Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy

Kevin A Hay, Laïla-Aïcha Hanafi, Daniel Li, Juliane Gust, W Conrad Liles, Mark M Wurfel, José A López, Junmei Chen, Dominic Chung, Susanna Harju-Baker, Sindhu Cherian, Xueyan Chen, Stanley R Riddell, David G Maloney, Cameron J Turtle, Kevin A Hay, Laïla-Aïcha Hanafi, Daniel Li, Juliane Gust, W Conrad Liles, Mark M Wurfel, José A López, Junmei Chen, Dominic Chung, Susanna Harju-Baker, Sindhu Cherian, Xueyan Chen, Stanley R Riddell, David G Maloney, Cameron J Turtle

Abstract

Lymphodepletion chemotherapy followed by infusion of CD19-specific chimeric antigen receptor-modified (CAR) T cells has produced impressive antitumor responses in patients with refractory CD19+ B-cell malignancies but is often associated with cytokine release syndrome (CRS). Our understanding of CRS continues to evolve, and identification of the kinetics of CRS and predictive clinical and laboratory biomarkers of severity are needed to evaluate strategies to mitigate toxicity. We report the clinical presentation of and identify biomarkers of severe CRS in 133 adult patients who received CD19 CAR T cells. CRS developed in 70% of patients, including 62.5% with grade 1 to 3 CRS (grade 1, 26%; grade 2, 32%; grade 3, 4.5%), 3.8% with grade 4, and 3.8% with grade 5. A majority of cases of grade ≥4 CRS occurred during CAR T-cell dose finding. Multivariable analysis of baseline characteristics identified high marrow tumor burden, lymphodepletion using cyclophosphamide and fludarabine, higher CAR T-cell dose, thrombocytopenia before lymphodepletion, and manufacturing of CAR T cells without selection of CD8+ central memory T cells as independent predictors of CRS. Severe CRS was characterized by hemodynamic instability, capillary leak, and consumptive coagulopathy. Angiopoietin-2 and von Willebrand factor, which are biomarkers of endothelial activation, were increased during severe CRS and also before lymphodepletion in patients who subsequently developed CRS. We describe a classification-tree algorithm to guide studies of early intervention after CAR T-cell infusion for patients at high risk of severe CRS. These data provide a framework for early intervention studies to facilitate safer application of effective CD19 CAR T-cell therapy.

© 2017 by The American Society of Hematology.

