ArticlePDF Available

Increase in tacrolimus trough levels after steroid withdrawal

Authors:

Abstract

Although there are experimental reports of cytochrome P450 3A4 iso-enzyme (CYP3A4) induction by glucocorticoids, there are no clinical reports about an interaction between tacrolimus and steroids. Therefore, tacrolimus trough level and dose were compared after dose-normalization before and after withdrawal of prednisolone. After withdrawal of 5 mg prednisolone, the median tacrolimus dose-normalized level increased by 14% in the retrospective ( n=54), and by 11% in the prospective ( n=8) part of the study. After withdrawal of 10 mg, this increase was 33% ( n=30) and 36% ( n=14), respectively. An additional pharmacokinetic part of the study ( n=8) revealed an 18% increase in AUC ( P=0.05) after withdrawal of 5 mg prednisolone, which is compatible with a reduced metabolism after steroid withdrawal. The significant increase in tacrolimus exposure after steroid withdrawal may on the one hand counteract the reduction in immunosuppression intended by steroid withdrawal, and, on the other hand, may result in an increase of serum creatinine which could be misinterpreted as rejection.
A preview of the PDF is not available
... Studies have demonstrated that steroid reduction/withdrawal typically increases tacrolimus trough levels soon after and may reduce tacrolimus dose requirements. [30][31][32] Herein, the daily dose of tacrolimus was similar between treatment arms during follow-up, suggesting that the initial steroid regimen did not impact long-term tacrolimus dosing requirements. Over the 5-y follow-up period, tacrolimus daily dose decreased from 0.07 mg/kg at 1 y to 0.06 mg/kg at 5 y posttransplantation. ...
Article
Full-text available
Unlabelled: Although prolonged-release tacrolimus (PR-T) is widely approved for posttransplantation immunosuppression in kidney recipients, large-scale studies are required to assess long-term outcomes. We present follow-up data from the Advagraf-based Immunosuppression Regimen Examining New Onset Diabetes Mellitus in Kidney Transplant Recipients (ADVANCE) trial, in which kidney transplant patients (KTPs) received corticosteroid minimization with PR-T. Methods: ADVANCE was a 24-wk, randomized, open-label, phase-4 study. De novo KTPs received PR-T with basiliximab and mycophenolate mofetil and were randomized to receive an intraoperative corticosteroid bolus plus tapered corticosteroids until day 10 (arm 1) or an intraoperative corticosteroid bolus (arm 2). In this 5-y, noninterventional follow-up, patients received maintenance immunosuppression according to standard practice. The primary endpoint was graft survival (Kaplan-Meier). Secondary endpoints included patient survival, biopsy-confirmed acute rejection-free survival, and estimated glomerular filtration rate (4-variable modification of diet in renal disease). Results: Follow-up study included 1125 patients. Overall graft survival at 1 and 5 y posttransplantation was 93.8% and 88.1%, respectively, and was similar between treatment arms. At 1 and 5 y, patient survival was 97.8% and 94.4%, respectively. Five-year graft and patient survival rates in KTPs who remained on PR-T were 91.5% and 98.2%, respectively. Cox proportional hazards analysis demonstrated similar risk of graft loss and death between treatment arms. Five-year biopsy-confirmed acute rejection-free survival was 84.1%. Mean ± standard deviation values of estimated glomerular filtration rate were 52.7 ± 19.5 and 51.1 ± 22.4 mL/min/1.73 m2 at 1 and 5 y, respectively. Fifty adverse drug reactions were recorded, probably tacrolimus-related in 12 patients (1.5%). Conclusions: Graft survival and patient survival (overall and for KTPs who remained on PR-T) were numerically high and similar between treatment arms at 5 y posttransplantation.
... 18 In contrast, another study reported a dose-dependent increase (11%-36%) in tacrolimus trough levels after withdrawal of prednisone (5 or 10 mg daily for 3 months) in 84 renal allograft recipients. 19 ...
Article
Full-text available
Global use of dexamethasone in COVID-19 patients has revealed a poor understanding of the drug–drug interaction (DDI) potential of dexamethasone, particularly with antiretroviral agents (ARVs). Dexamethasone is both a substrate and a dose-dependent inducer of cytochrome P450 3A4 (CYP3A4). As many ARVs are substrates and/or inhibitors or inducers of CYP3A4, there is concern about DDIs with dexamethasone either as a perpetrator or a victim. Assessment of DDIs that involve dexamethasone is complex as dexamethasone is used at a range of daily doses (generally 0.5 up to 40 mg) and a treatment course can be short, long, or intermittent. Moreover, DDIs with dexamethasone have been evaluated only for a limited number of drugs. Here, we summarize the available in vitro and in vivo data on the interaction potential of dexamethasone and provide recommendations for the management of DDIs with ARVs, considering various dexamethasone dosages and treatment durations.
... Pharmacokinetic interaction between corticosteroids and tacrolimus has been reported in renal transplant recipients, and induction of CYP3A enzymes and the efflux transporter P-glycoprotein was assumed to be the most likely mechanism of the interaction 63 . As a consequence of corticosteroid tapering and withdrawal, a significant increase in dose-corrected tacrolimus exposure has been demonstrated in CYP3A5 non-expresser kidney transplant patients [64][65][66] . However, no or negligible elevation in tacrolimus concentration (C 0 /D) was observed in CYP3A5*1 carriers that is consistent with the findings of the present study with heart transplant patients. ...
Article
Full-text available
High inter-individual variability in tacrolimus clearance is attributed to genetic polymorphisms of CYP3A enzymes. However, due to CYP3A phenoconversion induced by non-genetic factors, continuous changes in tacrolimus-metabolizing capacity entail frequent dose-refinement for optimal immunosuppression. In heart transplant recipients, the contribution of patients’ CYP3A-status ( CYP3A5 genotype and CYP3A4 expression) to tacrolimus blood concentration and dose-requirement was evaluated in the early and late post-operative period. In low CYP3A4 expressers carrying CYP3A5*3/*3 , the dose-corrected tacrolimus level was significantly higher than in normal CYP3A4 expressers or in those with CYP3A5*1 . Modification of the initial tacrolimus dose was required for all patients: dose reduction by 20% for low CYP3A4 expressers, a 40% increase for normal expressers and a 2.4-fold increase for CYP3A5*1 carriers. The perioperative high-dose corticosteroid therapy was assumed to ameliorate the low initial tacrolimus-metabolizing capacity during the first month. The fluctuation of CYP3A4 expression and tacrolimus blood concentration (C 0 /D) was found to be associated with tapering and cessation of corticosteroid in CYP3A5 non-expressers, but not in those carrying CYP3A5*1 . Although monitoring of tacrolimus blood concentration cannot be omitted, assaying recipients’ CYP3A-status can guide optimization of the initial tacrolimus dose, and can facilitate personalized tacrolimus therapy during steroid withdrawal in the late post-operative period.
