81
Views
0
CrossRef citations to date
0
Altmetric
Review

Pharmacotherapy considerations in patients who develop acute kidney injury during anti-cancer therapy

, , , , , & show all
Received 18 Feb 2024, Accepted 18 Apr 2024, Published online: 25 Apr 2024

References

  • Gupta S, Gudsoorkar P, Jhaveri KD. Acute kidney injury in critically Ill patients with cancer. Clin J Am Soc Nephrol. 2022;17(9):1385–1398.
  • Joseph A, Lafarge A, Azoulay E, et al. Acute kidney injury in cancer immunotherapy recipients. Cells. 2022;11(24):3991. doi: 10.3390/cells11243991
  • Tang J, Yang N, Pan S, et al. The renal damage and mechanisms relevant to antitumoral drugs. Front Oncol. 2023;13:1331671. doi: 10.3389/fonc.2023.1331671.
  • Rosner MH, Jhaveri KD, McMahon BA, et al. Onconephrology: the intersections between the kidney and cancer. CA Cancer J Clin. 2021;71(1):47–77. doi: 10.3322/caac.21636.
  • Navari RM, Aapro M, Longo DL. Antiemetic prophylaxis for chemotherapy-induced nausea and vomiting. Longo DL, editor. N Engl J Med. 2016;374(14):1356–1367. doi: 10.1056/NEJMra1515442
  • Nielsen DL, Juhl CB, Chen IM, et al. Immune checkpoint inhibitor–induced diarrhea and colitis: incidence and management. A systematic review and meta-analysis. Cancer Treat Rev. 2022;109:102440. doi: 10.1016/j.ctrv.2022.102440
  • Guise TA, Wysolmerski JJ, Solomon CG. Cancer-Associated Hypercalcemia. Solomon CG, editor. N Engl J Med. 2022;386(15):1443–1451. doi: 10.1056/NEJMcp2113128
  • Khairnar SI, Kulkarni YA, Singh K. Cardiotoxicity linked to anticancer agents and cardioprotective strategy. Arch Pharm Res. 2022;45(10):704–730. doi: 10.1007/s12272-022-01411-4
  • Corbacioglu S, Jabbour EJ, Mohty M. Risk factors for development of and progression of hepatic veno-occlusive Disease/Sinusoidal obstruction syndrome. Biol Blood Marrow Transplant. 2019;25(7):1271–1280. doi: 10.1016/j.bbmt.2019.02.018
  • Shah D, Soper B, Shopland L. Cytokine release syndrome and cancer immunotherapies – historical challenges and promising futures. Front Immunol. 2023;14:1190379. doi: 10.3389/fimmu.2023.1190379
  • Liu S, Zhao J, Wang F. Acute kidney injury in cancer patients. Clin Exp Nephrol. 2022;26(2):103–112. doi: 10.1007/s10157-021-02131-7
  • CalçCalçAs Marques R, Reis M, Pimenta G, et al. Severe acute kidney injury in hospitalized cancer patients: epidemiology and predictive model of renal replacement therapy and In-hospital mortality. Cancers (Basel). 2024;16(3):561. doi: 10.3390/cancers16030561
  • Abdelhafez M, Nayfeh T, Atieh A, et al. Diagnostic performance of fractional excretion of sodium for the differential diagnosis of acute kidney injury: a systematic review and meta-analysis. Clin J Am Soc Nephrol. 2022;17(6):785–797. doi: 10.2215/CJN.14561121
