Skip to main content

Advertisement

Log in

Prevention of complications from use of conventional immunosuppressants: a critical review

  • Review
  • Published:
Journal of Nephrology Aims and scope Submit manuscript

Abstract

Synthetic immunosuppressive drugs are largely used in immune-related renal diseases and in kidney transplantation. Most of these drugs have a low therapeutic index (the ratio that compares the blood concentration at which a drug becomes toxic and the concentration at which the drug is effective), which means that the drug should be dosed carefully and the patient monitored frequently. In this review, we consider the categories of synthetic immunosuppressive agents more frequently and conventionally used in clinical nephrology: glucocorticoids, Aalkylating agents (cyclophosphamide, chlorambucil), purine synthesis inhibitors (azathioprine, mycophenolate salts) and calcineurin inhibitors (cyclosporine, tacrolimus). For each category the possible side effects will be reviewed, the general and specific measures to prevent or treat the adverse events will be suggested, and the more common mistakes that may increase the risk of toxicity will be described. However, the efficacy and safety of immunosuppressive agents depend not only on the pharmacologic characteristics of single drugs but can be influenced also by the clinical condition and genetic characteristics of the patient, by the typology and severity of the underlying disease and by the interaction with other concomitantly used drugs.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Granner DK, Wang JC, Yamamoto KR (2015) Regulatory actions of glucocorticoid hormones: from organisms to mechanisms. Adv Exp Med Biol 872:3–31

    Article  CAS  PubMed  Google Scholar 

  2. Panettieri RA, Schaafsma D, Amrani Y, Koziol-White C, Ostrom R, Tliba O (2019) Non-genomic effects of glucocorticoids: an updated view. Trends Pharmacol Sci 40(1):38–49

    Article  CAS  PubMed  Google Scholar 

  3. Tornatore KM, Logue G, Venuto RC, Davis PJ (1997) Cortisol pharmacodynamics after methylprednisolone administration in young and elderly males. J Clin Pharmacol 37(4):304–311

    Article  CAS  PubMed  Google Scholar 

  4. Boonen E, Vervenne H, Meersseman P, Andrew R, Mortier L, Declercq PE, Vanwijngaerden YM, Spriet I, Wouters PJ, Vander Perre S, Langouche L, Vanhorebeek I, Walker BR, Van den Berghe G (2013) Reduced cortisol metabolism during critical illness. N Engl J Med 368(16):1477–1488

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Moroni G, Ponticelli C (2014) Rapidly progressive crescentic glomerulonephritis: early treatment is a must. Autoimmun Rev 13(7):723–729

    Article  PubMed  Google Scholar 

  6. Debono M, Ghobadi C, Rostami-Hodjegan A, Huatan H, Campbell MJ, Newell-Price J, Darzy K, Merke DP, Arlt W, Ross RJ (2009) Modified-release hydrocortisone to provide circadian cortisol profiles. J Clin Endocrinol Metab 94:1548–1554

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Peckett AJ, Wright DC, Riddell MC (2011) The effects of glucocorticoids on adipose tissue lipid metabolism. Metabolism 60:1500–1510

    Article  CAS  PubMed  Google Scholar 

  8. Shen Y, Roh HC, Kumari M, Rosen ED (2017) Adipocyte glucocorticoid receptor is important in lipolysis and insulin resistance due to exogenous steroids, but not insulin resistance caused by high fat feeding. Mol Metab 6(10):1150–1160

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kim HJ, Cha JY, Seok JW, Choi Y, Yoon BK, Choi H, Yu JH, Song SJ, Kim A, Lee H, Kim D, Han JY, Kim JW (2016) Dexras1 links glucocorticoids to insulin-like growth factor-1 signaling in adipogenesis. Sci Rep 6:28648

    Article  PubMed  PubMed Central  Google Scholar 

  10. Berthon BS, MacDonald-Wicks LK, Wood LG (2014) A systematic review of the effect of oral glucocorticoids on energy intake, appetite, and body weight in humans. Nutr Res 34:179–190

    Article  CAS  PubMed  Google Scholar 

  11. Cannon CP, Kumar A (2009) Treatment of overweight and obesity: lifestyle, pharmacologic, and surgical options. Clin Cornerstone 9(4):55–68

    Article  PubMed  Google Scholar 

  12. Liu XX, Zhu XM, Miao Q, Ye HY, Zhang ZY, Li YM (2014) Hyperglycemia induced by glucocorticoids in nondiabetic patients: a meta-analysis. Ann Nutr Metab 65(4):324–332

    Article  CAS  PubMed  Google Scholar 

  13. Burt MG, Willenberg VM, Petersons CJ, Smith MD, Ahern MJ, Stranks SN (2012) Screening for diabetes in patients with inflammatory rheumatological disease administered long-term prednisolone: a cross-sectional study. Rheumatology (Oxford) 51(6):1112–1119

    Article  CAS  Google Scholar 

  14. Trinkley KE, Anderson HD, Nair KV, Malone DC, Saseen JJ (2018) Assessing the incidence of acidosis in patients receiving metformin with and without risk factors for lactic acidosis. Ther Adv Chronic Dis 9(9):179–190

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Donnan K, Segar L (2019) SGLT2 inhibitors and metformin: dual antihyperglycemic therapy and the risk of metabolic acidosis in type 2 diabetes. Eur J Pharmacol 5(846):23–29

    Article  CAS  Google Scholar 

  16. Gunaratne K, Austin E, Wu PE (2018) Unintentional sulfonylurea toxicity due to a drug–drug interaction: a case report. BMC Res Notes 11(1):331

