BTK Inhibitor Downregulates IL-17 Secretion and Enhances CD20mb Sensitivity to ABC Type Diffuse Large B-cell Lymphoma via CYLD/NF-kB Signaling Pathway
DOI:
https://doi.org/10.54097/x2wh3a34Keywords:
Rituximab, Novel BTK Inhibitors, Diffuse Large B-cell lymphoma, IL-17, Th17, NF-κBAbstract
AIM: To explore whether two novel Bruton’s tyrosine kinase (BTK) inhibitors, acalabrutinib and zanubrutinib, in combination with Rituximab (RTX), enhance the cytotoxic effects on diffuse large B-cell lymphoma (DLBCL) cell lines and to elucidate the underlying mechanisms. Method: We selected the activated B-cell-like (ABC) DLBCL cell lines NU-DUL-1 and SU-DHL-2 as parental lines, and their corresponding Rituximab (CD20mb)-resistant lines NU-DUL-1-R as resistant strains. Using 20% fresh normal human serum as the source of complement, we employed 7-aminoactinomycin D (7-AAD) flow cytometric staining to assess the cytotoxic effects of BTK inhibitors combined with RTX via complement-dependent cytotoxicity (CDC) on tumor cells. Tumor cells were labeled with carboxyfluorescein diacetate N-succinimidyl ester (CFSE) and co-cultured with peripheral blood mononuclear cells (PBMCs). The cytotoxic effects of BTK inhibitors combined with RTX via antibody-dependent cell-mediated cytotoxicity (ADCC) were evaluated using 7-AAD and PE-Annexin V flow cytometric staining to determine whether sensitivity to RTX was increased. Flow cytometry was used to detect the expression of CD20 on DLBCL cell lines and the proportions of CD4+IL-17+ T cells in PBMNCs, granzyme B and TNF-α in CD8+ T cells in the PBMCs-tumor cell co-culture system. ELISA was employed to measure the concentrations of related cytokines. qPCR was used to assess the effects of BTK inhibitors on the mRNA expression of retinoic acid-related orphan receptor (RORC) and interleukin-17 (IL-17). Western blotting was performed to detect the protein expression levels of p-NF-κB-p65 in CD4+ T cells after treatment with BTK inhibitors. We also established a mouse model of BALB/c B-cell lymphoma using A20 cells overexpressing human CD20. Tumor volume was measured and recorded. Flow cytometry was used to detect the proportion of Th17 cells in peripheral blood and spleen, and ELISA was employed to measure the expression levels of IL-17 in serum. Results: (1) The novel BTK inhibitors had little effect on the expression of CD20 on the surface of NU-DUL-1, NU-DUL-1-R, and SU-DHL-2 cells. (2) When RTX exerted its CDC effect, compared with the monotherapy group, the combination of acatinib and RTX significantly enhanced the cytotoxic effects on NU-DUL-1, NU-DUL-1-R, and SU-DHL-2 cells (P<0.05), with a dose-dependent effect observed in NU-DUL-1 and NU-DUL-1-R cells (P<0.05). The combination of zanubrutinib and RTX also significantly enhanced the cytotoxic effects on NU-DUL-1 and NU-DUL-1-R cells (P<0.05), but the effect on SU-DHL-2 cells was not statistically significant at a zanubrutinib concentration of 10 µmol/L (P>0.05). (3) When RTX exerted its ADCC effect, compared with the monotherapy group, the combination of acalabrutinib and RTX significantly enhanced the cytotoxic effects on NU-DUL-1, NU-DUL-1-R, and SU-DHL-2 cells (P<0.05). The combination of zanubrutinib and RTX significantly enhanced the cytotoxic effects on NU-DUL-1 and SU-DHL-2 cells (P<0.05), but the effect on NU-DUL-1-R cells was not statistically significant at a zanubrutinib concentration of 3 µmol/L (P>0.05). (4) Flow cytometry results showed that both acalabrutinib and zanubrutinib significantly downregulated the proportion of Th17 cells and upregulated the secretion of granzyme B and TNF-α in CD8+ T cells (P<0.05). (5) Compared with the control group, RTX alone significantly upregulated the proportion of Th17 cells and increased IL-17 secretion. In contrast, the combination of acalabrutinib or zanubrutinib with RTX significantly downregulated the proportion of Th17 cells and reduced IL-17 secretion (P<0.05). (6) Western blot results showed that both acalabrutinib and zanubrutinib inhibited the phosphorylation of NF-κB-p65 in CD4+ T cells (P<0.05). (7) Both novel BTK inhibitors downregulated the expression of RORC and IL-17 mRNA in CD4+ T cells and reduced the secretion of IL-17, significantly inhibiting the differentiation of Th17 cells (P<0.05). (8) In vivo experiments demonstrated that zanubrutinib significantly reduced tumor volume and downregulated the proportion of Th17 cells and IL-17 levels in the spleen and peripheral blood of mice (P<0.05). Conclusions: BTK inhibitors reduce IL-17 secretion via the CYLD/NF-κB pathway and enhance the sensitivity of ABC - DLBCL to CD20 mb The combination of novel BTK inhibitors with RTX may be an effective strategy to overcome RTX resistance in ABC -DLBCL.
