Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 May 1.
Published in final edited form as: Curr Res Transl Med. 2018 Apr 3;66(2):50–52. doi: 10.1016/j.retram.2018.03.003

Insights into cytokine release syndrome and neurotoxicity after CD19-specific CAR-T cell therapy

Jordan Gauthier 1, Cameron J Turtle 1,2
PMCID: PMC5967886  NIHMSID: NIHMS957691  PMID: 29625831

Abstract

T-cells engineered to express CD19-specific chimeric antigen receptors (CD19 CAR-T cells) can achieve high response rates in patients with refractory/relapsed (R/R) CD19+ hematologic malignancies. Nonetheless, the efficacy of CD19-specific CAR-T cell therapy can be offset by significant toxicities, such as cytokine release syndrome (CRS) and neurotoxicity. In this report of our presentation at the 2018 Second French International Symposium on CAR-T cells (CAR-T day), we describe the clinical presentations of CRS and neurotoxicity in a cohort of 133 adults treated with CD19 CAR-T cells at Fred Hutchinson Cancer Research Center, and provide insights into the mechanisms contributing to these toxicities.

Keywords: review, chimeric antigen receptor therapy, hematological malignancies, solid tumors, adoptive t-cell therapy, cytokine release syndrome, neurotoxicity

Introduction

T-cells engineered to express CD19-specific chimeric antigen receptors (CD19 CAR-T cells) can achieve high response rates in patients with refractory/relapsed (R/R) CD19+ hematologic malignancies. Nonetheless, the efficacy of CAR T-cell therapy can be offset by toxicities, such as cytokine release syndrome (CRS) and neurotoxicity, which could hamper its widespread clinical application.

Efficacy of CD19 CAR-T cell therapy

CAR-T cell therapy has shown remarkable efficacy in patients with R/R CD19+ hematologic malignancies. In R/R ALL, we [1] and others [27] have reported minimal residual disease (MRD)-negative complete remission (CR) rates ranging from 60% [5] to 93% [1]. In R/R non-Hodgkin lymphoma (NHL), CAR T-cell therapy achieved best overall response rates (ORR) of 53% to 82% [812]; in R/R CLL patients, we [13] and others [14] have reported ORR of 74% and 57%, respectively.

CRS and neurotoxicity: clinical presentation and incidence

Upon recognition of CD19-expressing cells (normal B-cells or CD19+ tumor cells) CD19 CAR-T cells proliferate, exert cytotoxic effects against their target cells, and release cytokines that may trigger a systemic inflammatory response. This is thought to initiate CRS, which is characterized clinically by a variety of systemic symptoms and signs, such as fever, hypotension, capillary leak, coagulopathy and occasionally multiorgan failure. The presentation of CRS may differ between distinct CAR-T cell products, but generally occurs within a few days after CAR-T cell infusion. In most clinical trials of CAR-T cell therapy for ALL, the incidences of CRS and severe CRS were above 70% and 15%, respectively [1, 2, 46, 15, 16]. However, in NHL there may be more variability in the incidence of CRS, which was reported at 24% in the TRANSCEND trial [10] and 94% in the ZUMA-1 trial [12]. Comparable anti-tumor efficacy was noted in these trials. Additional studies will be required to confirm these findings and to investigate reasons for the apparent differences in the incidence of CRS.

The incidence of neurotoxicity, defined as any neurological symptoms occurring after CAR-T cells in the absence of another cause, also varies across studies, ranging from 13% [4] to 63% [7] in ALL, and from 7% [10] to 31% [12] in NHL. Neurotoxicity commonly presents with delirium, headache, decreased level of consciousness, or speech impairment. Focal neurologic deficits, seizures, and acute cerebral edema have been reported but are infrequent. Neurotoxicity usually develops after the onset of CRS and can present after its resolution. In almost all cases, neurotoxicity and CRS are reversible.

The FHCRC experience

Recently, we reported a clinical and biological description of CRS [17] and neurotoxicity [18] in 133 patients who underwent CD19 CAR T-cell therapy at our institution to treat ALL, NHL or CLL. The patients received cyclophosphamide +/− fludarabine-based lymphodepletion followed by the infusion of CD19 CAR-T cells formulated in a defined 1:1 ratio of CD4+:CD8+ CAR-T cells. CRS with fever preceded neurotoxicity in a majority of cases. The severity of neurotoxicity was associated with the severity of CRS and accordingly, an early onset of CRS predicted subsequent development of more severe neurotoxicity. CRS of any grade after CD19 CAR-T cell therapy occurred in 70% of patients. The median time to first fever was 2.2 days. We did not observe significant differences in the incidence, severity and clinical presentation of CRS between ALL, NHL and CLL patients, Underscoring the favorable outcome in most patients, only 20% of patients with CRS and/or neurotoxicity received tocilizumab and/or dexamethasone, and CRS was quickly reversible in a majority of cases with a median duration of three days. Ten patients (7.5%) developed grade ≥ 4 CRS and five of these patients died from complications associated with CRS and/or neurotoxicity. All patients with grade ≥ 4 CRS developed non-neurologic organ toxicities. Importantly, a majority of the grade ≥4 CRS and/or neurotoxicity observed in our study occurred during dose finding. Since completion of dose finding, the incidence of these toxicities has fallen.