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Villanueva G, Baldwin D (2005) Rosiglitazone therapy of posttransplant diabetes mellitus. Transplantation 80:1402–1405

    Article  CAS  PubMed  Google Scholar 

  18. Shorter DE, Armstrong PW (2000) Adverse effects of corticosteroids on the cardiovascular system. Can J Cardiol 16(4):505–511

    Google Scholar 

  19. Meng X, Chen X, Wu L, Zheng S (2017) The hyperlipidemia caused by overuse of glucocorticoid after liver transplantation and the immune adjustment strategy. J Immunol Res 2017:3149426

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Choi HK, Seeger JD (2005) Glucocorticoid use and serum lipid levels in US adults: the Third National Health and Nutrition Examination Survey. Arthritis Rheum 53:528–535

    Article  CAS  PubMed  Google Scholar 

  21. Liu D, Ahmet A, Ward L, Krishnamoorthy P, Mandelcorn ED, Leigh R, Brown JP, Cohen A, Kim H (2013) A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol 9(1):30

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT Jr, Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM, REDUCE-it investigators (2019) Cardiovascular risk reduction with Icosapent Ethyl Hypertriglyceridemia. N Engl J Med 380(1):11–22

    Article  CAS  PubMed  Google Scholar 

  23. Coelho MC, Dos Santos CV, Vieira Neto L, Gadelha MR (2014) Adverse effects of glucocorticoids: coagulopathy. Eur J Endocrinol 173:M11–M21

    Article  CAS  Google Scholar 

  24. Mantero F, Boscaro M (1992) Glucocorticoid-dependent hypertension. J Steroid Biochem Mol Biol 43(5):409–413

    Article  CAS  PubMed  Google Scholar 

  25. Sato A, Funder JW, Okubo M, Kubota E, Saruta T (1995) Glucocorticoid-induced hypertension in the elderly. Relation to serum calcium and family history of essential hypertension. Am J Hypertens 8(8):823–828

    Article  CAS  PubMed  Google Scholar 

  26. Yang S, Zhang L (2004) Glucocorticoids and vascular reactivity. Curr Vasc Pharmacol 2(1):1–12

    Article  PubMed  Google Scholar 

  27. Slama M, Susic D, Frohlich ED (2002) Prevention of hypertension. Curr Opin Cardiol 17(5):531–536

    Article  PubMed  Google Scholar 

  28. Anagnostis P, Athyros VG, Tziomalos K, Karagiannis A, Mikhailidis DP (2009) Clinical review: the pathogenetic role of cortisol in the metabolic syndrome: a hypothesis. J Clin Endocrinol Meatab 102(5):703–708

    CAS  Google Scholar 

  29. Walker BR (2007) Glucocorticoids and cardiovascular disease. Eur J Endocrinol 157:545–559

    Article  CAS  PubMed  Google Scholar 

  30. Borst O, Schaub M, Walker B, Schmid E, Münzer P, Voelkl J, Alesutan I, Rodríguez JM, Vogel S, Schoenberger T, Metzger K, Rath D, Umbach A, Kuhl D, Müller II, Seizer P, Geisler T, Gawaz M, Lang F (2015) Pivotal role of serum- and glucocorticoid-inducible kinase 1 in vascular inflammation and atherogenesis. Arterioscler Thromb Vasc Biol 35:547–557

    Article  CAS  PubMed  Google Scholar 

  31. Wei L, MacDonald TM, Walker BR (2004) Taking glucocorticoids by prescription is associated with subsequent cardiovascular disease. Ann Intern Med 141:764–770

    Article  PubMed  Google Scholar 

  32. Christiansen CF, Christensen S, Mehnert F, Cummings SR, Chapurlat RD, Sørensen HT (2009) Glucocorticoid use and risk of atrial fibrillation or flutter: a population-based, case–control study. Arch Intern Med 169:1677–1683

    Article  PubMed  Google Scholar 

  33. Moretti R, Torre P, Antonello RM, Zorzon M, Cazzato G (2000) Recurrent atrial fibrillation associated with pulse administration of high doses of methylprednysolone: a possible prophylactic treatment. Eur J Neurol 7(1):130

    Article  CAS  PubMed  Google Scholar 

  34. Stuck AE, Minder CE, Frei FJ (1989) Risk of infectious complications in patients taking glucocorticosteroids. Rev Infect Dis 11(6):954–963

    Article  CAS  PubMed  Google Scholar 

  35. Wolfe F, Caplan L, Michaud K (2006) Treatment for rheumatoid arthritis and the risk of hospitalization for pneumonia: associations with prednisone, disease-modifying antirheumatic drugs, and anti-tumor necrosis factor therapy. Arthritis Rheum 54(2):628–634

    Article  CAS  PubMed  Google Scholar 

  36. Caplan A, Fett N, Rosenbach M, Werth VP, Micheletti RG (2017) Prevention and management of glucocorticoid-induced side effects: A comprehensive review: Infectious complications and vaccination recommendations. J Am Acad Dermatol 76(2):191–198

    Article  PubMed  Google Scholar 

  37. Herrou J, De Lastours V (2018) Predictive factors of pneumocystis pneumonia in patients with rheumatic diseases exposed to prolonged high-dose glucocorticoids. Ann Rheum Dis. https://doi.org/10.1136/annrheumdis-2018-214718. (Epub ahead of print)

  38. Conn HO, Poynard T (1994) Corticosteroids and peptic ulcer: meta-analysis of adverse events during steroid therapy. J Intern Med 236:619–632

