582
Views
10
CrossRef citations to date
0
Altmetric
Review

Immunosuppressive therapy post-transplantation in children: what the clinician needs to know

Pages 139-154 | Received 19 Aug 2019, Accepted 08 Jan 2020, Published online: 23 Jan 2020

References

  • The NAPRTCS. 2014. Annual report. [cited 2019 Aug 15]. Available from: https://web.emmes.com/study/ped/index.htm.
  • Cooperative European Paediatric Renal Transplant Initiative (CERTAIN) registry. [cited 2019 Aug 15]. Available from: www.certain-registry.eu.
  • Peddi VR, Bryant M, Roy-Chaudhury P, et al. Safety, efficacy, and cost analysis of thymoglobulin induction therapy with intermittent dosing based on CD3+ lymphocyte counts in kidney and kidney-pancreas transplant recipients. Transplantation. 2002;73:1514–1518.
  • Stratta RJ, Sundberg AK, Farney AC, et al. Experience with alternate-day thymoglobulin induction in pancreas transplantation with portal-enteric drainage. Transplant Proc. 2005;37:3546–3548.
  • Wong W, Agrawal N, Pascual M, et al. Comparison of two dosages of thymoglobulin used as a short-course for induction in kidney transplantation. Transpl Int. 2006;19:629–635.
  • Gurk-Turner C, Airee R, Philosophe B, et al. Thymoglobulin dose optimization for induction therapy in high risk kidney transplant recipients. Transplantation. 2008;85:1425–1430.
  • Stevens RB, Mercer DF, Grant WJ, et al. Randomized trial of single-dose versus divided-dose rabbit anti-thymocyte globulin induction in renal transplantation: an interim report. Transplantation. 2008;85:1391–1399.
  • Goggins WC, Pascual MA, Powelson JA, et al. A prospective, randomized, clinical trial of intraoperative versus postoperative thymoglobulin in adult cadaveric renal transplant recipients. Transplantation. 2003;76:798–802.
  • Sterkers G, Baudouin V, Ansart-Pirenne H, et al. Duration of action of a chimeric interleukin-2 receptor monoclonal antibody, basiliximab, in pediatric kidney transplant recipients. Transplant Proc. 2000;32:2757–2759.
  • Höcker B, Kovarik JM, Daniel V, et al. Pharmacokinetics and immunodynamics of basiliximab in pediatric renal transplant recipients on mycophenolate mofetil comedication. Transplantation. 2008;86:1234–1240.
  • Grenda R, Watson A, Vondrak K, et al. A prospective, randomized, multicenter trial of tacrolimus-based therapy with or without basiliximab in pediatric renal transplantation. Am J Transplant. 2006;6:1666–1672.
  • Offner G, Toenshoff B, Höcker B, et al. Efficacy and safety of basiliximab in pediatric renal transplant patients receiving cyclosporine, mycophenolate mofetil, and steroids. Transplantation. 2008;86:1241–1248.
  • Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant. 2009;9(Suppl 3):S1–155.
  • Webster AC, Ruster LP, McGee R, et al. Interleukin 2 receptor antagonists for kidney transplant recipients. Cochrane Database Syst Rev. 2010;20:CD003897.
  • Hellemans R, Bosmans JL, Abramowicz D. Induction therapy for kidney transplant recipients: do we still need anti‐IL2 receptor monoclonal antibodies? Am J Transplant. 2017;17:22–27.
  • Dantal J, Hourmant M, Cantarovich D, et al. Effect of long-term immunosuppression in kidney-graft recipients on cancer incidence: randomised comparison of two cyclosporin regimens. Lancet. 1998;351:623–628.
  • Venkataramanan R, Jain A, Warty VW, et al. Pharmacokinetics of FK 506 following oral administration: a comparison of FK 506 and cyclosporine. Transplant Proc. 1991;23:931–933.
  • Andrews LM, Li Y, De Winter BCM, et al. Pharmacokinetic considerations related to therapeutic drug monitoring of tacrolimus in kidney transplant patients. Expert Opin Drug Metab Toxicol. 2017;13:1225–1236.
