Abstract
Chimeric antigen receptor (CAR) therapy targeting CD19 is an effective treatment for refractory B cell malignancies, especially acute lymphoblastic leukemia (ALL)1. Although a majority of patients will achieve a complete response following a single infusion of CD19-targeted CAR-modified T cells (CD19 CAR T cells)2,3,4, the broad applicability of this treatment is hampered by severe cytokine release syndrome (CRS), which is characterized by fever, hypotension and respiratory insufficiency associated with elevated serum cytokines, including interleukin-6 (IL-6)2,5. CRS usually occurs within days of T cell infusion at the peak of CAR T cell expansion. In ALL, it is most frequent and more severe in patients with high tumor burden2,3,4. CRS may respond to IL-6 receptor blockade but can require further treatment with high dose corticosteroids to curb potentially lethal severity2,3,4,5,6,7,8,9. Improved therapeutic and preventive treatments require a better understanding of CRS physiopathology, which has so far remained elusive. Here we report a murine model of CRS that develops within 2–3 d of CAR T cell infusion and that is potentially lethal and responsive to IL-6 receptor blockade. We show that its severity is mediated not by CAR T cell–derived cytokines, but by IL-6, IL-1 and nitric oxide (NO) produced by recipient macrophages, which enables new therapeutic interventions.
This is a preview of subscription content, access via your institution
Relevant articles
Open Access articles citing this article.
-
Involvement of inflammasomes in tumor microenvironment and tumor therapies
Journal of Hematology & Oncology Open Access 17 March 2023
-
CAR-cell therapy in the era of solid tumor treatment: current challenges and emerging therapeutic advances
Molecular Cancer Open Access 30 January 2023
-
Small-Molecule Compounds Boost CAR-T Cell Therapy in Hematological Malignancies
Current Treatment Options in Oncology Open Access 26 January 2023
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 per month
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Rent or buy this article
Get just this article for as long as you need it
$39.95
Prices may be subject to local taxes which are calculated during checkout




References
Sadelain, M., Rivière, I. & Riddell, S. Therapeutic T cell engineering. Nature 545, 423–431 (2017).
Davila, M. L. et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci. Transl. Med. 6, 224ra25 (2014).
Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).
Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).
Barrett, D. M., Teachey, D. T. & Grupp, S. A. Toxicity management for patients receiving novel T-cell engaging therapies. Curr. Opin. Pediatr. 26, 43–49 (2014).
Bonifant, C. L., Jackson, H. J., Brentjens, R. J. & Curran, K. J. Toxicity and management in CAR T-cell therapy. Mol. Ther. Oncolytics 3, 16011 (2016).
Brudno, J. N. & Kochenderfer, J. N. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 127, 3321–3330 (2016).
Oluwole, O. O. & Davila, M. L. At the bedside: clinical review of chimeric antigen receptor (CAR) T cell therapy for B cell malignancies. J. Leukoc. Biol. 100, 1265–1272 (2016).
Neelapu, S. S. et al. Chimeric antigen receptor T-cell therapy—assessment and management of toxicities. Nat. Rev. Clin. Oncol. 15, 47–62 (2018).
Lee, D. W. 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 385, 517–528 (2015).
Teachey, D. T. et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 6, 664–679 (2016).
Hay, K. A. et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood 130, 2295–2306 (2017).
Cray, C., Zaias, J. & Altman, N. H. Acute phase response in animals: a review. Comp. Med. 59, 517–526 (2009).
Meek, R. L., Eriksen, N. & Benditt, E. P. Murine serum amyloid A3 is a high density apolipoprotein and is secreted by macrophages. Proc. Natl Acad. Sci. USA 89, 7949–7952 (1992).
Vandevelde, M., Higgins, R. & Overmann A. Veterinary Neuropathology: Essentials of Theory and Practice. in Veterinary Neuropathology: Essentials of Theory and Practice 29 (Wiley, Hoboken, NJ, USA, 2012).
Gust, J. et al. Endothelial activation and blood–brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov. 7, 1404–1419 (2017).
Rincon, M. Interleukin-6: from an inflammatory marker to a target for inflammatory diseases. Trends Immunol. 33, 571–577 (2012).
Quezada, S. A., Jarvinen, L. Z., Lind, E. F. & Noelle, R. J. CD40/CD154 interactions at the interface of tolerance and immunity. Annu. Rev. Immunol. 22, 307–328 (2004).
Bossen, C. et al. Interactions of tumor necrosis factor (TNF) and TNF receptor family members in the mouse and human. J. Biol. Chem. 281, 13964–13971 (2006).
Tucker, T. A. & Schwiebert, L. M. CD40 ligation decreases its protein half-life at the cell surface. Eur. J. Immunol. 38, 864–869 (2008).
Wang, H. et al. A novel role of the scaffolding protein JLP in tuning CD40-induced activation of dendritic cells. Immunobiology 218, 835–843 (2013).
Wang, H. M. et al. Scaffold protein JLP is critical for CD40 signaling in B lymphocytes. J. Biol. Chem. 290, 5256–5266 (2015).
Elgueta, R. et al. Molecular mechanism and function of CD40/CD40L engagement in the immune system. Immunol. Rev. 229, 152–172 (2009).
Murray, P. J. & Wynn, T. A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 11, 723–737 (2011).