    Article  CAS  PubMed  Google Scholar 

  39. Broersen LH, Pereira AM, Jørgensen JO, Dekkers OM (2015) Adrenal insufficiency in corticosteroids use: systematic review and meta-analysis. J Clin Endocrinol Metab 100:2171–2180

    Article  CAS  PubMed  Google Scholar 

  40. West S, Kenedi C (2014) Strategies to prevent the neuropsychiatric side-effects of corticosteroids: a case report and review of the literature. Curr Opin Organ Transpl 19(2):201–208

    Article  CAS  Google Scholar 

  41. Frenkel B, White W, Tuckermann J (2015) Glucocorticoid-induced osteoporosis. Adv Exp Med Biol 872:179–215

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Buckley L, Humphrey MB (2018) Glucocorticoid-induced osteoporosis. N Engl J Med 379(26):2547–2556

    Article  PubMed  Google Scholar 

  43. Ding H, Wang T, Xu D, Cha B, Liu J, Li Y (2015) Dexamethasone-induced apoptosis of osteocytic and osteoblastic cells is mediated by TAK1 activation. Biochem Biophys Res Commun 460:157–163

    Article  CAS  PubMed  Google Scholar 

  44. Allen CS, Yeung JH, Vandermeer B, Homik J (2016) Bisphosphonates for steroid-induced osteoporosis. Cochrane Database Syst Rev 10:Cd001347

  45. Khaleeli AA, Edwards RH, Gohil K, McPhail G, Rennie MJ, Round J, Ross EJ (1983) Corticosteroid myopathy: a clinical and pathological study. Clin Endocrinol (Oxf) 18:155–166

    Article  CAS  Google Scholar 

  46. Pereira RM, Freire de Carvalho J (2011) Glucocorticoid-induced myopathy. Joint Bone Spine 78:41–44

    Article  CAS  PubMed  Google Scholar 

  47. Carli L, Tani C, Querci F, Della Rossa A, Vagnani S, Baldini C, Talarico R, d’Ascanio A, Neri R, Tavoni AG, Bombardieri S, Mosca M (2013) Analysis of the prevalence of cataracts and glaucoma in systemic lupus erythematosus and evaluation of the rheumatologists’ practice for the monitoring of glucocorticoid eye toxicity. Clin Rheumatol 32(7):1071–1073

    Article  CAS  PubMed  Google Scholar 

  48. Richmond E, Rogol AD (2016) Treatment of growth hormone deficiency in children, adolescents and at the transitional age. Best Pract Res Clin Endocrinol Metab 30(6):749–755

    Article  CAS  PubMed  Google Scholar 

  49. Luo X, Hou L, Liang L, Dong G, Shen S, Zhao Z, Gong CX, Li Y, Du ML, Su Z, Du H, Yan C (2017) Long-acting PEGylated recombinant human growth hormone (Jintrolong) for children with growth hormone deficiency: phase II and phase III multicenter, randomized studies. Eur J Endocrinol 177(2):195–205

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhai JL, Ge N, Zhen Y, Zhao Q, Liu C (2016) Corticosteroids significantly increase serum cystatin c concentration without affecting renal function in symptomatic heart failure. Clin Lab 62(1–2):203–207

    CAS  PubMed  Google Scholar 

  51. Reichert LJ, Koene RA, Wetzels JF (1999) Acute haemodynamic and proteinuric effects of prednisolone in patients with a nephrotic syndrome. Nephrol Dial Transpl 14(1):91

    Article  CAS  Google Scholar 

  52. Isidori AM, Venneri MA, Graziadio C, Simeoli C, Fiore D, Hasenmajer V, Sbardella E, Gianfrilli D, Pozza C, Pasqualetti P, Morrone S, Santoni A, Naro F, Colao A, Pivonello R, Lenzi A (2018) Effect of once-daily, modified-release hydrocortisone versus standard glucocorticoid therapy on metabolism and innate immunity in patients with adrenal insufficiency (DREAM): a single-blind, randomised controlled trial. Lancet Diab Endocrinol 6(3):173–185

    Article  CAS  Google Scholar 

  53. Ponticelli C, Locatelli F (2018) Glucocorticoids in the treatment of glomerular diseases: pitfalls and pearls. Clin J Am Soc Nephrol 13(5):815–822

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Liu J, Li X, Fan L, Yang J, Wang J, Sun J, Wang Z (2019) Proton pump inhibitors therapy and risk of bone diseases: an update meta-analysis. Life Sci 218:213–223

    Article  CAS  PubMed  Google Scholar 

  55. Helsby NA, Hui CY, Goldthorpe MA, Coller JK, Soh MC, Gow PJ, De Zoysa JZ, Tingle MD (2010) The combined impact of CYP2C19 and CYP2B6 pharmacogenetics on cyclophosphamide bioactivation. Br J Clin Pharmacol 70(6):844–853

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ponticelli C, Escoli R, Moroni G (2018) Does cyclophosphamide still play a role in glomerular diseases? Autoimmun Rev 17(10):1022–1027

    Article  CAS  PubMed  Google Scholar 

  57. Martin F, Lauwerys B, Lefèbvre C, Devogelaer JP, Houssiau FA (1997) Side-effects of intravenous cyclophosphamide pulse therapy. Lupus 6(3):254–257

    Article  CAS  PubMed  Google Scholar 

  58. Woytala PJ, Morgiel E, Łuczak A, Czesak-Woytala K, Wiland P (2016) The safety of intravenous cyclophosphamide in the treatment of rheumatic diseases. Adv Clin Exp Med 25(3):479–484