Figures

Figure 1.
Figure 1.
Presentation, management, and outcomes of patients with grade ≥4 CRS. Colors on the swimmer plot indicate the CRS grade on each day through 28 days after CAR T-cell infusion in all patients who developed grade ≥4 CRS. The duration of grade ≥3 neurotoxicity and interventions with tocilizumab and/or corticosteroids are indicated in the figure. ALL-2 developed dialysis-dependent acute kidney injury (AKI) through day 26 followed by resolution of CRS-associated organ toxicity (grade 0) on day 37. ALL-3 died 4 months after CAR T-cell infusion with irreversible neurotoxicity, despite resolution of fever and hypotension associated with CRS on day 13 after CAR T-cell infusion. NHL-1 had ongoing grade 1 AKI at last available laboratory value on day 83. Doses of medications: dexamethasone 10 mg intravenously or orally, methylprednisolone 1g intravenously, tocilizumab 4 to 8 mg/kg intravenously. NT, neurotoxicity.
Figure 2.
Figure 2.
Kinetics of presentation of CRS and neurotoxicity. (A) Cumulative incidence curve for first fever ≥38°C in patients with grade 1 to 3 (n = 82) or grade ≥4 CRS (n = 10). (B) Mean ± standard error of the mean (SEM) of the maximum temperature after CAR T-cell infusion. (C) Incidence and grading of neurotoxicity within each CRS grade. (D) The median time of onset of fever ≥38°C (red, n = 92) or neurotoxicity (blue, n = 53) after CAR T-cell infusion. One patient with grade 2 CRS who developed hypotension without fever is not included. Kruskal-Wallis test ***P < .0001; **P ranges from >.0001 to <.001; *P ranges from >.001 to <.005. d, days after CAR T-cell infusion; h, hours after CAR T-cell infusion; pre-chemo, before the start of lymphodepletion chemotherapy; pre-infusion, before CAR T-cell infusion.
Figure 3.
Figure 3.
Hemodynamic instability and clinical capillary leak in grade ≥4 CRS. (A-G) Mean ± SEM of (A) minimum systolic blood pressure, (B) minimum diastolic blood pressure, (C) maximum heart rate, (D) maximum respiratory rate, (E) minimum serum protein concentration, (F) minimum albumin concentration, and (G) weight gain from the start of lymphodepletion are shown at the indicated times after CAR T-cell infusion. Gray shading indicates the normal range. Kruskal-Wallis test: ***P < .0001; **P ranges from >.0001 to <.001; *P ranges from >.001 to <.005.
Figure 4.
Figure 4.
Hematopoietic toxicity, laboratory coagulopathy, and endothelial injury in grade ≥4 CRS. (A) Minimum absolute neutrophil count, (B) hematocrit, (C) hemoglobin, and (D) platelet count are shown for patients receiving cyclophosphamide/fludarabine lymphodepletion at the indicated times after CAR T-cell infusion (n = 104). (E) Total transfused units of packed red blood cells (pRBC), platelets (Plt), and cryoprecipitate (Cryo) in the first 28 days after CAR T-cell infusion. (F) Maximum PT, (G) maximum aPTT, (H) minimum fibrinogen, and (I) maximum D-dimer concentrations are shown at the indicated times after CAR T-cell infusion. (J) The fold change in VWF concentration in serum from a subset of patients at the peak of CAR T-cell expansion (n = 60; grade 0, n = 12; grade 1-3, n = 39; grade ≥4 CRS, n = 9) compared with the VWF concentration in pooled normal plasma (12.2 μg/mL; CRYOcheck, Precision Biologic, Dartmouth, NS, Canada). (K) The Ang-2:Ang-1 ratio at the peak of CAR T-cell expansion (n = 60; grade 0, n = 12; grade 1-3, n = 39; grade ≥4 CRS, n = 9). For (A-D) and (F-I), data represent the mean ± SEM. P values were determined using the Kruskal-Wallis test. For (E,J,K), each point represents data from 1 patient. The median and IQR are shown. P values were determined using the Wilcoxon test. Gray shading indicates the normal range. ***P < .0001; **P ranges from >.0001 to <.001; *P ranges from >.001 to <.005. Gr, grade.
Figure 5.
Figure 5.
CAR T-cell counts in blood and estimated probabilities of response or toxicity. (A-B) The absolute number and (C-D) percentage of CD8+ (left) and CD4+ (right) CAR T-cells in blood. The mean ± SEM of the maximum values are shown. P values were determined by using the Kruskal-Wallis test. (E-F) Estimated probabilities by logistic regression of grade ≥2 CRS and grade ≥3 neurotoxicity at (E) peak CD8+ and (F) CD4+ CAR T-cell counts in blood. (G-H) Estimated probabilities by logistic regression of bone marrow CR in ALL and CLL patients by flow cytometry and CR or overall response (OR) in NHL patients according to Cheson imaging criteria (2014) at peak (G) CD8+ and (H) CD4+ CAR T-cell counts in blood. Lymph node CR in CLL patients is not depicted because of the limited cohort size available for analysis. P values are color-coded to indicate the association between the CAR T-cell peak counts and outcomes. ***P < .0001; **P ranges from >.0001 to <.001; *P ranges from >.001 to <.005.
Figure 6.
Figure 6.
Biomarkers for early prediction of grade ≥4 CRS. (A-H) Concentrations of listed cytokines in serum obtained from patients at the indicated time points. P values were determined by using the Kruskal-Wallis test. (I) An algorithm for early identification of patients at high risk of grade ≥4 CRS using classification-tree modeling. Early high fever (≥38.9°C) within the first 36 hours after CAR T-cell infusion triggers evaluation of serum MCP-1 concentration. Patients with fever ≥38.9°C and serum MCP-1 ≥1343.5 pg/mL are at high risk for subsequent development of grade ≥4 CRS. ***P < .0001; **P ranges from >.0001 to <.001; *P ranges from >.001 to <.005.