... Relatedly, corticosteroids induce CYP3A4 and P-gp pathways that may potentially influence tacrolimus metabolism, yet the data are conflicting [35]. Increased tacrolimus levels upon the de-escalation of the dose or withdrawal of steroids have been reported [36,37]. In addition, by influencing the conversion of uridine diphosphoglucuronosyltransferase to the glucuronide metabolite of mycophenolicacid (MPA), tacrolimus may affect mycophenolicacid (MPA) levels [38]. ...
Article
Full-text available
Tacrolimus is a first-line calcineurin inhibitor (CNI) and an integral part of the immunosuppressive strategy in solid organ transplantation. Being a dose-critical drug, tacrolimus has a narrow therapeutic index that necessitates periodic monitoring to maintain the drug’s efficacy and reduce the consequences of overexposure. Tacrolimus is characterized by substantial intra- and inter-individual pharmacokinetic variability. At steady state, the tacrolimus blood concentration to daily dose ratio (C/D ratio) has been described as a surrogate for the estimation of the individual metabolism rate, where a low C/D ratio reflects a higher rate of metabolism. Fast tacrolimus metabolism (low C/D ratio) is associated with the risk of poor outcomes after transplantation, including reduced allograft function and survival, higher allograft rejection, CNI nephrotoxicity, a faster decline in kidney function, reduced death-censored graft survival (DCGS), post-transplant lymphoproliferative disorders, dyslipidemia, hypertension, and cardiovascular events. In this article, we discuss the potential role of the C/D ratio in a noninvasive monitoring strategy for identifying patients at risk for potential adverse events post-transplant.
Article
Full-text available
Background: Tacrolimus shows high variability in inter- and intra-individual pharmacokinetics (PK); therefore, it is important to develop an appropriate model for accurate therapeutic drug monitoring (TDM) procedures. This study aimed to develop a pharmacokinetic model for tacrolimus that can be used for TDM procedures in Korean adult transplant recipients by integrating published models with acquired real-world TDM data and evaluating clinically meaningful covariates. Methods: Clinical data of 1,829 trough blood samples from 269 subjects were merged with simulated datasets from published models and analyzed using a nonlinear mixed-effect model. The stochastic simulation and estimation (SSE) method was used to obtain the final parameter estimates. Results: The final estimated values for apparent clearance, the volume of distribution, and absorption rate were 21.2 L/h, 510 L, and 3.1/h, respectively. The number of post-operative days, age, body weight, and type of transplant organs were the major clinical factors affecting tacrolimus PK. Conclusion: A tacrolimus PK model that can incorporate published PK models and newly collected data from the Korean population was developed using the SSE method. Despite the limitations in model development owing to the nature of TDM data, the SSE method was useful in retrieving complete information from the TDM data by integrating published PK models while maintaining the variability of the model.
Article
Full-text available
Atherosclerosis is a major global health concern. The central modifiable risk factors and causative agents of the disease are high total and low-density lipoprotein (LDL) cholesterol. To reduce morbidity and mortality, a thorough understanding of the factors that influence an individual’s cholesterol status during the decades when the arteria-narrowing arteriosclerotic plaques are forming is critical. Several drugs are known to increase cholesterol levels; however, the mechanisms are poorly understood. Activation of pregnane X receptor (PXR), the major regulator of drug metabolism and molecular mediator of clinically significant drug–drug interactions, has been shown to induce hypercholesterolemia. As a major sensor of the chemical environment, PXR may in part mediate hypercholesterolemic effects of drug treatment. This review compiles the current knowledge of PXR in cholesterol homeostasis and discusses the role of PXR in drug-induced hypercholesterolemia.
Article
Background: Tacrolimus may be administered during hospitalization as an IV formulation or oral suspension. However, literature suggesting appropriate ratios for conversion from these formulations to capsules is limited. Objective: To evaluate conversion ratios after a switch in formulation of tacrolimus for solid-organ transplant recipients. Methods: This single-centre observational longitudinal study involved hospitalized patients who underwent a switch in formulation of tacrolimus according to 1 of 3 possible scenarios: IV to oral suspension, IV to capsule, or oral suspension to capsule. Data were collected from the earliest accessible electronic file (January 2009) to January 1, 2019. Conversion ratios were calculated for each of the 3 groups using data for blood concentrations and doses before and after the switch. The calculated ratios were then compared with recommended conversion ratios: 1:5 (i.e., 1 mg of IV tacrolimus is converted to 5 mg of oral tacrolimus, expressed as "5") for either of the switches involving an IV formulation and 1:1 (i.e., same amount, expressed as "1") for the switch from oral formulation to capsules. Results: For the group who underwent switching from the IV formulation to oral suspension, the mean calculated conversion ratio was 3.04, which was significantly different from the recommended ratio of 5. For the group who underwent switching from the IV formulation to capsules, the calculated conversion ratio was 5.18, which was not significantly different from the recommended ratio of 5. For the group who underwent switching from oral suspension to capsules, the calculated conversion ratio was 1.17, which was not significantly different from the recommended ratio of 1. Conclusion: In this small retrospective study of tacrolimus therapy, the calculated conversion ratio was significantly different from the recommended ratio for patients who were switched from IV administration to oral suspension, but not for those switched from IV administration or oral suspension to capsules. Therapeutic drug monitoring therefore appears indispensable, regardless of conversion ratios.
