Press

RESOURCES