    Article  PubMed  Google Scholar 

  59. Herbert LA, Rovin BH (2010) Oral cyclophosphamide is on the verge of extinction as therapy for severe autoimmune diseases (especially lupus): should nephrologists care? Nephron Clin Pract 117(1):c8–c14

    Article  CAS  Google Scholar 

  60. Appel GB, Contreras G, Dooley MA, Ginzler EM, Isenberg D, Jayne D, Li LS, Mysler E, Sánchez-Guerrero J, Solomons N, Wofsy D; Aspreva Lupus Management Study Group (2009) Mycophenolate mofetil versus cyclophosphamide for induction treatment of lupus nephritis. J Am Soc Nephrol 20(5):1103–1112

    Article  CAS  Google Scholar 

  61. Faurschou M, Sorensen IJ, Mellemkjaer L, Loft AG, Thomsen BS, Tvede N, Baslund B (2008) Malignancies in Wegener’s granulomatosis: incidence and relation to cyclophosphamide therapy in a cohort of 293 patients. J Rheumatol 35:100–105

    CAS  PubMed  Google Scholar 

  62. Yilmaz N, Emmungil H, Gucenmez S, Ozen G, Yildiz F, Balkarli A, Kimyon G, Coskun BN, Dogan I, Pamuk ON, Yasar S, Cetin GY, Yazici A, Ergulu Esmen S, Cagatay Y, Yilmaz S, Cefle A, Sayarlioglu M, Kasifoglu T, Karadag O, Pehlivan Y, Dalkilic E, Kisacik B, Cobankara V, Erken E, Direskeneli H, Aksu K, Yavuz S (2015) Incidence of cyclophosphamide-induced urotoxicity and protective effect of mesna in rheumatic diseases. J Rheumatol 42:1664–1666

    Article  CAS  Google Scholar 

  63. Kim S, Choi HJ, Jo CH, Park JS, Kwon TH, Kim GH (2015) Cyclophosphamide-induced vasopressin-independent activation of aquaporin-2 in the rat kidney. Am J Physiol Renal Physiol 309:F474–F483

    Article  CAS  PubMed  Google Scholar 

  64. Esposito P, Domenech MV, Serpieri N, Calatroni M, Massa I, Avella A, La Porta E, Estienne L, Caramella E, Rampino T (2017) Severe cyclophosphamide-related hyponatremia in a patient with acute glomerulonephritis. World J Nephrol 6:217–220

    Article  PubMed  PubMed Central  Google Scholar 

  65. Sterns RH, Silver SM (2006) Brain volume regulation in response to hypo-osmolality and its correction. Am J Med 119(Suppl 1):S12–S16

    Article  CAS  PubMed  Google Scholar 

  66. Spasovski G, Vanholder R, Allolio B, Annane D, Ball S, Bichet D, Decaux G, Fenske W, Hoorn EJ, Ichai C, Joannidis M, Soupart A, Zietse R, Haller M, van der Veer S, Van Biesen W, Nagler E; Hyponatraemia Guideline Development Group (2014) Clinical practice guideline on diagnosis and treatment of hyponatraemia. Nephrol Dial Transpl 29[Suppl 2]: i1–i39

  67. Appenzeller S, Blatyta PF, Costallat LT (2008) Ovarian failure in SLE patients using pulse cyclophosphamide: comparison of different regimes. Rheumatol Int 28:567–571

    Article  CAS  PubMed  Google Scholar 

  68. Tamirou F, Husson SN, Gruson D, Debiève F, Lauwerys BR, Houssiau FA (2017) Brief report: the euro-lupus low-dose intravenous cyclophosphamide regimen does not impact the ovarian reserve, as measured by serum levels of anti-müllerian hormone. Arthritis Rheumatol 69:1267–1271

    Article  CAS  PubMed  Google Scholar 

  69. Latta K, von Schnakenburg C, Ehrich JH (2001) A meta-analysis of cytotoxic treatment for frequently relapsing nephrotic syndrome in children. Ped Nephrol 16:271–282

    Article  CAS  Google Scholar 

  70. Meistrich ML (2009) Male gonadal toxicity. Pediatr Blood Cancer 53:261–266

    Article  PubMed  PubMed Central  Google Scholar 

  71. Ghobadi E, Moloudizargari M, Asghari MH, Abdollahi M (2017) The mechanisms of cyclophosphamide-induced testicular toxicity and the protective agents. Expert Opin Drug Metab Toxicol 13:525–536

    Article  CAS  PubMed  Google Scholar 

  72. Koyama H, WadaT Nishikawa Y, Iwanaga T, Aoki Y (1977) Cyclophosphamide-induced ovarian failure and its therapeutic significance in patients with breast cancer. Cancer 39(4):1403–1409

    Article  CAS  PubMed  Google Scholar 

  73. Hasky N, Uri-Belapolsky S, Goldberg K, Miller I, Grossman H, Stemmer SM, Ben-Aharon I, Shalgi R (2015) Gonadotrophin-releasing hormone agonists for fertility preservation: unraveling the enigma? Hum Reprod 30:1089–1101

    Article  CAS  PubMed  Google Scholar 

  74. Nishikawa T, Miyahara E, Kurauchi K, Watanabe E, Ikawa K, Asaba K, Tanabe T, Okamoto Y, Kawano Y (2015) Mechanisms of Fatal cardiotoxicity following high-dose cyclophosphamide therapy and a method for its prevention. PLoS ONE 10:e0131394