References

    1. Turtle CJ, Hanafi LA, Berger C, et al. . Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8(355):355ra116.
    1. Turtle CJ, Hanafi LA, Berger C, et al. . CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123-2138.
    1. Turtle CJ, Hanafi LA, Li D, et al. . CD19 CAR-T cells are highly effective in ibrutinib-refractory chronic lymphocytic leukemia [abstract]. Blood. 2016;128(22). Abstract 56.
    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(16):1509-1518.
    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(16):1507-1517.
    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(9967):517-528.
    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(6):540-549.
    1. 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.
    1. Davila ML, Rivière 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.
    1. Locke FL, Neelapu SS, Bartlett NL, et al. . Phase 1 Results of ZUMA-1: A Multicenter Study of KTE-C19 Anti-CD19 CAR T Cell Therapy in Refractory Aggressive Lymphoma. Mol Ther. 2017;25(1):285-295.
    1. Hay KA, Turtle CJ. Chimeric Antigen Receptor (CAR) T Cells: Lessons Learned from Targeting of CD19 in B-Cell Malignancies. Drugs. 2017;77(3):237-245.
    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(25):3322-3331.
    1. Brentjens RJ, Davila ML, Riviere I, et al. . CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra38.
    1. Brentjens RJ, Rivière I, Park JH, et al. . Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118(18):4817-4828.
    1. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725-733.
    1. Fitzgerald JC, Weiss SL, Maude SL, et al. . Cytokine release syndrome after chimeric antigen receptor T cell therapy for acute lymphoblastic leukemia. Crit Care Med. 2017;45(2):e124-e131.
    1. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016;127(26):3321-3330.
    1. Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 2014;20(2):119-122.
    1. Wang X, Chang WC, Wong CW, et al. . A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood. 2011;118(5):1255-1263.
    1. 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.
    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(6):664-679.
    1. Valentijn KM, Sadler JE, Valentijn JA, Voorberg J, Eikenboom J. Functional architecture of Weibel-Palade bodies. Blood. 2011;117(19):5033-5043.
    1. Fiedler U, Scharpfenecker M, Koidl S, et al. . The Tie-2 ligand angiopoietin-2 is stored in and rapidly released upon stimulation from endothelial cell Weibel-Palade bodies. Blood. 2004;103(11):4150-4156.
    1. Metcalf DJ, Nightingale TD, Zenner HL, Lui-Roberts WW, Cutler DF. Formation and function of Weibel-Palade bodies. J Cell Sci. 2008;121(Pt 1):19-27.
    1. Mikacenic C, Hahn WO, Price BL, et al. . Biomarkers of endothelial activation are associated with poor outcome in critical illness. PLoS One. 2015;10(10):e0141251.
    1. Page AV, Liles WC. Biomarkers of endothelial activation/dysfunction in infectious diseases. Virulence. 2013;4(6):507-516.
    1. Page AV, Tarr PI, Watkins SL, et al. . Dysregulation of angiopoietin 1 and 2 in Escherichia coli O157:H7 infection and the hemolytic-uremic syndrome. J Infect Dis. 2013;208(6):929-933.
    1. Page AV, Kotb M, McGeer A, Low DE, Kain KC, Liles WC. Systemic dysregulation of angiopoietin-1/2 in streptococcal toxic shock syndrome. Clin Infect Dis. 2011;52(8):e157-e161.
    1. Ricciuto DR, dos Santos CC, Hawkes M, et al. . Angiopoietin-1 and angiopoietin-2 as clinically informative prognostic biomarkers of morbidity and mortality in severe sepsis. Crit Care Med. 2011;39(4):702-710.
    1. Lovegrove FE, Tangpukdee N, Opoka RO, et al. . Serum angiopoietin-1 and -2 levels discriminate cerebral malaria from uncomplicated malaria and predict clinical outcome in African children. PLoS One. 2009;4(3):e4912-e4918.
    1. Darwish I, Liles WC. Emerging therapeutic strategies to prevent infection-related microvascular endothelial activation and dysfunction. Virulence. 2013;4(6):572-582.
    1. Ali SA, Shi V, Maric I, et al. . T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood. 2016;128(13):1688-1700.

Source: PubMed

3
Sottoscrivi