Effects of Tacrolimus Pharmacokinetic Variability on Acute Rejection and Long-Term Graft Function after Kidney Transplantation

Similar documents
SELECTED ABSTRACTS. All (n) % 3-year GS 88% 82% 86% 85% 88% 80% % 3-year DC-GS 95% 87% 94% 89% 96% 80%

High Postoperative Tacrolimus Variability Predisposes to Early Pancreas Graft Loss

Cover Page. The handle holds various files of this Leiden University dissertation.

Does the formulation of tacrolimus matter?

OUT OF DATE. Choice of calcineurin inhibitors in adult renal transplantation: Effects on transplant outcomes

Literature Review: Transplantation July 2010-June 2011

Diabetes Mellitus GUIDELINES UNGRADED SUGGESTIONS FOR CLINICAL CARE IMPLEMENTATION AND AUDIT BACKGROUND

Steroid Minimization: Great Idea or Silly Move?

HHS Public Access Author manuscript Kidney Int. Author manuscript; available in PMC 2015 September 01.

Pharmacogenetics to tailor Drug Exposure and Outcomes in Kidney Transplantation

Literature Review Transplantation

Incidence of Rejection in Renal Transplant Surgery in the LVHN Population Leading to Graft Failure: 6 Year Review

Cover Page. The handle holds various files of this Leiden University dissertation.

Risk Factors in Long Term Immunosuppressive Use and Advagraf. Daniel Serón Nephrology department Hospital Universitari Vall d Hebron

Date: 23 June Context and policy issues:

Progress in Pediatric Kidney Transplantation

Long-term prognosis of BK virus-associated nephropathy in kidney transplant recipients

The CARI Guidelines Caring for Australasians with Renal Impairment. Membranous nephropathy role of steroids GUIDELINES

Diltiazem use in tacrolimus-treated renal transplant recipients Kothari J, Nash M, Zaltzman J, Prasad G V R

Older Living Kidney Donors and Recipients. Charles Le University of Colorado 6/24/11

Current Trends in Kidney Transplantation: The Role of Nonadherence

kidney OPTN/SRTR 2012 Annual Data Report:

Reduced graft function (with or without dialysis) vs immediate graft function a comparison of long-term renal allograft survival

Controversies in Renal Transplantation. The Controversial Questions. Patrick M. Klem, PharmD, BCPS University of Colorado Hospital

Donor-derived Cell-free DNA Improves DSA-informed Diagnosis of ABMR in Kidney Transplant Patients

Case Report Beneficial Effect of Conversion to Belatacept in Kidney-Transplant Patients with a Low Glomerular-Filtration Rate

Chapter 6: Transplantation

An Economic Analysis of the Cost Effectiveness of Blood Gene Expression

Laboratory Monitoring of Cyclosporine Pre-dose Concentration (C 0 ) After Kidney Transplantation in Isfahan

Out of date SUGGESTIONS FOR CLINICAL CARE (Suggestions are based on level III and IV evidence)

CURRICULUM VITAE July 5, Name Chang-Kwon Oh. Date of Birth August 15, 1961

Innovation In Transplantation:

The New Kidney Allocation System: What You Need to Know. Anup Patel, MD Clinical Director Renal and Pancreas Transplant Division Barnabas Health

Clinical implementation of pharmacogenetics in kidney transplantation: calcineurin inhibitors in the starting blocks

Management of HBV in KidneyTransplanted Patients Dr.E.Nemati

Why Do We Need New Immunosuppressive Agents

Kidney transplantation 2016: current status and potential challenges

Brief Communication. Introduction

Predictors of cardiac allograft vasculopathy in pediatric heart transplant recipients

Correspondence should be addressed to Mirjam Tielen;

Clinical Prediction Models of Patient and Graft Survival in Kidney Transplant Recipients: A Systematic Review and Validation Study

Shin Hwang, Gi-Won Song, Dong-Hwan Jung, Gil-Chun Park, Chul-Soo Ahn, Deok-Bog Moon, Tae-Yong Ha, Ki-Hun Kim, and Sung-Gyu Lee

European Risk Management Plan. Measures impairment. Retreatment after Discontinuation

Recognition and Treatment of Chronic Allograft Dysfunction

Kidney Transplant Outcomes In Elderly Patients. Simin Goral MD University of Pennsylvania Medical Center Philadelphia, Pennsylvania

