25
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Corticosteroids do not influence the efficacy and kinetics of CAR-T cells for B-cell acute lymphoblastic leukemia

      letter

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Dear Editor, Chimeric antigen receptor (CAR) T-cell therapy has been demonstrated as a promising immunotherapeutic approach for treating the patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) 1–3 . However, cytokine release syndrome (CRS), the most prominent toxicity of CAR-T cell therapy, could be serious and even life-threatening 4,5 . Tocilizumab, an IL-6 receptor antagonist, has been widely used to treat CRS-related toxicities 5–7 , although corticosteroids could effectively abrogate CRS as well 4,5 , several earlier cases showed steroids may inhibit CAR T-cell persistence and their antimalignancy efficacy 7,8 , making corticosteroid therapy in CRS be often reserved for failure of tocilizumab or for neurologic toxicity since tocilizumab could not cross the blood-brain barrier. In our center, we use corticosteroids instead of tocilizumab as the first-line agent to manage CRS, here, we assessed the influence of steroids on the treatment effect and kinetics of CAR-T cells by comparing the difference between two groups of B-ALL patients who did (42 cases) or did not (26 cases) accept steroids. Relapsed/refractory B-ALL 9 patients treated by CAR-T therapy were from our clinical trials of ChiCTR-OIC-17013623 (CD19 for B-ALL), ChiCTR-ONC-17013648 (sequential CART for B-ALL after transplantation) and ChiCTR-OIC-17013523 (CD22 for B-ALL), these trials were approved by Beijing Boren Hospital institutional review board and informed consents were obtained. The lentiviral vectors encoding second generation CARs composed of CD3ζ and 4-1BB were used to produce CAR-T cells, a single dose or 2 fractionated doses (2 cases, within 3 days) of T-cells were infused. The construction of CD19- and CD22-specific CARs, the CAR-T cell manufacture and details of clinical protocols have been described in our published papers 10,11 and on the ChiCTR website. Treatment effects were evaluated on day 30 after T-cell infusion and then monthly in follow-up patients. Minimal residual disease (MRD) was detected by multiparameter flow cytometry (FCM) and real-time quantitative PCR for fusion genes. Extramedullary diseases (EMD) were examined using PET-CT, CT or MRI. The dynamic monitoring of CAR-T cells was performed through flow cytometric quantitation of FITC + CD3 + T cells 12 . B-cell aplasia (BCA) was assayed by FCM and defined as less than 3% CD19- or CD22-positive lymphocytes 6 . SPSS 23 software was used to analyze data and tests were two-sided, comparison of means was performed using T test or Mann Whitney U-test when continuous variables were abnormal distribution, categorical variables were compared by the chi-square test. P-value < 0.05 was considered to be statistically significant. Dexamethasone or methylprednisolone or both (alternately) were administrated for CRS when (1) continuous high fever not being released by antipyretics; (2) moderate-severe organ disfunction. Dexamethasone was applied in most cases especially for patients with neurologic symptoms; methylprednisolone was preferred for patients with pulmonary or liver dysfunction, and patients accepting high dose steroids. Steroids were usually started with low dose (dexamethasone 2–5 mg/dose) and would be increased if symptoms were not resolved, for severe CRS, steroids could be escalated up to dexamethasone 20 mg/m2/d or more higher up to methylprednisolone 10 mg/kg/d. Once CRS was improved, steroids were rapidly reduced and stopped. This study using corticosteroids to treat CRS was approved by Beijing Boren Hospital institutional review board. A total of 68 patients were included, patients followed up less than 1 month and could not be evaluated (went to other hospitals for transplantation or died within 1 month) were excluded. The median age was 15 (range, 2–55) years, with 28 (41.2%) adults and 40 (58.8%) children younger than 18 years, 22 (32.4%) patients presented with EMD, bone marrow blasts in patients without EMD varied between 5%–96.5%. Thirty-one (45.6%) cases had an HCT. Fifty-four (79.4%) patients received CD19- and 14 (20.6%) received CD22-specific CAR-T therapy. CRS occurred in 94.1% (64/68) of patients, 10 (14.7%) cases experienced grade I, 44 (64.7%) experienced grade II and 10 (14.7%) experienced grade III CRS 13 . Five (7.4%) patients presented grade II (4 cases) and III (1 case) neurologic toxicity. Graft-versus-host disease (GVHD) induced by CAR-T therapy occurred in 6 (19.4%) of 31 post-hematopoietic cell transplantation (post-HCT) patients (Table 1A; Supplementary Table 1). Table 1 Patient clinical data and treatment response. Variables No. (n = 68) % of patients (A) Clinical data Age  Children (<18 years) 40 58.8  Adults 28 41.2  EMD  Present 22 32.4  Absent 46 67.6 HCT  Pre 37 54.4  Post 31 45.6 CAR-T type  CD-19 54 79.4  CD-22a 14 20.6 CRS  0 4 5.9  I 10 14.7  II 44 64.7  III 10 14.7 Neurotoxicity (≥ grade 2) 5 7.4 GVHD after CART in post-HCT patients (n = 31) 6 19.4 Steroidsb  Not received 26 38.2  Received 42 61.8  High dosec 23 54.8  ≤7days 33 78.6  8–16 days 9 21.4 Treatment response Steroid group (n = 42) Non-steroid group (n = 26) No. % No. % p-value (B) Treatment response on D30 after T-cell infusion CR/CRi 40 95.2 24 92.3 0.344 MRD-CR 32 80.0 19 79.2 0.249 PR 2 4.8 1 3.8 NR 0 0.0 1 3.8 EMD extramedullary disease, HCT hematopoietic cell transplantation, CRS cytokine release syndrome, GVHD graft-versus-hos disease, CR complete remission, CRi CR with incomplete count recovery, MRD minimal residual disease, PR partial remission, NR no remission. a14 patients, who failed or relapsed after CD-19 CART, or had dim CD-19 but normal CD-22 antigen expression, received CD-22 specific CAR-T therapy. bUsed within 1 month after T cell infusion, for all of 10 patients with grade III CRS, 68.2% (30/44) with grade II CRS, and 2 cases with no CRS but GVHD (1 case) or neurotoxicity (1 case). c≥10 mg/m2/d dexamethasone or equivalent. Within 1 month post CAR-T cell infusion, 42 (61.8%) patients were administrated steroids, including all of 10 with grade III CRS, 68.2% (30/44) with grade II CRS and 2 patients with no CRS but GVHD (1 case) or neurotoxicity (1 case), the duration of steroid use was 1-16 days (78.6% ≤ 7 days). Whereas 26 (38.2%) patients did not accept any steroids only supportive cares. Fourteen cases received CD22 CAR-T cells happened to be equally distributed in steroid and non-steroid group (each has 7 patients). In steroid group, 23 (54.8%) patients were given high-dose steroids (defined as ≥ 10 mg/m2/d dexamethasone or equivalent), the average days of high-dose steroid administration was 4 (range, 1-10 days) and 91.3% (21/23) of patients took high-dose steroids no more than one week (Table 1A; Supplementary Tables 1, 2). After one month, 3 patients continued to receive steroids against GVHD (2 cases, both lasting 4 months) or neurologic toxicity (1 case, lasting 17 days); 1 patient without using steroids within one month accepted 12-day steroids for GVHD, who was counted in the steroid group in later follow-up of B-cell aplasia. We evaluated the impacts of steroids on treatment response. In the non-steroid group of 26 patients, 1 case had no response and 1 obtained partial remission (PR), the rest of 24 patients achieved complete remission (CR), the CR rate, composed of both CR and CR with incomplete count recovery (CRi) 9 , was 92.3% (24/26) and MRD negative CR was 79.2% (19/24). In the steroid group of 42 patients, 40 obtained CR and 2 were in PR, the rate of CR was 95.2% (40/42) and MRD negative CR was 80.0% (32/40). Obviously, there was no difference between steroid and non-steroid group in CR rate (p = 0.344) or in MRD–CR rate (p = 0.249) (Table 1B; Supplementary Table 1), this indicated that corticosteroids did not compromise the treatment effect of CAR-T cells for these patients. Then, we analyzed the expansion of CAR-T cells in peripheral blood (PB) by assessing the quantity