Higher bone marrow CD19+ tumor burden, severe thrombocytopenia, manufacturing of CAR-T cells from bulk CD8+ T cells rather than central memory CD8+ T cells, Cy/Flu lymphodepletion, and higher CAR-T cell dose were independently associated with all grades of CRS. Severe CRS was associated with higher and earlier CAR-T cell expansion in the blood.

The association between CAR T-cell expansion and toxicity could narrow the therapeutic index of CD19 CAR-T cell therapy. While decreasing the dose of CAR-T cells could prevent toxicity, this could be at the price of a diminished anti-tumor effect. To identify a therapeutic window of CAR-T cell dose leading to maximal efficacy with minimal toxicity, we used logistic regression to model the relationship between peak CAR-T cell counts in the blood and the probability of toxicity, as well as the relationship between peak CAR-T cell counts in the blood and disease response. We observed differences in the shape of these probability curves for disease response depending on the disease subtype. For bone marrow complete response in ALL, the curve was sigmoid, reaching a plateau at approximately 10 CD8+ CAR-T cells/uL, indicating that peak CD8+ CAR-T cell counts > 10 cells/uL might have provided only limited improvement in efficacy. In contrast, the logistic regression analysis showed an increase in CD8+ CAR-T cells beyond 10 cells/uL would likely increase the risks of severe neurotoxicity and CRS. Similar findings were noted for CD4+ CAR-T cells. In NHL patients, the probability curve for complete response did not plateau. Thus, any decrease in peak CAR-T cell counts to decrease toxicity would likely be accompanied by a decrease in efficacy. Other strategies to mitigate toxicity should be investigated in NHL, such as early interventions in patients at high risk for severe CRS. Using classification tree modeling we found that patients with high fever (≥ 38.9°C) and serum MCP-1 concentrations ≥ 1343.5 pg/mL within 36 hours of CAR-T cell infusion were at high risk of developing grade ≥4 CRS. This approach could predict grade 4–5 CRS with high sensitivity (100%) and specificity (84%). We also designed another model predicting grade 4–5 neurotoxicity that incorporated these parameters and serum IL-6 ≥ 16pg/ml. These approaches need to be validated to predict toxicity associated with other CAR-T cell products, in which the kinetics of cytokine levels and clinical presentation may differ.

Pathogenesis of CRS and neurotoxicity

The pathogenesis of CRS and neurotoxicity is incompletely understood. Analysis of patients treated in our study provided insights into the mechanisms contributing to these toxicities [17, 18]. The presence of hypotension, capillary leak, and a consumptive coagulopathy suggested that endothelial dysfunction might be present in severe CRS and/or neurotoxicity. We examined serum angiopoietin-2 and -1 and von Willebrand factor (VWF) concentrations in patients after CAR-T cell infusion and found evidence of endothelial activation during severe CRS and severe neurotoxicity. Serum from patients with severe toxicity contained high concentrations of endothelium-activating cytokines, and activated HUVEC cells in vitro, leading to binding of VWF and platelets. In some patients endothelial activation preceded lymphodepletion and CAR-T cell therapy, indicating that pre-existing endothelial activation might increase the risk of subsequent severe toxicity. We found increased permeability of the blood-brain barrier (BBB) in neurotoxicity and found that the BBB failed to protect the CSF from high concentrations of systemic cytokines. Furthermore, some cytokines could activate and induce stress in pericytes - key supporting components of the BBB. Evidence to support the role of endothelial activation and vascular dysfunction was found in autopsy data from a patient with fatal neurotoxicity, in whom we observed a multifocal thrombotic microangiopathy. It remains to be determined whether this mechanism contributes to the majority of cases of mild and reversible neurotoxicity. Development of robust animal models of neurotoxicity and CRS will be critical to elucidate the mechanisms responsible for severe toxicities after CAR-T cell therapy.

Conclusions

CRS and neurotoxicity after CD19 CAR-T cell therapy are usually of mild-moderate severity and reversible. However, some patients develop severe toxicity associated with robust CAR-T cell expansion and high cytokine concentrations in serum. The finding of endothelial activation in these patients may open new avenues to treat or prevent CAR-T cell associated CRS and/or neurotoxicity.

