126
Views
0
CrossRef citations to date
0
Altmetric
Case Report

Tocilizumab and CytoSorb for delayed severe cytokine release syndrome after ipilimumab plus nivolumab immunotherapy

ORCID Icon, & ORCID Icon
Received 30 Nov 2023, Accepted 17 Jun 2024, Published online: 17 Jul 2024

References

  • Foran AE, Nadel HR, Lee AF, et al. Nivolumab in the treatment of refractory pediatric Hodgkin lymphoma. J Pediatr Hematol Oncol. 2017;39(5):e263–e266. doi:10.1097/MPH.0000000000000703
  • Queirolo P, Boutros A, Tanda E, et al. Immune-checkpoint inhibitors for the treatment of metastatic melanoma: a model of cancer immunotherapy. Semin Cancer Biol. 2019;59:290–297. doi:10.1016/j.semcancer.2019.08.001
  • Hu Z, Li M, Chen Z, et al. Advances in clinical trials of targeted therapy and immunotherapy of lung cancer in 2018. Transl Lung Cancer Res. 2019;8(6):1091–1106. doi:10.21037/tlcr.2019.10.17
  • Xu W, Atkins MB, McDermott DF. Checkpoint inhibitor immunotherapy in kidney cancer. Nat Rev Urol. 2020;17(3):137–150. doi:10.1038/s41585-020-0282-3
  • June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018;379(1):64–73. doi:10.1056/NEJMra1706169
  • Tedesco VET, Mohan C. Biomarkers for predicting cytokine release syndrome following CD19-targeted CAR T cell therapy. J Immunol. 2021;206(7):1561–1568. doi:10.4049/jimmunol.2001249
  • Lee DW, Gardner R, Porter DL, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124(2):188–195. doi:10.1182/blood-2014-05-552729
  • Teachey DT, Rheingold SR, Maude SL, et al. Cytokine release syndrome after blinatumomab treatment related to abnormal macrophage activation and ameliorated with cytokine-directed therapy. Blood. 2013;121(26):5154–5157. doi:10.1182/blood-2013-02-485623
  • Gödel P, Shimabukuro-Vornhagen A, von Bergwelt-Baildon M. Understanding cytokine release syndrome. Intensive Care Med. 2018;44(3):371–373. doi:10.1007/s00134-017-4943-5
  • Shimabukuro-Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56. doi:10.1186/s40425-018-0343-9
  • Zhang X, Fu Z, Yan C. Cytokine release syndrome induced by pembrolizumab: a case report. Medicine (Baltimore). 2022;101(49):e31998. doi:10.1097/MD.0000000000031998
  • Dimitriou F, Matter AV, Mangana J, et al. Cytokine release syndrome during sequential treatment with immune checkpoint inhibitors and kinase inhibitors for metastatic melanoma. J Immunother. 2019;42(1):29–32. doi:10.1097/CJI.0000000000000236
  • Tay SH, Toh MMX, Thian YL, et al. Cytokine release syndrome in cancer patients receiving immune checkpoint inhibitors: a case series of 25 patients and review of the literature. Front Immunol. 2022;13:807050. doi:10.3389/fimmu.2022.807050
  • Liu LL, Skribek M, Harmenberg U, Gerling M. Systemic inflammatory syndromes as life-threatening side effects of immune checkpoint inhibitors: case report and systematic review of the literature. J Immunother Cancer. 2023;11(3):e005841. doi:10.1136/jitc-2022-005841
  • Kotch C, Barrett D, Teachey DT. Tocilizumab for the treatment of chimeric antigen receptor T cell-induced cytokine release syndrome. Expert Rev Clin Immunol. 2019;15(8):813–822. doi:10.1080/1744666X.2019.1629904
  • Frey N, Porter D. Cytokine release syndrome with chimeric antigen receptor T Cell therapy. Biol Blood Marrow Transplant. 2019;25(4):e123–e127. doi:10.1016/j.bbmt.2018.12.756
  • Yomota M, Mirokuji K, Sakaguchi M, et al. Cytokine release syndrome induced by immune-checkpoint inhibitor therapy for non-small-cell lung cancer. Intern Med. 2021;60(21):3459–3462. doi:10.2169/internalmedicine.5922-20
  • Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25(4):625–638. doi:10.1016/j.bbmt.2018.12.758
  • Paz-Ares L, Ciuleanu TE, Cobo M, et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): an international, randomised, open-label, Phase III trial. Lancet Oncol. 2021;22(2):198–211. doi:10.1016/S1470-2045(20)30641-0
  • Common Terminology Criteria for Adverse Events (CTCAE) v5.0. U.S. Department of Health and Human Services, National Cancer Institute. 2017. https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_8.5x11.pdf
  • ASSESSMENT REPORT FOR RoActemra. European Medicines Agency, evaluation of medicines for human use. 2009.
