100
Views
0
CrossRef citations to date
0
Altmetric
Review Articles

Immunosuppressants against acute kidney injury: what to prefer or to avoid?

, , , &
Pages 341-354 | Received 14 Aug 2023, Accepted 09 Mar 2024, Published online: 18 Mar 2024

References

  • Sawhney S, Bell S, Black C, et al. Harmonization of epidemiology of acute kidney injury and acute kidney disease produces comparable findings across four geographic populations. Kidney Int. 2022;101(6):1271–1281. doi:10.1016/j.kint.2022.02.033.
  • Patel M, Heipertz A, Joyce E, et al. Acute kidney disease predicts chronic kidney disease in pediatric non-kidney solid organ transplant patients. Pediatr Transplant. 26(6):e14172. doi:10.1111/petr.14172.
  • Fiorentino M, Bagagli F, Deleonardis A, et al. Acute kidney injury in kidney transplant patients in intensive care unit: from pathogenesis to clinical management. Biomedicines. 2023;11(5):1474. doi:10.3390/biomedicines11051474.
  • Anders H-J, Wilkens L, Schraml B, et al. One concept does not fit all: the immune system in different forms of acute kidney injury. Nephrol Dial Transplant. 2021;36(1):29–38. doi:10.1093/ndt/gfaa056.
  • Griffin BR, Bronsert M, Reece TB, et al. Creatinine elevations from baseline at the time of cardiac surgery are associated with postoperative complications. J Thorac Cardiovasc Surg. 2022;163(4):1378–1387. doi:10.1016/j.jtcvs.2020.03.174.
  • STARSurg Collaborative. Perioperative intravenous contrast administration and the incidence of acute kidney injury after major gastrointestinal surgery: prospective, multicentre cohort study. J British Surgery. 2020;107(8):1023–1032. doi:10.1002/bjs.11453.
  • Boyd A, Brown A, Patel J, et al. Basiliximab with delayed tacrolimus improves short-term renal outcomes post-liver transplantation—a real-world experience. Transplant Proc. 2021;53(5):1541–1547. doi:10.1016/j.transproceed.2021.04.001.
  • Xu Y, Huang Y, Zhang C, et al. Efficacy and safety of rituximab for primary nephrotic syndrome with acute kidney injury: a two-center prospective cohort study. Clin Immunol. 2023;246:109211. doi:10.1016/j.clim.2022.109211.
  • Banas B, Krämer BK, Krüger B, et al. Long-Term kidney transplant outcomes: role of prolonged-release tacrolimus. Transplant Proc. 2020;52(1):102–110. doi:10.1016/j.transproceed.2019.11.003.
  • Curci C, Sallustio F, Serino G, et al. Potential role of effector memory T cells in chronic T cell-mediated kidney graft rejection. Nephrol Dial Transplant. 2016;31(12):2131–2142. doi:10.1093/ndt/gfw245.
  • Yang L, Ning Q, Tang S-S Recent advances and next breakthrough in immunotherapy for cancer treatment. J Immunol Res. 2022;2022:8052212. doi:10.1155/2022/8052212.
  • Miano TA, Flesch JD, Feng R, et al. Early tacrolimus concentrations after lung transplant are predicted by combined clinical and genetic factors and associated with acute kidney injury. Clin Pharmacol Ther. 2020;107(2):462–470. doi:10.1002/cpt.1629.
  • Wang Y, Wei L, Guan Y, et al. Diabetes is a risk factor for high-dose methotrexate-associated AKI in lymphoma patients. Ren Fail. 2020;(1):1111–1117. doi:10.1080/0886022X.2020.1838926.
  • Lee CH, Gwon JG, Jung CW. Effectiveness of thymoglobulin induction therapy in kidney transplant from deceased donor with mild to moderate acute kidney injury. Transplant Proc. 2019;51(8):2611–2614. doi:10.1016/j.transproceed.2019.02.061.
  • Eriksson P, Wallin P, Sjöwall C. Clinical experience of sirolimus regarding efficacy and safety in systemic lupus erythematosus [original research]. Front Pharmacol. 2019;10:82. doi:10.3389/fphar.2019.00082.
  • Grins E, Ederoth P, Bjursten H, et al. Effect of cyclosporine on cytokine production in elective coronary artery bypass grafting: a Sub-analysis of the CiPRICS (cyclosporine to protect renal function in cardiac surgery) study. J Cardiothorac Vasc Anesth. 2022;36(7):1985–1994. doi:10.1053/j.jvca.2021.11.026.
  • Kovarik JM, Mueller EA, Van Bree JB, et al. Cyclosporine pharmacokinetics and variability from a microemulsion formulation—a multicenter investigation in kidney transplant patients. Transplantation. 1994;58(6):663. doi:10.1097/00007890-199409000-00004.
  • Tremblay S, Nigro V, Weinberg J, et al. A Steady-State head-to-Head pharmacokinetic comparison of all FK-506 (tacrolimus) formulations (ASTCOFF): an Open-Label, prospective, randomized, two-Arm, Three-Period crossover study. Am J Transplant. 2017;17(2):432–442. doi:10.1111/ajt.13935.
  • Zwart TC, Bezstarosti S, Achini FR, et al. Population pharmacokinetics of subcutaneous alemtuzumab in kidney transplantation. Br J Clin Pharmacol. 2023;89(4):1471–1485. doi:10.1111/bcp.15608.
  • Grim J, Chládek J, Martínková J. Pharmacokinetics and pharmacodynamics of methotrexate in non-Neoplastic diseases. Clin Pharmacokinet. 2003;42(2):139–151. doi:10.2165/00003088-200342020-00003.
  • Patocka J, Nepovimova E, Kuca K, et al. Cyclosporine A: chemistry and toxicity–a review. Curr Med Chem. 2021;28(20):3925–3934.
  • Ederoth P, Dardashti A, Grins E, et al. Cyclosporine before coronary artery bypass grafting does not prevent postoperative decreases in renal function: a randomized clinical trial. J. Anesthesiology. 2018;128(4):710–717. doi:10.1097/ALN.0000000000002104.
  • Wu Q, Wang X, Nepovimova E, et al. Mechanism of cyclosporine a nephrotoxicity: oxidative stress, autophagy, and signalings. Food Chem Toxicol. 2018;118:889–907. doi:10.1016/j.fct.2018.06.054.
  • Rao S, Bernshteyn M, Sohal R, et al. The management of erythrodermic psoriasis complicated by cyclosporine. Case Rep Dermatol Med. 2020;2020:5215478. doi:10.1155/2020/5215478.
  • Goździk M, Płuciennik A, Zawiasa-Bryszewska A, et al. Acute kidney injury following exposure to calcineurin inhibitors in a patient with idiopathic membranous nephropathy. Drug Saf Case Rep. 2019;6(1):9. doi:10.1007/s40800-019-0103-x.
  • Bacle A, Lhermitte R, Le DB, et al. Unexpected overdose of oral cyclosporine in a kidney transplant patient: a case report. Eur J Hosp Pharm. 2023;30(4):242–244. doi:10.1136/ejhpharm-2021-002730.
  • D’Oria R, Schipani R, Leonardini A, et al. The role of oxidative stress in cardiac disease: from physiological response to injury factor. Oxid Med Cell Longev. 2020;2020:5732929–5732956. doi:10.1155/2020/5732956.
  • Karimzadeh I, Jafari M, Davani-Davari D, et al. The pattern of cyclosporine nephrotoxicity and urinary kidney injury molecule 1 in allogenic hematopoietic stem cell transplant patients. Exp Clin Transplant. 2020;19(6):553–562. doi:10.6002/ect.2020.0123.
  • Ramírez-Bajo MJ, Martín-Ramírez J, Bruno S, et al. Nephroprotective potential of mesenchymal stromal cells and their extracellular vesicles in a murine model of chronic cyclosporine nephrotoxicity. Front Cell Dev Biol. 2020;8:296. doi:10.3389/fcell.2020.00296.
  • Broen JCA, van Laar JM. Mycophenolate mofetil, azathioprine and tacrolimus: mechanisms in rheumatology. Nat Rev Rheumatol. 2020;16(3):167–178. doi:10.1038/s41584-020-0374-8.
  • Sallustio BC. Monitoring intra-cellular tacrolimus concentrations in solid organ transplantation: use of peripheral blood mononuclear cells and graft biopsy tissue. Front Pharmacol. 2021;12:733285. doi:10.3389/fphar.2021.733285.
  • Nakata S, Kakimoto K, Numa K, et al. Risk factors for nephrotoxicity due to tacrolimus therapy for ulcerative colitis. Digestion. 2022;103(5):339–346. doi:10.1159/000524594.
  • Bentata Y. Tacrolimus: 20 years of use in adult kidney transplantation. What we should know about its nephrotoxicity. Artif Organs. 2020;44(2):140–152. doi:10.1111/aor.13551.
  • Claeys E, Vermeire K. Immunosuppressive drugs in organ transplantation to prevent allograft rejection: mode of action and side effects. J Immunological Sci. 2019;3(4):14–21. doi:10.29245/2578-3009/2019/4.1178.
  • Sprangers B, Decoo D, Dive D, et al. Management of adverse renal events related to alemtuzumab treatment in multiple sclerosis: a Belgian consensus. Acta Neurol Belg. 2018;118(2):143–151. doi:10.1007/s13760-017-0864-x.
  • Harrigan AM, West ML, Geldenhuys L, et al. Renal-Limited antiglomerular basement membrane disease related to alemtuzumab: a case report. Can J Kidney Health Dis. 2020;7. doi:10.1177/2054358120962680.
  • Chazin S, Saqqa O, Paramesh A, et al. Skin lesions and acute kidney injury in a kidney transplant recipient. Am J Transplant. 2021;21(6):2298–2300. doi:10.1111/ajt.16519.
  • Liou AA, Skiver BM, Yates E, et al. Acute thrombotic microangiopathy and cortical necrosis following administration of alemtuzumab: a case report. Am J Kidney Dis. 2019;73(5):615–619. doi:10.1053/j.ajkd.2018.09.013.
  • Guthoff M, Berger K, Althaus K, et al. Low-dose alemtuzumab induction in a tailored immunosuppression protocol for sensitized kidney transplant recipients. BMC Nephrol. 2020;21(1):178. doi:10.1186/s12882-020-01767-z.
  • van der Zwan M, Clahsen-Van Groningen MC, van den Hoogen MWF, et al. Comparison of alemtuzumab and anti-thymocyte globulin treatment for acute kidney allograft rejection. Front Immunol. 2020;11:1332. doi:10.3389/fimmu.2020.01332.
  • Saulite I, Guenova E, Hoetzenecker W. Adverse Reactions of Antibody-Therapy for Primary Cutaneous Lymphomas: rituximab, Brentuximab Vedotin, Alemtuzumab, and Mogamulizumab. Curr Probl Dermatol. 2018;53:70–81. doi:10.1159/000478079.
  • Cronstein BN, Aune TM. Methotrexate and its mechanisms of action in inflammatory arthritis. Nat Rev Rheumatol. 2020;16(3):145–154. doi:10.1038/s41584-020-0373-9.
  • Amitai I, Rozovski U, El-Saleh R, et al. Risk factors for high-dose methotrexate associated acute kidney injury in patients with hematological malignancies. Hematol Oncol. 2020;38(4):584–588. doi:10.1002/hon.2759.
  • Jessel S, Austin M, Kluger HM. Mycophenolate as primary treatment for immune checkpoint inhibitor induced acute kidney injury in a patient with concurrent immunotherapy-associated diabetes. Clin Oncol Case Rep 156. Eng.
  • Liang CA, Su YC, Lin SJ, et al. Risk factors for acute kidney injury after high-dose methotrexate therapy: a single-center study and narrative review. Eur J Clin Pharmacol. 2023;79(6):789–800. doi:10.1007/s00228-023-03491-7.
  • Attias P, Melica G, Boutboul D, et al. Epidemiology, risk factors, and outcomes of opportunistic infections after kidney allograft transplantation in the era of modern immunosuppression: a monocentric cohort study. J Clin Med. 2019;8(5):594. doi:10.3390/jcm8050594.
