Severe cytokine release syndrome is associated with hematologic toxicity following CD19 CAR T-cell therapy

Krishna R Juluri, Qian Vicky Wu, Jenna Voutsinas, Jue Hou, Alexandre V Hirayama, Erin Mullane, Nancy Miles, David G Maloney, Cameron J Turtle, Merav Bar, Jordan Gauthier, Krishna R Juluri, Qian Vicky Wu, Jenna Voutsinas, Jue Hou, Alexandre V Hirayama, Erin Mullane, Nancy Miles, David G Maloney, Cameron J Turtle, Merav Bar, Jordan Gauthier

Abstract

CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has demonstrated remarkable efficacy in patients with relapsed/refractory B-cell malignancies; however, it is associated with toxicities including cytokine release syndrome (CRS), neurotoxicity, and impaired hematopoietic recovery. The latter is associated with high-grade cytopenias requiring extended growth factor or transfusional support, potentially leading to additional complications such as infection or hemorrhage. To date, the factors independently associated with hematologic toxicity have not been well characterized. To address this deficit, we retrospectively analyzed 173 patients who received defined-composition CD19 CAR T-cell therapy in a phase 1/2 clinical trial (https://ichgcp.net/clinical-trials-registry/NCT01865617" title="See in ClinicalTrials.gov">NCT01865617), with primary end points of absolute neutrophil count and platelet count at day-28 after CAR T-cell infusion. We observed cumulative incidences of neutrophil and platelet recovery of 81% and 75%, respectively, at 28 days after infusion. Hematologic toxicity was noted in a significant subset of patients, with persistent neutropenia in 9% and thrombocytopenia in 14% at last follow-up. Using debiased least absolute shrinkage selector and operator regression analysis for high-dimensional modeling and considering patient-, disease-, and treatment-related variables, we identified increased CRS severity as an independent predictor for decreased platelet count and lower prelymphodepletion platelet count as an independent predictor of both decreased neutrophil and platelet counts after CD19 CAR T-cell infusion. Furthermore, multivariable models including CRS-related cytokines identified associations between higher peak serum concentrations of interleukin-6 and lower day-28 cell counts; in contrast, higher serum concentrations of transforming growth factor-β1 were associated with higher counts. Our findings suggest that patient selection and improved CRS management may improve hematopoietic recovery after CD19 CAR T-cell therapy.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Patient selection. Inclusion and exclusion criteria for analysis. Patients excluded from analysis at time of first competing event after CAR T-cell infusion, defined as relapse with marrow involvement, new cytotoxic therapy, second lymphodepletion or CAR T-cell infusion, death, or loss to follow-up.
Figure 2.
Figure 2.
Severity of hematologic toxicities. Percentage of patients with neutropenia (A), thrombocytopenia (B), and anemia (C) by post-CAR T-cell infusion day, stratified by CTCAE grade. Because of the variability in collection dates, for day = n, the minimum cell count falling within an arbitrary range of n was selected for each patient (ie, day 0 = days −7 and 0, day 14 = days 12-16, day-28 = days 23-33, day 60 = days 55-65, day 90 = days 85-95, day 120 = days 110-130, and day 180 = days 160-200). Patients were no longer included in this analysis pending receipt of a subsequent line of therapy, second CAR T-cell infusion, or withdrawal from the study.
Figure 2.
Figure 2.
Severity of hematologic toxicities. Percentage of patients with neutropenia (A), thrombocytopenia (B), and anemia (C) by post-CAR T-cell infusion day, stratified by CTCAE grade. Because of the variability in collection dates, for day = n, the minimum cell count falling within an arbitrary range of n was selected for each patient (ie, day 0 = days −7 and 0, day 14 = days 12-16, day-28 = days 23-33, day 60 = days 55-65, day 90 = days 85-95, day 120 = days 110-130, and day 180 = days 160-200). Patients were no longer included in this analysis pending receipt of a subsequent line of therapy, second CAR T-cell infusion, or withdrawal from the study.
Figure 3.
Figure 3.
Kinetics of hematopoietic recovery. Longitudinal ANC (A) and platelet counts (log scale) (B) are depicted in spaghetti plots, with the application of a nonparametric smoother to subjects grouped by CRS grade.
Figure 4.
Figure 4.
Multivariable analysis of factors associated with day-28 ANC and platelet counts. Forest plots of regression coefficients for day-28 neutrophil (A,C) or platelet (B,D) counts determined by high-dimensional inference for selected patient, disease, and treatment characteristics (A-B) or serum cytokine concentrations (C-D). Regression coefficient and associated 95% CI, denoted by circles and lines from a linear regression model, respectively. P-value of regression coefficient denoted by color gradient. CRS and ICANS variables are stratified by grade. Disease cohorts (CLL and NHL) and sex (female) are compared against a reference variable, ALL and male, respectively. All other variables are modeled as continuous variables. ALC, absolute lymphocyte count; IFN, interferon; MCP, monocyte chemoattractant protein; MIP, macrophage inflammatory protein; PT, prothrombin time; PTT, partial thromboplastin time, TIM, T-cell immunoglobulin and mucin domain-containing protein; TNF, tumor necrosis factor; sTNFR, soluble TNF-receptor.
Figure 5.
Figure 5.
Cumulative incidence of hematopoietic recovery. Cumulative incidence of neutrophil (A,C) and platelet (B,D) recovery, as defined by CIBMTR criteria. Cumulative incidence estimated by the Kalbfleisch and Prentice method with univariate comparisons across categories according to Gray’s test and stratified by entire cohort (A-B) or by pre-LD platelet count (C-D, grouped by quartiles). Shaded areas represent 95% CIs.