  • Perazella MA. The urine sediment as a biomarker of kidney disease. Am J Kidney Dis. 2015;66(5):748–755. doi: 10.1053/j.ajkd.2015.02.342
  • Chávez-Iñiguez JS, Navarro-Gallardo GJ, Medina-González R, et al. Acute kidney injury caused by obstructive nephropathy. Kershaw DB, editor. Int J Nephrol. 2020;2020:1–10. doi: 10.1155/2020/8846622
  • Perazella MA, Herlitz LC. The crystalline nephropathies. Kidney Int Rep. 2021;6(12):2942–2957. doi: 10.1016/j.ekir.2021.09.003
  • Mohidin B, Bass P, Salama A, et al. Malignant renal obstruction without dilatation. Acute Med J. 2019;18(2):121–123. doi: 10.52964/AMJA.0761
  • Rosner MH, Perazella MA, Ingelfinger JR. Acute kidney injury in patients with cancer. Ingelfinger JR, editor. N Engl J Med. 2017;376(18):1770–1781. doi: 10.1056/NEJMra1613984
  • Leung N, Rajkumar SV. Multiple myeloma with acute light chain cast nephropathy. Blood Cancer J. 2023;13(1):46. doi: 10.1038/s41408-023-00806-w
  • Gnanasampanthan S, Kousios A. Monoclonal gammopathies of renal significance. Clin Med. 2023;23(3):250–253. doi: 10.7861/clinmed.2023-RM3
  • Bai Y, Zheng Y, Zhang Q, et al. Renomegaly and acute kidney injury as primary manifestations of non-Hodgkin’s lymphoma: a report of three cases. Diagn Pathol. 2023;18(1):133. doi: 10.1186/s13000-023-01408-7
  • Jeyabalan A, Trivedi M. Paraneoplastic glomerular diseases. Adv Chronic Kidney Dis. 2022;29(2):116–126.e1.
  • Wilhelm D, Caster D, Coventry S, et al. A case of colon cancer and pauci-immune crescentic glomerulonephritis. Cureus [Internet]. 2022 [cited 2024 Jan 29]. Available from: https://www.cureus.com/articles/104756-a-case-of-colon-cancer-and-pauci-immune-crescentic-glomerulonephritis.
  • Weitz IC. Thrombotic microangiopathy in cancer. Semin Thromb Hemost. 2019;45(4):348–353. doi: 10.1055/s-0039-1687893
  • Izzedine H, Perazella MA. Thrombotic microangiopathy, cancer, and cancer drugs. Am J Kidney Dis Off J Natl Kidney Found. 2015;66(5):857–868. doi: 10.1053/j.ajkd.2015.02.340
  • Arnaud M, Loiselle M, Vaganay C, et al. Tumor lysis syndrome and AKI: beyond crystal mechanisms. J Am Soc Nephrol. 2022;33(6):1154. doi: 10.1681/ASN.2021070997
  • Abdel-Nabey M, Chaba A, Serre J, et al. Tumor lysis syndrome, acute kidney injury and disease-free survival in critically ill patients requiring urgent chemotherapy. Ann Intensive Care. 2022;12(1):15. doi: 10.1186/s13613-022-00990-1
  • Gupta S, Portales-Castillo I, Daher A, et al. Conventional chemotherapy nephrotoxicity. Adv Chronic Kidney Dis. 2021;28(5):402–414.e1. doi: 10.1053/j.ackd.2021.08.001.