Downloads
References
[1] Rodgers T D and Reagan P M. Targeting the B-cell receptor pathway: a review of current and future therapies for non-Hodgkin's lymphoma[J]. Expert opin emerg dr, 2018, 23 (2):111-122.
[2] Honigberg L A, Smith A M, Sirisawad M, et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy[J]. P natl acad sci USA, 2010, 107 (29):13075-80.
[3] Byrd J C, Furman R R, Coutre S E, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia[J]. New engl j med, 2013, 369 (1):32-42.
[4] Burger J A, Tedeschi A, Barr P M, et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia[J]. New engl j med, 2015, 373 (25):2425-37.
[5] Caimi P F, Hill B T, Hsi E D, et al. Clinical approach to diffuse large B cell lymphoma[J]. Blood rev, 2016, 30 (6):477-491.
[6] Burger J A, Keating M J, Wierda W G, et al. Safety and activity of ibrutinib plus rituximab for patients with high-risk chronic lymphocytic leukaemia: a single-arm, phase 2 study[J]. Lancet oncol, 2014, 15 (10):1090-9.
[7] O'Donnell M A and Ting A T. Chronicles of a death foretold: dual sequential cell death checkpoints in TNF signaling[J]. Cell cycle, 2010, 9 (6):1065-71.
[8] Davis R E, Ngo V N, Lenz G, et al. Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma[J]. Nature, 2010, 463 (7277):88-92.
[9] Massoumi R. Ubiquitin chain cleavage: CYLD at work[J]. Trends biochem sci, 2010, 35 (7):392-9.
[10] Bignell G R, Warren W, Seal S, et al. Identification of the familial cylindromatosis tumour-suppressor gene[J]. Nat genet, 2000, 25 (2):160-5.
[11] Miliani de Marval P, Lutfeali S, Jin J Y, et al. CYLD inhibits tumorigenesis and metastasis by blocking JNK/AP1 signaling at multiple levels[J]. Cancer Prev Res (Phila), 2011, 4 (6):851-9.
[12] Oshiumi H, Matsumoto M and Seya T. Ubiquitin-mediated modulation of the cytoplasmic viral RNA sensor RIG-I[J]. J biochem-tokyo, 2012, 151 (1):5-11.
[13] Reiley W, Zhang M, Wu X, et al. Regulation of the deubiquitinating enzyme CYLD by IkappaB kinase gamma-dependent phosphorylation[J]. Mol cell biol, 2005, 25 (10): 3886-95.
[14] Xu X, Kalac M, Markson M, et al. Reversal of CYLD phosphorylation as a novel therapeutic approach for adult T-cell leukemia/lymphoma (ATLL)[J]. Cell Death Dis, 2020, 11 (2): 94.
[15] Goy A, Ramchandren R, Ghosh N, et al. Ibrutinib plus lenalidomide and rituximab has promising activity in relapsed/refractory non-germinal center B-cell-like DLBCL[J]. Blood, 2019, 134 (13):1024-1036.
[16] Pavlasova G and Mraz M. The regulation and function of CD20: an "enigma" of B-cell biology and targeted therapy[J]. Haematol-hematol j, 2020, 105 (6):1494-1506.
[17] Compagno M, Lim W K, Grunn A, et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma[J]. Nature, 2009, 459 (7247):717-21.
[18] Ngo V N, Davis R E, Lamy L, et al. A loss-of-function RNA interference screen for molecular targets in cancer[J]. Nature, 2006, 441 (7089):106-10.
[19] Ngo V N, Young R M, Schmitz R, et al. Oncogenically active MYD88 mutations in human lymphoma[J]. Nature, 2011, 470 (7332):115-9.