  • Sami FL, Hammo H, Athavale A. Crystalline nephropathy with high-dose methotrexate in a patient with primary CNS lymphoma: a case report. Cureus. 2022;14(6):e26052. doi:10.7759/cureus.26052.
  • Kawano N, Matsumoto K, Takami A, et al. Pharmacokinetics of anti-thymocyte globulin in a patient with severe aplastic anemia treated with allogeneic bone marrow transplantation from a matched unrelated donor. (2432-7026 (Electronic)). eng.
  • Kovarik JM, Nashan B, Neuhaus P, et al. A population pharmacokinetic screen to identify demographic-clinical covariates of basiliximab in liver transplantation. Clin Pharmacol Ther. 2001;69(4):201–209. doi:10.1067/mcp.2001.114887.
  • Zhou H. Clinical pharmacokinetics of etanercept: a fully humanized soluble recombinant tumor necrosis factor receptor fusion protein. J Clin Pharmacol. 2005;45(5):490–497. doi:10.1177/0091270004273321.
  • Klotz U, Teml A, Schwab M. Clinical pharmacokinetics and use of infliximab. Clin Pharmacokinet. 2007;46(8):645–660. doi:10.2165/00003088-200746080-00002.
  • Toapanta N, Jiménez S, Molina-Gómez M, et al. Induction immunosuppression and outcome in kidney transplant recipients with early COVID-19 after transplantation. Clin Kidney J. 2022;15(11):2039–2045. doi:10.1093/ckj/sfac112.
  • Hamed KM, Dighriri IM, Baomar AF, et al. Overview of methotrexate toxicity: a comprehensive literature review. Cureus. 2022;14(9):e29518. doi:10.7759/cureus.29518.
  • Juliette R, Pierre-Joseph R, Gwenaëlle E, et al. LIS1, a glyco-humanized swine polyclonal anti-lymphocyte globulin, as a novel induction treatment in solid organ Transplantation. 2022; bioRxiv. doi:10.1101/2022.09.08.507114.
  • Akl A, Alobaidi S, Aboalsamh G. Living-Donor kidney transplant-associated thrombotic microangiopathy successfully treated with thymoglobulin: a case report. Exp Clin Transplant. 2019;17(Suppl 1):175–177. doi:10.6002/ect.MESOT2018.P51.
  • Ducloux D, Kazory A, Challier B, et al. Long-term toxicity of antithymocyte globulin induction may vary with choice of agent: a single-center retrospective study. Transplantation. 2004;77(7):1029–1033. doi:10.1097/01.tp.0000116442.81259.60.
  • Mo XD, Hong SD, Zhao YL, et al. Basiliximab for steroid-refractory acute graft-versus-host disease: a real-world analysis. Am J Hematol. 2022;97(4):458–469. doi:10.1002/ajh.26475.
  • Seo Y, Geum MJ, Lee KA, et al. Medication utilization analysis of basiliximab as a maintenance immunosuppressant in renal failure patients undergoing lung transplantation. Korean J Clin Pharm. 2020;30(3):149–160. doi:10.24304/kjcp.2020.30.3.149.
  • Kim KD, Lee KW, Kim SJ, et al. Safety and effectiveness of kidney transplantation using a donation after brain death donor with acute kidney injury: a retrospective cohort study. Sci Rep. 2021;11(1):5572. doi:10.1038/s41598-021-84977-1.
  • Kim HE, Paik HC, Jeong SJ, et al. Basiliximab induction with delayed calcineurin inhibitors for high-risk lung transplant candidates. Yonsei Med J. 2021;62(2):164–171. doi:10.3349/ymj.2021.62.2.164.
  • Bennett D, Fossi A, Marchetti L, et al. Postoperative acute kidney injury in lung transplant recipients. Interact Cardiovasc Thorac Surg. 2019;28(6):929–935. doi:10.1093/icvts/ivy355.
  • Lee YJ, Park BS, Park S, et al. Basiliximab-Induced non-cardiogenic pulmonary edema in a kidney transplant patient. Korean J Transplant. 2018;32(3):63–68. doi:10.4285/jkstn.2018.32.3.63.
  • Ammar A, Zafar Ahmed Mahmood H, Shahid Z, et al. Etanercept-associated nephropathy. Cureus. 2019;11(8):e5419. doi:10.7759/cureus.5419.
  • Xu YL, Zou P, Cao XJ. Advances in pharmacotherapy for acute kidney injury. Expert Opin Pharmacother. 2022;23(6):713–726. doi:10.1080/14656566.2022.2050214.
  • Abdelmageed MM, Kefaloyianni E, Arthanarisami A, et al. TNF or EGFR inhibition equally block AKI-to-CKD transition: opportunities for etanercept treatment. Nephrol Dial Transplant. 2023;38(5):1139–1150. doi:10.1093/ndt/gfac290.
  • Miao X, Huang Y, Liu T-T, et al. TNF-α/TNFR1 signaling is required for the full expression of acute and chronic itch in mice via peripheral and Central mechanisms. Neurosci Bull. 2018;34(1):42–53. doi:10.1007/s12264-017-0124-3.
  • McCoy IE, Hsu JY, Bonventre JV, et al. Acute kidney injury associates with long-term increases in plasma TNFR1, TNFR2, and KIM-1: findings from the CRIC study. JASN. 2022;33(6):1173–1181. doi:10.1681/ASN.2021111453.
  • Feist E, Baraliakos X, Behrens F, et al. Effectiveness of etanercept in rheumatoid arthritis: real-world data from the German non-interventional study ADEQUATE with focus on treat-to-Target and patient-reported outcomes. Rheumatol Ther. 2022;9(2):621–635. doi:10.1007/s40744-021-00418-5.
  • Subedi S, Gong Y, Chen Y, et al. Infliximab and biosimilar infliximab in psoriasis: efficacy, loss of efficacy, and adverse events. Drug Des Devel Ther. 2019;13:2491–2502. doi:10.2147/DDDT.S200147.
  • Melsheimer R, Geldhof A, Apaolaza I, et al. Remicade® (infliximab): 20 years of contributions to science and medicine. Biologics. 2019;13:139–178. doi:10.2147/BTT.S207246.
  • Zhou Y, Fan R, Botchway BOA, et al. Infliximab can improve traumatic brain injury by suppressing the tumor necrosis factor alpha pathway. Mol Neurobiol. 2021;58(6):2803–2811. doi:10.1007/s12035-021-02293-1.
  • Lin JS, Mamlouk O, Selamet U, et al. Infliximab for the treatment of patients with checkpoint inhibitor-associated acute tubular interstitial nephritis. Oncoimmunology. 2021;10(1):1877415. doi:10.1080/2162402X.2021.1877415.
  • Ozdemir A, Tumkaya L, Kalcan S, et al. The effects of TNF-alpha inhibitors on carbon tetrachloride-induced nephrotoxicity. Clin Exp Hypertens. 2022;44(3):291–296. doi:10.1080/10641963.2021.2018600.
  • Hsieh C-Y, Tsai T-F. Drug-induced neutropenia during treatment of non-Neoplastic dermatologic diseases: a review. Clin Drug Investig. 2020;40(10):915–926. doi:10.1007/s40261-020-00956-w.
  • Gibson DJ, Ward MG, Rentsch C, et al. Review article: determination of the therapeutic range for therapeutic drug monitoring of adalimumab and infliximab in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2020;51(6):612–628. doi:10.1111/apt.15643.
  • Hasskarl J. Everolimus. Recent Results Cancer Res. 2018;211:101–123. doi:10.1007/978-3-319-91442-8_8.
  • Desai V, Thakkar J, Wanchoo R, et al. 17 Targeted cancer therapies. Onco-Nephrology E-Book. 2019;:154.
  • Kadry Z, Stine JG, Dohi T, et al. Renal protective effect of everolimus in liver transplantation: a prospective randomized Open-Label trial. Transplant Direct. 2021;7(7):e709. doi:10.1097/TXD.0000000000001159.
