Response-adapted zanubrutinib and tislelizumab as a potential strategy to enhance CD19 CAR T-cell therapy in relapsed/refractory large B-cell lymphoma: A retrospective observational study

Rong Shen , Wei-Guo Cao , Li Wang , Ling-Shuang Sheng , Yi-Lun Zhang , Wen Wu , Peng-Peng Xu , Shu Cheng , Meng-Ke Liu , Yan Dong , Yue Wang , Xiang-Qin Weng , Xu-Feng Jiang , Qi Song , Hong-Mei Yi , Lei Li , Sheng Chen , Zi-Xun Yan , Wei-Li Zhao

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70310

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (4) : e70310 DOI: 10.1002/ctm2.70310
RESEARCH ARTICLE

Response-adapted zanubrutinib and tislelizumab as a potential strategy to enhance CD19 CAR T-cell therapy in relapsed/refractory large B-cell lymphoma: A retrospective observational study

Author information +
History +
PDF

Abstract

Background: CD19 chimeric antigen receptor (CAR) T-cell therapy is a potential treatment for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). The combination of targeted therapeutic strategies, particularly bruton tyrosine kinase inhibitor zanubrutinib and programmed death-1 inhibitor tislelizumab, may improve clinical outcomes and modulate the tumour microenvironment (TME).

Methods: We studied patients with R/R LBCL who received response-adapted zanubrutinib plus tislelizumab upon CD19 CAR T-cell therapy between June 2021 and March 2023. Patients were treated with zanubrutinib daily from leukapheresis to day 28 post-infusion; those achieving complete response continued zanubrutinib monotherapy for 3 months, while partial responders received combined zanubrutinib for 3 months and tislelizumab for up to 2 years. We evaluated the overall response rate (ORR), complete response rate (CRR), progression-free survival (PFS), overall survival (OS), and safety. DNA sequencing and RNA sequencing were performed on available tumour samples to analyse genetic aberrations and TME characteristics.

Results: A total of 54 patients with LBCL were included, with a median follow-up of 23.6 months. The ORR at day 28, month 3, and month 6 were 94% (CRR 66%), 87% (CRR 80%), and 80% (CRR 76%), respectively. The 2-year PFS and 2-year OS rates were 68% and 76%, respectively. Median PFS and median OS were not reached. Grade ≥ 3 cytokine release syndrome occurred in 9% of patients, with no grade ≥ 3 neurotoxicity observed. Genomic and transcriptomic data indicated that this regimen was effective across genetic subtypes and abrogated T-cell exhaustion within the TME. However, tumour-infiltrating M2 macrophages with dysregulated lipid metabolism were associated with poor clinical outcome.

Conclusions: Response-adapted zanubrutinib and tislelizumab potentially enhances the efficacy of CAR T-cell therapy with a favourable safety profile in R/R LBCL, effectively counteracting T-cell exhaustion. Future studies should focus on targeting M2 macrophages by reprogramming lipid metabolism to further attenuate the immunosuppressive TME.

Keywords

bruton tyrosine kinase inhibitor / chimeric antigen receptor T-cell therapy / large B-cell lymphoma / programmed death-1 inhibitor / tumour microenvironment

Cite this article

Download citation ▾
Rong Shen, Wei-Guo Cao, Li Wang, Ling-Shuang Sheng, Yi-Lun Zhang, Wen Wu, Peng-Peng Xu, Shu Cheng, Meng-Ke Liu, Yan Dong, Yue Wang, Xiang-Qin Weng, Xu-Feng Jiang, Qi Song, Hong-Mei Yi, Lei Li, Sheng Chen, Zi-Xun Yan, Wei-Li Zhao. Response-adapted zanubrutinib and tislelizumab as a potential strategy to enhance CD19 CAR T-cell therapy in relapsed/refractory large B-cell lymphoma: A retrospective observational study. Clinical and Translational Medicine, 2025, 15(4): e70310 DOI:10.1002/ctm2.70310

登录浏览全文

4963

注册一个新账户 忘记密码

References

[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.

[2]

Neelapu SS, Jacobson CA, Ghobadi A, et al. Five-year follow-up of ZUMA-1 supports the curative potential of axicabtagene ciloleucel in refractory large B-cell lymphoma. Blood. 2023; 141(19): 2307-2315.

[3]

Scholler N, Perbost R, Locke FL, et al. Tumor immune contexture is a determinant of anti-CD19 CAR T cell efficacy in large B cell lymphoma. Nat Med. 2022; 28(9): 1872-1882.

[4]

Yan ZX, Li L, Wang W, et al. Clinical efficacy and tumor microenvironment influence in a dose-escalation study of anti-CD19 chimeric antigen receptor T cells in refractory B-cell non-Hodgkin's lymphoma. Clin Cancer Res. 2019; 25(23): 6995-7003.