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Usui Y, Aida H, Kimula Y, Miura H, Aoyagi Y, Nakayama M, Takayama S (1992) A case of cyclophosphamide-induced interstitial pneumonitis diagnosed by bronchoalveolar lavage. Respiration 59(2):125–128

    Article  CAS  PubMed  Google Scholar 

  76. Barnes H, Holland AE, Westall GP, Goh NS, Glaspole IN (2018) Cyclophosphamide for connective tissue disease-associated interstitial lung disease. Cochrane Database Syst Rev 1:CD010908

  77. Ochoa R, Bejarano PA, Glück S, Montero AJ (2012) Pneumonitis and pulmonary fibrosis in a patient receiving adjuvant docetaxel and cyclophosphamide for stage 3 breast cancer: a case report and literature review. J Med Case Rep 6:413

  78. Shang W, Ning Y, Xu X et al (2015) Incidence of cancer in ANCA-associated vasculitis: a meta-analysis of observational studies. PLoS One 10:e0126016

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Hemminki K, Liu X, Ji J, Försti A (2016) Origin of B-cell neoplasms in autoimmune disease. PLoS One 11:e0158360

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Ponticelli C, Escoli R, Moroni G (2018) Fetal toxicity of immunosuppressive drugs in pregnancy. J Clin Med 17(10):1022–1027

    CAS  Google Scholar 

  81. Staatz CE, Tett SE (2007) Clinical pharmacokinetics and pharmacodynamics of mycophenolate in solid organ transplant recipients. Clin Pharmacokinet 46(1):13–58

    Article  CAS  PubMed  Google Scholar 

  82. Zhang D, Chow DS (2017) Clinical Pharmacokinetics of mycophenolic acid in hematopoietic stem cell transplantation recipients. Eur J Drug Metab Pharmacokinet 42(2):183–189

    Article  CAS  PubMed  Google Scholar 

  83. de Jong DJ, Goullet M, Naber TH (2004) Side effects of azathioprine in patients with Crohn’s disease. Eur J Gastroenterol Hepatol 16(2):207–212

    Article  PubMed  Google Scholar 

  84. Moran GW, Dubeau MF, Kaplan GG, Yang H, Eksteen B, Ghosh S, Panaccione R (2015) Clinical predictors of thiopurine-related adverse events in Crohn’s disease. World J Gastroenterol 21(25):7795–7804

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Regueiro M, Mardini H (2002) Determination of thiopurine methyltransferase genotype or phenotype optimizes initial dosing of azathioprine for the treatment of Crohn’s disease. J Clin Gastroenterol 35(3):240–244

    Article  CAS  PubMed  Google Scholar 

  86. Connell WR, Kamm MA, Ritchie JK, Lennard-Jones JE (1993) Bone marrow toxicity caused by azathioprine in inflammatory bowel disease: 27 years of experience. Gut 34(8):1081–1085

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Corominas H, Domènech M, González-Juan D, González-Suárez B, Díaz C, Pujol J, Vázquez G, Baiget M (2000) Aplasia after azathioprine administration: role of the thiopurine methyltransferase genetic polymorphism. Med Clin (Barc) 115(8):299–301

    Article  CAS  Google Scholar 

  88. Pruijt JF, Haanen JB, Hollander AA, den Ottolander GJ (1996) Azathioprine-induced pure red-cell aplasia. Nephrol Dial Transpl 11(7):1371–1373

    Article  CAS  Google Scholar 

  89. Ohmann EL, Burckart GJ, Brooks MM, Chen Y, Pravica V, Girnita DM, Zeevi A, Webber SA (2010) Genetic polymorphisms influence mycophenolate mofetil-related adverse events in pediatric heart transplant patients. J Heart Lung Transpl 29(5):509–516

    Article  Google Scholar 

  90. Varnell CD, Fukuda T, Kirby CL, Martin LJ, Warshaw BL, Patel HP, Chand DH, Barletta GM, Van Why SK, VanDe Voorde RG, Weaver DJ, Wilson A, Verghese PS, Vinks AA, Greenbaum LA, Goebel J, Hooper DK (2010) Mycophenolate mofetil-related leukopenia in children and young adults following kidney transplantation: influence of genes and drugs. Pediatr Transpl 21(7). https://doi.org/10.1111/petr.13033

    Article  CAS  Google Scholar 

  91. Rerolle JP, Szelag JC, Le Meur Y (2007) Unexpected rate of severe leucopenia with the association of mycophenolate mofetil and valganciclovir in kidney transplant recipients. Nephrol Dial Transpl 22(2):671–672

    Article  CAS  Google Scholar 

  92. Hardinger KL, Brennan DC, Lowell J, Schnitzler MA (2004) Long-term outcome of gastrointestinal complications in renal transplant patients treated with mycophenolate mofetil. Transpl Int 17(10):609–616

    Article  PubMed  Google Scholar 

  93. Manger B, Hiepe F, Schneider M, Worm M, Wimmer P, Paulus EM, Schwarting A (2015) Impact of switching from mycophenolate mofetil to enteric-coated mycophenolate sodium on gastrointestinal side effects in patients with autoimmune disease: a Phase III, open-label, single-arm, multicenter study. Clin Exp Gastroenterol 8:205–213

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Siramolpiwat S, Sakonlaya D (2017) Clinical and histologic features of Azathioprine-induced hepatotoxicity. Scand J Gastroenterol 52:876–880

    Article  CAS  PubMed  Google Scholar 

  95. Musumba CO (2013) Review article: the association between nodular regenerative hyperplasia, inflammatory bowel disease and thiopurine therapy. Aliment Pharmacol Ther 38:1025–37

