Impact of cytokine release syndrome on cardiac function following CD19 CAR-T cell therapy in children and young adults with hematological malignancies

Haneen Shalabi, Vandana Sachdev, Amita Kulshreshtha, Julia W Cohen, Bonnie Yates, Doug R Rosing, Stanislav Sidenko, Cindy Delbrook, Crystal Mackall, Brandon Wiley, Daniel W Lee, Nirali N Shah, Haneen Shalabi, Vandana Sachdev, Amita Kulshreshtha, Julia W Cohen, Bonnie Yates, Doug R Rosing, Stanislav Sidenko, Cindy Delbrook, Crystal Mackall, Brandon Wiley, Daniel W Lee, Nirali N Shah

Abstract

Background: Chimeric antigen receptor (CAR) T-cell-associated cytokine release syndrome (CRS) may present with tachycardia, hemodynamic instability and reduced cardiac function. Pediatric CAR studies examining cardiac toxicity are limited.

Methods: We report on cardiac toxicity observed in children and young adults with hematologic malignancies enrolled in a CD19-28ζ CAR T-cell phase I trial (NCT01593696). All patients had a formal baseline echocardiogram. Real-time studies included echocardiograms on intensive care unit (ICU) transfer, and serial troponin and pro-B-type natriuretic peptide (pro-BNP) in the select patients.

Results: From July 2012 to March 2016, 52 patients, with a median age of 13.4 years (range 4.2-30.3) were treated. CRS developed in 37/52 (71%), which was grade 3-4 CRS in nine patients (17%). The median prior anthracycline exposure was 205 mg/m2 (range 70-620 mg/m2) in doxorubicin equivalents. The median baseline left ventricle ejection fraction (LVEF) and baseline LV global longitudinal strain (GLS) were 60% (range 50%-70%) and 16.8% (range 14.1%-23.5%, n=37) respectively. The majority, 78% (29/37), of patients had a reduced GLS <19% at baseline, and 6% (3/52) of patients had baseline LVEF <53%. ICU transfers occurred in 21 patients, with nine requiring vasoactive hemodynamic support and three necessitating >1 vasopressor. Six (12%) patients developed cardiac dysfunction (defined by >10% absolute decrease in LVEF or new-onset grade 2 or higher LV dysfunction, per CTCAE v4), among whom 4 had grade 3-4 CRS. Troponin elevations were seen in 4 of 13 patients, all of whom had low LVEF. Pro-BNP was elevated from baseline in 6/7 patients at the onset of CRS, with higher levels correlating with more severe CRS. Cardiac dysfunction fully resolved in all but two patients by day 28 post-CAR.

Conclusion: Cardiac toxicity related to CD19-28ζ CAR T-cell-associated CRS was generally reversible by day 28 postinfusion. Implementation of more frequent monitoring with formal echocardiograms incorporating systemic analysis of changes in GLS, and cardiac biomarkers (troponin and proBNP) may help to earlier identify those patients at highest risk of severe cardiac systolic dysfunction, facilitating earlier interventions for CRS to potentially mitigate acute cardiac toxicity.

Keywords: chimeric antigen; cytokines; hematologic neoplasms; immunotherapy; pediatrics; receptors.

Conflict of interest statement

Competing interests: CM is a founder, holds equity in and is a consultant for Lyell Immunopharma which is developing CAR T-cell therapeutics, consults for Neoimmune Tech, Apricity and Nektar, holds equity in Apricity and Allogene and has received royalties from NIH for a CD22-CAR licensed to Juno. DWL consults for Juno Therapeutics/Celgene, Harpoon Therapeutics and Amgen and his institution receives clinical trial support from Kite Pharma.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY. Published by BMJ.

Figures

Figure 1
Figure 1
(A) Left ventricular EF (%) in all patients with CRS. Each line represents a patient’s EF during the course of CRS as determined by echocardiogram. Black lines indicate those patients with CRS who had preserved EF; without cardiac dysfunction. Red lines indicate those patients with CRS who had decreased EF and experienced cardiac dysfunction. (B) Cumulative anthracycline dose for all patients treated on CD19-28ζ CAR T-cell trial stratified by those who had decreased EF (min: 100 mg/m2, max: 570 mg/m2) and those that had preserved EF (min: 70 mg/m2, max: 620 mg/m2) as compared with their baseline echocardiograms. (C) Pre-CAR and Post-CAR T-cell therapy proBNP levels in seven patients. Two patients with the highest post-therapy pro-BNP levels (depicted by the *) had decreased EF and cardiac dysfunction. CAR, chimeric antigen receptor; CRS, cytokine release syndrome; pro-BNP, pro-B-type natriuretic peptide.
Figure 2
Figure 2
Cardiac screening and monitoring algorithm for pediatric and young adult patients undergoing CAR T-Cell Therapy. *Obtain cardiology consult in patients with a history of heart failure (preserved or reduced ejection fraction (EF), known cardiomyopathy, cardiac arrhythmias, prior chest/mediastinum radiotherapy, baseline BNP or troponin elevated above upper limit of normal. Consider cardiology consult in patients with asymptomatic LV dysfunction (LVEF 300 mg/m2; $Monitor for conduction abnormalities and QT prolongation; #High-risk patients include those that required cardiology consult prior to infusion; ∧Consider cardiac MR in patients with new or worsened LV dysfunction (LVEF <53% or drop in GLS >15% from baseline) that has persisted after CAR T-cell therapy. BNP, B-type natriuretic peptide; CAR, chimeric antigen receptor; CRS, cytokine release syndrome; GLS, global longitudinal strain; TTE, transthoracic echocardiogram.

