Therapeutic efficacy of anti-CD19 CAR-T cells in a mouse model of systemic lupus erythematosus

Xuexiao Jin, Qin Xu, Chengfei Pu, Kaixiang Zhu, Cheng Lu, Yu Jiang, Lei Xiao, Yongmei Han, Linrong Lu, Xuexiao Jin, Qin Xu, Chengfei Pu, Kaixiang Zhu, Cheng Lu, Yu Jiang, Lei Xiao, Yongmei Han, Linrong Lu

Abstract

Dysregulated B-cell activation plays pivotal roles in systemic lupus erythematosus (SLE), which makes B-cell depletion a potential strategy for SLE treatment. The clinical success of anti-CD19 CAR-T cells in treating B-cell malignancies has attracted the attention of researchers. In this study, we aimed to investigate the feasibility of applying anti-CD19 CAR-T cell therapy to SLE treatment in a mouse disease model. We constructed murine anti-CD19 CARs with either CD28 or 4-1BB as the intracellular costimulatory motif and evaluated the therapeutic function of the corresponding CAR-T cells by infusing them into MRL-lpr mice. Furthermore, anti-CD19 CAR-T cells were transferred to MRL-lpr mice before the onset of disease to determine their role in SLE prevention. According to our observations, compared with antibody treatment, the adoptive transfer of our anti-CD19 CAR-T cells showed a more sustained B-cell-depletion effect in MRL-lpr mice. The transfer of syngeneic anti-CD19 CAR-T cells not only prevented disease pathogenesis before the onset of disease symptoms but also displayed therapeutic benefits at a later stage after disease progression. We also tried to optimize the treatment strategy and found that compared with CAR-T cells with the CD28 costimulatory motif, CAR-T cells with the 4-1BB costimulatory motif showed better therapeutic efficiency without cell enrichment. Taken together, these results show that anti-CD19 CAR-T cell therapy was effective in the prevention and treatment of a murine model of SLE, indicating its potential for clinical use in patients.

Keywords: Autoimmune disease; B cells; Systemic lupus erythematosus; T cells; Treatment.

Conflict of interest statement

The authors declare no competing interests.

© 2020. CSI and USTC.

