Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2023

Open Access 01.12.2023 | Correspondence

Decitabine in combination with fludarabine and cyclophosphamide as a lymphodepletion regimen followed by CD19/CD22 bispecific targeted CAR T-cell therapy significantly improves survival in relapsed/refractory B-ALL patients

verfasst von: Yunju Ma, Haiping Dai, Qingya Cui, Sining Liu, Liqing Kang, Xiaying Lian, Wei Cui, Jia Yin, Lingling Liu, Mengjie Cai, Lei Yu, Depei Wu, Xiaowen Tang

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2023

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Relapse is a major limitation of chimeric antigen receptor (CAR) T-cell therapy. Here, we speculated that decitabine (DAC) in combination with fludarabine and cyclophosphamide (FC) as a lymphodepletion regimen may improve the efficacy of CD19/CD22 CAR T-cell therapy. Fourteen of 26 patients with relapsed/refractory B cell acute lymphoblastic leukemia (r/r B-ALL) without remission before lymphodepletion treatment were treated with DAC (total dose 100 mg/m2 in 3 days) followed by the FC regimen (DAC group), while twelve patients received the FC regimen (CON group). On Day 28 after CAR T-cells infusion, no significant differences in complete remission (CR) and minimal residual disease negative CR rates were found between both groups. However, there were significant differences in overall survival (OS) and leukemia-free survival (LFS) between two groups: 3-year OS, 92.3% (DAC) versus 41.7% (CON), P = 0.005 and 3-year LFS, 92.9% (DAC) versus 27.3% (CON), P < 0.001. There was no significant difference in the incidence of cytokine release syndrome between both groups. Median time to platelet and neutrophil counts recovery was similar in both groups. All adverse events were reversible and manageable. In conclusion, DAC in combination with the FC lymphodepletion regimen may be a new treatment option that can improve the efficacy of CAR T-cell therapy in r/r B-ALL.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40164-023-00397-z.
Yunju Ma, Haiping Dai, Qingya Cui have contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
To the editor
Antigen escape-mediated relapse is a major limitation of chimeric antigen receptor (CAR) T-cell therapy. One strategy to overcome antigen escape following CAR T-cell therapy is to generate T cells simultaneously targeting both CD19 and CD22. Although some clinical trials, using CD19/CD22 bispecific CAR T-cell therapy, have demonstrated promising therapeutic efficacy. CD19/CD22 CAR T-cell therapy has not induced durable remissions or reduced the relapse rate in patients with relapsed/refractory B-cell acute lymphoblastic leukemia (r/r B-ALL) [1, 2]. Relapse after CD19/CD22 CAR T-cell therapy is often associated with poor persistence of CAR T- cells in part caused by T cell exhaustion and the immunosuppressive microenvironment, which suggests the need for novel strategies to improve CD19/CD22 CAR T-cell therapy.
The hypomethylating agent decitabine (DAC) has been demonstrated to reverse T cell exhaustion, increase antigen expression, enhance T cell activation and modify the tumor microenvironment [37]. In addition, DAC combined with cytotoxic chemotherapy represents a promising strategy for the treatment of patients with high tumor burden [8], which is a significant predictor of poor prognosis in B-ALL. Therefore, we speculated that DAC in combination with fludarabine and cyclophosphamide (FC) as a lymphodepletion regimen may synergize and improve the efficacy of CAR T-cell therapy.
We retrospectively analyzed 26 r/r B-ALL patients without remission before lymphodepletion treatment who were enrolled in a phase 1/2 clinical trial of CD19/CD22 CAR T-cell therapy (NCT03614858) from October 2017 to May 2021 at the First Affiliated Hospital of Soochow University (Additional file 1: Figure S1). Fourteen patients received DAC combined with the FC regimen (DAC group) while twelve patients were treated with FC alone (CON group) followed by CAR T-cell therapy. The patients received DAC combined with FC depending on disease characteristics such as TP53 mutation, comorbidities, patients' wishes, and economic burden. Patients received the following lymphodepletion regimen: FC (fludarabine 30 mg/m2/day and cyclophosphamide 300 mg/m2/day) on days -5 to -3, with or without DAC (total dose 100 mg/m2 from day -6 to -4; Additional file 2: Figure S2).
There were more patients (42.9%) who relapsed after allogeneic hematopoietic stem cell transplantation (allo-HSCT) prior to CAR T-cell therapy in the DAC group than in the CON group (P = 0.017) (Table 1). On Day 28 after CAR T-cells infusion, no significant difference in minimal residual disease negative CR rates was found between both groups (Additional file 3: Table S1). Among the nontransplant patients in the DAC group, only one patient (16.7%, 1/6) relapsed. However, 1 of four nontransplant patients in the CON group had no response after CAR T-cell therapy and 3 patients relapsed after CAR T-cell treatment (Fig. 1A).
Table 1
Baseline Characteristics of Patients
Baseline characteristics of patients
Characteristic
DAC group (14)
Control group (12)
P value
Gender
 Male
10 (71.4%)
5 (41.7%)
0.233
 Female
4 (28.6%)
7 (58.3%)
Age, median
26.5 (8–52)
31 (16–74)
0.226
Ph + ALL, n (%)
2 (14.3%)
5 (41.7%)
0.190
Ph-like ALL, n (%)
3 (21.4%)
2 (16.7%)
1.000
White blood cell ≥ 50 × 10^9/L, n (%)
6 (42.9%)
2 (16.7%)
0.216
Extramedullary leukemia, n (%)
2 (14.3%)
0 (0%)
0.483
Monosomal karyotype, n (%)
0 (0%)
1 (8.3%)
0.462
Complex karyotype, n (%)
2 (14.3%)
2 (16.7%)
1.000
KMT2A rearranged, n (%)
1 (7.1%)
0 (0%)
1.000
TP53 mutation or deletion, n (%)
2 (14.3%)
0 (0%)
0.483
T315I mutation, n (%)
1 (7.1%)
2 (16.7%)
0.580
Poor-risk cytogenetics, n (%)
8 (57.1%)
8 (66.7%)
0.701
Prior cycles of therapy, median (range)
4 (2–16)
6.5 (1–20)
0.251
  < 5 cycles of therapy, n (%)
9 (64.3%)
4 (33.3%)
0.235
  ≥ 5 and < 10 lines of therapy, n (%)
2 (14.3%)
5 (41.7%)
  ≥ 10 cycles of therapy, n (%)
3 (21.4%)
3 (25%)
Primary refractory to chemotherapy, n (%)
2 (14.3%)
3 (25%)
0.635
Numbers of relapses, median (range)
1 (0–2)
1 (0–2)
0.685
Relapse after previous HSCT, n (%)
 Yes
6 (42.9%)
0 (0%)
0.017
 No
8 (57.1%)
12 (100%)
Blasts in BM before lymphodepletion treatment, median (range)
35.75 (5–82)
43.75 (6–85.5)
0.487
LDH, pre-lymphodepletion, median (range)
173.8 (106.5–644)
168.7 (83.6–722.2)
0.956
Ferritin, pre-lymphodepletion, median (range)
774.945 (234.06–2718.84)
1614.66 (411.63–2663.69)
0.140
CRP, pre-lymphodepletion, median (range)
5.115 (0.28–15.36)
1.195 (0.12–15.36)
0.170
ECOG, pre-lymphodepletion, median (range)
2 (1–3)
2 (1–3)
0.609
CAR T-cell dose, 10^7/kg, median (range)
1 (0.5–2)
1 (0.5–2.5)
0.950
Source of CAR T-cells, n (%)
 Autologous
10 (71.4%)
12 (100%)
0.100
 Donor-derived allogenic
4 (28.6%)
0 (0%)
Transduction rate (%), median
31.435 (13.02–64.35)
44.685 (6.07–63.92)
0.643
With a median follow-up of 13 months, there were significant differences in overall survival (OS) and leukemia-free survival (LFS) between both groups (Fig. 1B, C). Patients who underwent allo-HSCT after CAR T-cell therapy in the DAC or the CON group had higher OS and LFS than those without allo-HSCT, although the number of patients in some subgroups was relatively small (Fig. 1D-E). CAR T- cells copies in the peripheral blood were detected by qPCR at several indicated time points after infusion in all patients (Fig. 1F).
There was no statistically significant difference in the incidence of cytokine release syndrome between both groups (Additional file 4: Table S2). All adverse events were reversible and manageable. The medians of peak type 1 helper T (Th1) cytokines (IL-2 and IFN-γ) concentrations were higher in the DAC group than in the CON group. However, in regard to type 2 helper T (Th2) cytokines, the peak value of serum IL-4 after CAR T-cells infusion was significantly higher in the CON group than in the DAC group (P = 0.029; Additional file 5: Table S3).
There are several potential mechanisms underlying the therapeutic benefit of DAC-based lymphodepletion prior to CAR T-cell therapy: 1. DAC can upregulate CD19 expression to make leukemia cells more susceptible to CAR T-cell therapy [5]. 2. DAC can inhibit the methylation of tumor suppressor genes associated with B-ALL and induce leukemia cell apoptosis at high doses. 3. DAC pretreatment can modify the immunosuppressive tumor microenvironment to enhance CAR T-cell efficacy and endogenous immunity, leading to long-term antileukemia immunity [9]. In our study, an increased level of IL-4 was detected in the CON group compared with the DAC group, which suggested that DAC depolarized Th2 cells and inhibited tumor growth [10].
In summary, our data demonstrated that the combination of DAC and FC as a conditioning regimen was safe and effective for Chinese patients with r/r B-ALL. DAC in combination with the FC lymphodepletion regimen may be a new treatment option that can improve the efficacy of CAR T-cell therapy in r/r B-ALL. Moreover, CD19/CD22 CAR T-cell therapy as a bridge to allo-HSCT could be a promising strategy for r/r B-ALL patients to achieve prolonged OS and LFS. However, due to the small sample size and retrospective nature of this study, large-scale randomized controlled clinical trials should be prospectively conducted to confirm our results. Further studies are warranted to determine the key factors and pathways that underlie the synergistic antitumor effect of DAC and CAR T-cells.

Acknowledgements

The authors would like to thank all members of the study team, the patient and their family, and Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd.

Declarations

This study was approved by the Ethics Committee of the First Affiliated Hospital of Soochow University (2022445) and was conducted in accordance with the Declaration of Helsinki principles. All patients provided written informed consent.
Written informed consent was obtained from the patient and his parents.

Competing interests

All authors declare there are no competing interest to report.
Open AccessThis 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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Dai H, Wu Z, Jia H, Tong C, Guo Y, Ti D, et al. Bispecific CAR-T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J Hematol Oncol. 2020;13(1):30–9.CrossRefPubMedPubMedCentral Dai H, Wu Z, Jia H, Tong C, Guo Y, Ti D, et al. Bispecific CAR-T cells targeting both CD19 and CD22 for therapy of adults with relapsed or refractory B cell acute lymphoblastic leukemia. J Hematol Oncol. 2020;13(1):30–9.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Hu Y, Zhou Y, Zhang M, Ge W, Li Y, Yang L, et al. CRISPR/Cas9-engineered universal CD19/CD22 dual-targeted CAR-T cell therapy for relapsed/refractory B-cell acute lymphoblastic leukemia. Clin Cancer Res. 2021;27(10):2764–72.CrossRefPubMed Hu Y, Zhou Y, Zhang M, Ge W, Li Y, Yang L, et al. CRISPR/Cas9-engineered universal CD19/CD22 dual-targeted CAR-T cell therapy for relapsed/refractory B-cell acute lymphoblastic leukemia. Clin Cancer Res. 2021;27(10):2764–72.CrossRefPubMed
3.
Zurück zum Zitat Nahas MR, Stroopinsky D, Rosenblatt J, Cole L, Pyzer AR, Anastasiadou E, et al. Hypomethylating agent alters the immune microenvironment in acute myeloid leukaemia (AML) and enhances the immunogenicity of a dendritic cell/AML vaccine. Br J Haematol. 2019;185(4):679–90.CrossRefPubMedPubMedCentral Nahas MR, Stroopinsky D, Rosenblatt J, Cole L, Pyzer AR, Anastasiadou E, et al. Hypomethylating agent alters the immune microenvironment in acute myeloid leukaemia (AML) and enhances the immunogenicity of a dendritic cell/AML vaccine. Br J Haematol. 2019;185(4):679–90.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Wang Y, Tong C, Dai H, Wu Z, Han X, Guo Y, et al. Low-dose decitabine priming endows CAR T cells with enhanced and persistent antitumour potential via epigenetic reprogramming. Nat Commun. 2021;12(1):409.CrossRefPubMedPubMedCentral Wang Y, Tong C, Dai H, Wu Z, Han X, Guo Y, et al. Low-dose decitabine priming endows CAR T cells with enhanced and persistent antitumour potential via epigenetic reprogramming. Nat Commun. 2021;12(1):409.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Li S, Xue L, Wang M, Qiang P, Xu H, Zhang X, et al. Decitabine enhances cytotoxic effect of T cells with an anti-CD19 chimeric antigen receptor in treatment of lymphoma. Onco Targets Ther. 2019;12:5627–38.CrossRefPubMedPubMedCentral Li S, Xue L, Wang M, Qiang P, Xu H, Zhang X, et al. Decitabine enhances cytotoxic effect of T cells with an anti-CD19 chimeric antigen receptor in treatment of lymphoma. Onco Targets Ther. 2019;12:5627–38.