Significance of Basiliximab Induction Therapy in Standard-Risk Renal Transplant in Tacrolimus Era: A Meta-Analysis

TDM. Measurement techniques used to determine cyclosporine level include:

Hasan Fattah 3/19/2013

DRUG LEVEL MONITORING AND ADJUSTMENT Silvio Sandrini, Brescia, Italy Chairs: Ryszard Grenda, Warsaw, Poland Julio Pascual, Barcelona, Spain

Acute rejection and late renal transplant failure: Risk factors and prognosis

Should Pediatric Patients Wait for HLA-DR-Matched Renal Transplants?

Should red cells be matched for transfusions to patients listed for renal transplantation?

Increased Early Rejection Rate after Conversion from Tacrolimus in Kidney and Pancreas Transplantation

General Introduction. 1 general introduction 13

ASBMT and Marrow Transplantation

Ten-year outcomes in a randomized phase II study of kidney transplant recipients administered belatacept 4-weekly or 8-weekly

Substance Use Among Potential Kidney Transplant Candidates and its Impact on Access to Kidney Transplantation: A Canadian Cohort Study

Efficacy and Safety of Thymoglobulin and Basiliximab in Kidney Transplant Patients at High Risk for Acute Rejection and Delayed Graft Function

ORIGINAL ARTICLE. Eric F. Martin, 1 Jonathan Huang, 3 Qun Xiang, 2 John P. Klein, 2 Jasmohan Bajaj, 4 and Kia Saeian 1

RENAL EVENING SPECIALTY CONFERENCE

Transplant Nephrology Update: Focus on Outcomes and Increasing Access to Transplantation

Liver Transplantation: The End of the Road in Chronic Hepatitis C Infection

CKD in Other Organ Transplants

BK virus infection in renal transplant recipients: single centre experience. Dr Wong Lok Yan Ivy

Narender Goel et al. Middletown Medical PC, Montefiore Medical Center & Albert Einstein College of Medicine, New York

TITLE: Proton Pump Inhibitors (PPIs) in Renal Transplant Patients: Evidence for PPI of Choice

Home Hemodialysis or Transplantation of the Treatment of Choice for Elderly?

A clear path forward COMING SOON THE LATEST INNOVATION IN KIDNEY TRANSPLANT SURVEILLANCE CAN DRIVE BETTER OUTCOMES FOR YOUR PATIENTS

EARLY VERSUS LATE STEROID WITHDRAWAL Julio Pascual, Barcelona, Spain Chairs: Ryszard Grenda, Warsaw, Poland

Therapeutic drug monitoring

Chapter 4: Steroid-resistant nephrotic syndrome in children Kidney International Supplements (2012) 2, ; doi: /kisup.2012.

Long-Term Renal Allograft Survival in the United States: A Critical Reappraisal

Transplant Success in Sensitized Patients Receiving a Standardized Desensitization Therapy: 3 Year Outcomes

Biological Basis for Increased Risk of Graft Loss in African American (AA)-APOL1 and Beyond

Pathological back-ground of renal transplant pathology and important mile-stones of the Banff classification

The CARI Guidelines Caring for Australasians with Renal Impairment. Idiopathic membranous nephropathy: use of other therapies GUIDELINES

Systematic Reviews and Meta- Analysis in Kidney Transplantation

Takashi Yagisawa 1,2*, Makiko Mieno 1,3, Norio Yoshimura 1,4, Kenji Yuzawa 1,5 and Shiro Takahara 1,6

Introduction and Overview of the Current Landscape on Organ Donation and Transplantation in Canada Jag Gill, MD

Serum samples from recipients were obtained within 48 hours before transplantation. Pre-transplant

James E. Cooper, M.D. Assistant Professor, University of Colorado at Denver Division of Renal Disease and Hypertension, Kidney and PancreasTransplant

BK Virus (BKV) Management Guideline: July 2017

Lucia Cea Soriano 1, Saga Johansson 2, Bergur Stefansson 2 and Luis A García Rodríguez 1*

Guideline 2: Treatment of HCV infection in patients with CKD Kidney International (2008) 73 (Suppl 109), S20 S45; doi: /ki.2008.