  • Tanaka T, Narazaki M, Kishimoto T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy. 2016;8(8):959–970. doi:10.2217/imt-2016-0020
  • Frey NV, Porter DL. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program. 2016;2016(1):567–572. doi:10.1182/asheducation-2016.1.567
  • Ciner AT, Hochster HS, August DA, et al. Delayed cytokine release syndrome after neoadjuvant nivolumab: a case report and literature review. Immunotherapy. 2021;13(13):1071–1078. doi:10.2217/imt-2020-0329
  • Oda H, Ishihara M, Miyahara Y, et al. First case of cytokine release syndrome after nivolumab for gastric cancer. Case Rep Oncol. 2019;12(1):147–156. doi:10.1159/000496933
  • Ceschi A, Noseda R, Palin K, Verhamme K. Immune checkpoint inhibitor-related cytokine release syndrome: analysis of WHO global pharmacovigilance database. Front Pharmacol. 2020;11:557. doi:10.3389/fphar.2020.00557
  • Hellwig Y, Yoo YE, Reß ML, et al. Fulminant skin GvHD with a cytokine pattern resemblant of cytokine release syndrome successfully treated with multimodal immunosuppression including tocilizumab. Pediatr Blood Cancer. 2015;62(11):2033–2035. doi:10.1002/pbc.25595
  • Abboud R, Keller J, Slade M, et al. Severe cytokine-release syndrome after T cell-replete peripheral blood haploidentical donor transplantation is Associated with poor survival and Anti-IL-6 therapy is safe and well tolerated. Biol Blood Marrow Transplant. 2016;22(10):1851–1860. doi:10.1016/j.bbmt.2016.06.010
  • Gupta S, Seethapathy H, Strohbehn IA, et al. Acute kidney injury and electrolyte abnormalities after chimeric antigen receptor T-cell (CAR-T) therapy for diffuse large B-Cell lymphoma. Am J Kidney Dis. 2020;76(1):63–71. doi:10.1053/j.ajkd.2019.10.011
  • Song J, Park DW, Moon S, et al. Diagnostic and prognostic value of interleukin-6, pentraxin 3, and procalcitonin levels among sepsis and septic shock patients: a prospective controlled study according to the Sepsis-3 definitions. BMC Infect Dis. 2019;19(1):968. doi:10.1186/s12879-019-4618-7
  • Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 2016;6(6):664–679. doi:10.1158/2159-8290.CD-16-0040
  • Hay KA, Hanafi LA, Li D, et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017;130(21):2295–2306. doi:10.1182/blood-2017-06-793141
  • Zhang Y, Zhang W, Dai H, et al. An analytical biomarker for treatment of patients with recurrent B-ALL after remission induced by infusion of anti-CD19 chimeric antigen receptor T (CAR-T) cells. Sci China Life Sci. 2016;59(4):379–385. doi:10.1007/s11427-016-5035-4
  • Wang Z, Han W. Biomarkers of cytokine release syndrome and neurotoxicity related to CAR-T cell therapy. Biomark Res. 2018;6:4. doi:10.1186/s40364-018-0116-0
  • Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7(303):303ra139. doi:10.1126/scitranslmed.aac5415
  • Powell MZ, Mara KC, Bansal R, et al. Procalcitonin as a biomarker for predicting bacterial infection in chimeric antigen receptor T-cell therapy recipients. Cancer Med. 2023;12(8):9228–9235. doi:10.1002/cam4.5665
  • Arkader R, Troster EJ, Lopes MR, et al. Procalcitonin does discriminate between sepsis and systemic inflammatory response syndrome. Arch Dis Child. 2006;91(2):117–120. doi:10.1136/adc.2005.077446
  • Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra25. doi:10.1126/scitranslmed.3008226
  • Frey N. Cytokine release syndrome: who is at risk and how to treat. Best Pract Res Clin Haematol. 2017;30(4):336–340. doi:10.1016/j.beha.2017.09.002
  • Persic V, Jerman A, Malgaj Vrecko M, et al. Effect of CytoSorb coupled with hemodialysis on interleukin-6 and hemodynamic parameters in patients with systemic inflammatory response syndrome: a retrospective cohort study. J Clin Med. 2022;11(24):7500. doi:10.3390/jcm11247500
  • Becker S, Lang H, Vollmer Barbosa C, et al. Efficacy of CytoSorb(R): a systematic review and meta-analysis. Crit Care. 2023;27(1):215. doi:10.1186/s13054-023-04492-9
  • Technical data CytoSorb 300 CytoSorbents Europe GmbH2021 [cited 2023 2023-11-11]. https://cytosorb-therapy.com/en/the-adsorber/technical-data/
  • Scharf C, Liebchen U, Paal M, et al. Blood purification with a cytokine adsorber for the elimination of myoglobin in critically ill patients with severe rhabdomyolysis. Crit Care. 2021;25(1):41. doi:10.1186/s13054-021-03468-x
  • Williams DM. Clinical pharmacology of corticosteroids. Respir Care. 2018;63(6):655–670. doi:10.4187/respcare.06314
  • Rotz SJ, Leino D, Szabo S, et al. Severe cytokine release syndrome in a patient receiving PD-1-directed therapy. Pediatr Blood Cancer. 2017;64(12):1–5. doi:10.1002/pbc.26642
  • Nassiri AA, Hakemi MS, Miri MM, et al. Blood purification with CytoSorb in critically ill COVID-19 patients: a case series of 26 patients. Artif Organs. 2021;45(11):1338–1347. doi:10.1111/aor.14024
  • Hensel M, Volk T, Docke WD, et al. Hyperprocalcitonemia in patients with noninfectious SIRS and pulmonary dysfunction associated with cardiopulmonary bypass. Anesthesiology. 1998;89(1):93–104. doi:10.1097/00000542-199807000-00016
  • Samsudin I, Vasikaran SD. Clinical utility and measurement of procalcitonin. Clin Biochem Rev. 2017;38(2):59–68.
  • Vasishta S, Patel S. Elevated procalcitonin in acute pseudogout flare: a case report. Cureus. 2019;11(6):e4853. doi:10.7759/cureus.4853
  • Rule JA, Hynan LS, Attar N, et al. Procalcitonin identifies cell injury, not bacterial infection, in acute liver failure. PLOS ONE. 2015;10(9):e0138566. doi:10.1371/journal.pone.0138566
  • Becker KL, Snider R, Nylen ES. Procalcitonin in sepsis and systemic inflammation: a harmful biomarker and a therapeutic target. Br J Pharmacol. 2010;159(2):253–264. doi:10.1111/j.1476-5381.2009.00433.x
  • Redl H, Schlag G, Togel E, et al. Procalcitonin release patterns in a baboon model of trauma and sepsis: relationship to cytokines and neopterin. Crit Care Med. 2000;28(11):3659–3663. doi:10.1097/00003246-200011000-00021
  • Redl H, Schiesser A, Togel E, et al. Possible role of TNF on procalcitonin release in a baboon model of sepsis. Shock. 2001;16(1):25–27. doi:10.1097/00024382-200116010-00005
  • Diorio C, Shaw PA, Pequignot E, et al. Diagnostic biomarkers to differentiate sepsis from cytokine release syndrome in critically ill children. Blood Adv. 2020;4(20):5174–5183. doi:10.1182/bloodadvances.2020002592