Investigation of the risk factors to predict cytokine release syndrome in relapsed or refractory B-cell acute lymphoblastic leukemia patients receiving IL-6 knocking down anti-CD19 chimeric antigen receptor T-cell therapy

Wen-Jie Gong, Yan Qiu, Ming-Hao Li, Li-Yun Chen, Yan-Yan Li, Jing-Qiu Yu, Li-Qing Kang, Ai-Ning Sun, De-Pei Wu, Lei Yu, Sheng-Li Xue, Wen-Jie Gong, Yan Qiu, Ming-Hao Li, Li-Yun Chen, Yan-Yan Li, Jing-Qiu Yu, Li-Qing Kang, Ai-Ning Sun, De-Pei Wu, Lei Yu, Sheng-Li Xue

Abstract

CD19 chimeric antigen receptor-T (CAR-T) cell therapy has achieved remarkable results in patients with relapsed or refractory B-cell acute lymphoblastic leukemia (r/r B-ALL). However, the cytokine release syndrome (CRS) was presented in most patients as common toxicity and severe CRS (sCRS) characterized by the sharp increase in interleukin-6 (IL-6) could be life-threatening. We conducted a phase II clinical trial of ssCAR-T-19 cells, anti-CD19 CAR-T cells with shRNA targeting IL-6, in 61 patients with r/r B-ALL. This trial was registered at www.clinicaltrials.gov as #NCT03275493. Fifty-two patients achieved CR while nine patients were considered NR. The median duration of response (DOR) and overall survival (OS) were not reached (>50 months). CRS developed in 81.97% of patients, including 54.10% with grades 1 to 2 (grade 1, 31.15%; grade 2, 22.95%) and 27.87% with grades 3 to 4 (grade 3, 26.23%; grade 4, 1.64%). sCRS occurs earlier than mild CRS (mCRS). A multivariable analysis of baseline characteristics identified high bone marrow disease burden and poor genetic risk before infusion as independent risk factors for sCRS. After infusion, patients with sCRS exhibited larger expansion of ssCAR-T-19 cells, higher peak levels of IL-6, IL-10, and IFN-γ, and suffered more severe hematological and non-hematological toxicities compared with those with mCRS.

Keywords: IL-6 knocking down; chimeric antigen receptor T-cell therapy; cytokine release syndrome; relapsed or refractory B-cell acute lymphoblastic leukemia; risk factors.

Conflict of interest statement

Authors M-HLi, L-QKang, and LYu were employed by the Shanghai Unicar-Therapy Bio-Medicine Technology Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Gong, Qiu, Li, Chen, Li, Yu, Kang, Sun, Wu, Yu and Xue.