  • López FN, Alfocea PA, Suazo EJO, et al. Impact of everolimus-based immunosuppression on renal function in liver transplant recipients. Transplantation Proceedings. 2020.
  • Shoji MK, Shishido S, Freitag SK. The use of sirolimus for treatment of orbital lymphatic malformations: a systematic review. Ophthalmic Plast Reconstr Surg. 2020;36(3):215–221. doi:10.1097/IOP.0000000000001518.
  • Newitt VN. UCSF oncologist strives to stem the rise of cancer in Tanzania. Oncology Times. 2023;21:21,22,29.
  • Xu KY, Shameem R, Wu S. Risk of hyperglycemia attributable to everolimus in cancer patients: a meta-analysis. Acta Oncol. 2016; 55(9–10):1196–1203. doi:10.3109/0284186X.2016.1168939.
  • Tedesco‐Silva H, del Carmen Rial M, Cruz Santiago J, et al. Optimizing the clinical utility of sirolimus‐based immunosuppression for kidney transplantation. Clin Transplant. 2019;33(2):e13464. doi:10.1111/ctr.13464.
  • Tsai KF, Li LC, Hsu CN, et al. Effects of conversion from calcineurin inhibitors to sirolimus or everolimus on renal function and possible mechanisms in liver transplant recipients. J Clin Pharmacol. 2019;59(3):326–334. doi:10.1002/jcph.1334.
  • Nguyen LS, Vautier M, Allenbach Y, et al. Sirolimus and mTOR inhibitors: a review of side effects and specific management in solid organ transplantation. Drug Saf. 2019;42(7):813–825. doi:10.1007/s40264-019-00810-9.
  • Paluri RK, Sonpavde G, Morgan C, et al. Renal toxicity with mammalian target of rapamycin inhibitors: a meta-analysis of randomized clinical trials. Oncol Rev. 2019;13(2):455. doi:10.4081/oncol.2019.455.
  • Saggi SJ, Nath S, Culas R, et al. Early experience with methylprednisolone on SARS-CoV-2 infection in the African American population, a retrospective analysis. Clin Med Insights Circ Respir Pulm Med. 2020;14. doi:10.1177/1179548420980699.
  • Garg AX, Chan MTV, Cuerden MS, et al. Effect of methylprednisolone on acute kidney injury in patients undergoing cardiac surgery with a cardiopulmonary bypass pump: a randomized controlled trial. CMAJ. 2019;191(9):E247–E256. doi:10.1503/cmaj.181644.
  • Sakemi T, Yamaguchi M, Fujimi S, et al. Effects of the methylprednisolone pulse therapy on renal function. Am J Nephrol. 1991;11(1):48–53. doi:10.1159/000168272.
  • Fouad D, Shuker E, Farhood M. Renal toxicity of methylprednisolone in male Wistar rats and the potential protective effect by boldine supplementation. J King Saud Unive Sci. 2023;35(1):102381. doi:10.1016/j.jksus.2022.102381.
  • Dumortier J, Cottin J, Lavie C, et al. Methylprednisolone liver toxicity: a new case and a French regional pharmacovigilance survey. Clin Res Hepatol Gastroenterol. 2017;41(4):497–501. doi:10.1016/j.clinre.2017.03.008.
  • Sajgure A, Kulkarni A, Joshi A, et al. Safety and efficacy of mycophenolate in COVID-19: a nonrandomised prospective study in Western India. Lancet Reg Health Southeast Asia. 2023;11:100154. doi:10.1016/j.lansea.2023.100154.
  • Nakano K, Nishizawa M, Fukuda N, et al. Mycophenolate mofetil as a successful treatment of corticosteroid-resistant immune checkpoint inhibitor-induced hepatitis. Oxf Med Case Reports. 2020;2020(4):omaa027. doi:10.1093/omcr/omaa027.
  • Alam A, Van Zyl JS, Hall SA, et al. Impact of risk-stratified mycophenolate dosing in heart transplantation. Clin Transplant. 2021;35(11):e14445. doi:10.1111/ctr.14445.