Update on Transplant Glomerulopathy

Pediatric Kidney Transplantation

Risk factors associated with the deterioration of renal function after kidney transplantation

CHAPTER 5 RENAL TRANSPLANTATION. Editor: Dr Goh Bak Leong

Kidney Transplant Outcomes for Prolonged Cold Ischemic Times in the Context of Kidney Paired Donation

Le migliori strategie immunosoppressive per il paziente con re-trapianto Prof. Maurizio Salvadori FIRENZE

Assessment of Deceased Donor Kidneys Using a Donor Scoring System

Outpatient Management of Delayed Graft Function Is Associated With Reduced Length of Stay Without an Increase in Adverse Events

Risk factors in the progression of BK virus-associated nephropathy in renal transplant recipients

A clear path forward AVAILABLE NOW THE LATEST INNOVATION IN KIDNEY TRANSPLANT SURVEILLANCE CAN DRIVE BETTER OUTCOMES FOR YOUR PATIENTS

For more information about how to cite these materials visit

J Am Soc Nephrol 14: , 2003

Pancreas After Islet Transplantation: A First Report of the International Pancreas Transplant Registry

Kidney and liver organ transplantation in persons with human immunodeficiency virus

This assessment report is based on evidence submitted by Astellas Pharma Ltd. on 18 January 2010.

Short-term and Long-term Survival of Kidney Allograft Cure Model Analysis

Transcription:

: Journal of Student Solutions to Pharmacy Challenges Volume 1 Issue 1 Article 4 2017 Effects of Tacrolimus Pharmacokinetic Variability on Acute Rejection and Long-Term Graft Function after Kidney Transplantation Stephan Seibert University of Minnesota - Twin Cities, seibe058@umn.edu Follow this and additional works at: http://pubs.lib.umn.edu/advances Recommended Citation Seibert, Stephan (2017) "Effects of Tacrolimus Pharmacokinetic Variability on Acute Rejection and Long-Term Graft Function after Kidney Transplantation," Advances in Pharmacy: Journal of Student Solutions to Pharmacy Challenges: Vol. 1 : Iss. 1, Article 4. Available at: http://pubs.lib.umn.edu/advances/vol1/iss1/4 This work is licensed under a Creative Commons Attribution-Noncommercial 4.0 License Advances in Pharmacy: Journal of Student Solutions to Pharmacy Challenges is published by the University of Minnesota Libraries Publishing.

Effects of Tacrolimus Pharmacokinetic Variability on Acute Rejection and Long-Term Graft Function after Kidney Transplantation Stephan R. Seibert 1 1 University of Minnesota College of Pharmacy, Duluth, MN, USA June 2017 Abstract Tacrolimus (TAC) is used for immunosuppression after kidney transplantation. Non-optimal TAC therapy contributes to renal toxicity, graft damage, and rejection. This paper reviews publications associating TAC intra-patient pharmacokinetic variability with long- and short-term transplant outcomes. The literature was reviewed performing systematic searches of MEDLINE and EMBASE databases from 1990 to May 2016 using appropriate Medical Subject Headings (MeSH) and key words. Titles and abstracts of hits were scanned for relevance, resulting in nine articles included in this analysis. Included articles were evaluated for content and relevance, and summarized. All studies were relatively small (N <400) non-randomized retrospective chart reviews. Only two articles did not show significant association between variability of TAC trough concentrations and at least one outcome after kidney transplantation. Four studies analyzing acute rejection (AR) showed significant association, while two did not. All studies analyzing graft loss or composite outcomes including graft loss showed significant association. Other studies showed association with donor-specific-antibody (DSA) development, but no association with renal function decline or overall patient survival. This review indicates an overall trend towards worse transplant outcomes in patients with higher TAC pharmacokinetic variability, however the size and design of the studies limit generalizability. Larger studies with more robust design are needed and should include genetic subgroup analysis and identification of sources of TAC variability to come to a definitive conclusion. Ideally, a dosing protocol incorporating wide varieties of genetic and clinical factors should be developed to optimize TAC dosing in transplant patients. 1 Introduction Tacrolimus (TAC) has become the main calcineurin inhibitor used in immunosuppressive therapy after kidney transplantations, largely replacing cyclosporine in clinical use. (1) Despite its clinical benefits, TAC therapy is not without problems. TAC has a narrow therapeutic window and requires therapeutic drug monitoring of blood trough concentrations. Frequent dose adjustments are common in order to avoid nephrotoxicity associated with concentrations that are too high and, conversely, insufficient immunosuppression which has been shown to increase the risk of adverse outcomes such as acute rejection (AR) at sub-therapeutic concentrations.(2) Outcomes and survival of kidney transplant patients have consistently improved over recent decades. Especially AR has become relatively rare and occurs in only less than 10% of recipients. Long-term graft function remains a problem, however, and approximately half of all kidney allografts are lost by 10 years after transplantation. (3) It is unclear what exactly causes this graft loss, but it is likely due to a variety of contributing factors and insufficient immunosuppression may play a large role. 1