References

    1. Hay KA. Cytokine release syndrome and neurotoxicity after CD19 chimeric antigen receptor-modified (CAR-) T cell therapy. Br J Haematol 2018;183:364–74. 10.1111/bjh.15644
    1. Grupp SA, Kalos M, Barrett D, et al. . Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013;368:1509–18. 10.1056/NEJMoa1215134
    1. Maude SL, Frey N, Shaw PA, et al. . Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371:1507–17. 10.1056/NEJMoa1407222
    1. Maude SL, Teachey DT, Porter DL, et al. . CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 2015;125:4017–23. 10.1182/blood-2014-12-580068
    1. Maude SL, Laetsch TW, Buechner J, et al. . Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 2018;378:439–48. 10.1056/NEJMoa1709866
    1. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. . T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 2015;385:517–28. 10.1016/S0140-6736(14)61403-3
    1. Park JH, Rivière I, Gonen M, et al. . Long-Term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 2018;378:449–59. 10.1056/NEJMoa1709919
    1. Gardner RA, Finney O, Annesley C, et al. . Intent-To-Treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 2017;129:3322–31. 10.1182/blood-2017-02-769208
    1. 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;6:224ra225. 10.1126/scitranslmed.3008226
    1. Frey NV. Chimeric antigen receptor T cells for acute lymphoblastic leukemia. Am J Hematol 2019;94:S24–7. 10.1002/ajh.25442
    1. Maude SL, Barrett D, Teachey DT, et al. . Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J 2014;20:119–22. 10.1097/PPO.0000000000000035
    1. 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;6:664–79. 10.1158/-16-0040
    1. Lee DW, Gardner R, Porter DL, et al. . Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124:188–95. 10.1182/blood-2014-05-552729
    1. Kochenderfer JN, Dudley ME, Kassim SH, et al. . Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol 2015;33:540–9. 10.1200/JCO.2014.56.2025
    1. Hay KA, Hanafi L-A, Li D, et al. . Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood 2017;130:2295–306. 10.1182/blood-2017-06-793141
    1. Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev 2019;34:45–55. 10.1016/j.blre.2018.11.002
    1. Alvi RM, Frigault MJ, Fradley MG, et al. . Cardiovascular events among adults treated with chimeric antigen receptor T-cells (CAR-T). J Am Coll Cardiol 2019;74:3099–108. 10.1016/j.jacc.2019.10.038
    1. Ganatra S, Carver JR, Hayek SS, et al. . Chimeric antigen receptor T-cell therapy for cancer and heart: JACC Council perspectives. J Am Coll Cardiol 2019;74:3153–63. 10.1016/j.jacc.2019.10.049
    1. Asnani A. Cardiotoxicity of immunotherapy: incidence, diagnosis, and management. Curr Oncol Rep 2018;20:44. 10.1007/s11912-018-0690-1
    1. Kochenderfer JN, Somerville RPT, Lu T, et al. . Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J Clin Oncol 2017;35:1803–13. 10.1200/JCO.2016.71.3024
    1. Burstein DS, Maude S, Grupp S, et al. . Cardiac profile of chimeric antigen receptor T cell therapy in children: a single-institution experience. Biol Blood Marrow Transplant 2018;24:1590–5. 10.1016/j.bbmt.2018.05.014
    1. Guha A, Armanious M, Fradley MG. Update on cardio-oncology: novel cancer therapeutics and associated cardiotoxicities. Trends Cardiovasc Med 2019;29:29–39. 10.1016/j.tcm.2018.06.001
    1. 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;25:625–38. 10.1016/j.bbmt.2018.12.758
    1. Lang RM, Badano LP, Mor-Avi V, et al. . Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American Society of echocardiography and the European association of cardiovascular imaging. J Am Soc Echocardiogr 2015;28:e14:1–39. 10.1016/j.echo.2014.10.003
    1. Levy PT, Machefsky A, Sanchez AA, et al. . Reference ranges of left ventricular strain measures by two-dimensional Speckle-Tracking echocardiography in children: a systematic review and meta-analysis. J Am Soc Echocardiogr 2016;29:e206:209–25. 10.1016/j.echo.2015.11.016
    1. Kleinman K. Harriet Lane handbook : a manual for pediatric house officers. 22nd edition Philadelphia: ELSEVIER, 2021.
    1. Plana JC, Galderisi M, Barac A, et al. . Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of echocardiography and the European association of cardiovascular imaging. J Am Soc Echocardiogr 2014;27:911–39. 10.1016/j.echo.2014.07.012
    1. Sawaya H, Sebag IA, Plana JC, et al. . Assessment of echocardiography and biomarkers for the extended prediction of cardiotoxicity in patients treated with anthracyclines, taxanes, and trastuzumab. Circ Cardiovasc Imaging 2012;5:596–603. 10.1161/CIRCIMAGING.112.973321
    1. Clasen SC, Scherrer-Crosbie M. Applications of left ventricular strain measurements to patients undergoing chemotherapy. Curr Opin Cardiol 2018;33:493–7. 10.1097/HCO.0000000000000541
    1. Gardner RA, Ceppi F, Rivers J, et al. . Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without attenuation of antileukemic efficacy. Blood 2019;134:2149–58. 10.1182/blood.2019001463

Source: PubMed

3
Suscribir