  • Stoleru G, Bhattacharya S, Patel P, et al. S3308 characterization of mycophenolate mofetil gastrointestinal toxicity and risk factors for severe disease and poor prognosis. Am J Gastroenterol. 2021;116(1):S1363–S1364. doi:10.14309/01.ajg.0000786764.63177.77.
  • Kiang TKL, Ensom MHH. Exposure-toxicity relationships of mycophenolic acid in adult kidney transplant patients. Clin Pharmacokinet. 2019;58(12):1533–1552. doi:10.1007/s40262-019-00802-z.
  • Siewe N, Friedman A. Cancer therapy with immune checkpoint inhibitor and CSF-1 blockade: a mathematical model. J Theor Biol. 2023;556:111297. doi:10.1016/j.jtbi.2022.111297.
  • Pal SK, Albiges L, Tomczak P, et al. Atezolizumab plus cabozantinib versus cabozantinib monotherapy for patients with renal cell carcinoma after progression with previous immune checkpoint inhibitor treatment (CONTACT-03): a multicentre, randomised, open-label, phase 3 trial. Lancet. 402(10397):185–195. doi:10.1016/S0140-6736(23)00922-4.
  • Garcia P, Stedman MR, Merlo S, et al. Kidney function while on immune checkpoint inhibitors: trends in incidence of acute kidney injury, and its causes and outcomes. J Onco-Nephrol. 2023;7(2):57–65. doi:10.1177/23993693231161875.
  • Korman AJ, Garrett-Thomson SC, Lonberg N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat Rev Drug Discov. 2022;21(7):509–528. doi:10.1038/s41573-021-00345-8.
  • Abdelrahim M, Mamlouk O, Lin H, et al. Incidence, predictors, and survival impact of acute kidney injury in patients with melanoma treated with immune checkpoint inhibitors: a 10-year single-institution analysis. OncoImmunology. 2021;10(1):1927313. doi:10.1080/2162402X.2021.1927313.
  • Lemoine M, Dilly B, Curie A, et al. Ipilimumab-induced renal granulomatous arteritis: a case report. BMC Nephrol. 2019;20(1):366. doi:10.1186/s12882-019-1552-2.
  • Person F, Chahoud-Schriefer T, Fehrle W, et al. Severe acute kidney injury due to nivolumab/ipilimumab-induced granulomatosis and fibrinoid vascular necrosis. J Immunother. 2020;43(1):29–31. doi:10.1097/Cji.0000000000000296.
  • Gebauer E, Bechtel-Walz W, Schell C, et al. Development of nivolumab/Ipilimumab-Associated autoimmune nephritis during steroid therapy. Case Rep Nephrol Dial. 2021;11(3):270–274. doi:10.1159/000517502.
  • Biewenga M, van der Kooij MK, Wouters MWJM, et al. Checkpoint inhibitor induced hepatitis and the relation with liver metastasis and outcome in advanced melanoma patients. Hepatol Int. 2021;15(2):510–519. doi:10.1007/s12072-021-10151-4.
  • Tsui E, Gonzales JA. Retinal vasculitis associated with ipilimumab. Ocul Immunol Inflamm. 2020;28(6):868–870. doi:10.1080/09273948.2019.1610460.
  • Domenico M, Diana G, Maria Grazia V, et al. Nivolumab serum concentration in metastatic melanoma patients could be related to outcome and enhanced immune activity: a gene profiling retrospective analysis. J ImmunoTher Cancer. 2022;10(11):e005132. doi:10.1136/jitc-2022-005132.
  • Okawa S, Fujiwara K, Shimonishi A, et al. Rapidly progressive acute kidney injury associated with nivolumab treatment. Case Rep Oncol. 2020;13(1):85–90. doi:10.1159/000505235.
  • Ryuzaki M, Tokuyama H, Uchiyama K, et al. Acute interstitial nephritis with karyomegalic epithelial cells after nivolumab treatment—two case reports. Clinical Med Insights: Case Report. 2019;12:1179547619853647.
  • Cruz-Whitley J, Giehl N, Jen K-Y, et al. Membranoproliferative glomerulonephritis associated with nivolumab therapy. Case Rep Nephrol. 2020;:2638283–2638284. doi:10.1155/2020/2638283.