  • Motwani SS, McMahon GM, Humphreys BD, et al. Development and validation of a risk prediction model for acute kidney injury after the first course of cisplatin. J Clin Oncol. 2018;36(7):682–688. doi: 10.1200/JCO.2017.75.7161
  • Romani AMP. Cisplatin in cancer treatment. Biochem Pharmacol. 2022;206:115323. doi: 10.1016/j.bcp.2022.115323
  • Motwani SS, Sandhu SK, Kitchlu A. Cisplatin nephrotoxicity: novel insights into mechanisms and preventative strategies. Semin Nephrol. 2022;42(6):151341. doi: 10.1016/j.semnephrol.2023.151341
  • Bonilla M, Workeneh BT, Uppal NN. Hypomagnesemia in patients with cancer: the forgotten Ion. Semin Nephrol. 2022;42(6):151347. doi: 10.1016/j.semnephrol.2023.151347
  • Volovat S, Apetrii M, Stefan A, et al. Cisplatin and AKI: an ongoing battle with new perspectives—a narrative review. Int Urol Nephrol. 2022;55(5):1205–1209. doi: 10.1007/s11255-022-03418-8
  • Yamada S, Yazawa M, Yamamoto M, et al. A case of biopsy-proven oxaliplatin-induced acute tubulointerstitial nephritis with thrombocytopenia and anemia. CEN Case Rep. 2019;8(3):188–193. doi: 10.1007/s13730-019-00390-8
  • Moradbeygi F, Ghasemi Y, Farmani AR, et al. Glucarpidase (carboxypeptidase G2): biotechnological production, clinical application as a methotrexate antidote, and placement in targeted cancer therapy. Biomed Pharmacother. 2023;166:115292. doi: 10.1016/j.biopha.2023.115292
  • Sakran R, Milo G, Jabareen A, et al. Effective elimination of high-dose methotrexate by repeated hemodiafiltration and high-flux hemodialysis in patients with acute kidney injury. J Oncol Pharm Pract. 2022;28(2):508–515. doi: 10.1177/10781552211052564
  • Visser S, Huisbrink J, Van ‘T Veer NE, et al. Renal impairment during pemetrexed maintenance in patients with advanced nonsmall cell lung cancer: a cohort study. Eur Respir J. 2018;52(4):1800884. doi: 10.1183/13993003.00884-2018
  • Fung E, Anand S, Bhalla V. Pemetrexed-induced nephrogenic diabetes insipidus. Am J Kidney Dis. 2016;68(4):628–632. doi: 10.1053/j.ajkd.2016.04.016
  • Chauvet S, Courbebaisse M, Ronco P, et al. Pemetrexed-induced acute kidney injury leading to chronic kidney disease. Clin Nephrol. 2014;82 (2014):402–406. doi: 10.5414/CN107921
  • Horino T, Inotani S, Ishihara M, et al. Gemcitabine‐induced renal thrombotic microangiopathy. Nephrology. 2022;27(8):724–725. doi: 10.1111/nep.14043
  • Mateo FJP, Guasch AD, Pineda JAG, et al. Capecitabine-related thrombotic microangiopathy. J Gastrointest Cancer [Internet]. 2024 [cited 2024 Jan 29]. Available from: https://link.springer.com. doi: 10.1007/s12029-023-00993-6
  • Witte WP, Bruins P, Boerma D, et al. Risk of acute kidney injury following mytomicin C resorption during hyperthermic intraperitoneal chemotherapy. MRAJ. 2023 [cited 2024 Jan 29];11(7.2). Available from https://esmed.org/MRA/mra/article/view/4181. doi: 10.18103/mra.v11i7.2.4181
  • Ensergueix G, Pallet N, Joly D, et al. Ifosfamide nephrotoxicity in adult patients. Clin Kidney J. 2020;13(4):660–665. doi: 10.1093/ckj/sfz183
  • Akilesh S, Juaire N, Duffield JS, et al. Chronic ifosfamide toxicity: kidney pathology and pathophysiology. Am J Kidney Dis. 2014;63(5):843–850. doi: 10.1053/j.ajkd.2013.11.028
  • Shabbir-Moosajee M, Jehangir S, Sawani S, et al. Safety and efficacy of bendamustine in the conditioning regimen for autologous stem cell transplantation in patients with relapsed/refractory lymphoma. Blood Res. 2019;54(2):108–113. doi: 10.5045/br.2019.54.2.108
  • Nikolova T, Roos WP, Krämer OH, et al. Chloroethylating nitrosoureas in cancer therapy: DNA damage, repair and cell death signaling. Biochim Biophys Acta BBA - Rev Cancer. 2017;1868(1):29–39. doi: 10.1016/j.bbcan.2017.01.004
  • Grignani G, Le Cesne A, Martín-Broto J. Trabectedin as second-line treatment in advanced soft tissue sarcoma: quality of life and safety outcomes. Future Oncol. 2022;18(30s):13–22. doi: 10.2217/fon-2022-0518
  • Glezerman I, Salvatore S, Tap W, et al. Pegylated liposomal doxorubicin causes kidney-limited thrombotic microangiopathy. Am J Kidney Dis. 2024;83(4):S549–553. doi: 10.1053/j.ajkd.2023.08.014
  • Mo Z, Deng Y, Bao Y, et al. Evaluation of cardiotoxicity of anthracycline‐containing chemotherapy regimens in patients with bone and soft tissue sarcomas: a study of the FDA adverse event reporting system joint single‐center real‐world experience. Cancer Med. 2023;12(24):21709–21724. doi: 10.1002/cam4.6730
  • Rashidi A, Wanchoo R, Izzedine H. How I manage hypertension and Proteinuria associated with VEGF inhibitor. Clin J Am Soc Nephrol CJASN. 2023;18(1):121–123. doi: 10.2215/CJN.05610522
  • Eremina V, Baelde HJ, Quaggin SE. Role of the VEGF–a signaling pathway in the glomerulus: evidence for crosstalk between components of the glomerular filtration barrier. Nephron Physiol. 2007;106(2):32–37. doi: 10.1159/000101798
  • Pfister F, Amann K, Daniel C, et al. Characteristic morphological changes in anti-VEGF therapy-induced glomerular microangiopathy. Histopathology. 2018;73(6):990–1001. doi: 10.1111/his.13716
  • Onteddu NK, Mudupula Vemula SS, Areddy VR, et al. Bevacizumab-induced nephropathy presenting as Crescentic Glomerulopathy. Cureus. 2023;15:e48787. doi: 10.7759/cureus.48787
  • Tanihara Y, Masuda S, Inui K-I. Inhibitory effects of vandetanib on creatinine transport via renal organic cation transporter OCT2. Eur J Pharm Sci Off J Eur Fed Pharm Sci. 2021;158:105666. doi: 10.1016/j.ejps.2020.105666
  • Mir O, Coriat R, Boudou-Rouquette P, et al. Sorafenib-induced diarrhea and hypophosphatemia: mechanisms and therapeutic implications. Ann Oncol Off J Eur Soc Med Oncol. 2012;23(1):280–281. doi: 10.1093/annonc/mdr525.
  • Strasma A, Coke H, Mamlouk O, et al. Lupus-like glomerulonephritis associated with regorafenib, a multikinase inhibitor. Kidney Med. 2021;3(2):294–298. doi: 10.1016/j.xkme.2020.11.011
  • Singh AK, Hussain S, Ahmed R, et al. Impact of imatinib treatment on renal function in chronic myeloid leukaemia patients. Nephrol Carlton Vic. 2022;27(4):318–326. doi: 10.1111/nep.14014
  • Troxell ML, Higgins JP, Kambham N. Antineoplastic treatment and renal injury: an update on renal Pathology Due to Cytotoxic and targeted therapies. Adv Anat Pathol. 2016;23(5):310–329. doi: 10.1097/PAP.0000000000000122