Article
Objective To evaluate the literature on a potential dexamethasone–direct oral anticoagulant (DOAC) drug interaction and provide management considerations with COVID hypercoagulability. Data Sources A search of EMBASE, PubMed, and Google Scholar (January 1990 to May 2021), limited to the English language, using applicable search terms resulted in 137 articles, with 21 relevant articles included. Regulatory agency and clinical guidance documents were also reviewed. Study Selection and Data Extraction Included articles describe in vitro or in vivo animal or human data for dexamethasone induction of cytochrome P450 (CYP) 3A4 or P-glycoprotein (P-gp). Data Synthesis Dexamethasone has the potential to interact with the DOACs via CYP3A4 and/or P-gp induction. Only apixaban and rivaroxaban have CYP3A4 metabolism. Dexamethasone can increase CYP3A4 activity by up to 70% and reduce the area under the concentration-time curve (AUC) of CYP3A4 substrates by >40%, which is consistent with criteria for a weak CYP inducer. In rodents, dexamethasone P-gp induction is associated with AUC reductions of 20% to 50%. Human data are lacking. Relevance to Patient Care and Clinical Practice Severe COVID-19 infection is associated with hypercoagulability. Although heparins are the preferred anticoagulants for hospitalized COVID-19 patients, DOACs are being utilized. Dexamethasone is recommended for hospitalized COVID-19 patients requiring supplemental oxygen. The concurrent use of dexamethasone and apixaban or rivaroxaban in such patients carries the potential for reduced anticoagulant effect during a state of heightened thrombotic risk. Conclusions Concurrent use of dexamethasone and apixaban or rivaroxaban in hospitalized COVID-19 patients with laboratory evidence of COVID coagulopathy should be avoided until higher-quality data are available.
Article
Full-text available
The cytochrome P450 (CYP) enzyme family is the most important enzyme system catalyzing the phase 1 metabolism of pharmaceuticals and other xenobiotics such as herbal remedies and toxic compounds in the environment. The inhibition and induction of CYPs are major mechanisms causing pharmacokinetic drug–drug interactions. This review presents a comprehensive update on the inhibitors and inducers of the specific CYP enzymes in humans. The focus is on the more recent human in vitro and in vivo findings since the publication of our previous review on this topic in 2008. In addition to the general presentation of inhibitory drugs and inducers of human CYP enzymes by drugs, herbal remedies, and toxic compounds, an in-depth view on tyrosine-kinase inhibitors and antiretroviral HIV medications as victims and perpetrators of drug–drug interactions is provided as examples of the current trends in the field. Also, a concise overview of the mechanisms of CYP induction is presented to aid the understanding of the induction phenomena.
Article
Full-text available
Tacrolimus is metabolized by CYP3A4 and CYP3A5 enzymes. Patients expressing CYP3A5 (in Caucasian patients about 15% of the population but more frequent in African Americans and Asians) have a dose requirement that is around 50% higher than non-expressers to reach the target concentration. CYP3A5 expressers can be considered fast metabolizers. The trough concentration/dose (C0/D) ratio of tacrolimus has recently been proposed as a prognostic marker for poor outcome after kidney transplantation. Patients with a low C0/D ratio (also referred to as fast metabolizers) seem to have more tacrolimus-related nephrotoxicity, more BK-viremia, and a lower graft survival. At first sight, the expression of CYP3A5 and a low C0/D ratio seem to be overlapping factors, both pointing towards patients in whom a higher tacrolimus dose is needed to reach the tacrolimus target concentration. However, there are important differences, and these differences may explain why the impact of the C0/D ratio on long term outcome is stronger than for CYP3A5 genotype status. Patients with a low C0/D ratio require a high tacrolimus dose and are exposed to high tacrolimus peak concentrations. The higher peak exposure to tacrolimus (and/or its metabolites) may explain the higher incidence of nephrotoxicity, BK-viremia and graft loss. A potential confounder is the concurrent maintenance treatment of corticosteroids, as steroids are sometimes continued in patients at high immunological risk. Steroids induce the metabolism of tacrolimus via pregnane X receptor mediated increased CYP3A4 expression, resulting in lower tacrolimus C0/D ratio in high risk patients. Also non-adherence may result in lower C0/D ratio which is also associated with poor outcome. The C0/D ratio of tacrolimus does seem to identify a group of patients with increased risk of poor outcome after kidney transplantation. Our recommendation is to monitor tacrolimus peak concentrations in these patients, and if these are high then target slightly lower pre-dose concentrations. Another possibility would be to switch to a prolonged release formulation or to dose the drug more frequently, in smaller doses, to avoid high peak concentrations.