Figures

Figure 1
Figure 1
Duration of response and overall survival. (A) Kaplan–Meier estimates of the duration of response (DOR) in patients who achieve CR after ssCAR-T-19 infusion with censoring patients at subsequent allogeneic stem-cell transplant. (B) Kaplan–Meier estimate of overall survival (OS) in all the patients without censoring of patients at subsequent allogeneic stem-cell transplant. Median DOR in patients who achieved CR and median OS in all the patients were not reached. Dashed lines in (A) and (B) denote the 95% confidence interval.
Figure 2
Figure 2
Analysis of patient characteristics associated with the severity of CRS. (A, B) Comparison of bone marrow disease burden or MRD between patients with mCRS and sCRS. All the patients were evaluated for bone marrow disease burden and MRD before ssCAR-T-19 cell infusion. Mean values were calculated for each group. P-values were calculated using Mann–Whitney U test. **means p <0.01, *p-values <0.05. (C, D) The test for linear trend between bone marrow disease burden or MRD and the severity of CRS. Bone marrow disease burden and MRD were divided into four groups and the CRS were graded into five levels. The size of the circle represented the number of patients. Mantel–Haenszel chi-square test was used to calculate the linear trend.
Figure 3
Figure 3
The ROC curve for the two-variable regression model. Sixty-one r/r B-ALL patients receiving ssCAR-T-19 therapy were enrolled in the forward-selected logistic regression model. The model was used to predict which patients would develop sCRS after ssCAR-T-19 cell infusion. The logit(p) function transformed the logistic regression score into the predicted probability of the case model. Logit (p) = ln (p/1 − p). The ROC curve was drawn using the logistic regression score. The severity of CRS was predicted using bone marrow disease burden and genetic risk of patients with r/r B-ALL before ssCAR-T-19 infusion. The sensitivity was 70.6%, the specificity was 86.4%, and AUC was 0.785.
Figure 4
Figure 4
Peak cytokines, CRP, ferritin after ssCAR-T-19 infusion. (A–C) Levels of peak cytokines, CRP and ferritin of the patients after ssCAR-T-19 infusion. P-values were calculated using t-test. ****means p-values <0.0001, ***p <0.001, and **p <0.01. (A) IL6, IL10, and IFN-γ, (B) CRP and, (C) ferritin are shown in patients with mCRS and sCRS, respectively. The error bars in (A–C) represent mean ± SD.
Figure 5
Figure 5
Hematologic and non-hematologic toxicities after ssCAR-T-19 infusion. (A–G) Hematologic and non-hematologic toxicities after ssCAR-T-19 infusion. P-values were calculated using t-test. ****means p-values <0.0001, ***p <0.001, **p <0.01, and *p <0.05. (A) Minimum absolute neutrophil count, (B) hemoglobin (Hb) and platelet count (PLT), (C) maximum PT and APTT, (D) minimum fibrinogen (FIB), (E) maximum total bilirubin (TB), serum aspartate aminotransferase (AST), and gamma-glutamyl transpeptidase (GGT), (F) creatinine (Cr), and (G) N-terminal pro-brain natriuretic peptide (NT-proBNP) is shown in patients with mCRS and sCRS, respectively. The upper and lower boundaries of each box indicate the 25th and 75th percentiles. The middle horizontal lines represent the median values and the whiskers mean the minimum and maximum.
Figure 6
Figure 6
Expansion and persistence of ssCAR-T-19 cell in peripheral blood. The copies of ssCAR-T-19 cells in peripheral blood measured by qRT-PCR after infusion. Forty-eight patients with complete expansion data were included. Patients who achieved CR were shown in black while patients who achieved NR were shown in red.
Figure 7
Figure 7
Relationship between peak copies of ssCAR-T-19 cell, bone marrow disease burden and CRS grade in r/r B-LL patients. (A) Comparison of ssCAR-T-19 cell expansion between patients with mCRS and sCRS. Mean values were calculated for each group. P-value was calculated using a Mann–Whitney U test. **means p-value <0.01. (B) Relationship between peak copies of ssCAR-T-19 cell, bone marrow disease burden, and CRS grade in r/r B-ALL patients. The bone marrow disease burden was divided into four groups. The y-axis represented the peak copies of the ssCAR-T-19 cells within 14 days after infusion. Mean values were calculated for each group and error bars indicate standard deviation. The different shapes of icons represented the grade of CRS. Red represented patients who achieved CR while blue represented patients who had no remission.

References

    1. Li L, Liu J, Xu M, Yu H, Lv C, Cao F, et al. . Treatment response, survival, safety, and predictive factors to chimeric antigen receptor T cell therapy in Chinese relapsed or refractory b cell acute lymphoblast leukemia patients. Cell Death Dis (2020) 11(3):207. doi: 10.1038/s41419-020-2388-1
    1. 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
    1. Ying Z, Huang XF, Xiang X, Liu Y, Kang X, Song Y, et al. . A safe and potent anti-CD19 CAR T cell therapy. Nat Med (2019) 25(6):947–53. doi: 10.1038/s41591-019-0421-7
    1. O'Leary MC, Lu X, Huang Y, Lin X, Mahmood I, Przepiorka D, et al. . FDA Approval summary: Tisagenlecleucel for treatment of patients with relapsed or refractory b-cell precursor acute lymphoblastic leukemia. Clin Cancer Res (2019) 25(4):1142–6. doi: 10.1158/1078-0432.CCR-18-2035
    1. Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, et al. . Lisocabtagene maraleucel for patients with relapsed or refractory large b-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet (London England) (2020) 396(10254):839–52. doi: 10.1016/s0140-6736(20)31366-0
    1. Bouchkouj N, Kasamon YL, de Claro RA, George B, Lin X, Lee S, et al. . FDA Approval summary: Axicabtagene ciloleucel for relapsed or refractory Large b-cell lymphoma. Clin Cancer Res (2019) 25(6):1702–8. doi: 10.1158/1078-0432.CCR-18-2743
    1. Wang M, Munoz J, Goy A, Locke FL, Jacobson CA, Hill BT, et al. . KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med (2020) 382(14):1331–42. doi: 10.1056/NEJMoa1914347
    1. Fowler NH, Dickinson M, Dreyling M, Martinez-Lopez J, Kolstad A, Butler J, et al. . Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial. Nat Med (2022) 28(2):325–32. doi: 10.1038/s41591-021-01622-0
    1. Zhong XS, Matsushita M, Plotkin J, Riviere I, Sadelain M. Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther (2010) 18(2):413–20. doi: 10.1038/mt.2009.210
    1. Mestermann K, Giavridis T, Weber J, Rydzek J, Frenz S, Nerreter T, et al. . The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci Trans Med (2019) 11(499). doi: 10.1126/scitranslmed.aau5907
    1. Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, et al. . Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discovery (2016) 6(6):664–79. doi: 10.1158/-16-0040
    1. Park JH, Romero FA, Taur Y, Sadelain M, Brentjens RJ, Hohl TM, et al. . Cytokine release syndrome grade as a predictive marker for infections in patients with relapsed or refractory b-cell acute lymphoblastic leukemia treated with chimeric antigen receptor T cells. Clin Infect Dis an Off Publ Infect Dis Soc America (2018) 67(4):533–40. doi: 10.1093/cid/ciy152
    1. 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. N Engl J Med (2017) 377(26):2531–44. doi: 10.1056/NEJMoa1707447
    1. 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. N Engl J Med (2018) 378(5):439–48. doi: 10.1056/NEJMoa1709866
    1. Schmidts A, Wehrli M, Maus MV. Toward better understanding and management of CAR-T cell-associated toxicity. Annu Rev Med (2021) 72:365–82. doi: 10.1146/annurev-med-061119-015600
    1. 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
    1. Shimabukuro-Vornhagen A, Godel P, Subklewe M, Stemmler HJ, Schlosser HA, Schlaak M, et al. . Cytokine release syndrome. J Immunother Cancer (2018) 6(1):56. doi: 10.1186/s40425-018-0343-9
    1. Gust J, Hay KA, Hanafi LA, 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) 7(12):1404–19. doi: 10.1158/-17-0698
    1. Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. . Chimeric antigen receptor T-cell therapy — assessment and management of toxicities. Nat Rev Clin Oncol (2017) 15(1):47–62. doi: 10.1038/nrclinonc.2017.148
    1. Kotch C, Barrett D, Teachey DT. Tocilizumab for the treatment of chimeric antigen receptor T cell-induced cytokine release syndrome. Expert Rev Clin Immunol (2019) 15(8):813–22. doi: 10.1080/1744666X.2019.1629904
    1. Porter DL, Hwang WT, 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. Sci Trans Med (2015) 7(303):303ra139. doi: 10.1126/scitranslmed.aac5415
    1. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. . Efficacy and toxicity management of 19-28z CAR T cell therapy in b cell acute lymphoblastic leukemia. Sci Trans Med (2014) 6(224):224ra25. doi: 10.1126/scitranslmed.3008226
    1. Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J, 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):4817–28. doi: 10.1182/blood-2011-04-348540
    1. Lee DW, Santomasso BD, Locke FL, Ghobadi A, Turtle CJ, Brudno JN, et al. . ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood marrow Transplant J Am Soc Blood Marrow Transplant (2019) 25(4):625–38. doi: 10.1016/j.bbmt.2018.12.758
    1. von Stackelberg A, Locatelli F, Zugmaier G, Handgretinger R, Trippett TM, Rizzari C, et al. . Phase I/Phase II study of blinatumomab in pediatric patients with Relapsed/Refractory acute lymphoblastic leukemia. J Clin Oncol Off J Am Soc Clin Oncol (2016) 34(36):4381–9. doi: 10.1200/jco.2016.67.3301