Figures

Fig. 1
Fig. 1
CAR-T cell construction. a Flow cytometry showing mCherry-CAR expression on mouse primary T cells 2 days after transduction. bd Effector cells, sorted CAR-T cells cultured for another 3–5 days; target cells, freshly sorted splenocytes labeled with CellTrace Violet. After coculture at the indicated ratios for 4 h, the percentage of lysed B cells (b) and levels of intracellular IFNγ expressed by effector cells (c) were analyzed by flow cytometry. d The supernatant IFNγ concentration after 24 h of coculture was analyzed by ELISA. The data are representative of at least two independent experiments with similar results
Fig. 2
Fig. 2
Efficacy of CAR-T cells in an SLE mouse model. a Working scheme of CAR-T cell manufacture and adoptive transfer in a mouse experiment. The percentage of blood CD19+ cells at 1 week (b, c) or 9 weeks (d) after transfer was analyzed by flow cytometry; the percentage of blood CD138+ cells at 1 week (e) or 9 weeks (f) after transfer was analyzed by flow cytometry. Live CD45+ cells were gated and plotted (bf). g Effector cells, lymphocytes isolated from TBI + CAR-T cell-treated mice 9 weeks after transfer; target cells, fresh splenocytes labeled with CellTrace Violet; the percentage of lysed B cells was analyzed by flow cytometry after 24 h of coculture. The data are representative of at least two independent experiments with similar results. The data were analyzed by Student’s t test, and significance is indicated by *P < 0.05, ****P < 0.0001, and ns no significant differences
Fig. 3
Fig. 3
CAR-T cell treatment shows therapeutic benefits in mouse SLE. Mice received different treatments at 13 weeks of age and were sacrificed at 22 weeks of age for subsequent examinations. a The survival rate of MRL-lpr mice that received different treatments; the difference in survival between the TBI + PBS group and TBI + CAR-T group was compared by logrank (Mantel–Cox test) calculation. b The mouse skin lesions were scored, and the scores were plotted. c The ratios of spleen weight to total body weight were determined. d Representative hematoxylin and eosin (H&E)-stained sections of glomerular areas of the kidneys. Black arrow, immune complex formation; red arrow, infiltration of numerous lymphocytes. Original magnification, ×400. Bars represent 50 µm. e Statistical histology scores of H&E-stained kidney sections. f C3 and IgG deposition in the glomerulus was detected by immunofluorescence and is presented in a merged manner; frozen sections of the kidneys were stained with anti-mouse-C3-APC, anti-mouse-IgG-FITC, and DAPI. Original magnification, ×400. Bars represent 50 µm. The data are representative of at least two independent experiments with similar results. b, c The data were analyzed by Student’s t test, and significance is indicated by *P < 0.05
Fig. 4
Fig. 4
1D3-4-1BB CAR-T cells partially deplete B cells. a Diagram of the DNA encoding 1D3-4-1BB CAR. It encodes the antibody light chains (blue boxes), including their native signal peptides (white box), joined by a flexible linker (blue bar) to the VH and fused to the transmembrane region of CD8a (navy blue), signaling domains of 4-1BB (red box) and cytosolic domain of CD3ζ (orange box). The construct also contains IRES-driven EGFP to allow the detection of transduced cells. b Flow cytometry of GFP+ expression on mouse primary T cells after lentiviral transduction. Cells were FVD− TCRβ+ gated. c Unsorted 4-1BB CAR-T cells (effector) and CellTrace Violet-labeled splenocytes (target) were cocultured for 4 h and analyzed by flow cytometry. Blood was collected from the femoral arteries of MRL-lpr mice at 1 week (d, f) or 9 weeks (e, g) after transfer, and CD19+ and CD138+ percentages among FVD− CD45+ cells were analyzed by flow cytometry. CD4/CD8 ratios in the blood were also analyzed by flow cytometry at 1 week (h) or 9 weeks (i) after transfer. The data are representative of at least two independent experiments with similar results. The data were analyzed by Student’s t test, and significance is indicated by *P < 0.05
Fig. 5
Fig. 5
Therapeutic effects of 4-1BB CAR-T cell treatment. Mice that received low-dose TBI as preconditioning and adoptive transfer with 4-1BB CAR-T cells or PBS at 13 weeks of age were sacrificed at 22 weeks of age for subsequent examinations. a The survival rate of the mice; the difference in survival between the two groups was compared by logrank (Mantel–Cox test) calculation. b Total body weight and spleen weight. Sera were isolated from MRL-lpr mice; anti-dsDNA antibody (c) and ANA (d) levels were detected by ELISA. e Urine protein to creatinine ratios were analyzed by a chemistry analyzer. f Representative H&E-stained sections of glomerular areas of the kidneys. Black arrow, immune complex formation; red arrow, infiltration of numerous lymphocytes. Original magnification, ×400. Bars represent 50 µm. g C3 and IgG deposition in the glomerulus was detected by immunofluorescence and is presented in a merged manner; frozen sections of the kidney were stained with anti-mouse-C3-APC, anti-mouse-IgG-FITC, and DAPI. Original magnification, ×400. Bars represent 50 µm. The data are representative of at least two independent experiments with similar results. b, d The data were analyzed by Student’s t test, and significance is indicated by *P < 0.05