  • De Jonge H, De Loor H, Verbeke K, et al. In vivo CYP3A4 activity, CYP3A5 genotype, and hematocrit predict tacrolimus dose requirements and clearance in renal transplant patients. Clin Pharmacol Ther. 2012;92:366–375.
  • Tang JT, Andrews LM, Van Gelder T, et al. Pharmacogenetic aspects of the use of tacrolimus in renal transplantation: recent developments and ethnic considerations. Expert Opin Drug Metab Toxicol. 2016;12:555–565.
  • Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet. 2001;27:383–391.
  • Picard N, Bergan S, Marquet P, et al. Pharmacogenetic biomarkers predictive of the pharmacokinetics and pharmacodynamics of immunosuppressive drugs. Ther Drug Monit. 2016;38(Suppl 1):S57–69.
  • Billing H, Höcker B, Fichtner A, et al. Single-nucleotide polymorphism of CYP3A5 impacts the exposure to tacrolimus in pediatric renal transplant recipients: a pharmacogenetic substudy of the TWIST trial. Ther Drug Monit. 2017;39:21–28.
  • Andrews LM, De Winter BC, Tang JT, et al. Overweight kidney transplant recipients are at risk of being overdosed following standard bodyweight-based tacrolimus starting dose. Transplant Direct. 2017;3:e129.
  • Trompeter R, Filler G, Webb NJ, et al. Randomized trial of tacrolimus versus cyclosporin microemulsion in renal transplantation. Pediatr Nephrol. 2002;7:141–149.
  • Filler G, Webb NJ, Milford DV, et al. Four-year data after pediatric renal transplantation: a randomized trial of tacrolimus vs. cyclosporin microemulsion. Pediatr Transplant. 2005;9:498–503.
  • Tanabe K. Calcineurin inhibitors in renal transplantation: what is the best option? Drugs. 2003;3:1535–1548.
  • Habbig S, Volland R, Krupka K, et al. Dyslipidemia after pediatric renal transplantation-the impact of immunosuppressive regimens. Pediatr Transplant. 2017;21:e12914.
  • Tönshoff B, Höcker B. Treatment strategies in pediatric solid organ transplant recipients with calcineurin inhibitor-induced nephrotoxicity. Pediatr Transplant. 2006;10:721–729.
  • Grotz WH, Breitenfeldt MK, Braune SW et al. Calcineurin-inhibitor induced pain syndrome (CIPS): a severe disabling complication after organ transplantation. Transpl Int. 2001;14:16–23.
  • Nankivell BJ, Borrows RJ, Fung CL, et al. The natural history of chronic allograft nephropathy. N Engl J Med. 2003;349:2326–2333.
  • Ojo AO, Held PJ, Port FK, et al. Chronic renal failure after transplantation of a nonrenal organ. N Engl J Med. 2003;349:931–940.
  • Höcker B, Tönshoff B. Treatment strategies to minimize or prevent chronic allograft dysfunction in pediatric renal transplant recipients: an overview. Paediatr Drugs. 2009;11:381–396.
  • Höcker B, Tönshoff B. Calcineurin inhibitor-free immunosuppression in pediatric renal transplantation: a viable option? Paediatr Drugs. 2011;13:49–69.
  • Neu AM, Ho PL, RN F, et al. Tacrolimus vs. cyclosporine A as primary immunosuppression in pediatric renal transplantation: a NAPRTCS study. Pediatr Transplant. 2003;7:217–222.
  • Ekberg H, Tedesco-Silva H, Demirbas A, et al. ELITE-symphony study. Reduced exposure to calcineurin inhibitors in renal transplantation. N Engl J Med. 2007 Dec;20(357):2562–2575.
  • Kahan BD, Keown P, Levy GA, et al. Therapeutic drug monitoring of immunosuppressant drugs in clinical practice. Clin Ther. 2002;24:330–350.
  • Weber LT, Armstrong VW, Shipkova M, et al. Cyclosporin A absorption profiles in pediatric renal transplant recipients predict the risk of acute rejection. Ther Drug Monit. 2004;26:415–424.
  • Riva N, Guido PC, Ibañez J, et al. Therapeutic monitoring of pediatric renal transplant patients with conversion to generic cyclosporin. Int J Clin Pharm. 2014;36:779–786.