[20] Da Roit F, Engelberts P J, Taylor R P, et al. Ibrutinib interferes with the cell-mediated anti-tumor activities of therapeutic CD20 antibodies: implications for combination therapy[J]. Haematol-hematol j, 2015, 100 (1):77-86.
[21] Younes A, Sehn L H, Johnson P, et al. Randomized Phase III Trial of Ibrutinib and Rituximab Plus Cyclophosphamide, Doxorubicin, Vincristine, and Prednisone in Non-Germinal Center B-Cell Diffuse Large B-Cell Lymphoma[J]. J clin oncol, 2019, 37 (15):1285-1295.
[22] Syed Y Y. Zanubrutinib: First Approval[J]. Drugs, 2020, 80 (1):91-97.
[23] Byrd J C, Harrington B, O'Brien S, et al. Acalabrutinib (ACP-196) in Relapsed Chronic Lymphocytic Leukemia[J]. New engl j med, 2016, 374 (4):323-32.
[24] Tam C and Thompson P A. BTK inhibitors in CLL: second-generation drugs and beyond[J]. Blood adv, 2024, 8 (9):2300-2309.
[25] Yin Q, Sivina M, Robins H, et al. Ibrutinib Therapy Increases T Cell Repertoire Diversity in Patients with Chronic Lymphocytic Leukemia[J]. J immunol, 2017, 198 (4):1740-1747.
[26] Xu X, Wei T, Zhong W, et al. Down-regulation of cylindromatosis protein phosphorylation by BTK inhibitor promotes apoptosis of non-GCB-diffuse large B-cell lymphoma [J]. Cancer Cell Int, 2021, 21 (1):195.
[27] Han S, Guan-lun G, Yan-li X, et al. Acalabrutinib enhances killing effect of rituximab biosimilar HLX01 on rituximab-resistant non-GCB DLBCL cell line[J]. Chinese Journal of Pathophysiology, 2022, 38 (01):11-19.
[28] Crump M, Neelapu S S, Farooq U, et al. Outcomes in refractory diffuse large B-cell lymphoma: results from the international SCHOLAR-1 study[J]. Blood, 2017, 130 (16):1800-1808.
[29] Voso M T, Pantel G, Rutella S, et al. Rituximab reduces the number of peripheral blood B-cells in vitro mainly by effector cell-mediated mechanisms[J]. Haematol-hematol j, 2002, 87 (9):918-25.
[30] Berinstein N L, Grillo-López A J, White C A, et al. Association of serum Rituximab (IDEC-C2B8) concentration and anti-tumor response in the treatment of recurrent low-grade or follicular non-Hodgkin's lymphoma[J]. Ann oncol, 1998, 9 (9):995-1001.
[31] Li Q, Xu X, Zhong W, et al. IL-17 induces radiation resistance of B lymphoma cells by suppressing p53 expression and thereby inhibiting irradiation-triggered apoptosis[J]. Cell mol immunol, 2015, 12 (3):366-72.
[32] Zhong W, Xu X, Zhu Z, et al. Increased interleukin-17A levels promote rituximab resistance by suppressing p53 expression and predict an unfavorable prognosis in patients with diffuse large B cell lymphoma[J]. Int j oncol, 2018, 52 (5):1528-1538.
[33] Zhong W, Zhu Z, Xu X, et al. Human bone marrow-derived mesenchymal stem cells promote the growth and drug-resistance of diffuse large B-cell lymphoma by secreting IL-6 and elevating IL-17A levels[J]. J Exp Clin Cancer Res, 2019, 38 (1):73.
[34] Zhong W and Li Q. Rituximab or irradiation promotes IL-17 secretion and thereby induces resistance to rituximab or irradiation[J]. Cell mol immunol, 2017, 14 (12):1020-1022.
[35] Massoumi R, Chmielarska K, Hennecke K, et al. Cyld inhibits tumor cell proliferation by blocking Bcl-3-dependent NF-kappaB signaling[J]. Cell, 2006, 125 (4):665-77.
[36] Oh H and Ghosh S. NF-κB: roles and regulation in different CD4(+) T-cell subsets[J]. Immunol rev, 2013, 252 (1):41-51.
[37] Sun D, Luo F, Xing J C, et al. 1,25(OH)2 D3 inhibited Th17 cells differentiation via regulating the NF-κB activity and expression of IL-17[J]. Cell proliferat, 2018, 51 (5): e12461.
Downloads
Published
Issue
Section
License

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