References

    1. Rahman A, Isenberg DA. Systemic lupus erythematosus. N. Engl. J. Med. 2008;358:929–939. doi: 10.1056/NEJMra071297.
    1. Arbuckle MR, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N. Engl. J. Med. 2003;349:1526–1533. doi: 10.1056/NEJMoa021933.
    1. Xiong W, Lahita RG. Pragmatic approaches to therapy for systemic lupus erythematosus. Nat. Rev. Rheumatol. 2014;10:97–107. doi: 10.1038/nrrheum.2013.157.
    1. Isenberg D, et al. Efficacy and safety of atacicept for prevention of flares in patients with moderate-to-severe systemic lupus erythematosus (SLE): 52-week data (APRIL-SLE randomised trial) Ann. Rheum. Dis. 2015;74:2006–2015. doi: 10.1136/annrheumdis-2013-205067.
    1. Furie RA, et al. A phase 2, randomised, placebo-controlled clinical trial of blisibimod, an inhibitor of B cell activating factor, in patients with moderate-to-severe systemic lupus erythematosus, the PEARL-SC study. Ann. Rheum. Dis. 2015;74:1667–1675. doi: 10.1136/annrheumdis-2013-205144.
    1. Vital EM, et al. B cell biomarkers of rituximab responses in systemic lupus erythematosus. Arthritis Rheum. 2011;63:3038–3047. doi: 10.1002/art.30466.
    1. Jonsdottir T, Sundelin B, Welin Henriksson E, van Vollenhoven RF, Gunnarsson I. Rituximab-treated membranous lupus nephritis: clinical outcome and effects on electron dense deposits. Ann. Rheum. Dis. 2011;70:1172–1173. doi: 10.1136/ard.2010.129288.
    1. Arce-Salinas CA, Rodriguez-Garcia F, Gomez-Vargas JI. Long-term efficacy of anti-CD20 antibodies in refractory lupus nephritis. Rheumatol. Int. 2012;32:1245–1249. doi: 10.1007/s00296-010-1755-0.
    1. Gregersen JW, Jayne DR. B-cell depletion in the treatment of lupus nephritis. Nat. Rev. Nephrol. 2012;8:505–514. doi: 10.1038/nrneph.2012.141.
    1. Odendahl M, et al. Disturbed peripheral B lymphocyte homeostasis in systemic lupus erythematosus. J. Immunol. 2000;165:5970–5979. doi: 10.4049/jimmunol.165.10.5970.
    1. Mei HE, Schmidt S, Dorner T. Rationale of anti-CD19 immunotherapy: an option to target autoreactive plasma cells in autoimmunity. Arthritis Res. Ther. 2012;14(Suppl 5):S1. doi: 10.1186/ar3909.
    1. Jyothi MD, Flavell RA, Geiger TL. Targeting autoantigen-specific T cells and suppression of autoimmune encephalomyelitis with receptor-modified T lymphocytes. Nat. Biotechnol. 2002;20:1215–1220. doi: 10.1038/nbt758.
    1. Ellebrecht CT, et al. Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease. Science. 2016;353:179–184. doi: 10.1126/science.aaf6756.
    1. Zhang L, et al. Chimeric antigen receptor (CAR) T cells targeting a pathogenic MHC class II:peptide complex modulate the progression of autoimmune diabetes. J. Autoimmun. 2019;96:50–58. doi: 10.1016/j.jaut.2018.08.004.
    1. Fransson M, et al. CAR/FoxP3-engineered T regulatory cells target the CNS and suppress EAE upon intranasal delivery. J. Neuroinflammation. 2012;9:112. doi: 10.1186/1742-2094-9-112.
    1. Blat D, Zigmond E, Alteber Z, Waks T, Eshhar Z. Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells. Mol. Ther. 2014;22:1018–1028. doi: 10.1038/mt.2014.41.
    1. MacDonald KG, et al. Alloantigen-specific regulatory T cells generated with a chimeric antigen receptor. J. Clin. Investig. 2016;126:1413–1424. doi: 10.1172/JCI82771.
    1. Kansal R, et al. Sustained B cell depletion by CD19-targeted CAR T cells is a highly effective treatment for murine lupus. Sci Transl Med. 2019;11:eaav1648. doi: 10.1126/scitranslmed.aav1648.
    1. Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood. 2010;116:3875–3886. doi: 10.1182/blood-2010-01-265041.
    1. Feucht J, et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat. Med. 2019;25:82–88. doi: 10.1038/s41591-018-0290-5.
    1. Cheadle EJ, et al. Natural expression of the CD19 antigen impacts the long-term engraftment but not antitumor activity of CD19-specific engineered T cells. J. Immunol. 2010;184:1885–1896. doi: 10.4049/jimmunol.0901440.
    1. Abraham PM, Quan SH, Dukala D, Soliven B. CD19 as a therapeutic target in a spontaneous autoimmune polyneuropathy. Clin. Exp. Immunol. 2014;175:181–191. doi: 10.1111/cei.12215.
    1. Hofmann K, Clauder AK, Manz RA. Targeting B cells and plasma cells in autoimmune diseases. Front. Immunol. 2018;9:835. doi: 10.3389/fimmu.2018.00835.
    1. Zhao Z, et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015;28:415–428. doi: 10.1016/j.ccell.2015.09.004.
    1. Li S, et al. CD33-specific chimeric antigen receptor T cells with different co-stimulators showed potent anti-leukemia efficacy and different phenotype. Hum. Gene Ther. 2018;29:626–639. doi: 10.1089/hum.2017.241.
    1. Priceman SJ, et al. Co-stimulatory signaling determines tumor antigen sensitivity and persistence of CAR T cells targeting PSCA+ metastatic prostate cancer. Oncoimmunology. 2018;7:e1380764. doi: 10.1080/2162402X.2017.1380764.
    1. Du H, et al. Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells. Cancer Cell. 2019;35:221–37 e8. doi: 10.1016/j.ccell.2019.01.002.
    1. Hamieh M, et al. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature. 2019;568:112–116. doi: 10.1038/s41586-019-1054-1.
    1. van Bekkum DW. Effectiveness and risks of total body irradiation for conditioning in the treatment of autoimmune disease with autologous bone marrow transplantation. Rheumatology. 1999;38:757–761. doi: 10.1093/rheumatology/38.8.757.
    1. Zhao J, et al. P2X7 blockade attenuates murine lupus nephritis by inhibiting activation of the NLRP3/ASC/caspase 1 pathway. Arthritis Rheum. 2013;65:3176–3185. doi: 10.1002/art.38174.

Source: PubMed

3
구독하다