  • Furuto Y, Hashimoto H, Namikawa A, et al. Focal segmental glomerulosclerosis lesion associated with inhibition of tyrosine kinases by lenvatinib: a case report. BMC Nephrol. 2018;19(1):273. doi: 10.1186/s12882-018-1074-3
  • Izzedine H, Escudier B, Rouvier P, et al. Acute tubular necrosis associated with mTOR inhibitor therapy: a real entity biopsy-proven. Ann Oncol. 2013;24(9):2421–2425. doi: 10.1093/annonc/mdt233
  • 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
  • Estrada CC, Maldonado A, Mallipattu SK. Therapeutic inhibition of VEGF signaling and associated Nephrotoxicities. J Am Soc Nephrol JASN. 2019;30(2):187–200. doi: 10.1681/ASN.2018080853
  • Porta C, Cosmai L, Gallieni M, et al. Renal effects of targeted anticancer therapies. Nat Rev Nephrol. 2015;11(6):354–370. doi: 10.1038/nrneph.2015.15
  • Cosmai L, Gallieni M, Porta C. Renal toxicity of anticancer agents targeting HER2 and EGFR. J Nephrol. 2015;28(6):647–657. doi: 10.1007/s40620-015-0226-9
  • Petrelli F, Borgonovo K, Cabiddu M, et al. Risk of anti-EGFR monoclonal antibody-related hypomagnesemia: systematic review and pooled analysis of randomized studies. Expert Opin Drug Saf. 2012;Suppl 11(sup1):S9–19. doi: 10.1517/14740338.2011.606213
  • Luo W, Li Y, Ye F, et al. Anti-EGFR monoclonal antibody plus chemotherapy for treating advanced non-small cell lung cancer: a meta-analysis. Medicine (Baltimore). 2021;100(47):e27954. doi: 10.1097/MD.0000000000027954
  • Jhaveri KD, Sakhiya V, Fishbane S. Nephrotoxicity of the BRAF inhibitors vemurafenib and Dabrafenib. JAMA Oncol. 2015;1(8):1133–1134. doi: 10.1001/jamaoncol.2015.1713
  • Sanagawa A, Hotta Y, Mori N, et al. BRAF/MEK inhibitor-associated nephrotoxicity in a real-world setting and human kidney cells. Anticancer Drugs. 2021;32(10):1076–1083. doi: 10.1097/CAD.0000000000001106
  • Seethapathy H, Bates H, Chute DF, et al. Acute kidney injury following encorafenib and binimetinib for metastatic melanoma. Kidney Med. 2020;2(3):373–375. doi: 10.1016/j.xkme.2020.01.012
  • Reid A, Vidal L, Shaw H, et al. Dual inhibition of ErbB1 (EGFR/HER1) and ErbB2 (HER2/neu). Eur J Cancer Oxf Engl 1990. 2007;43(3):481–489. doi: 10.1016/j.ejca.2006.11.007
  • Gemmete JJ, Mukherji SK. Trastuzumab (Herceptin): Fig 1. AJNR Am J Neuroradiol. 2011;32(8):1373–1374. doi: 10.3174/ajnr.A2619
  • Hurvitz SA, Hegg R, Chung W-P, et al. Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer: updated results from DESTINY-Breast03, a randomised, open-label, phase 3 trial. Lancet. 2023;401(10371):105–117. doi: 10.1016/S0140-6736(22)02420-5
  • Hakroush S, Wulf S, Gallwas J, et al. Case report: collapsing focal segmental glomerulosclerosis after initiation of ado-trastuzumab emtansine therapy. Front Oncol. 2021;11:796223. doi: 10.3389/fonc.2021.796223
  • Jhaveri K, Drago JZ, Shah PD, et al. A phase I study of Alpelisib in combination with trastuzumab and LJM716 in patients with PIK3CA -mutated HER2-positive metastatic breast cancer. Clin Cancer Res. 2021;27(14):3867–3875. doi: 10.1158/1078-0432.CCR-21-0047
  • Kalantri P, Lomashvili K Deruxtecan-Induced Reversible Fanconi Syndrome. Cureus [Internet]. 2023 [cited 2024 Apr 15]. Available from: https://www.cureus.com/articles/157520-deruxtecan-induced-reversible-fanconi-syndrome.