of CAR-T cells using FCM (Fig. 1a) on day 7, 11, 15, 20 and 30 after cell infusion (Fig. 1b; Supplementary Table 3). Unexpectedly, the average CAR-T cell numbers in steroid group were significantly higher than those in non-steroid group at all time points from day11 (p = 0.0302 on D11; p = 0.0053 on D15; p = 0.0045 on D20 and p = 0.0028 on D30), except for day 7 when CAR-T cells began to expand (p = 0.9815). These data demonstrated that steroids did not suppress the proliferation of CAR-T cells in PB, on the contrary, the T-cell expansion in steroid group was much greater, the reasonable explanation is that higher expansion of CAR-T cells led to the higher grade of CRS which hence needed to be controlled by steroids. Fig. 1 Kinetics of CAR-T cells in relapsed/refractory B-ALL patients treated or not treated with steroids (detected by flow cytometry). a The representative flow cytometry plots showing CAR-T cells. b CAR-T cell numbers in peripheral blood (PB) on day 7, 11, 15, 20 and D30. c Percentages of patients with detectable CAR-T cells in bone marrow (BM) and cerebrospinal fluid (CSF), assayed once or twice between day 14 to day 35. d Percentages of patients with B-cell aplasia (BCA) at 2 and 3 months. Based on Maude SL et al (N Engl J Med. 2014;371:1507-1517), BCA was defined as less than 3% CD19 or CD22 (4 cases) positive lymphocytes. Since CAR-T cells can distribute to bone marrow (BM) and cerebrospinal fluid (CSF) 12,14,15 , we investigated the existence of CAR-T cells in BM and CSF by detecting them once or twice during day 14-35 after cell infusion. In steroid group, the percentages of patients with detectable CAR-T cells were 85.2% (23/27) in BM and 68.6% (24/35) in CSF; while in non-steroid group, the percentages of patients with CAR-T cells were 78.6% (11/14) in BM and 57.9% (11/19) in CSF, there were no significant differences between two groups (p = 0.923 in BM and p = 0.433 in CSF, respectively) (Fig. 1c; Supplementary Table 4). This implied that steroids did not interfere the trafficking of T-cells to BM and CSF. The persistence of functional CAR-T cells was usually assessed by B-cell aplasia (BCA) 6,12 , we hence monitored BCA monthly in evaluable patients. Since most of this cohort of patients underwent transplantation or received other therapies after CART, BCA was only observed in some patients and the number of patients gradually decreased over time. At 2- and 3-month, 100% (18/18, 2 after CD22) and 84.6% (11/13, 1 after CD22) of patients presented BCA in steroid group, compared to 75% (6/8, 2 post CD22) and 50% (4/8, 1 post CD22) of patients in non-steroid group, the percentages of patients with BCA in steroid group were higher than those in non-steroid group but there were no significant differences between 2 groups (p = 0.086 at 2 months; p = 0.146 at 3 months) (Fig. 1d). In later time points of 4–6 months, although limited cases (all were after CD19) left in each group which could not be compared, in the steroid group, 100% of patients (4-month, 7/7; 5-month, 7/7; 6-month, 5/5) still maintained BCA and CR (Supplementary Table 5). This evidence indicated that the functional CAR-T cells could exist a longer time without being impacted by corticosteroids. In conclusion, our data revealed that corticosteroids even high-dose steroids did not influence treatment outcomes of CAR-T cells and their proliferation and duration. The possibility of our results differing from previous observation could be: we used high-dose steroids for short term (average 4 days; 91.3% ≤ 7 days) whereas they used longer time (Davila ML et al mentioned 3 cases accepted 7-21 days of high-dose steroids 7 ). Very recently, two studies showed that early intervention with corticosteroids for CRS 16 or neurotoxicity 17 does not impact on the antitumor potency of CD19 CAR -T cells, which support our results, the potential mechanisms need to be further explored. Supplementary information supplimentary table1 supplimentary table2 supplimentary table3 supplimentary table4 supplimentary table5 ethics approval