References

    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, Hay KA, Hanafi LA, et al. . Durable Molecular Remissions in Chronic Lymphocytic Leukemia Treated With CD19-Specific Chimeric Antigen Receptor-Modified T Cells After Failure of Ibrutinib. J Clin Oncol. 2017;35(26):3010-3020.
    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. Wang M, Munoz J, Goy A, et al. . KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N Engl J Med. 2020;382(14):1331-1342.
    1. Locke FL, Ghobadi A, Jacobson CA, et al. . 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(1):31-42.
    1. Neelapu SS, Locke FL, Bartlett NL, et al. . Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N Engl J Med. 2017;377(26):2531-2544.
    1. Schuster SJ, Bishop MR, Tam CS, et al. ; JULIET Investigators . Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med. 2019;380(1):45-56.
    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. Abramson JS, Palomba ML, Gordon LI, et al. . Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839-852.
    1. Hirayama AV, Gauthier J, Hay KA, et al. . High rate of durable complete remission in follicular lymphoma after CD19 CAR-T cell immunotherapy. Blood. 2019;134(7):636-640.
    1. Cordeiro A, Bezerra ED, Hirayama AV, et al. . Late Events after Treatment with CD19-Targeted Chimeric Antigen Receptor Modified T Cells. Biol Blood Marrow Transplant. 2020;26(1):26-33.
    1. Hirayama AV, Turtle CJ. Toxicities of CD19 CAR-T cell immunotherapy. Am J Hematol. 2019;94(S1):S42-S49.
    1. Hill JA, Li D, Hay KA, et al. . Infectious complications of CD19-targeted chimeric antigen receptor-modified T-cell immunotherapy. Blood. 2018; 131(1):121-130.
    1. Logue JM, Zucchetti E, Bachmeier CA, et al. . Immune reconstitution and associated infections following axicabtagene ciloleucel in relapsed or refractory large B-cell lymphoma. Haematologica. 2021;106(4)978-986.
    1. Brudno JN, Maric I, Hartman SD, et al. . T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma. J Clin Oncol. 2018;36(22):2267-2280.
    1. Shalabi H, Shah NN, Fry TJ, Yates B, Delbrook C. Chimeric antigen receptor induced cytopenia differs from chemotherapy induced myelosuppression [abstract]. Blood. 2017;130(suppl 1). Abstract 5048.
    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(5):439-448.
    1. Wudhikarn K, Palomba ML, Pennisi M, et al. . Infection during the first year in patients treated with CD19 CAR T cells for diffuse large B cell lymphoma. Blood Cancer J. 2020;10(8):79.
    1. Hay KA, Hanafi LA, Li D, et al. . Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017;130(21):2295-2306.
    1. Fried S, Avigdor A, Bielorai B, et al. . Early and late hematologic toxicity following CD19 CAR-T cells. Bone Marrow Transplant. 2019;54(10):1643-1650.
    1. Nahas GR, Komanduri KV, Pereira D, et al. . Incidence and risk factors associated with a syndrome of persistent cytopenias after CAR-T cell therapy (PCTT). Leuk Lymphoma. 2020;61(4):940-943.