  • Song SM, Jeon J, Jang HR, et al. Acute kidney injury in bortezomib-treated patients with multiple myeloma. Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc - Eur Ren Assoc. 2023;38(9):2077–2085. doi: 10.1093/ndt/gfad016
  • Klomjit N, Evans R, Le TK, et al. Frequency and characteristics of chemotherapy‐associated thrombotic microangiopathy: analysis from a large pharmacovigilance database. Am J Hematol [Internet]. 2023 [cited 2024 Apr 15];98(12). Available from. doi: 10.1002/ajh.27101
  • Lodhi A, Kumar A, Saqlain MU, et al. Thrombotic microangiopathy associated with proteasome inhibitors. Clin Kidney J. 2015;8(5):632–636. doi: 10.1093/ckj/sfv059
  • Moscvin M, Liacos CI, Chen T, et al. Mutations in the alternative complement pathway in multiple myeloma patients with carfilzomib-induced thrombotic microangiopathy. Blood Cancer J. 2023;13(1):31. doi: 10.1038/s41408-023-00802-0
  • Bonilla M, Jhaveri KD, Izzedine H. Anaplastic lymphoma kinase inhibitors and their effect on the kidney. Clin Kidney J. 2022;15(8):1475–1482. doi: 10.1093/ckj/sfac062
  • Izzedine H, Brocheriou I, Amoura Z, et al. Acute tubular injury and renal arterial myocyte vacuolization following crizotinib administration. Kidney Int Rep. 2021;6(2):526–528. doi: 10.1016/j.ekir.2020.11.029
  • Chen MF, Harada G, Liu D, et al. Brief report: tyrosine kinase inhibitors for lung cancers that inhibit MATE-1 can lead to “false” decreases in renal function. J Thorac Oncol. 2024;19(1):153–159. doi: 10.1016/j.jtho.2023.09.1444
  • Yasuma T, Kobayashi T, D’Alessandro-Gabazza CN, et al. Renal injury during Long-Term Crizotinib Therapy. Int J Mol Sci. 2018;19(10):2902. doi: 10.3390/ijms19102902
  • Da Silva A, Chretien B, Alexandre J, et al. Abemaciclib‐induced reversible grade 4 nephrotoxicity. Breast J. 2020;26(11):2329–2330. doi: 10.1111/tbj.13980
  • Gulturk I, Yilmaz M, Ozmen A, et al. Ribociclib induced acute kidney injury: A case report. J Oncol Pharm Pract. 2021;27(8):2023–2026. doi: 10.1177/10781552211007202
  • Hanna PE, Strohbehn IA, Moreno D, et al. Adverse kidney outcomes of CDK 4/6 inhibitors for metastatic breast cancer. NPJ Breast Cancer. 2023;9(1):70. doi: 10.1038/s41523-023-00576-5
  • Meraz-Muñoz A, Amir E, Ng P, et al. Acute kidney injury associated with immune checkpoint inhibitor therapy: incidence, risk factors and outcomes. J Immunother Cancer. 2020;8(1):e000467. doi: 10.1136/jitc-2019-000467
  • Kitchlu A, Jhaveri KD, Wadhwani S, et al. A systematic review of immune checkpoint inhibitor–associated glomerular disease. Kidney Int Rep. 2021;6(1):66–77. doi: 10.1016/j.ekir.2020.10.002
  • Liu C, Wei W, Yang L, et al. Incidence and risk factors of acute kidney injury in cancer patients treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Front Immunol. 2023;14:1173952. doi: 10.3389/fimmu.2023.1173952.
  • Liu F, Wang Z, Li X, et al. Comparative risk of acute kidney injury among cancer patients treated with immune checkpoint inhibitors. Cancer Commun. 2023;43(2):214–224. doi: 10.1002/cac2.12396
  • Sprangers B, Leaf DE, Porta C, et al. Diagnosis and management of immune checkpoint inhibitor-associated acute kidney injury. Nat Rev Nephrol. 2022;18(12):794–805. doi: 10.1038/s41581-022-00630-8
  • Schneider BJ, Naidoo J, Santomasso BD, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO guideline update. J Clin Oncol. 2021;39(36):4073–4126. doi: 10.1200/JCO.21.01440.
  • Tanemoto M, Iida Y, Kasai T. Short-term steroid therapy for immune-checkpoint-inhibitor-related acute kidney injury. Int Urol Nephrol. 2023;56(3):1199–1200. doi: 10.1007/s11255-023-03734-7
  • Seethapathy H, Herrmann SM, Rashidi A. Immune checkpoint inhibitor–associated AKI: debates in diagnosis, management, and rechallenge. Semin Nephrol. 2022;42(6):151346. doi: 10.1016/j.semnephrol.2023.151346
  • Zhou P, Gao Y, Kong Z, et al. Immune checkpoint inhibitors and acute kidney injury. Front Immunol. 2024;15:1353339. doi: 10.3389/fimmu.2024.1353339
  • Singh S, Khasbage S, Kaur RJ, et al. Chimeric antigen receptor T cell: a cancer immunotherapy. Indian J Pharmacol. 2022;54(3):226–233. doi: 10.4103/ijp.ijp_531_20
  • Zhou H, Yang M, Cui L, et al. Chimeric antigen receptor T cell therapy and nephrotoxicity: from diagnosis to treatment strategies. Int Immunopharmacol. 2020;89:107072. doi: 10.1016/j.intimp.2020.107072
  • Manohar S, Jhaveri KD, Perazella MA. Immunotherapy-related acute kidney injury. Adv Chronic Kidney Dis. 2021;28(5):429–437.e1.