Article
Tacrolimus (FK-506) is an immunosuppressant agent that acts by a variety of different mechanisms which include inhibition of calcineurin. It is used as a therapeutic alternative to cyclosporin, and therefore represents a cornerstone of immunosuppressive therapy in organ transplant recipients. Tacrolimus is now well established for primary immunosuppression in liver and kidney transplantation, and experience with its use in other types of solid organ transplantation, including heart, lung, pancreas and intestinal, as well as its use for the prevention of graft-versus-host disease in allogeneic bone marrow transplantation (BMT), is rapidly accumulating. Large randomised nonblind multicentre studies conducted in the US and Europe in both liver and kidney transplantation showed similar patient and graft survival rates between treatment groups (although rates were numerically higher with tacrolimus-versus cyclosporin-based immunosuppression in adults with liver transplants), and a consistent statistically significant advantage for tacrolimus with respect to acute rejection rate. Chronic rejection rates were also significantly lower with tacrolimus in a large randomised liver transplantation trial, and a trend towards a lower rate of chronic rejection was noted with tacrolimus in a large multicentre renal transplantation study. In general, a similar trend in overall efficacy has been demonstrated in a number of additional clinical trials comparing tacrolimus-with cyclosporin-based immunosuppression in various types of transplantation. One notable exception is in BMT, where a large randomised trial showed significantly better 2-year patient survival with cyclosporin over tacrolimus, which was primarily attributed to patients with advanced haematological malignancies at the time of (matched sibling donor) BMT. These survival results in BMT require further elucidation. Tacrolimus has also demonstrated efficacy in various types of transplantation as rescue therapy in patients who experience persistent acute rejection (or significant adverse effects) with cyclosporin-based therapy, whereas cyclosporin has not demonstrated a similar capacity to reverse refractory acute rejection. A corticosteroid-sparing effect has been demonstrated in several studies with tacrolimus, which may be a particularly useful consideration in children receiving transplants. The differences in the tolerability profiles of tacrolimus and cyclosporin may well be an influential factor in selecting the optimal treatment for patients undergoing organ transplantation. Although both drugs have a similar degree of nephrotoxicity, cyclosporin has a higher incidence of significant hypertension, hypercholesterolaemia, hirsutism and gingival hyperplasia, while tacrolimus has a higher incidence of diabetes mellitus, some types of neurotoxicity (e.g. tremor, paraesthesia), diarrhoea and alopecia. Conclusion: Tacrolimus is an important therapeutic option for the optimal individualisation of immunosuppressive therapy in transplant recipients. Pharmacodynamic Properties Tacrolimus (FK-506) is a macrolide immunosuppressant that acts by a variety of different mechanisms which include inhibition of calcineurin. The drug inhibits T lymphocyte activation and transcription of cytokine genes including that for interleukin-2. Tacrolimus inhibits cell-mediated and, to a lesser extent, humoral immune responses. Cytokines produced by T helper (Th)1 cells are preferentially suppressed over those produced by Th2 cells. The mechanism of action of tacrolimus is largely similar to that of cyclosporin, but tacrolimus is 10 to 100 times more potent. The drugs both inhibit calcineurin but do so via formation of complexes with different immunophilins: tacrolimus binds to FK-506 binding protein 12, whereas cyclosporin binds to cyclophilin A. The drugs appear to differ in their effects on patterns of Th2 cell cytokine expression and possibly some aspects of humoral immunity. Furthermore, lymphocyte sensitivity to the drugs may differ between patients. In animal models, tacrolimus had an organ-specific effect in stimulating hepatic regeneration after partial hepatectomy, and attenuated hepatic ischaemic or reperfusion injury. Tacrolimus does not appear to cause postoperative cholestasis in liver transplant recipients, and postoperative disturbances in biliary secretion and flow rates may recover more rapidly with tacrolimus than cyclosporin. Like cyclosporin, tacrolimus has nephrotoxic effects that appear to be mechanistically related to its immunosuppressive activity, possibly involving inhibition of calcineurin. Tacrolimus suppresses insulin production at the trans-criptional level and appears to be more diabetogenic than cyclosporin in some patients. In patients with liver transplants, tacrolimus reduced β cell secretory reserve, and was associated with significant insulin resistance and impaired β cell-α cell axis. Further clarification is required of the comparative effects of tacrolimus and cyclosporin on factors involved in cardiac transplant-associated coronary artery disease. Although tacrolimus has been associated with a lower incidence of positivity for anti-endothelial cell antibodies than cyclosporin, a higher incidence of pathological microvascular endothelial dysfunction has also been reported. Tacrolimus may have an in vitro antithrombotic effect. Findings have been conflicting regarding the comparative effect of tacrolimus and cyclosporin on endothelium-independent microcirculatory responses 1 year after cardiac transplantation. Tacrolimus and cyclosporin appear to have similar effects on most aspects of cardiac function in renal or liver transplant recipients. Pharmacokinetic Propertie Like cyclosporin, the pharmacokinetic properties of tacrolimus can vary widely between individuals and dosage regimens are titrated according to whole-blood trough drug concentrations. Oral bioavailability of tacrolimus is about 20 to 25%, and food appears to have a significant effect in reducing the rate and extent of absorption. The drug binds extensively to erythrocytes, and whole-blood concentrations of tacrolimus are approximately 15 to 35 times those measured in plasma. Tacrolimus is almost completely metabolised prior to elimination. Metabolism is via 3A4 isoenzymes of the cytochrome P450 (CYP) system, primarily in the liver but also in the intestinal mucosa, and a number of metabolites are formed. At least 1 metabolite appears to be active, although the immunosuppressive activity of tacrolimus is primarily due to the parent drug. Elimination half-life of tacrolimus has been reported to be approximately 12 hours in liver transplant recipients and 19 hours in renal transplant recipients. Less than 1% of an intravenous dose of tacrolimus is eliminated unchanged in the urine. The main route of elimination for tacrolimus and its metabolites is via the biliary tract. Like cyclosporin, tacrolimus is subject to a number of pharmacokinetic (and pharmacodynamic) drug interactions of potential clinical significance, including those involving other drugs metabolised by the CYP enzyme system. Therapeutic Efficacy In most clinical trials, tacrolimus-based primary immunosuppression initially included concomitant administration of corticosteroids, typically with azathioprine or mycophenolate mofetil, and sometimes with adjunctive antilymphocyte antibody induction therapy. In general, rescue therapy with tacrolimus usually involved simple conversion from cyclosporin to tacrolimus without modification of concomitant drug therapy. Clinical trials comparing tacrolimus-with cyclo-sporin-based immunosuppression were conducted in a nonblind fashion, presumably because of the need to monitor whole-blood