  • Yamada H, Okajima F, Onda T, et al. New-onset graves’ disease after the initiation of nivolumab therapy for gastric cancer: a case report. BMC Endocr Disord. 2020;20(1):132. doi:10.1186/s12902-020-00613-5.
  • Tanaka R, Maruyama H, Tomidokoro Y, et al. Nivolumab-induced chronic inflammatory demyelinating polyradiculoneuropathy mimicking rapid-onset guillain–barré syndrome: a case report. Jpn J Clin Oncol. 2016;46(9):875–878. doi:10.1093/jjco/hyw090.
  • Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res. 2020;10(3):727–742.
  • Bonilla M, Bijol V, Corona AGDL, et al. A case of immune-complex mediated glomerulonephritis associated with pembrolizumab. J Onco-Nephrol. 2021;6(1–2):79–83. doi:10.1177/23993693211064627.
  • Basnet S, Dhital R, Tharu B. Acute tubulointerstitial nephritis: a case report on rare adverse effect of pembrolizumab. Medicina (Kaunas). 2019;55(5):176. doi:10.3390/medicina55050176.
  • Izzedine H, Mathian A, Champiat S, et al. Renal toxicities associated with pembrolizumab. Clin Kidney J. 2019;12(1):81–88. doi:10.1093/ckj/sfy100.
  • Ishibuchi K, Iwakura T, Kaneko M, et al. Pembrolizumab-associated nephrotic syndrome recovered from transient hemodialysis in a patient with lung cancer. CEN Case Rep. 2020;9(3):215–219. doi:10.1007/s13730-020-00462-0.
  • De Giglio A, Grandinetti V, Aprile M, et al. Patterns of renal toxicity from the combination of pemetrexed and pembrolizumab for advanced nonsquamous non-small-cell lung cancer (NSCLC): a single-center experience. Lung Cancer. 2022;174:91–96. doi:10.1016/j.lungcan.2022.10.007.
  • Dalvin LA, Shields CL, Orloff M, et al. Checkpoint inhibitor immune therapy: systemic indications and ophthalmic side effects. Retina. 2018;38(6):1063–1078. doi:10.1097/IAE.0000000000002181.
  • OConnor C, Jordan E, O’Connell M. Atypical stevens–johnson syndrome caused by pembrolizumab in the treatment of metastatic melanoma: are corticosteroids a safe treatment option? J Dermatol Dermatol Surg. 2020;24/(2):137. doi:10.4103/jdds.jdds_73_20.
  • Zakharova E, Makarova T, Vorobyeva OA. Case of atezolizumab-associated tubulointerstitial nephritis. Journal of Onco-Nephrology. 2022;6(1-2):56–58. doi:10.1177/23993693221080479.
  • Izzedine H, Mathian A, Albiges L, et al. Association of thrombotic microangiopathy with atezolizumab therapy in cancer patients. Eur J Cancer. 2021;149:34–36. doi:10.1016/j.ejca.2021.02.033.
  • Taki T, Oda N, Fujioka Y, et al. Successful treatment of non-small-cell lung cancer with atezolizumab following tubulointerstitial nephritis due to pembrolizumab. Intern Med. 2020;59(13):1639–1642. doi:10.2169/internalmedicine.4260-19.
  • Kawataki M, Nakanishi Y, Yokoyama T, et al. Hypothyroidism as an immune-related adverse event caused by atezolizumab in a patient with muscle spasms: a case report. Respir Med Case Rep. 2022. doi:10.1016/j.rmcr.2022.101585.
  • Syed YY. Durvalumab: first global approval. Drugs. 2017;77(12):1369–1376. doi:10.1007/s40265-017-0782-5.
  • Janagam P. Durvalumab-Induced iga nephropathy in stage iii non-small cell lung cancer after chemoradiation. Chest. 2021;60(4):A1578. doi:10.1016/j.chest.2021.07.1443.
  • Swanson LA, Kassab I, Tsung I, et al. Liver injury during durvalumab-based immunotherapy is associated with poorer patient survival: a retrospective analysis. Front Oncol. 2022;12:984940. doi:10.3389/fonc.2022.984940.

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.