  • van de Donk NWCJ, Zweegman S. T-cell-engaging bispecific antibodies in cancer. Lancet Lond Engl. 2023;402(10396):142–158. doi: 10.1016/S0140-6736(23)00521-4
  • Crombie JL, Graff T, Falchi L, et al. Consensus recommendations on the management of toxicity associated with CD3×CD20 bispecific antibody therapy. Blood. 2024;143(16):1565–1575. doi: 10.1182/blood.2023022432
  • Acharya R, Horn B, Zeng X, et al. Collapsing focal segmental glomerulosclerosis and acute kidney injury associated with chimeric antigen receptor T-Cell (CAR-T) therapy: a case report. Kidney Med. 2021;3(6):1086–1090. doi: 10.1016/j.xkme.2021.06.011
  • Fry A, Saleemi A, Griffiths M, et al. Acute renal failure following intravesical bacille Calmette-Guérin chemotherapy for superficial carcinoma of the bladder. Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc - Eur Ren Assoc. 2005;20(4):849–850. doi: 10.1093/ndt/gfh688
  • Borówka M, Łącki-Zynzeling S, Nicze M, et al. Adverse renal effects of anticancer immunotherapy: a review. Cancers (Basel). 2022;14(17):4086. doi: 10.3390/cancers14174086
  • Borden EC. Interferons α and β in cancer: therapeutic opportunities from new insights. Nat Rev Drug Discov. 2019;18(3):219–234. doi: 10.1038/s41573-018-0011-2
  • Fishman M, Dutcher JP, Clark JI, et al. Overall survival by clinical risk category for high dose interleukin-2 (HD IL-2) treated patients with metastatic renal cell cancer (mRCC): data from the PROCLAIMSM registry. J Immunother Cancer. 2019;7(1):84. doi: 10.1186/s40425-019-0567-3
  • Ren Z, Zhang X, Fu Y-X. Facts and hopes on chimeric cytokine agents for cancer immunotherapy. Clin Cancer Res Off J Am Assoc Cancer Res. 2024;OF1–OF14. doi: 10.1158/1078-0432.CCR-23-1160
  • Renaghan AD, Jaimes EA, Malyszko J, et al. Acute kidney injury and CKD associated with hematopoietic stem cell transplantation. Clin J Am Soc Nephrol CJASN. 2020;15(2):289–297. doi: 10.2215/CJN.08580719
  • Schoettler ML, Carreras E, Cho B, et al. Harmonizing definitions for diagnostic criteria and prognostic assessment of transplantation-associated thrombotic microangiopathy: a report on behalf of the European Society for Blood and Marrow transplantation, American Society for transplantation and Cellular Therapy, Asia-Pacific Blood and Marrow transplantation Group, and Center for International Blood and Marrow Transplant Research. Transplant Cell Ther. 2023;29(3):151–163. doi: 10.1016/j.jtct.2022.11.015.