Ohashi, Y. et al. Hypotension and reduced nitric oxide–elicited vasorelaxation in transgenic mice overexpressing endothelial nitric oxide synthase. J. Clin. Invest. 102, 2061–2071 (1998).
Titheradge, M. A. Nitric oxide in septic shock. Biochim. Biophys. Acta 1411, 437–455 (1999).
Moore, W. M. et al. L-N6-(1-iminoethyl)lysine: a selective inhibitor of inducible nitric oxide synthase. J. Med. Chem. 37, 3886–3888 (1994).
Garvey, E. P. et al. 1400W is a slow, tight binding, and highly selective inhibitor of inducible nitric-oxide synthase in vitro and in vivo. J. Biol. Chem. 272, 4959–4963 (1997).
Dinarello, C. A. Immunological and inflammatory functions of the interleukin-1 family. Annu. Rev. Immunol. 27, 519–550 (2009).
Saini, A. S., Shenoy, G. N., Rath, S., Bal, V. & George, A. Inducible nitric oxide synthase is a major intermediate in signaling pathways for the survival of plasma cells. Nat. Immunol. 15, 275–282 (2014).
Sims, J. E. & Smith, D. E. The IL-1 family: regulators of immunity. Nat. Rev. Immunol. 10, 89–102 (2010).
Zhao, Z. et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell 28, 415–428 (2015).
van der Stegen, S. J. et al. Preclinical in vivo modeling of cytokine release syndrome induced by ErbB-retargeted human T cells: identifying a window of therapeutic opportunity? J. Immunol. 191, 4589–4598 (2013).
Dinarello, C. A., Simon, A. & van der Meer, J. W. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat. Rev. Drug Discov. 11, 633–652 (2012).
Shultz, L. D. et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J. Immunol. 154, 180–191 (1995).
Serreze, D. V., Gaedeke, J. W. & Leiter, E. H. Hematopoietic stem-cell defects underlying abnormal macrophage development and maturation in NOD/Lt mice: defective regulation of cytokine receptors and protein kinase C. Proc. Natl Acad. Sci. USA 90, 9625–9629 (1993).
Ohteki, T., Suzue, K., Maki, C., Ota, T. & Koyasu, S. Critical role of IL-15–IL-15R for antigen-presenting cell functions in the innate immune response. Nat. Immunol. 2, 1138–1143 (2001).
Gutierrez, E. G., Banks, W. A. & Kastin, A. J. Blood-borne interleukin-1 receptor antagonist crosses the blood–brain barrier. J. Neuroimmunol. 55, 153–160 (1994).
Liu, J., Zhao, M. L., Brosnan, C. F. & Lee, S. C. Expression of type II nitric oxide synthase in primary human astrocytes and microglia: role of IL-1β and IL-1 receptor antagonist. J. Immunol. 157, 3569–3576 (1996).
Tarassishin, L., Suh, H. S. & Lee, S. C. LPS and IL-1 differentially activate mouse and human astrocytes: role of CD14. Glia 62, 999–1013 (2014).
Gade, T. P. et al. Targeted elimination of prostate cancer by genetically directed human T lymphocytes. Cancer Res. 65, 9080–9088 (2005).
Maher, J., Brentjens, R. J., Gunset, G., Rivière, I. & Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ /CD28 receptor. Nat. Biotechnol. 20, 70–75 (2002).
Rivière, I., Brose, K. & Mulligan, R. C. Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells. Proc. Natl Acad. Sci. USA 92, 6733–6737 (1995).
Gong, M. C. et al. Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen. Neoplasia 1, 123–127 (1999).
Acknowledgements
We thank the Alexander S. Onassis Public Benefit Foundation for their support (T.G.). We thank the following MSK core facilities for their outstanding support: flow cytometry core facility, laboratory of comparative pathology, animal facility, integrated genomics operation and bioinformatics core. We thank G. Gunset, Z. Zhao, A. Dobrin and P. Lindenbergh for their assistance with some experiments. This study was supported by Juno Therapeutics and the MSK Cancer Center Support Grant/Core Grant (P30 CA008748).
Author information
Authors and Affiliations
Contributions
T.G. designed the study, performed experiments, analyzed and interpreted the data and wrote the manuscript. S.J.C.v.d.S. helped design and perform in vivo experiments. J.E. performed in vivo experiments. M.H. performed in vivo experiments. A.P. performed and interpreted histopathological studies. M.S. designed the study, analyzed and interpreted the data and wrote the manuscript.
Corresponding author
Ethics declarations
Competing interests
A patent application on CRS prevention listing T.G. and M.S. as co-inventors has been filed by MSK.
Additional information
Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
About this article
Cite this article
Giavridis, T., van der Stegen, S.J.C., Eyquem, J. et al. CAR T cell–induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 24, 731–738 (2018). https://doi.org/10.1038/s41591-018-0041-7
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41591-018-0041-7
This article is cited by
-
CAR-cell therapy in the era of solid tumor treatment: current challenges and emerging therapeutic advances
Molecular Cancer (2023)
-
Involvement of inflammasomes in tumor microenvironment and tumor therapies
Journal of Hematology & Oncology (2023)
-
Advancing CAR T cell therapy through the use of multidimensional omics data
Nature Reviews Clinical Oncology (2023)
-
Humanized mouse models for immuno-oncology research
Nature Reviews Clinical Oncology (2023)
-
RUNX3 improves CAR-T cell phenotype and reduces cytokine release while maintaining CAR-T function
Medical Oncology (2023)