    Article  CAS  Google Scholar 

  96. Hantash B, Fiorentino D (2006) Liver enzyme abnormalities in patients with atopic dermatitis treated with mycophenolate mofetil. Arch Dermatol 142:109–110

    PubMed  Google Scholar 

  97. Loupy A, Anglichaeu D, Mamzer-Bruneel MF, Martinez F, Thervet E, Legendre C, Serpaggi J, Pol S (2006) Mycophenolate sodium-induced hepatotoxicity: first report. Transplantation 82(4):581

    Article  CAS  PubMed  Google Scholar 

  98. Kawasaki Y (2009) Mizoribine: a new approach in the treatment of renal diseases. Clin Dev Immunol 2009:681482

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Floyd A, Pedersen L, Nielsen GL, Thorlacius-Ussing O, Sorensen HT (2003) Risk of acute pancreatitis in users of azathioprine: a population-based case-control study. Am J Gastroenterol 98:1305–1308

    CAS  PubMed  Google Scholar 

  100. Elli A, Aroldi A, Montagnino G, Tarantino A, Ponticelli C (1998) Mycophenolate mofetil and cough. Transplantation 66(3):409

    Article  CAS  PubMed  Google Scholar 

  101. Maes B, Oellerich M, Ceuppens JL, Armstrong VW, Evenepoel P, Kuypers D, Messiaen T, Shipkova M, Wieland E, Vanrenterghem Y (2002) A new acute inflammatory syndrome related to the introduction of mycophenolate mofetil in patients with Wegener’s granulomatosis. Nephrol Dial Transpl 17:923–926

    Article  Google Scholar 

  102. Moreso F, Seron D, Morales JM, Cruzado JM, Gil-Vernet S, Pérez JL, Fulladosa X, Andrés A, Grinyó JM (1998) Incidence of leukopenia and cytomegalovirus disease in kidney transplants treated with mycophenolate mofetil combined with low cyclosporine and steroid doses. Clin Transpl 12:198–205

    CAS  Google Scholar 

  103. Borni-Duval C, Caillard S, Olagne J, Perrin P, Braun-Parvez L, Heibel F, Moulin B (2013) Risk factors for BK virus infection in the era of therapeutic drug monitoring. Transplantation 95(12):1498–1505

    Article  CAS  PubMed  Google Scholar 

  104. Aleissa M, Nicol P, Godeau M, Tournier E, de Bellissen F, Robic MA, Livideanu CB, Mazereeuw-Hautier J, Paul C (2017) Azathioprine hypersensitivity syndrome: two cases of febrile neutrophilic dermatosis induced by azathioprine. Case Rep Dermatol 9:6–11

    Article  PubMed  PubMed Central  Google Scholar 

  105. Volgger B, Marth C, Zeimet A, Müller-Holzner E, Ruth N, Dapunt O (1997) Fulminant course of a microinvasive vulvar carcinoma in an immunosuppressed woman. Gynecol Oncol 65(1):177–179

    Article  CAS  PubMed  Google Scholar 

  106. Weaver JL (2012) Establishing the carcinogenic risk of immunomodulatory drugs. Toxicol Pathol 40:267–271

    Article  CAS  PubMed  Google Scholar 

  107. Perez HC, Benavides X, Perez JS, Pabon MA, Tschen J, Maradei-Anaya SJ, Lopez L, Lozano E (2017) Basic aspects of the pathogenesis and prevention of non-melanoma skin cancer in solid organ transplant recipients: a review. Int J Dermatol 56(4):370–378

    Article  CAS  PubMed  Google Scholar 

  108. Casetta I, Iuliano G, Filippini G (2007) Azathioprine for multiple sclerosis. Cochrane Database Syst Rev 4:CD003982

  109. Dun B, Sharma A, Teng Y, Liu H, Purohit S, Xu H, Zeng L, She JX (2013) Mycophenolic acid inhibits migration and invasion of gastric cancer cells via multiple molecular pathways. PLoS One 8:e81702

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Ponticelli C, Moroni G (2015) Immunosuppression in pregnant women with systemic lupus erythematosus. Expert Rev Clin Immunol 11(5):549–552

    Article  CAS  PubMed  Google Scholar 

  111. O’Keefe SJ, Tamura J, Kincaid RL, Tocci MJ, O’Neill EA (1992) FK-506- and CsA-sensitive activation of the interleukin-2 promoter by calcineurin. Nature 357:692–694

    Article  PubMed  Google Scholar 

  112. Staatz CE, Goodman LK, Tett SE (2010) Effect of CYP3A and ABCB1 single nucleotide polymorphisms on the pharmacokinetics and pharmacodynamics of calcineurin inhibitors: part I. Clin Pharmacokinet 49(3):141–175

    Article  CAS  PubMed  Google Scholar 

  113. Vanhove T, Annaert P, Kuypers DR (2016) Clinical determinants of calcineurin inhibitor disposition: a mechanistic review. Drug Metab Rev 48(1):88–112

    Article  CAS  PubMed  Google Scholar 

  114. Vanhove T, Remijsen Q, Kuypers D, Gillard P (2017) Drug-drug interactions between immunosuppressants and antidiabetic drugs in the treatment of post-transplant diabetes mellitus. Transpl Rev (Orlando) 31(2):69–77

    Article  Google Scholar 

  115. Cattran DC, Alexopoulos E, Heering P, Hoyer PF, Johnston A, Meyrier A, Ponticelli C, Saito T, Choukroun G, Nachman P, Praga M, Yoshikawa N (2007) Cyclosporin in idiopathic glomerular disease associated with the nephrotic syndrome: workshop recommendations. Kidney Int 72(12):1429–1447

