CAR T Cell Therapy-Related Cardiovascular Outcomes and Management: Systemic Disease or Direct Cardiotoxicity?

Arjun K Ghosh, Daniel H Chen, Avirup Guha, Strachan Mackenzie, J Malcolm Walker, Claire Roddie, Arjun K Ghosh, Daniel H Chen, Avirup Guha, Strachan Mackenzie, J Malcolm Walker, Claire Roddie

Abstract

CD19-specific chimeric antigen receptor (CAR) T cell therapies have shown remarkable early success in highly refractory and relapsing hematological malignancies. However, this potent therapy is accompanied by significant toxicity. Cytokine release syndrome and neurotoxicity are the most widely reported, but the true extent and characteristics of cardiovascular toxicity remain poorly understood. Thus far, adverse cardiovascular effects observed include sinus tachycardia and other arrhythmias, left ventricular systolic dysfunction, profound hypotension, and shock requiring inotropic support. The literature regarding cardiovascular toxicities remains sparse; prospective studies are needed to define the cardiac safety of CAR T cell therapies to optimally harness their potential. This review summarizes the current understanding of the potential cardiovascular toxicities of CD19-specific CAR T cell therapies, outlines a proposed cardiac surveillance protocol for patients receiving this new treatment, and provides a roadmap of the future direction of cardio-oncology research in this area.

Keywords: ALL, acute lymphoblastic leukemia; CAR, chimeric antigen receptor; CI, confidence interval; CMR, cardiac magnetic resonance imaging; CR, complete response; CRS, cytokine release syndrome; IL, interleukin; LVSD, left ventricular systolic dysfunction; TTE, transthoracic echocardiography; cardiomyopathy; cytokine release syndrome; immunotherapy; leukemia; lymphoma.

© 2020 The Authors.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Manufacturing CAR T Cell Therapy Autologous T cells are collected from the patient. A chimeric receptor antibody (CAR) that recognizes CD19 is inserted into the cell surface using lentiviral or retroviral vectors. The CAR T cells undergo ex vivo rapid multiplication to generate therapeutic quantities. The patient undergoes lymphodepleting chemotherapy (usually a combination of fludarabine and cyclophosphamide) before receiving an infusion of the CAR T cells to reduce the native population of T cells, which in turn promotes CAR T-cell expansion.
Figure 2
Figure 2
CAR T Cell Toxicities Significant multiorgan toxicities can be associated with chimeric receptor antibody (CAR) T-cell therapy.
Figure 3
Figure 3
Management of CRS Low-grade cytokine release syndrome (CRS) can be managed with supportive care alone. Tocilizumab, an interleukin (IL)-6 receptor blocker, is considered as a first line of treatment in patients with grade 2 or higher CRS. When this approach is ineffective, a dose of corticosteroids is the second-line treatment. ASTCT = American Society for Transplantation and Cellular Therapy.
Central Illustration
Central Illustration
A Single Institution Proposed Cardiac Screening and Monitoring in Patients Undergoing Chimeric Antigen Receptor T Cell Therapy Patient journey through cardio-oncology screening and monitoring as part of the chimeric receptor antibody (CAR) T cell therapy program at the University College of London Hospital. CMR = cardiac magnetic resonance; ECG = electrocardiography; NT-proBNP = N-terminal pro–B-type natriuretic peptide; TnT = troponin T; TTE = transthoracic echocardiography.