          Related collections

          Most cited references7

          • Record: found
          • Abstract: found
          • Article: not found

          Intent to treat leukemia remission by CD19CAR T cells of defined formulation and dose in children and young adults

          Publisher's Note: There is an [Related article:] Inside Blood Commentary on this article in this issue. Defined-composition manufacturing platform of CD19 CAR T cells contributes to >90% intent-to-treat complete remission rate. Uniformity of durable persistence of CAR T cells and mitigation of antigen escape are key aspects for further optimization. Transitioning CD19-directed chimeric antigen receptor (CAR) T cells from early-phase trials in relapsed patients to a viable therapeutic approach with predictable efficacy and low toxicity for broad application among patients with high unmet need is currently complicated by product heterogeneity resulting from transduction of undefined T-cell mixtures, variability of transgene expression, and terminal differentiation of cells at the end of culture. A phase 1 trial of 45 children and young adults with relapsed or refractory B-lineage acute lymphoblastic leukemia was conducted using a CD19 CAR product of defined CD4/CD8 composition, uniform CAR expression, and limited effector differentiation. Products meeting all defined specifications occurred in 93% of enrolled patients. The maximum tolerated dose was 10 6 CAR T cells per kg, and there were no deaths or instances of cerebral edema attributable to product toxicity. The overall intent-to-treat minimal residual disease–negative (MRD − ) remission rate for this phase 1 study was 89%. The MRD − remission rate was 93% in patients who received a CAR T-cell product and 100% in the subset of patients who received fludarabine and cyclophosphamide lymphodepletion. Twenty-three percent of patients developed reversible severe cytokine release syndrome and/or reversible severe neurotoxicity. These data demonstrate that manufacturing a defined-composition CD19 CAR T cell identifies an optimal cell dose with highly potent antitumor activity and a tolerable adverse effect profile in a cohort of patients with an otherwise poor prognosis. This trial was registered at www.clinicaltrials.gov as #NCT02028455.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            High efficacy and safety of low dose CD19-directed CAR-T cell therapy in 51 refractory or relapsed B acute lymphoblastic leukemia patients.

            Refractory or relapsed B lymphoblastic leukemia (B-ALL) patients have a dismal outcome with current therapy. We treated 42 primary refractory/hematological relapsed (R/R) and 9 refractory minimal residual disease by flow cytometry (FCM-MRD(+)) B-ALL patients with optimized second generation CD19-directed CAR-T cells. The CAR-T cell infusion dosages were initially ranged from 0.05 to 14 × 10(5)/kg and were eventually settled at 1 × 10(5)/kg for the most recent 20 cases. 36/40 (90%) evaluated R/R patients achieved complete remission (CR) or CR with incomplete count recovery (CRi), and 9/9 (100%) FCM-MRD(+) patients achieved MRD(-). All of the most recent 20 patients achieved CR/CRi. Most cases only experienced mild to moderate CRS. 8/51 cases had seizures that were relieved by early intervention. 23 of 27 CR/CRi patients bridged to allogeneic hematopoietic stem cell transplantation (allo-HCT) remained in MRD(-) with a median follow-up time of 206 (45-427) days, whereas 9 of 18 CR/CRi patients without allo-HCT relapsed. Our results indicate that a low CAR-T cell dosage of 1 × 10(5)/kg, is effective and safe for treating refractory or relapsed B-ALL, and subsequent allo-HCT could further reduce the relapse rate.Leukemia accepted article preview online, 11 May 2017. doi:10.1038/leu.2017.145.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without attenuation of antileukemic efficacy

              Gardner et al report that early intervention with tocilizumab and steroids at the first signs of mild cytokine release syndrome (CRS) following CD19 chimeric antigen receptor (CAR) T-cell infusion for B-cell acute lymphocytic leukemia reduces the development of life-threatening severe CRS without having a negative impact on antileukemic effect.
                Bookmark

                Author and article information

                Contributors
                tongcr@borenhospital.com
                Journal
                Blood Cancer J
                Blood Cancer J
                Blood Cancer Journal
                Nature Publishing Group UK (London )
                2044-5385
                6 February 2020
                6 February 2020
                February 2020
                : 10
                : 2
                : 15
                Affiliations
                [1 ]Department of Hematology, Beijing Boren Hospital, Beijing, China
                [2 ]Cytology Laboratory, Beijing Boren Hospital, Beijing, China
                [3 ]Department of Bone Marrow Transplantation, Beijing Boren Hospital, Beijing, China
                [4 ]ISNI 0000 0004 0459 167X, GRID grid.66875.3a, Division of Hematopathology, Department of Laboratory Medicine and Pathology, , Mayo Clinic, ; Rochester, MN USA
                [5 ]ISNI 0000000123704535, GRID grid.24516.34, Clinical Translational Research Center, Shanghai Pulmonary Hospital, , Tongji University School of Medicine, ; Shanghai, China
                Article
                280
                10.1038/s41408-020-0280-y
                7005173
                32029707
                8ca9f5f1-26f0-4cd5-9c70-5db5eb05ea9f
                © The Author(s) 2020

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 15 September 2019
                : 21 January 2020
                Categories
                Correspondence
                Custom metadata
                © The Author(s) 2020

                Oncology & Radiotherapy
                cancer therapy,haematological cancer
                Oncology & Radiotherapy
                cancer therapy, haematological cancer

                Comments

                Comment on this article