    Article  CAS  PubMed  Google Scholar 

  116. Naesens M, Kuypers DR, Sarwal M (2009) Calcineurin inhibitor nephrotoxicity. Clin J Am Soc Nephrol 4(2):481–508

    Article  CAS  PubMed  Google Scholar 

  117. Burdmann EA, Andoh TF, Yu L, Bennett WM (2003) Cyclosporine nephrotoxicity. Semin Nephrol 23:465–476

    Article  CAS  PubMed  Google Scholar 

  118. Griffiths MH, Crowe AV, Papadaki L, Banner NR, Yacoub MH, Thompson FD, Neild GH (1996) Cyclosporin nephrotoxicity in heart and lung transplant patients. QJM 89(10):751–763

    Article  CAS  PubMed  Google Scholar 

  119. Bennett WM, DeMattos A, Meyer MM, Andoh T, Barry JM (1996) Chronic cyclosporine nephropathy: the Achilles’ heel of immunosuppressive therapy. Kidney Int 50(4):1089–1100

    Article  CAS  PubMed  Google Scholar 

  120. Issa N, Kukla A, Ibrahim HN (2013) Calcineurin inhibitor nephrotoxicity: a review and perspective of the evidence. Am J Nephrol 37(6):602–612

    Article  CAS  PubMed  Google Scholar 

  121. Curtis JJ (2002) Hypertensinogenic mechanism of the calcineurin inhibitors. Curr Hypertens Rep 4(5):377–380

    Article  PubMed  Google Scholar 

  122. Blankenstein KI, Borschewski A, Labes R et al (2017) Calcineurin inhibitor cyclosporine A activates renal Na-K-Cl cotransporters via local and systemic mechanisms. Am J Physiol Renal Physiol 312:F489–F501

    Article  CAS  PubMed  Google Scholar 

  123. Robert N, Wong GW, Wright JM (2010) Effect of cyclosporine on blood pressure. Cochrane Database Syst Rev 1:CD007893

  124. Grześk E, Malinowski B, Wiciński M et al (2016) Cyclosporine-A, but not tacrolimus significantly increases reactivity of vascular smooth muscle cells. Pharmacol Rep 68:201–205

    Article  PubMed  CAS  Google Scholar 

  125. Klein IH, Abrahams A, van Ede T, Hene RJ, Koomans HA, Ligtenberg G (2002) Different effects of tacrolimus andcyclosporine on renal hemodynamic and blood pressure in healthy subjects. Transplantation 73:732

    Article  CAS  PubMed  Google Scholar 

  126. Ponticelli C, Cucchiari D (2017) Renin-angiotensin system inhibitors in kidney transplantation: a benefit-risk assessment. J Nephrol 30(2):155–157

    Article  CAS  PubMed  Google Scholar 

  127. Kuypers DR, Neumayer HH, Fritsche L, Budde K, Rodicio JL, Vanrenterghem Y (2004) Lacidipine Study Group. Calcium channel blockade and preservation of renal graft function in cyclosporine-treated recipients: a prospective randomized placebo-controlled 2-year study. Transplantation 78(8):1204–1211

    Article  CAS  PubMed  Google Scholar 

  128. Øzbay LA, Smidt K, Mortensen DM Carstens J, Jørgensen KA, Rungby J (2011) Cyclosporin and tacrolimus impair insulin secretion and transcriptional regulation in INS-1E beta-cells. Br J Pharmacol 162: 136–146

    Article  CAS  Google Scholar 

  129. Chakkera HA, Kudva Y, Kaplan B (2017) Calcineurin inhibitors: pharmacologic mechanisms impacting both insulin resistance and insulin secretion leading to glucose dysregulation and diabetes mellitus. Clin Pharmacol Ther 101:114–120

    Article  CAS  PubMed  Google Scholar 

  130. Li Z, Sun F, Zhang Y, Chen H, He N, Chen H, Song P, Wang Y, Yan S, Zheng S (2015) Tacrolimus induces insulin resistance and increases the glucose absorption in the jejunum: a potential mechanism of the diabetogenic effects. PLoS One 10:e0143405

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Triñanes J, Rodriguez-Rodriguez AE, Brito-Casillas Y, Wagner A, De Vries APJ, Cuesto G, Acebes A, Salido E, Torres A, Porrini E (2017) Deciphering tacrolimus-induced toxicity in pancreatic β cells. Am J Transpl 17(11):2829–2840

    Article  CAS  Google Scholar 

  132. Rao SR, Sundararajan S, Subbarayan R, Murugan Girija D (2017) Cyclosporine-A induces endoplasmic reticulum stress and influences pro-apoptotic factors in human gingival fibroblasts. Mol Cell Biochem 429:179–185

    Article  CAS  PubMed  Google Scholar 

  133. Nash MM, Zalztman JS (1998) Efficacy of azithromycin in the treatment of cyclosporine-induced gingival hyperplasia in renal transplant recipients. Transplantation 65(12):1611–1615

    Article  CAS  PubMed  Google Scholar 

  134. Hirsch R, Deng H, Laohachai MN (2012) Azithromycin in periodontal treatment: more than an antibiotic. J Periodontal Res 47(2):137–148

    Article  PubMed  CAS  Google Scholar 

  135. Sen A, Callisen H, Libricz S, Patel B (2019) Complications of solid organ transplantation: cardiovascular, neurologic, renal, and gastrointestinal. Crit Care Clin 35(1):169–186