    1. Kantarjian H, Stein A, Gökbuget N, Fielding AK, Schuh AC, Ribera JM, et al. . Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med (2017) 376(9):836–47. doi: 10.1056/NEJMoa1609783
    1. Ortiz-Maldonado V, Rives S, Castella M, Alonso-Saladrigues A, Benitez-Ribas D, Caballero-Banos M, et al. . CART19-BE-01: A multicenter trial of ARI-0001 cell therapy in patients with CD19(+) Relapsed/Refractory malignancies. Mol Ther (2021) 29(2):636–44. doi: 10.1016/j.ymthe.2020.09.027
    1. Gu R, Liu F, Zou D, Xu Y, Lu Y, Liu B, et al. . Efficacy and safety of CD19 CAR T constructed with a new anti-CD19 chimeric antigen receptor in relapsed or refractory acute lymphoblastic leukemia. J Hematol Oncol (2020) 13(1):122. doi: 10.1186/s13045-020-00953-8
    1. 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 (2015) 385(9967):517–28. doi: 10.1016/s0140-6736(14)61403-3
    1. 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
    1. Curran KJ, Margossian SP, Kernan NA, Silverman LB, Williams DA, Shukla N, et al. . Toxicity and response after CD19-specific CAR T-cell therapy in pediatric/young adult relapsed/refractory b-ALL. Blood (2019) 134(26):2361–8. doi: 10.1182/blood.2019001641
    1. Turtle CJ, Hanafi LA, 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. J Clin Invest (2016) 126(6):2123–38. doi: 10.1172/jci85309
    1. Levine JE, Grupp SA, Pulsipher MA, Dietz AC, Rives S, Myers GD, et al. . Pooled safety analysis of tisagenlecleucel in children and young adults with b cell acute lymphoblastic leukemia. J Immunother Cancer (2021) 9(8). doi: 10.1136/jitc-2020-002287
    1. Shah BD, Ghobadi A, Oluwole OO, Logan AC, Boissel N, Cassaday RD, et al. . KTE-X19 for relapsed or refractory adult b-cell acute lymphoblastic leukaemia: Phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet (London England) (2021) 398(10299):491–502. doi: 10.1016/s0140-6736(21)01222-8
    1. Yan Z, Zhang H, Cao J, Zhang C, Liu H, Huang H, et al. . Characteristics and risk factors of cytokine release syndrome in chimeric antigen receptor T cell treatment. Front Immunol (2021) 12:611366. doi: 10.3389/fimmu.2021.611366
    1. Sterner RM, Sakemura R, Cox MJ, Yang N, Khadka RH, Forsman CL, et al. . GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood (2019) 133(7):697–709. doi: 10.1182/blood-2018-10-881722
    1. Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, et al. . Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med (2018) 24(6):739–48. doi: 10.1038/s41591-018-0036-4
    1. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med (2018) 24(6):731–8. doi: 10.1038/s41591-018-0041-7
    1. Kang L, Tang X, Zhang J, Li M, Xu N, Qi W, et al. . Interleukin-6-knockdown of chimeric antigen receptor-modified T cells significantly reduces IL-6 release from monocytes. Exp Hematol Oncol (2020) 9:11. doi: 10.1186/s40164-020-00166-2
    1. Tan AHJ, Vinanica N, Campana D. Chimeric antigen receptor-T cells with cytokine neutralizing capacity. Blood Adv (2020) 4(7):1419–31. doi: 10.1182/bloodadvances.2019001287
    1. Xue L, Yi Y, Xu Q, Wang L, Yang X, Zhang Y, et al. . Chimeric antigen receptor T cells self-neutralizing IL6 storm in patients with hematologic malignancy. Cell Discovery (2021) 7(1):84. doi: 10.1038/s41421-021-00299-6
    1. Liu ZF, Chen LY, Wang J, Kang LQ, Tang H, Zhou Y, et al. . Successful treatment of acute b lymphoblastic leukemia relapse in the skin and testicle by anti-CD19 CAR-T with IL-6 knocking down: A case report. biomark Res (2020) 8:12. doi: 10.1186/s40364-020-00193-5
    1. Chen LY, Kang LQ, Zhou HX, Gao HQ, Zhu XF, Xu N, et al. . Successful application of anti-CD19 CAR-T therapy with IL-6 knocking down to patients with central nervous system b-cell acute lymphocytic leukemia. Transl Oncol (2020) 13(11):100838. doi: 10.1016/j.tranon.2020.100838
    1. Frey NV, Shaw PA, Hexner EO, Pequignot E, Gill S, Luger SM, et al. . Optimizing chimeric antigen receptor T-cell therapy for adults with acute lymphoblastic leukemia. J Clin Oncol Off J Am Soc Clin Oncol (2020) 38(5):415–22. doi: 10.1200/jco.19.01892
    1. Tedesco VE, Mohan C. Biomarkers for predicting cytokine release syndrome following CD19-targeted CAR T cell therapy. J Immunol (2021) 206(7):1561–8. doi: 10.4049/jimmunol.2001249
    1. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. . Current concepts in the diagnosis and management of cytokine release syndrome. Blood (2014) 124(2):188–95. doi: 10.1182/blood-2014-05-552729
    1. Jiang H, Liu L, Guo T, Wu Y, Ai L, Deng J, et al. . Improving the safety of CAR-T cell therapy by controlling CRS-related coagulopathy. Ann Hematol (2019) 98(7):1721–32. doi: 10.1007/s00277-019-03685-z
    1. Santomasso BD, Park JH, Salloum D, Riviere I, Flynn J, Mead E, et al. . Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with b-cell acute lymphoblastic leukemia. Cancer Discovery (2018) 8(8):958–71. doi: 10.1158/-17-1319

Source: PubMed

3
Tilaa