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat You L, Han Q, Zhu L, Zhu Y, Bao C, Yang C, et al. Decitabine-mediated epigenetic reprograming enhances anti-leukemia efficacy of CD123-targeted chimeric antigen receptor T-cells. Front Immunol. 2020;11:1787.CrossRefPubMedPubMedCentral You L, Han Q, Zhu L, Zhu Y, Bao C, Yang C, et al. Decitabine-mediated epigenetic reprograming enhances anti-leukemia efficacy of CD123-targeted chimeric antigen receptor T-cells. Front Immunol. 2020;11:1787.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Prinzing B, Zebley CC, Petersen CT, Fan Y, Anido AA, Yi Z, et al. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity. Sci Transl Med. 2021;13(620):272.CrossRef Prinzing B, Zebley CC, Petersen CT, Fan Y, Anido AA, Yi Z, et al. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity. Sci Transl Med. 2021;13(620):272.CrossRef
8.
Zurück zum Zitat Benton CB, Thomas DA, Yang H, Ravandi F, Rytting M, O’Brien S, et al. Safety and clinical activity of 5-aza-2’-deoxycytidine (decitabine) with or without Hyper-CVAD in relapsed/refractory acute lymphocytic leukaemia. Br J Haematol. 2014;167(3):356–65.CrossRefPubMedPubMedCentral Benton CB, Thomas DA, Yang H, Ravandi F, Rytting M, O’Brien S, et al. Safety and clinical activity of 5-aza-2’-deoxycytidine (decitabine) with or without Hyper-CVAD in relapsed/refractory acute lymphocytic leukaemia. Br J Haematol. 2014;167(3):356–65.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Li X, Zhang Y, Chen M, Mei Q, Liu Y, Feng K, et al. Increased IFNgamma(+) T cells are responsible for the clinical responses of low-dose DNA-Demethylating agent decitabine antitumor therapy. Clin Cancer Res. 2017;23(20):6031–43.CrossRefPubMed Li X, Zhang Y, Chen M, Mei Q, Liu Y, Feng K, et al. Increased IFNgamma(+) T cells are responsible for the clinical responses of low-dose DNA-Demethylating agent decitabine antitumor therapy. Clin Cancer Res. 2017;23(20):6031–43.CrossRefPubMed
10.
Zurück zum Zitat Hogg SJ, Beavis PA, Dawson MA, Johnstone RW. Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov. 2020;19(11):776–800.CrossRefPubMed Hogg SJ, Beavis PA, Dawson MA, Johnstone RW. Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov. 2020;19(11):776–800.CrossRefPubMed
Metadaten
Titel
Decitabine in combination with fludarabine and cyclophosphamide as a lymphodepletion regimen followed by CD19/CD22 bispecific targeted CAR T-cell therapy significantly improves survival in relapsed/refractory B-ALL patients
verfasst von
Yunju Ma
Haiping Dai
Qingya Cui
Sining Liu
Liqing Kang
Xiaying Lian
Wei Cui
Jia Yin
Lingling Liu
Mengjie Cai
Lei Yu
Depei Wu
Xiaowen Tang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2023
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00397-z

Weitere Artikel der Ausgabe 1/2023

Experimental Hematology & Oncology 1/2023 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.