trough drug concentrations to optimise the clinical management of patients. In the following sections, cyclosporin refers to the standard formulation of the drug (the microemulsion formulation is specified when applicable). Hepatic Transplantation As primary immunosuppression in adults with hepatic transplantation, tacrolimus-based regimens achieved similar patient and graft survival rates to cyclosporin-based regimens, with a trend towards higher rates with tacrolimus, and significantly lower rates of acute rejection. This was demonstrated in 2 large multicentre randomised trials, one of which also showed significantly lower rates of chronic rejection at 1 and 3 years post-transplantation among patients receiving tacrolimus-based immunosuppression. In general, similar overall efficacy trends were noted in more recent smaller randomised studies comparing tacrolimus with the cyclosporin microemulsion formulation. Results of these studies showed patient and graft survival rates of approximately 75 to 100% and 70 to 95%, respectively, after 6 to 30 months of tacrolimus-based therapy; acute rejection rates varied widely between studies. Long term survival data from a large cohort of 1000 patients treated with tacrolimus-based immunosuppression after liver transplantation indicate 6-year patient and graft survival rates of 68 and 63%, respectively. Several studies demonstrated that corticosteroid therapy could be successfully withdrawn in approximately 70 to 90% of liver transplant recipients treated with tacrolimus-based immunosuppressive therapy. Tacrolimus is also effective as rescue therapy in adult patients with persistent acute or chronic rejection or drug-related toxicity with cyclosporin-based primary immunosuppression after hepatic transplantation. This has been demonstrated in a number of noncomparative studies, the largest involving a group of 475 patients with 2-year patient and graft survival rates of 80 and 73%, respectively, after conversion from cyclosporin to tacrolimus because of acute or chronic rejection. In general, results of studies with tacrolimus-based primary immunosuppressive and rescue therapy for paediatric liver transplantation have been very similar to those of studies in adult patients. Results of the only prospective randomised comparison between tacrolimus (n = 30; mean age 3.5 years) and cyclosporin (n = 21; mean age 3.2 years) as primary immunosuppression showed similar 1-year patient (80 vs 81%) and graft (70 vs 71%) survival rates, and a trend favouring tacrolimus for acute rejection rate (52 vs 79%). Importantly, some studies demonstrated that corticosteroid therapy can be successfully discontinued in approximately 70 to 85% of children receiving tacrolimus as primary immunosuppressive or rescue therapy. Renal Transplantation Tacrolimus-based regimens achieved similar patient and graft survival rates and lower rates of acute rejection compared with cyclosporin-based regimens when used as primary immunosuppression in adults with renal transplantation. This was demonstrated in 2 large multicentre randomised trials; 1-year patient survival rates were ≈95% and corresponding graft survival rates were ≈85 to 90% for both treatment groups. A statistically significant advantage favouring tacrolimus for acute rejections rates was noted in both studies (26 vs 46% and 31 vs 46%; both p < 0.001). One of the multicentre studies (European data) showed a nonsignificant trend towards a lower rate of chronic rejection among tacrolimus recipients than cyclosporin recipients at 4 years post-transplantation (5.5 vs 11.3%). Long term (3-year) survival data from one of the trials (US data) showed similar rates of patient (≈90%) and graft (≈80%) survival for both treatment groups. In this study, approximately 25% of patients were African-American, and results in this high-risk subgroup mirrored those for all patients in each treatment group. Numerous noncomparative, retrospective or meta-analytical trials, as well as a few small-to moderate-sized randomised comparisons with cyclosporin microemulsion, have also been conducted with tacrolimus-based regimens for primary immunosuppression in adult renal transplant recipients, and results generally support those of the large multicentre studies. Rescue therapy with tacrolimus, primarily in patients who developed acute rejection while receiving cyclosporin-based primary immunosuppression (n = 40 to 169), was associated with patient survival rates >90% and corresponding graft survival rates >70% after 1 to 3 years of follow-up after conversion. In general, results of studies with tacrolimus-based primary immunosuppressive and rescue therapy for paediatric renal transplantation have been very similar to those of studies in adult patients. In the largest study, 81 children (82 transplants) received tacrolimus-based therapy as primary immunosuppression. One-year patient and graft survival rates approached 100%, and 4-year rates were 94 and 84%, respectively. About two-thirds of patients were successfully withdrawn from corticosteroid therapy. Heart Transplantation In prospective studies comparing tacrolimus-with cyclosporin-based primary immunosuppressive regimens in heart transplant recipients, patient survival rates were similar between treatment groups and there was a consistent trend towards more favourable acute rejection rates with tacrolimus. One-and ≈2-year patient survival rates were ≈80 to 90% for both treatment groups, and a large non-randomised comparison also showed similar 5-year patient survival rates between tacrolimus and cyclosporin treatment groups (76 vs 71%). In a moderate-sized randomised study of 73 patients, the mean number of acute rejection episodes per patient was significantly lower among tacrolimus than cyclosporin recipients (1.33 vs 1.87; p < 0.01). A number of small studies (n < 20) of tacrolimus as rescue therapy in adults showed that at least 70% of patients had either no rejection episodes or only mild rejection after conversion from cyclosporin to tacrolimus (follow-up periods were usually at least 6 months). Tacrolimus has had limited use in paediatric heart transplantation. Tacrolimus-based primary immunosuppression was associated with good patient survival rates in a group of 26 children (≈80% at 1 and 3 years post-transplantation). In addition, rates of moderate to severe acute rejection were lower and corticosteroid withdrawal rates were much higher than those in historical controls treated with cyclosporin-based therapy. The results of a nonrandomised study of 40 paediatric heart transplant recipients showed that the presence of 2 HLA-DR loci donor/ recipient mismatches increased the risk of high-grade rejection in children receiving cyclosporin-based therapy, whereas the risk of rejection was not increased in those receiving tacrolimus-based therapy. Tacrolimus-treated children with 2 HLA-DR mismatches had a significantly lower risk of severe rejection than cyclosporin-treated children with only 1 HLA-DR mismatch. In small studies (n < 25) of tacrolimus as rescue therapy, graft loss was not reported (follow-up periods up to 40 months) and corticosteroid dosages were reduced or discontinued in most children. Lung Transplantation Tacrolimus-and cyclosporin-based primary immunosuppressive regimens were associated with similar 1-(83 vs 71%) and 2-year (76 vs 66%) patient survival rates, as well as similar proportions of patients free from acute rejection (14 vs 11.5%), in a prospective randomised study of 133 lung transplant recipients. Results for all of these end-points tended to favour tacrolimus, and the trial showed a significantly lower incidence of obliterative bronchiolitis among tacrolimus than cyclosporin recipients (21.7 vs 38%; p < 0.05). In general, the use of tacrolimus as rescue therapy in small numbers of patients with lung transplantation (n ≤15) was associated with a reduced incidence of acute rejection after conversion from cyclosporin, and at least two-thirds of patients remained alive during mean follow-up periods of approximately 6 to 18 months. Pancreas or Kidney and Pancreas Transplantation Numerous studies have been conducted demonstrating the efficacy of tacrolimus as primary immunosuppression after solitary pancreas transplantation or simultaneous pancreas and kidney transplantation (SPK); however, no large randomised trial has prospectively compared tacrolimus-with cyclosporin-based regimens in this clinical setting. Nevertheless, data from 2 large (n > 200) retrospective analyses indicate significantly better pancreas graft survival with tacrolimus-based therapy in patients with solitary pancreas or SPK transplantation, and patient and renal graft survival was better with tacrolimus than cyclosporin in SPK recipients. For example, a multicentre matched-pair analysis comparing tacrolimus-with cyclosporin-based therapy in SPK recipients at 18 months post-transplant showed pancreas graft survival rates of 88 versus 71%, renal graft survival rates of 94 versus 77% and patient survival rates of 97 versus 83% (p ≤0.002 for all comparisons). Results of 2 moderate-sized studies of tacrolimus as rescue therapy in SPK recipients showed patient survival rates approaching 100% and pancreas and renal graft survival rates of about 90% (follow-up period ≤1 year after conversion from cyclosporin to tacrolimus). Intestinal Transplantation Several reports involving small numbers of patients indicate that tacrolimus is effective in this clinical setting. Data from the International Transplant Registry (n = 170) indicate that, depending on the subgroup of intestinal transplant recipients, tacrolimus-based primary immunosuppression is associated with 1-and 3-year patient survival rates of 59 to 83% and 40 to 47%, respectively, and 1-and 3-year graft survival rates of 51 to 65% and 29 to 38%, respectively. In general, patient and graft survival rates were as good as or better than those achieved with cyclosporin-based regimens. Among cyclosporin recipients, 1-and 3-year patient survival rates were 41 to 57% and 28 to 50%, respectively, and 1-and 3-year graft survival rates were 17 to 44% and 11 to 41%, respectively. Bone Marrow Transplantation Three randomised comparative trials have consistently demonstrated a lower incidence of grade II to IV acute graft-versus-host disease (GVHD) with tacrolimus-than cyclosporin-based therapy after allogeneic bone marrow transplantation (BMT). However, the largest of the trials (n = 329) also showed that 2-year overall survival (57 vs 47%; p < 0.05) and disease-free survival (50 vs 41%; p = 0.01) were significantly better among cyclosporin-than tacrolimus-treated patients with haematological malignancy who received BMT from matched sibling donors. These differences were attributed primarily to patients with advanced haematological malignancy at the time of BMT. These survival data in BMT require confirmation. Tacrolimus has also been used with some success in the treatment of patients who developed acute or chronic GVHD or significant toxicity while receiving cyclosporin-based immunosuppressive therapy after BMT, but data are preliminary. Tolerability The principal adverse effects associated with tacrolimus treatment include nephrotoxicity, neurotoxicity, disturbances in glucose metabolism, gastrointestinal (GI) disturbance and hypertension. Susceptibility to infection and malignancy is also increased. All of these adverse effects also occur with cyclosporin, although the incidence of some adverse effects differs between the drugs (see later in this section). Tacrolimus is rarely associated with the cyclosporin-specific adverse effects hirsutism, gingivitis and gum hyperplasia, but it may cause alopecia and pruritus in some patients. Many of the adverse effects of tacrolimus are dose-related; nephrotoxicity, neurotoxicity, glucose metabolism disturbances, GI disturbances and infections may occur more frequently or be more severe at higher whole-blood tacrolimus concentrations. Importantly, these adverse events can often be managed by dosage reductions. Concomitant drugs such as corticosteroids may also contribute to some adverse effects. In the major trials in patients undergoing liver or kidney transplants, withdrawal rates because of adverse events tended to be higher with tacrolimus than cyclosporin. Nephrotoxicity occurred in as many as half of patients treated with either tacrolimus or cyclosporin. Neurotoxicity associated with tacrolimus most frequently manifests as tremor, headache, insomnia and paraesthesia, and some neurological effects (including tremor and paraesthesia) may be more problematic with tacrolimus than with cyclosporin. Diabetes mellitus and/or hyperglycaemia also tended to occur more frequently with tacrolimus than with cyclosporin in the major trials in kidney or liver transplant recipients. In 2 large multicentre randomised kidney transplantation trials, the incidence of new-onset type 1 diabetes mellitus was 20 vs 4% in the US trial and 8 vs 2% in the European study. However, about one-quarter to one-third of affected tacrolimus recipients were able to discontinue insulin therapy within 1 year. Furthermore, tacrolimus has generally not been more diabetogenic than cyclosporin in cardiac transplant trials. Also, at least 1 recent study in renal transplant recipients showed a lower incidence of post-transplantation diabetes mellitus with tacrolimus than in previous reports, suggesting that, with more experience, it may be possible to reduce the risk of developing this complication. Other metabolic disturbances that can occur with tacrolimus include hyper-kalaemia and hypomagnesaemia. A number of studies have shown that tacrolimus has less adverse effect than cyclosporin on lipid profiles and/or the general cardiovascular risk profile. In particular, significantly lower serum levels of total cholesterol, triglycerides and/or low density lipoprotein-cholesterol have been reported with tacrolimus. Hypertension occurred in up to half of patients treated with tacrolimus in major trials, but it was normally mild to moderate in severity, whereas hypertension can be more severe with cyclosporin. In cardiac transplant recipients, hypertension requiring treatment occurred more frequently with cyclosporin-than tacrolimus-based regimens. GI disturbance, including diarrhoea, nausea and constipation, occurs commonly in patients treated with tacrolimus; diarrhoea is more frequent with tacrolimus than with cyclosporin. Infection rates were similar in tacrolimus-and cyclosporin-treated groups in the major clinical trials in kidney or liver transplant recipients. The tolerability profile of tacrolimus in children is generally similar to that in adults. However, children are at increased risk of potentially fatal Epstein-Barr virus-related post-transplant lymphoproliferative disorders (PTLD). The inci-dence of PTLD in paediatric liver transplant recipients may be higher with tacrolimus-than cyclosporin-based immunosuppression. From the reported ex-periences (in >10 patients) of using tacrolimus in primary liver transplantation in children, the incidence of PTLD usually ranged from 3 to 11%, although higher values have been reported. The incidence of PTLD in paediatric patients converted to tacrolimus therapy appears to be higher than that in primary therapy, but this may be associated with high cumulative dosages of immunosuppressive agents required to treat intractable rejection. The risks of tacrolimus treatment during pregnancy appear to be no greater than those with cyclosporin, and it has been suggested that tacrolimus may be associated with a lower incidence of maternal hypertension or pre-eclampsia. Dosage and Administration The dosage recommendations outlined in this section focus on the use of tacrolimus in the US and UK in patients who have undergone liver or kidney transplantation. Although treatment regimens can vary between countries and individual transplantation centres, it is likely that tacrolimus is used in a similar manner for immunosuppression following other types of transplantation. Whenever possible, tacrolimus should be initiated using the oral route of administration. For patients unable to take tacrolimus orally, therapy may be initiated by continuous intravenous infusion. In the US, the recommended intravenous starting dose is 0.03 to 0.05 mg/kg/day for adults receiving liver or kidney transplantation and for children receiving liver transplantation; no specific recommendation for paediatric kidney transplantation is provided in US prescribing information. In the UK, initial intravenous dose recommendations for adults are 0.01 to 0.05 mg/kg/day for liver and 0.05 to 0.10 mg/kg/day for kidney transplantation; corresponding recommendations for children are 0.05 mg/kg/day for liver and 0.1 mg/kg/day for kidney transplantation. Conversion from intravenous to oral therapy should occur as soon as is clinically feasible, usually within 2 to 3 days. Whether administered by the oral or intravenous route, the initial dose of tacrolimus should begin approximately 6 hours after the completion of liver transplant surgery and within 24 hours of kidney transplantation surgery. Oral tacrolimus is administered in 2 divided daily doses at 12-hour intervals. In adults, the recommended starting oral dosage of tacrolimus as primary immunosuppression is 0.10 to 0.15 mg/kg/day (US) or 0.10 to 0.20 mg/kg/day (UK) for liver transplantation and 0.2 mg/kg/day (US) or 0.15 to 0.30 mg/kg/day (UK) for kidney transplantation. Initial recommended dosage in children receiving liver transplantation is 0.15 to 0.20 mg/kg/day (US) or 0.3 mg/kg/day (UK). In the UK, 0.3 mg/kg/day is the recommended initial dose of tacrolimus in paediatric renal transplant recipients; US prescribing information does not provide a corresponding recommendation for this patient population. During maintenance therapy the dose of tacrolimus can often be reduced. In general, children require higher doses than adults to achieve similar blood concentrations of tacrolimus. Likewise, African-American patients typically require higher tacrolimus doses than Caucasian patients (at least in kidney transplantation) to achieve similar blood concentrations of the drug. Patients with hepatic or renal dysfunction should receive doses at the lowest value of the recommended intravenous and oral dosage ranges (and further dosage reductions may be required). When tacrolimus is used as rescue therapy in patients not responding to (or not tolerating) cyclosporin-based therapy, treatment should begin with the same initial dosage as for primary therapy in that particular allograft (UK recommendation). Tacrolimus should not be started until approximately 24 hours after discontinuation of cyclosporin therapy.
Article
In the Leiden cohort of cadaveric renal transplants, the authors compared the risk factors of a low intercept, defined as a creatinine clearance lower than 50 ml/min at 6 months, and of a negative slope of the reciprocal creatinine concentrations after 6 months. Two hundred of 654 grafts (31%) failed to reach optimal function because of old donor age, female gender of the donor, histoincompatibility, delayed graft function, or acute rejection episodes in the first 6 months. Forty-four percent of all grafts displayed progressive deterioration of function over time. The association with younger recipient age, sensitization, class 1 histoincompatibility, baseline immunosuppression, and late acute rejection episodes suggests an underlying immunological process, very likely activated through the indirect antigen-presenting route. The negative impact of proteinuria and diastolic hypertension at 6 months on the slope is compatible with their role as progression factors. Although associations are not necessarily causally related and interventions do not necessarily result in an improvement in outcome, it is conceivable that better matching, optimal immunosuppression, and a more aggressive anti-hypertensive and antiproteinuric treatment result in improvement of long-term graft survival.
Article
Diabetogenic effects have been ascribed to several drugs currently used for immunosuppression following organ transplantations, including corticosteroids, cyclosporin and tacrolimus (FK-506). Azathioprine appears to be devoid of adverse effects on carbohydrate metabolism. The pathogenesis of immunosuppression-associated diabetes mellitus has not been clearly defined, and may be multifactorial in organ transplant recipients. Metabolic similarities between post-transplant diabetes and non-insulin-dependent diabetes mellitus include defective insulin secretion and impaired insulin action in target tissues. The predominant effect of corticosteroids is induction of a state of insulin resistance. Cyclosporin and tacrolimus have been shown to inhibit endogenous insulin secretion and may also have adverse effects on tissue sensitivity to insulin. Postoperative diabetes mellitus developing de novo is a frequent complication of organ transplantation. Treatment with diet, oral antidiabetic agents or insulin may be necessary. Postoperative diabetes may be a transient phenomenon in some patients, whereas others may require long term insulin treatment. Although clinically overt diabetes is readily diagnosed, the prevalence of subclinical degrees of glucose intolerance may be higher than is currently recognised. The long term clinical implications of immunosuppression-associated glucose intolerance and diabetes are uncertain and rely on extrapolations from studies in non-transplant populations. Patients with impaired glucose tolerance may have an increased probability of progression to diabetes mellitus, whereas long term diabetes carries the risk of tissue damage from specific microvascular complications, i.e. diabetic retinopathy, neuropathy and nephropathy. Epidemiological and experimental studies have implicated glucose intolerance and hyperinsulinaemia as risk factors for atherosclerosis. Hypertension and atherogenic plasma lipid profiles are also frequently encountered in transplant recipients treated with cyclosporin, tacrolimus and corticosteroids. Thus, patients treated with these drugs, particularly in combination, may possess a multiplicity of risk factors for macrovascular disease. These factors may be relevant to the development of accelerated atherosclerosis that occurs in renal and cardiac transplant recipients. However, their contribution to post-transplant macrovascular disease is uncertain at present. Carefully designed prospective studies will be necessary to determine the natural history of postoperative diabetes in organ transplant recipients. We recommend that future clinical studies of immunosuppressive agents should avoid arbitrary diagnostic criteria for diabetes and should incorporate rigorous methods for the assessment of glucose tolerance, insulin secretion and insulin action. Modifications of existing immunosuppressive drug regimens may reduce the incidence or severity of postoperative diabetes. Elucidation of the molecular mechanisms responsible for this metabolic complication should provide a more logical basis for prevention and treatment.