TAC pharmacokinetics are influenced by genetic factors. Several single-nucleotide-polymorphisms (SNPs) are associated with altered TAC trough levels. (4) These SNPs contribute to inter-patient variability in TACs pharmacokinetics and partially determine initial starting doses, although there are other factors associated with intra-patient pharmacokinetic variability post-transplant including drug-drug interactions, food-drug interactions, changes in hepatic metabolism, and nonadherence. (5) In recent years, intra-patient variability of TAC concentrations post-transplant has been investigated and proposed as a significant risk factor for long term complications such as late rejection, graft loss, donorspecific-antibody (DSA) development, and declining kidney function. Understanding the variables associated with intra-patient variability and strategies to reduce post-transplant pharmacokinetic variability may improve short- and long-term outcomes. The long-term goal is to develop a comprehensive understanding of the influencing factors on TAC pharmacokinetics in renal transplant patients and their relationship to a recipients genetic make-up to optimize individual immunosuppressive therapy and enhance patient outcomes. Specifically, this paper aims to perform a systematic review of the literature related to the effects of TAC pharmacokinetic variability on short- and long-term outcomes after kidney transplantation. Databases were systematically searched for relevant publications, the literature was critically evaluated, and a summary of our findings is presented here. 2 Methods 2.1 Database Search A review of the existing literature on TAC pharmacokinetic variability in kidney transplant patients and its association with short- and long-term outcomes was performed by systematically searching MEDLINE and EMBASE databases from 1990 to May 2016. Searches were performed using the following focused and auto-exploded Medical Subject Heading (MeSH) terms and key words: (1) exp *Tacrolimus/; (2) exp *Kidney Transplantation/; (3) variability.mp; and (4) variation.mp. Boolean operators were used to produce the final search algorithm: 1 AND 2 AND (3 OR 4). Results were limited to studies on human subjects and manuscripts in English language, which produced 171 hits in MEDLINE and 246 hits in EMBASE, for a total of 417 hits. EndNote Basic (Thomson Reuters, PA, USA) was used to store and organizes articles. After removal of duplicate hits using EndNotes Find Duplicate function as well as manual screening of titles and authors, 280 individual hits remained. 2.2 Inclusions and Reference Search Each of the 280 hits was evaluated for inclusion by manually reading all titles and abstracts. Articles were included if the respective study or analysis attempted to relate a statistical measure of variability (standard deviation, intra-patient variability, or coefficient of variation) in TAC trough concentrations to objective short- and long-term outcomes after kidney transplantation. This method resulted in an inclusion of 8 articles. Furthermore, the reference sections of these articles were screened for additional relevant literature, resulting in the inclusion of 1 additional article, for a total of 9 articles used in our analysis. 2.3 Evaluation Each included article was evaluated for several characteristics, including type of study design, timeframe during which TAC trough concentrations were measured post-transplant, number of subjects, specifics about patient populations (such as ethnicities and age ranges), statistical measure of TAC trough variability, types of measured outcomes, and primary results. Studies were also evaluated for their level of evidence using the Oxford 2011 Levels of Evidence grading scheme. Aip June 2017 2

3 Results 3.1 Included Studies The study selection process is summarized in figure 1. After removal of duplicate hits and manual screening of abstracts for relevance, eight full text articles plus one additional article pulled from their references were reviewed, for a total of nine articles included in our qualitative analysis. Each of those studies is summarized in table 1, listing year of publication, primary author, study design, level of evidence, number of included subjects, characteristics about the study population, type of statistical measure of variability used, time frame of measured TAC trough concentrations, outcomes assessed, and main findings. 3.2 Study Designs and Characteristics Included studies were similar in design and strength of evidence. All studies were retrospective cohort analyses of transplant patients receiving TAC maintenance therapy. Studies were based on chart reviews focusing on the relationship between TAC pharmacokinetic variability and prognosis of outcomes after transplant. Applying the Oxford 2011 Levels of Evidence grading scheme, all studies displayed evidence of level 3 out of 5 (1 being the strongest and 5 being the weakest evidence). All studies had relatively small sample sizes, ranging from 46 patients to 394 patients. None of the studies were multi-center trials, and study participants were relatively homogeneous within each study, representing local populations from the US, Canada, the Netherlands, Spain, Ireland, and Korea. Three of the nine studies focused only on pediatric patients, while the other six studies were limited to adult patients. Eight of the studies analyzed only kidney transplants, while one study of Canadian pediatric patients included kidney, lung, and liver transplants. Pharmacokinetic variability in TAC trough concentrations among transplant patients was the main focus of all studies, however the time frames of included TAC levels varied greatly among studies: Five studies analyzed the variability of TAC trough concentrations within the first year after transplant, starting at 1, 3, 4, or 6 months after transplant. One study included all measurements between 1 month and 2 years after transplant. One study focused on later measurements between 1 year after transplant and last followup. Another study included all TAC trough concentrations within 1 year of study begin, at which point all included patients were at least 3 months post-transplant. Lastly, one study conducted two separate analyzes, one of them including all TAC levels within 6 months prior to an acute rejection event, the other analyzing measurements between 6 months after transplant and last follow-up. Each study used one of three statistical measures of variation to assess the degree of TAC pharmacokinetic variability among patients: Two studies used a simple standard deviation (SD), three studies used intrapatient variability (IPV), and four studies used a coefficient of variation (CV). There was no trend between the publication dates of articles and type of variability measures used or between the type of variability measure and its significance for outcomes. Each measure of variability differs slightly. Their definitions are given in equations 1 through 3. 3.3 Study Outcomes and Findings The reviewed articles included a wide variety of outcome measures commonly seen in the progression of kidney transplant recipients. Most commonly, events of AR were the primary measured outcome, which was the case in six studies. Other analyzed outcomes were graft loss (two studies), DSA development (one study), kidney function decline (one study), overall survival (one study) or some form of composite including a variety of measures like AR, graft loss, kidney function decline, chronic nephropathy and glomerulopathy, or death (three studies). The majority of studies analyzed correlations with more than one outcome and only three of the studies focused solely on AR. The majority of reviewed studies found a statistically significant correlation (p<0.05) between higher levels of the respective measure of TAC pharmacokinetic variability and at least one negative outcome after transplantation. Only one study found no correlation with any outcome. Interestingly, of the six studies that Aip June 2017 3

analyzed AR, four found a significant correlation with variability measures, while two did not. Furthermore, only the Korean study analyzed subgroups based on genotype. In this study, transplant recipients who expressed CYP3A5, an enzyme involved in the metabolism of TAC, showed an association between TAC IPV and AR, but patients that did not express CYP3A5 due to a polymorphism showed no association. All studies that analyzed graft loss or composite outcomes including graft loss demonstrated significant associations with TAC variability. Lastly, DSA development demonstrated association, while kidney function decline by itself and overall patient survival did not. 4 Discussion and Conclusion While insufficient immunosuppression of transplant recipients is a known determinant of poor outcomes, only few studies have looked at the association between pharmacokinetic variability of immunosuppressive drug regimens and outcomes after kidney transplantation. A systematic review of the literature only found nine relevant studies published between 1990 and 2016 attempting to associate TAC variability with kidney transplant outcomes. Additionally, all of these studies were non-random retrospective chart reviews from highly specific populations and had relatively small sample sizes, the largest including 394 patients, limiting their generalizability. Seven of the nine studies found some form of statistically significant association between TAC pharmacokinetic variability and negative transplant outcomes. Most importantly, all studies analyzing graft loss (two) or a composite outcome including graft loss (three) found significant association with TAC variability. Those that analyzed AR found mixed results, with four studies finding significant association and two finding no association. Additionally, one study found association with DSA development, while another study found no association with kidney function decline. The most recent study, which also had the largest sample size, found no association with overall patient survival, suggesting that high degrees of TAC variability may lead to adverse graft outcomes but not necessarily to the death of the patient if dialysis or re-transplant are available as rescue options. This systematic review shows that there may be an overall trend towards worse outcomes for kidney transplant patients demonstrating high levels of TAC pharmacokinetic variability, however the scarceness, retrospective designs, homogeneity, and small sizes of the reviewed studies limits the generalizability of this conclusion. All studies focused on TAC trough concentrations and did not assess variability in administered TAC doses, which could be a separate source of pharmacokinetic variability. Additionally, only one of the nine studies attempted subgroup analysis based on recipients genotype, and only included a single polymorphism. In summary, larger studies with more robust designs are needed to draw definitive conclusions about the interaction of TAC pharmacokinetic variability and outcomes after kidney transplantation. Future studies should consist of larger prospective cohorts from multiple centers and populations and should streamline TAC measurement time frames as well as the statistic variability measure, preferably using the CV, as it is arguably the most robust of the three measures. (6) Additionally, future research should include analysis of genetics backgrounds of recipients, as several SNPs in TAC metabolizing enzymes have been identified and associated with TAC pharmacokinetics. (4, 7, 8) Lastly, attempts should be made to identify and measure definitive sources of TAC pharmacokinetic variability, such as non-adherence or drug-drug interactions. Ideally, a dosing equation or protocol should be developed incorporating a wide range of genetic and clinical factors to optimize TAC dosing. 5 Acknowledgments I would like to thank my mentor, Dr. Pamala Jacobson, for her inspiration and support with this review and the follow-up analysis of patient data. I also thank the DeKAF Genomics team for sharing their data with me and allowing me to participate in their research group. Aip June 2017 4

6 References 1. Matas AJ, Smith JM, Skeans MA, et al. OPTN/SRTR 2011 Annual Data Report: Kidney. 2. Israni AK, Riad SM, Leduc R, et al. Tacrolimus trough levels after month 3 as a predictor of acute rejection following kidney transplantation: a lesson learned from DeKAF Genomics. Transpl Int. 2013;26:982-989. 3. Lamb KE, Lodhi S, Meier-Kriesche HU. Long-Term Renal Allograft Survival in the United States: A Critical Reappraisal. American Journal of Transplantation. 2011;11:450-462. 4. Oetting WS, Schladt DP, Guan W, et al. Genomewide Association Study of Tacrolimus Concentrations in African American Kidney Transplant Recipients Identifies Multiple CYP3A5 Alleles. Am J Transplant. 2016;16:574-582. 5. Shuker N, van Gelder T, Hesselink DA. Intra-patient variability in tacrolimus exposure: causes, consequences for clinical management. Transplant Rev (Orlando). 2015;29:78-84. 6. Hsiau M, Fernandez HE, Gjertson D, Ettenger RB, Tsai EW. Monitoring nonadherence and acute rejection with variation in blood immunosuppressant levels in pediatric renal transplantation. Transplantation. 2011;92:918-922. 7. Pulk RA, Schladt DS, Oetting WS, et al. Multigene predictors of tacrolimus exposure in kidney transplant recipients. Pharmacogenomics. 2015;16:841-854. 8. Jacobson P, Miller M, Schladt D, et al. Variants associated with tacrolimus troughs in European American kidney transplant recipients: A genome wide association study. American Journal of Transplantation. 2015;15:no pagination. 9. Borra LCP, Roodnat JI, Kal JA, Mathot RAA, Weimar W, Van Gelder T. High within-patient variability in the clearance of tacrolimus is a risk factor for poor long-term outcome after kidney transplantation. Nephrology Dialysis Transplantation. 2010;25:2757-2763. 10. Pollock-Barziv SM, Finkelstein Y, Manlhiot C, et al. Variability in tacrolimus blood levels increases the risk of late rejection and graft loss after solid organ transplantation in older children. Pediatr Transplant. 2010;14:968-975. 11. Ro H, Min SI, Yang J, et al. Impact of tacrolimus intraindividual variability and CYP3A5 genetic polymorphism on acute rejection in kidney transplantation. Ther Drug Monit. 2012;34:680-685. 12. Prytula AA, Bouts AH, Mathot RAA, et al. Intra-patient variability in tacrolimus trough concentrations and renal function decline in pediatric renal transplant recipients. Pediatric transplantation. 2012;16:613-618. 13. Sapir-Pichhadze R, Wang Y, Famure O, Li Y, Kim SJ. Time-dependent variability in tacrolimus trough blood levels is a risk factor for late kidney transplant failure. Kidney Int. 2014;85:1404-1411. 14. Schmid S, Bryce R, Reeder B, et al. Volatility of serum creatinine relative to tacrolimus levels predicts kidney transplant rejection. Annals of transplantation : quarterly of the Polish Transplantation Society. 2014;19:403-406. 15. Rodrigo E, Segundo DS, Fernandez-Fresnedo G, et al. Within-Patient Variability in Tacrolimus Blood Levels Predicts Kidney Graft Loss and Donor-Specific Antibody Development. Transplantation. 2015. 16. O Regan JA, Canney M, Connaughton DM, et al. Tacrolimus trough-level variability predicts longterm allograft survival following kidney transplantation. Journal of Nephrology. 2016;29:269-276. Aip June 2017 5

Tables Year Primary Author Design 2010 Borra 9 Retro. 2010 Pollock- BarZiv 10 Retro. 2011 Hsiau 6 Retro. 2012 Ro 11 Retro. 2012 Prytula 12 Retro. Evidence Level N Population Var. Measure TAC Measurement Outcomes Level 3 297 Dutch IPV 6 12 mo. pt. DCGF (graft loss, Adults chronic allograft nephropathy, or doubling of scr, > 12 mo. pt.) Level 3 144 Canadian SD 6 mo. prior to Late AR Pediatrics late rejection (> 6 mo. pt.) (8 18 or last follow yo.); up heart, kidney, 6 mo. pt. Graft Loss lung, or last follow-up (> 12 mo. pt.) liver transplants Level 3 46 US Pediatrics (2 22 yo.) Level 3 249 Korean Adults Level 3 69 Dutch Pediatrics (3 18 yo.) Results High TAC IPV associated with more DCGF (p = 0.003) Higher TAC SD associated with more late rejection (p =0.02) Increase in TAC SD > 2 associated with more graft loss (p = 0.003) CV 1 12 mo. pt. AR High TAC CV associated with more AR (p = 0.005) IPV 6 12 mo. pt. AR High TAC IPV associated with more AR in CYP3A5 expressers (p = 0.001) but not in non-expressers CV 0 12 mo. after study begin, at least 3 mo. pt. egfr Decline; Late AR (4 year follow-up) No sig. association between TAC CV and egfr decline (p = 0.337); High TAC CV associated with more late AR (p = 0.045)

2014 Sapir- Pichhadze 13 Retro. 2014 Schmid 14 Retro. 2015 Rodrigo 15 Retro. 2016 O Regan 16 Retro. Level 3 356 Canadian Adults Level 3 81 Canadian Adults Level 3 310 Spanish Adults SD 12 mo. pt. last follow-up CV 1 mo. 2 years pt. Composite Endpoint (late AR, transplant glomerulopathy, graft loss, death) AR Increase in TAC SD associated with worse outcomes (p = 0.01) No sig. association between TAC CV and AR (p = 0.65) CV 4 12 mo. pt. DCGL, dndsa, AR High TAC CV (> 30%) associated with more DCGL (p = 0.004) and dndsa (p = 0.002), but not AR (p = 0.327) Level 3 394 Irish Adults IPV 3 12 mo. pt. Graft Loss, Patient Survival (> 12 mo. pt.) High IPV associated with more graft loss (p = 0.019), but not patient survival (p = 0.23) Table 1. Summary of Reviewed Articles TAC Tacrolimus CV Coefficient of Variability IPV Intra-Individual Variability SD Standard Deviation AR Acute Rejection DCGL Death Censored Graft Loss dndsa De Novo Donor Specific Antibodies egfr Estimated Glomerular Filtration Rate mo. month pt. post transplant yo. years old

8 Figures Figure 1: Study Section Flow Diagram Aip June 2017 8

9 Equations SD = 1 n n (x i x) 2 i=1 Equation 1. Standard Deviation IP V = 1 n n i=1 x 1 x x Equation 2. Intra-Patient Variability CV = SD x Equation 3. Coefficient of Variation Aip June 2017 9