    Article  PubMed  Google Scholar 

  136. Ponticelli C, Campise R (2005) Neurological complications in kidney transplant recipients. J Nephrol 18:521–528

    PubMed  Google Scholar 

  137. Song T, Rao Z, Tan Q, Qiu Y, Liu J, Huang Z, Wang X, Lin T (2016) Calcineurin inhibitors associated posterior reversible encephalopathy syndrome in solid organ transplantation: report of 2 cases and literature review. Medicine (Baltimore) 95:e3173

    Article  Google Scholar 

  138. Kockx M, Glaros E, Leung B, Ng TW, Berbée JF, Deswaerte V, Nawara D, Quinn C, Rye KA, Jessup W, Rensen PC, Meikle PJ, Kritharides L (2016) Low-density lipoprotein receptor-dependent and low-density lipoprotein receptor-independent mechanisms of cyclosporin a-induced dyslipidemia. Arterioscler Thromb Vasc Biol 36(7):1338–1349

    Article  CAS  PubMed  Google Scholar 

  139. Suk HY, Zhou C, Yang TT, Zhu H, Yu RY, Olabisi O, Yang X, Brancho D, Kim JY, Scherer PE, Frank PG, Lisanti MP, Calvert JW, Lefer DJ, Molkentin JD, Ghigo A, Hirsch E, Jin J, Chow CW (2013) Ablation of calcineurin Aβ reveals hyperlipidemia and signaling cross-talks with phosphodiesterases. J Biol Chem 288:3477–3488

    Article  CAS  PubMed  Google Scholar 

  140. Fuhrmann A, Lopes P, Sereno J, Pedro J, Espinoza DO, Pereira MJ, Reis F, Eriksson JW, Carvalho E (2014) Molecular mechanisms underlying the effects of cyclosporin A and sirolimus on glucose and lipid metabolism in liver, skeletal muscle and adipose tissue in an in vivo rat model. Biochem Pharmacol 88:216–228

    Article  CAS  PubMed  Google Scholar 

  141. Brocks DR, Chaudhary HR, Ben-Eltriki M, Elsherbiny ME, El-Kadi AO (2014) Effects of serum lipoproteins on cyclosporine. A cellular uptake and renal toxicity in vitro. Can J Physiol Pharmacol 92:140–148

    Article  CAS  PubMed  Google Scholar 

  142. Clarke H, Ryan MP (1999) Cyclosporine A-induced alterations in magnesium homeostasis in the rat. Life Sci 64(15):1295–1306

    Article  CAS  PubMed  Google Scholar 

  143. Lea JP, Sands JM, McMahon SJ, Tumlin J (1994) Evidence that the inhibition of Na+/K(+)-ATPase activity by FK506 involves calcineurin. Kidney Int 46:647–652

    Article  CAS  PubMed  Google Scholar 

  144. Osorio JM, Bravo J, Pérez A, Ferreyra C, Osuna A (2010) Magnesemia in renal transplant recipients: relation with immunosuppression and posttransplant diabetes. Transpl Proc 42(8):2910–2913

    Article  CAS  PubMed  Google Scholar 

  145. Stamp L, Searle M, O’Donnell J, Chapman P (2005) Gout in solid organ transplantation: a challenging clinical problem. Drugs 65(18):2593–2611

    Article  CAS  PubMed  Google Scholar 

  146. Piccoli GB, Cabiddu G, Attini R, Gerbino M, Todeschini P, Perrino ML, Mone AM, Piredda GB, Gnappi E, Caputo F et al (2016) Pregnancy outcomes after kidney graft in Italy: are the changes over time the result of different therapies or of different policies? A nationwide survey (1978–2013). Nephrol Dial Transpl 31:1957–1965

    Article  Google Scholar 

  147. Thiagarajan KM, Arakali SR, Mealey KJ, Cardonick EH, Gaughan WJ, Davison JM, Moritz MJ, Armenti VT (2013) Safety considerations: breastfeeding after transplant. Prog. Transpl 23:137–146

    Article  Google Scholar 

  148. Euvrard S,Morelon E, Rostaing L, Goffin E, Brocard A, Tromme I, Broeders N, del Marmol V, Chatelet V, Dompmartin A, Kessler M, Serra AL, Hofbauer GF, Pouteil-Noble C, Campistol JM, Kanitakis J, Roux AS, Decullier E, Dantal J; TUMORAPA Study Group (2014). Sirolimus and secondary skin-cancer prevention in kidney transplantation. N Engl J Med 367(4):329–339

  149. Liacini A, Seamone ME, Muruve DA, Tibbles LA (2010) Anti-BK virus mechanisms of sirolimus and leflunomide alone and in combination: toward a new therapy for BK virus infection. Transplantation 90:1450–1457

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudio Ponticelli.

Ethics declarations

Conflict of interest

Dr Ponticelli has no conflict of interest to disclose. Dr. Glassock is a compensated consultant to Bristol-Myers Squibb, Genentech, Chemocentryx, Mallinckrodt, Apellis, Ionis, Achillion, Omeros. He also receives a stipend from Wolters -Kluwer (UpToDate) and Karger Publications (American Journal of Nephrology and Nephrology Viewpoints) for Editorial Services. He owns stock in REATA, Inc. (Bardoxolone).

Ethical approval

This article does not contain any studies with human participants performed by any of the authors.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ponticelli, C., Glassock, R.J. Prevention of complications from use of conventional immunosuppressants: a critical review. J Nephrol 32, 851–870 (2019). https://doi.org/10.1007/s40620-019-00602-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40620-019-00602-5

Keywords

Navigation