References

    1. Park J.H., Geyer M.B., Brentjens R.J. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood. 2016;127:3312.
    1. Brown C.E., Mackall C.L. CAR T cell therapy: inroads to response and resistance. Nature Rev Immunol. 2019;19:73–74.
    1. Salter A.I., Pont M.J., Riddell S.R. Chimeric antigen receptor–modified T cells: CD19 and the road beyond. Blood. 2018;131:2621.
    1. Maude S., Barrett D.M. Current status of chimeric antigen receptor therapy for haematological malignancies. Br J Haematol. 2016;172:11–22.
    1. Subklewe M., von Bergwelt-Baildon M., Humpe A. Chimeric antigen receptor T cells: a race to revolutionize cancer therapy. Transfus Med Hemother. 2019;46:15–24.
    1. Brudno J.N., Kochenderfer J.N. Chimeric antigen receptor T-cell therapies for lymphoma. Nat Rev Clin Oncol. 2017;15:31.
    1. Crump M., Neelapu S.S., Farooq U. Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study. Blood. 2017;130:1800–1808.
    1. Maude S.L., Laetsch T.W., Buechner J. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439–448.
    1. Locke F.L., Ghobadi A., Jacobson C.A. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019;20:31–42.
    1. Maude S.L., Frey N., Shaw P.A. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517.
    1. Schuster S.J., Bishop M.R., Tam C. Sustained disease control for adult patients with relapsed or refractory diffuse large B-cell lymphoma: an updated analysis of JULIET, a global pivotal phase 2 trial of tisagenlecleucel. Blood. 2018;132:1684.
    1. Schuster S.J., Bishop M.R., Tam C.S. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380:45–56.
    1. Neelapu S.S., Locke F.L., Bartlett N.L. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–2544.
    1. Schuster S.J., Svoboda J., Chong E.A. Chimeric antigen receptor T cells in refractory B-cell lymphomas. N Engl J Med. 2017;377:2545–2554.
    1. Lee D.W., Gardner R., Porter D.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124:188–195.
    1. Neelapu S.S., Tummala S., Kebriaei P. Chimeric antigen receptor T-cell therapy—assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15:47–62.
    1. Lee D.W., Santomasso B.D., Locke F.L. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019;25:625–638.
    1. Brudno J.N., Kochenderfer J.N. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016;127:3321.
    1. Frey N. Cytokine release syndrome: who is at risk and how to treat. Best Prac Res Clin Oncol. 2017;30:336–340.
    1. Santomasso B., Bachier C., Westin J., Rezvani K., Shpall E.J. The Other Side of CAR T-Cell Therapy: Cytokine Release Syndrome, Neurologic Toxicity, and Financial Burden. American Society of Clinical Oncology Educational Book. 2019:433–444.
    1. June C.H., Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018;379:64–73.
    1. Belin C., Simard C., Dos Santos A. JS1.4 Neurological follow-up of neurotoxicity after CAR T cell therapy in lymphoma patients: a French neurological multi-center survey. J Neurooncol. 2019;21 iii5–iii5.
    1. Gust J., Finney O.C., Li D. Glial injury in neurotoxicity after pediatric CD19-directed chimeric antigen receptor T cell therapy. Ann Neurol. 2019;86:42–54.
    1. Burstein D.S., Maude S., Grupp S., Griffis H., Rossano J., Lin K. Cardiac profile of chimeric antigen receptor T cell therapy in children: a single-institution experience. Biol Blood Marrow Transplant. 2018;24:1590–1595.
    1. Fitzgerald J.C.M.D.P., Weiss S.L.M.D.M., Maude S.L.M.D.P. Cytokine release syndrome after chimeric antigen receptor T cell therapy for acute lymphoblastic leukemia. Crit Care Med. 2017;45:e124–e131.
    1. Alvi R.M., Frigault M.J., Fradley M.G. Cardiovascular events among adults treated with chimeric antigen receptor T-cells (CAR-T) J Am Coll Cardiol. 2019;74:3099–3108.
    1. Asnani A. Cardiotoxicity of immunotherapy: incidence, diagnosis, and management. Curr Oncol Rep. 2018;20:44.
    1. Porter D.L., Hwang W.T., Frey N.V. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7:303ra139.
    1. Cordeiro A., Bezerra E.D., Hirayama A.V. Late events after treatment with CD19-targeted chimeric antigen receptor modified T cells. Biol Blood Marrow Transplant. 2020;26:26–33.
    1. Hu Y., Feng J., Shao M., Huang H. Profile of capillary-leak syndrome in patients received chimeric antigen receptor T cell therapy. Blood. 2018;132:5204.
    1. Medina de Chazal H., Del Buono M.G., Keyser-Marcus L. Stress cardiomyopathy diagnosis and treatment: JACC State-of-the-Art Review. J Am Coll Cardiol. 2018;72:1955–1971.
    1. Linette G.P., Stadtmauer E.A., Maus M.V. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122:863.
    1. Plana J.C., Thavendiranathan P., Bucciarelli-Ducci C., Lancellotti P. Multi-modality imaging in the assessment of cardiovascular toxicity in the cancer patient. J Am Coll Cardiol Img. 2018;11:1173.
    1. Seraphim A., Westwood M., Bhuva A.N. Advanced imaging modalities to monitor for cardiotoxicity. Curr Treat Options Oncol. 2019;20:73.
    1. Yu A.F., Ky B. Roadmap for biomarkers of cancer therapy cardiotoxicity. Heart. 2016;102:425.
    1. Michel L., Rassaf T., Totzeck M. Biomarkers for the detection of apparent and subclinical cancer therapy-related cardiotoxicity. J Thorac Dis. 2018:S4282–S4295.
    1. Tan L.L., Lyon A.R. Role of biomarkers in prediction of cardiotoxicity during cancer treatment. Curr Treat Options Cardiovasc Med. 2018;20:55.
    1. Leger Kasey J., Leonard D., Nielson D., de Lemos James A., Mammen Pradeep P.A., Winick Naomi J. Circulating microRNAs: potential markers of cardiotoxicity in children and young adults treated with anthracycline chemotherapy. J Am Heart Assoc. 2017;6 e004653.
    1. Armenian S.H., Lacchetti C., Barac A. Prevention and monitoring of cardiac dysfunction in survivors of adult cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2017;35:893–911.
    1. Cameron B.J., Gerry A.B., Dukes J. Identification of a titin-derived HLA-A1–presented peptide as a cross-reactive target for engineered MAGE A3–directed T cells. Sci Transl Med. 2013;5
    1. Charrot S., Hallam S. CAR-T cells: future perspectives. HemaSphere. 2019;3:e188.
    1. Aghajanian H., Kimura T., Rurik J.G. Targeting cardiac fibrosis with engineered T cells. Nature. 2019;573:430–433.
    1. Ganatra S., Carver J.R., Hayek S.S. Chimeric antigen receptor T-cell therapy for cancer and heart: JACC Council Perspectives. J Am Coll Cardiol. 2019;74:3153–3163.
    1. Hippisley-Cox J., Coupland C., Brindle P. Development and validation of QRISK3 risk prediction algorithms to estimate future risk of cardiovascular disease: prospective cohort study. BMJ. 2017;357:j2099.
    1. Makita S., Imaizumi K., Kurosawa S., Tobinai K. Chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma: opportunities and challenges. Drugs Context. 2019;8:212567.

Source: PubMed

3
Abonnere