Article
Background: Current immunosuppressive protocols fail to prevent chronic rejection often manifested as graft vascular disease (GVD) in solid organ transplant recipients. Several new immunosuppressants including sirolimus, a dual function growth factor antagonist, have been discovered, but studies of drug efficacy have been hampered by the lack of a model of GVD in primates, as a prelude to clinical trials. As described earlier, we have developed a novel non-human primate model of GVD where progression of GVD is quantified by intravascular ultrasound (IVUS). Methods: Twelve cynomolgus monkeys underwent aortic transplantation from blood group compatible but mixed lymphocyte reaction-mismatched donors. To allow the development of GVD in the allograft, no treatment was administered for the first 6 weeks. Six monkeys were treated orally with sirolimus from day 45 after transplantation to day 105. Results: Progression of GVD measured as change in intimal area from day 42 to 105 was halted in sirolimus-treated monkeys compared to untreated monkeys (P<0.001, general linear model). On day 105, the intimal area +/- SEM was 3.7+/-1.0 and 6.4+/-0.5 mm2, respectively (P<0.05, t test). The magnitude of allograft intimal area on day 105 correlated inversely with sirolimus trough levels (R2=0.67, P<0.05). Regression of the intimal area was seen in four of six sirolimus-treated monkeys, which was significantly different from the untreated monkeys (P<0.05). Conclusions: Our results in the first non-human primate model of GVD showed that treatment with sirolimus not only halted the progression of preexisting GVD but also was associated with partial regression. Sirolimus trough blood levels were correlated with efficacy. Therefore, sirolimus has the potential to control clinical chronic allograft rejection.
Article
Background. Corticosteroids have always been an integral part of immunosuppressive regimens in renal transplantation. The primary goal of this analysis was to assess the safety of steroid withdrawal in our pediatric renal transplant recipients receiving tacrolimus-based immunosuppression. Methods. Between December 1989 and December 1996, 82 renal transplantations were performed in pediatric patients receiving tacrolimus-based immunosuppression. Two of these patients lost their grafts within 3 weeks of transplantation (and were still on steroids at the time of graft loss), and were excluded from further analysis. Seventy-four patients (92.5%) were taken off prednisone a median of 5.7 months after transplantation. Of these 74, 56 (70%) remained off prednisone (OFF), and 18 (22.5%) were restarted on prednisone a median of 14.8 months after discontinuing steroids (OFF --> ON). 6(7.5%) were never taken off prednisone (ON). The mean follow-up was 59+/-23 months. Results. The 1-, 3-, and 5-year actuarial patient survival rates in the OFF group were 100%, 98%, and 96%, respectively; in the OFF --> ON group, they were 100%, 100%, and 100%, and in the ON group, they were 100%, 83%, and 83%. The 1-, 3-, and 5- year actuarial graft survival rates in the OFF group were 100%, 95%, and 82%, respectively; in the OFF --> ON group, they were 100%, 89%, and 83%; and in the ON group, they were 100%, 50%, and 33%. Two of the six graft losses in the OFF group, three out of four in the OFF --> ON Group, and two out of five in the ON group, were to chronic rejection. A time-dependent Cox regression analysis showed that the hazard for graft failure for those who came and stayed off prednisone was 0.178 relative to those who were never withdrawn from prednisone (P=0.005). Patients who were 10 years of age or younger were withdrawn from prednisone earlier (median: 5 months) than those older than 10 years (median: 7.5 months, P=0.02). In addition, patients who never had acute rejection were withdrawn from steroids earlier (median: 5 months) than those who had one or more episodes of acute rejection (median: 7.6 months, P=0.001). There was no effect of donor age, race, sex, recipient race, sex, cadaveric versus living donor, 48-hr graft function, panel reactive antibody, and total HLA mismatches or matches on the likelihood of being weaned off steroids. Serum creatinine at most recent follow-up in the OFF group was 1.2+/-0.5 mg/dl; in the OFF a ON group, it was 1.8+/-0.9 mg/dl, and in the ON group it was 2.0 mg/dl (P<0.003). The incidence of rejection in the OFF, OFF --> ON, and ON groups was 39%, 77%, and 100%, respectively (P<0.05). Conclusion. These data suggest that steroid withdrawal in pediatric renal transplant patients receiving tacrolimus-based immunosuppression is associated with reasonable short- and medium-term patient and graft survival, and acceptable renal function. Patients who discontinue and then resume steroids had patient and graft survival rates comparable with those in patients who discontinue and stay off steroids, but had a higher serum creatinine and a higher incidence of rejection.
Article
Background. Tacrolimus (FK506), a macrolide molecule that potently inhibits the expression of interleukin 2 by T lymphocytes, represents a potential major advance in the management of rejection following solid-organ transplantation. This randomized, open-label study compared the efficacy and safety of tacrolimus-based versus cyclosporine-based immunosuppression in patients receiving cadaveric kidney transplants. Methods. A total of 412 patients were randomized to tacrolimus (n=205) or cyclosporine (n=207) after cadaveric renal transplantation and were followed for 1 year for patient and graft survival and the incidence of acute rejection. Results. One-year patient survival rates were 95.6% for tacrolimus and 96.6% for cyclosporine (P=0.576). Corresponding 1-year graft survival rates were 91.2% and 87.9% (P=0.289). There was a significant reduction in the incidence of biopsy-confirmed acute rejection in the tacrolimus group (30.7%) compared with the cyclosporine group (46.4%, P=0.001), which was confirmed by blinded review, and in the use of antilymphocyte therapy for rejection (10.7% and 25.1%, respectively; P<0.001). Impaired renal function, gastrointestinal disorders, and neurological complications were commonly reported in both treatment groups, but tremor and paresthesia were more frequent in the tacrolimus group. The incidence of posttransplant diabetes mellitus was 19.9% in the tacrolimus group and 4.0% in the cyclosporine group (P<0.001), and was reversible in some patients. Conclusions. Tacrolimus is more effective than cyclosporine in preventing acute rejection in cadaveric renal allograft recipients, and significantly reduces the use of antilymphocyte antibody preparations. Tacrolimus was associated with a higher incidence of neurologic events, which were rarely treatment limiting, and with posttransplant diabetes mellitus, which was reversible in some patients.