  • Allison AC, Eugui EM. Mechanisms of action of mycophenolate mofetil in preventing acute and chronic allograft rejection. Transplant. 2005;80:181–190.
  • van Leuven SI, Kastelein JJ, Allison AC, et al. Mycophenolate mofetil (MMF): firing at the atherosclerotic plaque from different angles? Cardiovasc Res. 2006;69:341–347.
  • van Gelder T, Silva HT, de Fijter JW, et al. Comparing mycophenolate mofetil regimens for de novo renal transplant recipients: the fixed-dose concentration-controlled trial. Transplantation. 2008;86:1043–1051.
  • Höcker B, van Gelder T, Martin-Govantes J, et al. Comparison of MMF efficacy and safety in paediatric vs. adult renal transplantation: subgroup analysis of the randomised, multicentre FDCC trial. Nephrol Dial Transplant. 2011;26:1073–1079.
  • Shipkova M, Armstrong VW, Oellerich M, et al. Mycophenolate mofetil in organ transplantation: focus on metabolism, safety and tolerability. Expert Opin Drug Metab Toxicol. 2005;1:505–526.
  • Cattaneo D, Perico N, Gaspari F, et al. Glucocorticoids interfere with mycophenolate mofetil bioavailability in kidney transplantation. Kidney Int. 2002;62:1060–1067.
  • Bunchman T, Navarro M, Broyer M, et al. The use of mycophenolate mofetil suspension in pediatric renal allograft recipients. Pediatr Nephrol. 2001;16:978–984.
  • Höcker B, Weber LT, Bunchman T, et al. Mycophenolate mofetil suspension in pediatric renal transplantation: three-year data from the tricontinental trial. Pediatr Transplant. 2005;9:504–511.
  • Staskewitz A, Kirste G, Tönshoff B, et al. Mycophenolate mofetil in pediatric renal transplantation without induction therapy: results after 12 months of treatment. Transplantation. 2001;71:638–644.
  • Jungraithmayr T, Staskewitz A, Kirste G, et al. Pediatric renal transplantation with mycophenolate mofetil-based immunosuppression without induction: results after three years. Transplantation. 2003;75:454–461.
  • Cransberg K, Marlies Cornelissen EA, Davin JC, et al. Improved outcome of pediatric kidney transplantations in the Netherlands–effect of the introduction of mycophenolate mofetil? Pediatr Transplant. 2005;9:1004–1011.
  • Ferraris JR, Ghezzi LF, Vallejo G, et al. Improved long-term allograft function in pediatric renal transplantation with mycophenolate mofetil. Pediatr Transplant. 2005;9:178–182.
  • Ferraris JR, Tambutti ML, Redal MA, et al. Conversion from azathioprine [correction of azathioprina] to mycophenolate mofetil in pediatric renal transplant recipients with chronic rejection. Transplantation. 2000;70:297–301.
  • Henne T, Latta K, Strehlau J, et al. Mycophenolate mofetil-induced reversal of glomerular filtration loss in children with chronic allograft nephropathy. Transplantation. 2003;76:1326–1330.
  • Clayton PA, McDonald SP, Chapman JR, et al. Mycophenolate versus azathioprine for kidney transplantation: a 15-year follow-up of a randomized trial. Transplantation. 2012;94:152–158.
  • Harden PN, Sherston SN. Optimal management of young adult transplant recipients: the role of integrated multidisciplinary care and peer support. Ann Saudi Med. 2013;33:489–491.
  • Weber LT, Shipkova M, Armstrong VW, et al. The pharmacokinetic-pharmacodynamic relationship for total and free mycophenolic acid in pediatric renal transplant recipients: a report of the German study group on mycophenolate mofetil therapy. J Am Soc Nephrol. 2002;13:759–768.
  • van Gelder T, Le Meur Y, Shaw LM, et al. Therapeutic drug monitoring of mycophenolate mofetil in transplantation. Ther Drug Monit. 2006;28:145–154.
