Cytokine release syndrome in cancer immunotherapy with chimeric antigen receptor engineered T cells

2014 ◽  
Vol 343 (2) ◽  
pp. 172-178 ◽  
Author(s):  
Xiao-Jun Xu ◽  
Yong-Min Tang
2014 ◽  
Vol 32 (15_suppl) ◽  
pp. 3022-3022
Author(s):  
Michel Sadelain ◽  
Maud Condomines ◽  
Zeguo Zhao ◽  
Renier J. Brentjens ◽  
Isabelle Riviere

Author(s):  
Bill X. Wu ◽  
No-Joon Song ◽  
Brian P. Riesenberg ◽  
Zihai Li

Abstract The use of chimeric antigen receptor (CAR) T cell technology as a therapeutic strategy for the treatment blood-born human cancers has delivered outstanding clinical efficacy. However, this treatment modality can also be associated with serious adverse events in the form of cytokine release syndrome. While several avenues are being pursued to limit the off-target effects, it is critically important that any intervention strategy has minimal consequences on long term efficacy. A recent study published in Science Translational Medicine by Dr. Hudecek’s group proved that dasatinib, a tyrosine kinase inhibitor, can serve as an on/off switch for CD19-CAR-T cells in preclinical models by limiting toxicities while maintaining therapeutic efficacy. In this editorial, we discuss the recent strategies for generating safer CAR-T cells, and also important questions surrounding the use of dasatinib for emergency intervention of CAR-T cell mediated cytokine release syndrome.


Hematology ◽  
2014 ◽  
Vol 2014 (1) ◽  
pp. 559-564 ◽  
Author(s):  
Shannon L. Maude ◽  
Elizabeth J. Shpall ◽  
Stephan A. Grupp

Abstract Relapsed and refractory leukemias pose substantial challenges in both children and adults, with very little progress being made in more than a decade. Targeted immunotherapy using chimeric antigen receptor (CAR)-modified T cells has emerged as a potent therapy with an innovative mechanism. Dramatic clinical responses with complete remission rates as high as 90% have been reported using CAR-modified T cells directed against the B-cell-specific antigen CD19 in patients with relapsed/refractory acute lymphoblastic leukemia. Supraphysiologic T-cell proliferation, a hallmark of this therapy, contributes to both efficacy and the most notable toxicity, cytokine release syndrome, posing a unique challenge for toxicity management. Further studies are necessary to identify additional targets, standardize approaches to cytokine release syndrome management, and determine the durability of remissions.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2622-2622
Author(s):  
Runxia Gu ◽  
Fang Liu ◽  
Dehui Zou ◽  
Bingcheng Liu ◽  
Wei Liu ◽  
...  

Data from systemic clinical trials about chimeric antigen receptor-modified T cell therapy against CD19 (CD19 CAR) differ in CAR design, T-cell activation and transduction methods at different institutions. However, according to these clinical trials, the single-chain fragment variable (scFv) sequence specific for tumor antigen were mostly derived from the FMC63 or SJ25C1 clones. Our previous study showed that the CD19 CAR constructed in our laboratory derived from clone HI19α (HI19α-4-1BB-ζ CAR) was highly effective in preclinical models. Herein, we conducted a single-arm, phase I clinical trial to evaluate the safety and efficacy of HI19α-4-1BB-ζ CAR (CNCT19) in patients with relapsed/refractory acute lymphoblastic leukemia (R/R B-ALL). From November 2016 through December 2018, 20 R/R B-ALL patients were enrolled into this clinical trial. Complete remission (CR) or complete remission with incomplete count recovery (CRi) was achieved in 100% (18/18) of patients that could be evaluated on day 28 after infusion, which accounted for 90% of all 20 enrolled patients. After a median follow-up of 17.0 months (range, 0.2 - 19.8), the median overall survival (OS) for the entire cohort of patients was 9.6 months (95% CI 4.2 - 15.0), and was not reached for 14 patients bridge to allogeneic transplantation. The median relapse free survival (RFS) of all patients was 9.0 months (95% CI, 6.7 - 11.2). Two patients died within 28 days due to cytokine release syndrome (CRS), while other patients experienced controllable cytokine-release syndrome and neurotoxicity. In order to better understand the correlation between T cell subsets and long-term response, we consistently evaluated the T cell phenotype and expansion kinetics in peripheral blood after CART infusion. The results revealed that the percentage of CD8+ naïve T cells (TN) collected from peripheral blood 20min after CAR infusion, were significantly lower in patients who relapsed from CART therapy than patients with continues CR (p=0.003), while central memory T cells (TCM), effective memory T cells (TEM) and effector T cells (TE) had similar proliferation kinetics between these two groups. In addition, multivariate analysis indicated that low percentage of CD8+TN cells was an independent factor associated with shorter RFS (p=0.033, 95% CI 0.031-0.861). This report is the first trial to provide evidence that CNCT19, a CD19 CAR constructed of a new anti-CD19 chimeric antigen receptor HI19α, has potent antileukemia activities in patients with R/R B-ALL. Furthermore, our results indicate the phenotype and kinetics of T cells are possible biomarkers to predict the long-term prognosis of CART treatment. Disclosures Lv: Juventas Cell Therapy Ltd.: Employment.


Leukemia ◽  
2016 ◽  
Vol 31 (1) ◽  
pp. 246-248 ◽  
Author(s):  
M Ruella ◽  
S S Kenderian ◽  
O Shestova ◽  
M Klichinsky ◽  
J J Melenhorst ◽  
...  

2013 ◽  
Vol 6 (1) ◽  
Author(s):  
Ethan Q Han ◽  
Xiu-ling Li ◽  
Chun-rong Wang ◽  
Tian-fang Li ◽  
Shuang-yin Han

Immunotherapy ◽  
2020 ◽  
Vol 12 (14) ◽  
pp. 1047-1052 ◽  
Author(s):  
Shuning Wei ◽  
Runxia Gu ◽  
Yingxi Xu ◽  
Xiaoyu Liu ◽  
Yanyan Xing ◽  
...  

Aim: Although numerous pro-inflammatory cytokines promote signaling via intracellular pathways involving Janus kinases, it remains unclear if ruxolitinib, a Janus kinase1/2 inhibitor, provides control of cytokine-release syndrome (CRS) without toxicity against therapeutic T cells. Materials & methods: We report successful clinical experience using ruxolitinib as adjuvant therapy to treat steroid-refractory CRS, which was related to CD22/CD19 chimeric antigen receptor-modified T cell sequential infusion, in a patient with Philadelphia chromosome-like acute lymphoblastic leukemia. Results: His symptoms improved rapidly after first dose of ruxolitinib; this was associated with reduced levels of circulating pro-inflammatory indicators. He eventually achieved minimal residual disease negative remission. Discussion: This is the first case in which ruxolitinib was used to treat steroid-refractory CRS; furthermore, this intervention had no apparent impact on the antileukemic actions of the chimeric antigen receptor-modified T cells. Our results suggest that adjuvant ruxolitinib therapy may be an alternative therapeutic approach for the management of CRS.


Sign in / Sign up

Export Citation Format

Share Document