    1. Jain T, Knezevic A, Pennisi M, et al. . Hematopoietic recovery in patients receiving chimeric antigen receptor T-cell therapy for hematologic malignancies. Blood Adv. 2020;4(15):3776-3787.
    1. Center for International Blood & Marrow Transplant Research. Cellular Therapy Manuals: Peripheral Blood Count Recovery. Milwaukee, WI: CIBMTR; Last modified 22 January 2021. Available at: /.
    1. Lee DW, Gardner R, Porter DL, et al. . Current concepts in the diagnosis and management of cytokine release syndrome [published correction appears in Blood. 2015;126(8):1048]. Blood. 2014;124(2):188-195.
    1. Tibshirani R. Regression Shrinkage and Selection via the Lasso. J R Stat Soc B. 1996;58(1):267-288.
    1. Schemper M, Smith TL. A note on quantifying follow-up in studies of failure time. Control Clin Trials. 1996;17(4):343-346.
    1. Kalbfleisch JD, Prentice RL. The statistical analysis of failure time data. New York: John Wiley & Sons, Inc.; 1980.
    1. Fine JP, Gray RJ. A Proportional Hazards Model for the Subdistribution of a Competing Risk. J Am Stat Assoc. 1999;94(446):496-509.
    1. Gray RJ. A Class of $K$-Sample Tests for Comparing the Cumulative Incidence of a Competing Risk. Ann Stat. 1988;16(3):1141-1154.
    1. Fu Z, Parikh CR, Zhou B. Penalized variable selection in competing risks regression. Lifetime Data Anal. 2017;23(3):353-376.
    1. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood. 2016;127(26):3321-3330.
    1. Rodríguez MdC, Bernad A, Aracil M. Interleukin-6 deficiency affects bone marrow stromal precursors, resulting in defective hematopoietic support. Blood. 2004;103(9):3349-3354.
    1. Feng X, Scheinberg P, Wu CO, et al. . Cytokine signature profiles in acquired aplastic anemia and myelodysplastic syndromes. Haematologica. 2011;96(4):602-606.
    1. Tie R, Li H, Cai S, et al. . Interleukin-6 signaling regulates hematopoietic stem cell emergence. Exp Mol Med. 2019;51(10):1-12.
    1. Zhao JL, Ma C, O’Connell RM, et al. . Conversion of danger signals into cytokine signals by hematopoietic stem and progenitor cells for regulation of stress-induced hematopoiesis. Cell Stem Cell. 2014;14(4):445-459.
    1. Assoian RK, Komoriya A, Meyers CA, Miller DM, Sporn MB. Transforming growth factor-beta in human platelets. Identification of a major storage site, purification, and characterization. J Biol Chem. 1983;258(11):7155-7160.
    1. Oh SA, Li MO. TGF-β: guardian of T cell function. J Immunol. 2013;191(8):3973-3979.
    1. Blank U, Karlsson S. TGF-β signaling in the control of hematopoietic stem cells. Blood. 2015;125(23):3542-3550.
    1. Challen GA, Boles NC, Chambers SM, Goodell MA. Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-beta1. Cell Stem Cell. 2010;6(3):265-278.
    1. Santomasso BD, Park JH, Salloum D, et al. . Clinical and Biological Correlates of Neurotoxicity Associated with CAR T-cell Therapy in Patients with B-cell Acute Lymphoblastic Leukemia. Cancer Discov. 2018;8(8):958-971.
    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.

Source: PubMed

3
Abonnere