  • Tönshoff B, David-Neto E, Ettenger R, et al. Pediatric aspects of therapeutic drug monitoring of mycophenolic acid in renal transplantation. Transplant Rev (Orlando). 2011;25:78–89.
  • Le Meur Y, Büchler M, Thierry A, et al. Individualized mycophenolate mofetil dosing based on drug exposure significantly improves patient outcomes after renal transplantation. Am J Transplant. 2007;7:2496–2503.
  • Elion GB. The pharmacology of azathioprine. Ann NY Acad Sci. 1993;685:400–407.
  • Mudigonda T, Levender MM, O’Neill JL, et al. Incidence, risk factors, and preventative management of skin cancers in organ transplant recipients: a review of single- and multicenter retrospective studies from 2006 to 2010. Dermatol Surg. 2013;39:345–364.
  • Mittal A, Colegio OR. Skin cancers in organ transplant recipients. Am J Transplant. 2017;17:2509–2530.
  • Mahalati K, Kahan BD. Clinical pharmacokinetics of sirolimus. Clin Pharmacokinet. 2001;40:573–585.
  • Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmacokinetics of everolimus. Clin Pharmacokinet. 2004;43:83–95.
  • Crowe A, Bruelisauer A, Duerr L, et al. Absorption and intestinal metabolism of SDZ-RAD and rapamycin in rats. Drug Metab Dispos. 1999;27:627–632.
  • Rapamune. Summary of product characteristics. New York: Pfizer; Last updated 2011.
  • Certican. Basic prescribing information. Basel, Switzerland: Novartis Pharma AG; 2012.
  • Ganschow R, Pape L, Sturm E, et al. Growing experience with mTOR inhibitors in pediatric solid organ transplantation. Pediatr Transplant. 2013;17:694–706.
  • Ettenger R, Hoyer PF, Grimm P, et al. Multicenter trial of everolimus in pediatric renal transplant recipients: results at three year. Pediatr Transplant. 2008;12:456–463.
  • Pape L, Ahlenstiel T, Ehrich JH, et al. Reversal of loss of glomerular filtration rate in children with transplant nephropathy after switch to everolimus and low-dose cyclosporine A. Pediatr Transplant. 2007;11:291–295.
  • Kovarik JM, Curtis JJ, Hricik DE, et al. Differential pharmacokinetic interaction of tacrolimus and cyclosporine on everolimus. Transplant Proc. 2006;38:3456–3458.
  • Kovarik JM, Kalbag J, Figuerredo J, et al. Differential influence of two cyclosporine formulations on everolimus pharmacokinetics: A clinically relevant pharmacokinetic interaction. J Clin Pharmacol. 2002;42:95–99.
  • Brandhorst G, Tenderich G, Zittermann A, et al. Everolimus exposure in cardiac transplant recipients is influenced by concomitant calcineurin inhibitor. Ther Drug Monit. 2008;30:113–116.
  • Tönshoff B, Ettenger R, Dello Strologo L, et al. Early conversion of pediatric kidney transplant patients to everolimus with reduced tacrolimus and steroid elimination: results of a randomized trial. Am J Transplant. 2019;19:811–822.
  • Tedesco-Silva H, Felipe C, Ferreira A, et al. Reduced incidence of cytomegalovirus infection in kidney transplant recipients receiving everolimus and reduced tacrolimus doses. Am J Transplant. 2015;15:2655–2664.
  • Höcker B, Zencke S, Pape L, et al. Impact of everolimus and low-dose cyclosporin on cytomegalovirus replication and disease in pediatric renal transplantation. Am J Transplant. 2016;16:921–929.
  • Oliveira PF, Cheng CY, Alves MG. Emerging role for mammalian target of rapamycin in male fertility. Trends Endocrinol Metab. 2017;28:165–167.
  • Grimbert P, Thaunat O. mTOR inhibitors and risk of chronic antibody-mediated rejection after kidney transplantation: where are we now? Transpl Int. 2017;30:647–657.
  • Franchimont D. Overview of the actions of glucocorticoids on the immune response: a good model to characterize new pathways of immunosuppression for new treatment strategies. Ann N Y Acad Sci. 2004;1024:124–137.