  • Stewart GD, Klatte T, Cosmai L, et al. The multispeciality approach to the management of localised kidney cancer. Lancet Lond Engl. 2022;400:523–534. doi: 10.1016/S0140-6736(22)01059-5
  • Hu SL. The Nephrologist’s management of renal cell carcinoma after kidney surgery. Semin Nephrol. 2020;40(1):59–68. doi: 10.1016/j.semnephrol.2019.12.007
  • Saltarella I, Altamura C, Campanale C, et al. Anti-angiogenic activity of drugs in multiple myeloma. Cancers (Basel). 2023;15:1990. doi: 10.3390/cancers15071990
  • de Roij van Zuijdewijn C, van Dorp W, Florquin S, et al. Bisphosphonate nephropathy: a case series and review of the literature. Br J Clin Pharmacol. 2021;87(9):3485–3491. doi: 10.1111/bcp.14780
  • Jani C, Ueberroth J, Page S, et al. Acute tubular necrosis caused by zoledronic acid infusion in a patient with osteoporosis. Am J Ther. 2021;29:e146–e148. doi: 10.1097/MJT.0000000000001380
  • Neyra JA, Vaidya OU, Hendricks A, et al. Collapsing focal segmental glomerulosclerosis resulting from a single dose of zoledronate. Nephron Extra. 2014;4(3):168–174. doi: 10.1159/000366450
  • Glezerman IG, Sternlicht H. Hypercalcemia of malignancy and new treatment options. Ther Clin Risk Manag. 2015;1779. doi: 10.2147/TCRM.S83681
  • Murakami N, Mulvaney P, Danesh M, et al. A multi-center study on safety and efficacy of immune checkpoint inhibitors in cancer patients with kidney transplant. Kidney Int. 2021;100(1):196–205. doi: 10.1016/j.kint.2020.12.015
  • Chang F-C, Chen T-W, Huang T-M, et al. Spectrum of cancer patients receiving renal biopsy. J Formos Med Assoc Taiwan Yi Zhi. 2022;121(1):152–161. doi: 10.1016/j.jfma.2021.02.009
  • Alonso F, Martín de Francisco ÁLM, Auñón P, et al. Efectos renales adversos por inhibidores del check-point (ICP) en pacientes con cáncer. Recomendaciones del grupo de Onconefrología de la Sociedad Española de Nefrología (SEN). Nefrología. 2023;43(5):622–635. doi: 10.1016/j.nefro.2022.12.004.
  • Niculae A, Gherghina M-E, Peride I, et al. Pathway from acute kidney injury to chronic kidney disease: molecules involved in renal fibrosis. Int J Mol Sci. 2023;24(18):14019. doi: 10.3390/ijms241814019
  • Siddiq S, Hussain Z, Mubarakali J, et al. Incidence, patterns, and characteristics of patients with acute kidney injury requiring renal replacement therapy in cancer settings. Cureus. 2023;15:e48627. doi: 10.7759/cureus.48627
  • Janus N, Launay-Vacher V. Pharmacokinetic/Pharmacodynamic considerations for cancer patients undergoing hemodialysis. Expert Opin Drug Metab Toxicol. 2017;13(6):617–623. doi: 10.1080/17425255.2017.1292252
  • Duerr M, Glander P, Diekmann F, et al. Increased incidence of angioedema with ACE inhibitors in combination with mTOR inhibitors in kidney transplant recipients. Clin J Am Soc Nephrol CJASN. 2010;5(4):703–708. doi: 10.2215/CJN.07371009
  • Trivedi M, Nair S. Onconephrology: new frontiers necessitate new skills? Indian J Nephrol. 2023;33:409–410. doi: 10.4103/ijn.ijn_217_23
  • Ashammakhi N, Wesseling-Perry K, Hasan A, et al. Kidney-on-a-chip: untapped opportunities. Kidney Int. 2018;94(6):1073–1086. doi: 10.1016/j.kint.2018.06.034
  • Tanase DM, Gosav EM, Radu S, et al. The predictive role of the biomarker kidney molecule-1 (KIM-1) in acute kidney injury (AKI) cisplatin-induced nephrotoxicity. Int J Mol Sci. 2019;20(20):5238. doi: 10.3390/ijms20205238
  • Rossiter A, La A, Koyner JL, et al. New biomarkers in acute kidney injury. Crit Rev Clin Lab Sci. 2024;61(1):23–44. doi: 10.1080/10408363.2023.2242481
  • Selamet U, Ahdoot RS, Salasnek R, et al. Onconephrology: mitigation of renal injury in chemotherapy administration. Curr Opin Nephrol Hypertens. 2024;33(2):257–266. doi: 10.1097/MNH.0000000000000960

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.