[5]

Gunderson AJ, Kaneda MM, Tsujikawa T, et al. Bruton tyrosine kinase-dependent immune cell cross-talk drives pancreas cancer. Cancer Discov. 2016; 6(3): 270-285.

[6]

Fan F, Yoo HJ, Stock S, et al. Ibrutinib for improved chimeric antigen receptor T-cell production for chronic lymphocytic leukemia patients. Int J Cancer. 2021; 148(2): 419-428.

[7]

Chong EA, Alanio C, Svoboda J, et al. Pembrolizumab for B-cell lymphomas relapsing after or refractory to CD19-directed CAR T-cell therapy. Blood. 2022; 139(7): 1026-1038.

[8]

Hirayama AV, Kimble EL, Wright JH, et al. Timing of anti-PD-L1 antibody initiation affects efficacy/toxicity of CD19 CAR T-cell therapy for large B-cell lymphoma. Blood Adv. 2024; 8(2): 453-467.

[9]

Wang L, Yang J, Jin B, et al. Efficacy and safety of chimeric antigen receptor T cells therapy combined with zanubrutinib in the treatment of relapsed/refractory diffuse large B-cell lymphoma. Blood. 2023; 142(Supplement 1): 4841-4841.

[10]

Lu Y, Liu H, Ye SG, et al. [Efficacy and safety analysis of the zanubrutinib-based bridging regimen in chimeric antigen receptor T-cell therapy for relapsed/refractory diffuse large B-cell lymphoma]. Zhonghua Xue Ye Xue Za Zhi. 2023; 44(10): 813-819.

[11]

Cheson BD, Fisher RI, Barrington SF, et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014; 32(27): 3059-3068.

[12]

Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant. 2019; 25(4): 625-638.

[13]

Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy—assessment and management of toxicities. Nat Rev Clin Oncol. 2018; 15(1): 47-62.

[14]

Shen R, Fu D, Dong L, et al. Simplified algorithm for genetic subtyping in diffuse large B-cell lymphoma. Signal Transduct Target Ther. 2023; 8(1): 145.

[15]

Ye X, Wang L, Nie M, et al. A single-cell atlas of diffuse large B cell lymphoma. Cell Rep. 2022; 39(3): 110713.

[16]

Zheng L, Qin S, Si W, et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science. 2021; 374(6574): abe6474.

[17]

Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005; 102(43): 15545-15550.

[18]

Wang Y, Shi Q, Shi Z-Y, et al. Biological signatures of International Prognostic Index in diffuse large B-cell lymphoma. Blood Advances. 2024; 8(7): 1587-1599.

[19]

Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov. 2022; 21(11): 799-820.

[20]

Ruella M, Korell F, Porazzi P, Maus MV. Mechanisms of resistance to chimeric antigen receptor-T cells in haematological malignancies. Nat Rev Drug Discov. 2023; 22(12): 976-995.

[21]

Luo W, Li C, Wu J, et al. Bruton tyrosine kinase inhibitors preserve anti-CD19 chimeric antigen receptor T-cell functionality and reprogram tumor micro-environment in B-cell lymphoma. Cytotherapy. 2023; 25(7): 739-749.

[22]

Beltra JC, Manne S, MS Abdel-Hakeem, et al. Developmental relationships of four exhausted CD8(+) T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity. 2020; 52(5): 825-841.e8.

[23]

Qin JS, Johnstone TG, Baturevych A, et al. Antitumor potency of an anti-CD19 chimeric antigen receptor T-cell therapy, lisocabtagene maraleucel in combination with ibrutinib or acalabrutinib. J Immunother. 2020; 43(4): 107-120.

[24]

Pittet MJ, Michielin O, Migliorini D. Clinical relevance of tumour-associated macrophages. Nat Rev Clin Oncol. 2022; 19(6): 402-421.

[25]

Molgora M, Esaulova E, Vermi W, et al. TREM2 modulation remodels the tumor myeloid landscape enhancing anti-PD-1 immunotherapy. Cell. 2020; 182(4): 886-900.e17.

[26]

Chen J, Zhu T, Jiang G, Zeng Q, Li Z, Huang X. Target delivery of a PD-1-TREM2 scFv by CAR-T cells enhances anti-tumor efficacy in colorectal cancer. Mol Cancer. 2023; 22(1): 131.

[27]

Jaitin DA, Adlung L, Thaiss CA, et al. Lipid-associated macrophages control metabolic homeostasis in a Trem2-dependent manner. Cell. 2019; 178(3): 686-698.e14.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

6

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/