  • Ingulli E, Tejani A. Steroid withdrawal after renal transplantation. In: Tejani AH, Fine RM, editors. Pediatric renal transplantation. New York: Wiley-Liss; 1994. p. 221–238.
  • Benfield MR, Bartosh S, Ikle D, et al. A randomized double-blind, placebo controlled trial of steroid withdrawal after pediatric renal transplantation. Am J Transplant. 2010;10:81–88.
  • Sutherland S, Li L, Concepcion W, et al. Steroid-free immunosuppression in pediatric renal transplantation: rationale for and outcomes following conversion to steroid based therapy. Transplantation. 2009;87:1744–1748.
  • Barletta GM, Kirk E, Gardner JJ, et al. Rapid discontinuation of corticosteroids in pediatric renal transplantation. Pediatr Transplant. 2009;13:571–578.
  • Höcker B, Weber LT, Feneberg R, et al. Prospective, randomized trial on late steroid withdrawal in pediatric renal transplant recipients under cyclosporine microemulsion and mycophenolate mofetil. Transplantation. 2009;87:934–941.
  • Höcker B, Weber L, Feneberg R, et al. Improved growth and cardiovascular risk after late steroid withdrawal: 2-year results of a prospective, randomized trial in paediatric renal transplantation. Nephrol Dial Transplant. 2010;25:617–624.
  • Sarwal MM, Vidhun JR, Alexander SR, et al. Continued superior outcomes with modification and lengthened follow-up of a steroid-avoidance pilot with extended daclizumab induction in pediatric renal transplantation. Transplantation. 2003;76:1331–1339.
  • Shapiro R, Ellis D, Tan HP, et al. Antilymphoid antibody preconditioning and tacrolimus monotherapy for pediatric kidney transplantation. J Pediatr. 2006;148:813–818.
  • Chavers BM, Chang C, Gillingham KJ, et al. Pediatric kidney transplantation using a novel protocol of rapid (6-day) discontinuation of prednisolone: 2-year results. Transplantation. 2009;88:237–241.
  • Grenda R, Watson A, Trompeter R, et al. A randomized trial to assess the impact of early steroid withdrawal on growth in pediatric renal transplantation: the TWIST Study. Am J Transplant. 2010;10:828–836.
  • Webb N, Douglas S, Rajai A, et al. Corticosteroid-free kidney transplantation improves growth: two-year follow-up of the TWIST randomised controlled trial. Transplantation. 2015;99:1178–1185.
  • Sarwal MM, Ettenger RB, Dharnidharka V, et al. Complete corticosteroid avoidance is effective and safe in children with renal transplants: a multicentre randomized trial with three year follow-up. Am J Transplant. 2012;12:2719–2729.
  • Pape L, Offner G, Kreuzer M, et al. De novo therapy with everolimus, low-dose cyclosporine A, basiliximab and steroid elimination in pediatric kidney transplantation. Am J Transplant. 2010;10:2349–2354.
  • van Gelder T. What is the future of generics in transplantation? Transplantation. 2015;99:2269–2273.
  • Suresh S, Upton J, Green M, et al. Live vaccines after pediatric solid organ transplant: proceedings of a consensus meeting, 2018. Pediatr Transplant. 2019;23:e13571.
  • Dharnidharka VR, Lamb KE, Zheng J, et al. Across all solid organs, adolescent age recipients have worse transplant organ survival than younger age children: a US national registry analysis. Pediatr Transplant. 2015;19:471–476.
  • Rubik J, Debray D, Iserin F, et al. Comparative pharmacokinetics of tacrolimus in stable pediatric allograft recipients converted from immediate-release tacrolimus to prolonged-release tacrolimus formulation. Pediatr Transplant. 2019;23:e13391.
  • Rubik J, Debray D, Kelly D, et al. Efficacy and safety of prolonged-release tacrolimus in stable pediatric allograft recipients converted from immediate-release tacrolimus - a phase 2, open-label, single-arm, one-way crossover study. Transpl Int. 2019;32:1182–1193.
  • Noble J, Jouve T, Janbon B, et al. Belatacept in kidney transplantation and its limitations. Expert Rev Clin Immunol. 2019;15:359–367.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.