Acknowledgments

We thank the FHCRC Cell Processing Facility and Seattle Cancer Care Alliance (SCCA) Cell Therapy Laboratory, and the staff of the Program in Immunology and SCCA Immunotherapy Clinic. Funding for this study was provided by: NCI R01 CA136551; NIDDK P30 DK56465; NCI P30 CA15704; Life Science Discovery Fund; Bezos Family Foundation; Juno Therapeutics, Inc.

Footnotes

Disclosure of conflicts of interest

CT: Juno Therapeutics, Seattle Genetics, Precision Biosciences, Adaptive Biotechnologies, Bluebird Bio, Celgene, Gilead Sciences (Consulting or Advisory Role); Juno Therapeutics (Research Funding)

JG: Juno Therapeutics (Research Funding)

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Turtle CJ, Hanafi L-AA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. The Journal of Clinical Investigation. 2016;126(6):2123–38. doi: 10.1172/JCI85309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gardner RA, Finney O, Annesley C, Brakke H, Summers C, Leger K, et al. Intent to treat leukemia remission by CD19CAR T cells of defined formulation and dose in children and young adults. Blood. 2017;129(25):3322–31. doi: 10.1182/blood-2017-02-769208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–17. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. New England Journal of Medicine. 2018;378(5):439–48. doi: 10.1056/NEJMoa1709866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, 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 (London, England) 2015;385(9967):517–28. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Park JH, Riviere I, Gonen M, Wang X, Senechal B, Curran KJ, et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N Engl J Med. 2018;378(5):449–59. doi: 10.1056/NEJMoa1709919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Shah BD, Stock W, Wierda WG, Oluwole O, Holmes H, Schiller GJ, et al. Phase 1 Results of ZUMA-3: KTE-C19, an Anti-CD19 Chimeric Antigen Receptor (CAR) T Cell Therapy, in Adult Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia (R/R ALL) ASH Abstract. 2017 [Google Scholar]
  • 8.Turtle CJ, Hanafi L-AA, Berger C, Hudecek M, Pender B, Robinson E, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Science translational medicine. 2016;8(355):355ra116. doi: 10.1126/scitranslmed.aaf8621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, et al. Primary Analysis of Juliet: A Global, Pivotal, Phase 2 Trial of CTL019 in Adult Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma. ASH Abstract. 2017 [Google Scholar]
  • 10.Abramson JS, Palomba M, Gordon LI, Lunning MA, Arnason JE, Wang M, et al. High durable CR rates in relapsed/refractory (R/R) aggressive B-NHL treated with the CD19-directed CAR T cell product JCAR017 (TRANSCEND NHL 001): defined composition allows for dose-finding and definition of pivotal cohort. ASH Abstract. 2017 [Google Scholar]
  • 11.Kochenderfer JN, Somerville R, Lu T, Shi V, Bot A, Rossi J, et al. Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated With High Serum Interleukin-15 Levels. Journal of Clinical Oncology. 2017;35(16):1803–13. doi: 10.1200/JCO.2016.71.3024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. The New England Journal of Medicine. 2017;377:2531–44. doi: 10.1056/NEJMoa1707447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Turtle CJ, Hay KA, Hanafi L-A, Li D, Cherian S, Chen X, et al. Durable Molecular Remissions in Chronic Lymphocytic Leukemia Treated With CD19-Specific Chimeric Antigen Receptor–Modified T Cells After Failure of Ibrutinib. Journal of Clinical Oncology. 2017 doi: 10.1200/JCO.2017.72.8519. Online Publication. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Porter DL, Hwang TW, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Science Translational Medicine. 2015;7(303):303ra139. doi: 10.1126/scitranslmed.aac5415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England journal of medicine. 2014;371(16):1507–17. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Maude SL, Barrett DM, Rheingold SR, Aplenc R, Teachey DT, Callahan C, et al. Efficacy of Humanized CD19-Targeted Chimeric Antigen Receptor (CAR)-Modified T Cells in Children and Young Adults with Relapsed/Refractory Acute Lymphoblastic Leukemia. ASH Abstract. 2016;128(22):217. [Google Scholar]
  • 17.Hay KA, Hanafi LA, Li D, Gust J, Liles WC, Wurfel MM, et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017;130(21):2295–306. doi: 10.1182/blood-2017-06-793141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gust J, Hay KA, Hanafi L-A, Li D, Myerson D, Gonzalez-Cuyar LF, et al. Endothelial Activation and Blood–Brain Barrier Disruption in Neurotoxicity after Adoptive Immunotherapy with CD19 CAR-T Cells. Cancer Discovery. 2017 doi: 10.1158/2159-8290.CD-17-0698. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES