Comparison of seven CD19 CAR designs in engineering NK cells for enhancing anti-tumour activity

Yao Wang , Jianhuan Li , Zhiqian Wang , Yanhong Liu , Tongjie Wang , Mengyun Zhang , Chengxiang Xia , Fan Zhang , Dehao Huang , Leqiang Zhang , Yaoqin Zhao , Lijuan Liu , Yanping Zhu , Hanmeng Qi , Xiaofan Zhu , Wenbin Qian , Fangxiao Hu , Jinyong Wang

Cell Proliferation ›› 2024, Vol. 57 ›› Issue (11) : e13683

PDF
Cell Proliferation ›› 2024, Vol. 57 ›› Issue (11) : e13683 DOI: 10.1002/cpr.13683
ORIGINAL ARTICLE

Comparison of seven CD19 CAR designs in engineering NK cells for enhancing anti-tumour activity

Author information +
History +
PDF

Abstract

Chimeric antigen receptor-natural killer (CAR-NK) cell therapy is emerging as a promisingcancer treatment, with notable safety and source diversity benefits over CAR-Tcells. This study focused on optimizing CAR constructs for NK cells to maximize theirtherapeutic potential. We designed seven CD19 CAR constructs and expressed themin NK cells using a retroviral system, assessing their tumour-killing efficacy and persistence.Results showed all constructs enhanced tumour-killing and prolonged survivalin tumour-bearing mice. In particular, CAR1 (CD8 TMD-CD3ζ SD)-NK cells showedsuperior efficacy in treating tumour-bearing animals and exhibited enhanced persistencewhen combined with OX40 co-stimulatory domain. Of note, CAR1-NK cellswere most effective at lower effector-to-target ratios, while CAR4 (CD8 TMD-OX40CD- FcϵRIγ SD) compromised NK cell expansion ability. Superior survival rates werenoted in mice treated with CAR1-, CAR2 (CD8 TMD- FcϵRIγ SD)-, CAR3 (CD8 TMDOX40CD- CD3ζ SD)- and CAR4-NK cells over those treated with CAR5 (CD28TMD- FcϵRIγ SD)-, CAR6 (CD8 TMD-4-1BB CD-CD3ζ 1-ITAM SD)- and CAR7 (CD8TMD-OX40 CD-CD3ζ 1-ITAM SD)-NK cells, with CAR5-NK cells showing the weakestanti-tumour activity. Increased expression of exhaustion markers, especially inCAR7-NK cells, suggests that combining CAR-NK cells with immune checkpointinhibitors might improve anti-tumour outcomes. These findings provide crucialinsights for developing CAR-NK cell products for clinical applications.

Cite this article

Download citation ▾
Yao Wang, Jianhuan Li, Zhiqian Wang, Yanhong Liu, Tongjie Wang, Mengyun Zhang, Chengxiang Xia, Fan Zhang, Dehao Huang, Leqiang Zhang, Yaoqin Zhao, Lijuan Liu, Yanping Zhu, Hanmeng Qi, Xiaofan Zhu, Wenbin Qian, Fangxiao Hu, Jinyong Wang. Comparison of seven CD19 CAR designs in engineering NK cells for enhancing anti-tumour activity. Cell Proliferation, 2024, 57(11): e13683 DOI:10.1002/cpr.13683

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

GrossG, Gorochov G, WaksT, EshharZ. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant Proc. 1989;21:127-130.

[2]

TranAC, ZhangDH, ByrnR, Roberts MR. Chimeric zeta-receptors direct human natural-killer (Nk) effector function to permit killing of Nk-resistant tumor-cells and Hiv-infected T-lymphocytes. J Immunol. 1995;155:1000-1009.

[3]

KlichinskyM, RuellaM, ShestovaO, et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat Biotechnol. 2020;38:947-953.

[4]

BoyiadzisMM, Dhodapkar MV, BrentjensRJ, et al. Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. J Immunother Cancer. 2018;6:137.

[5]

RafiqS, Hackett CS, BrentjensRJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol. 2020;17:147-167.

[6]

WangJ, ChenS, XiaoW, et al. CAR-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J Hematol Oncol. 2018;11:7.

[7]

MajznerRG, Rietberg SP, SotilloE, et al. Tuning the antigen density requirement for CAR T-cell activity. Cancer Discov. 2020;10:702-723.

[8]

RomeoC, SeedB. Cellular immunity to HIV activated by CD4 fused to T cell or Fc receptor polypeptides. Cell. 1991;64:1037-1046.

[9]

LiSS, TaoZF, XuYX, 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.

[10]

ImaiC, MiharaK, AndreanskyM, et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18:676-684.

[11]

TanJW, JiaYJ, ZhouMX, et al. Chimeric antigen receptors containing the OX40 signalling domain enhance the persistence of T cells even under repeated stimulation with multiple myeloma target cells. J Hematol Oncol. 2022;15:39.

[12]

López-CantilloG, Urueña C, CamachoBA, Ramírez-SeguraC. CAR-T cell performance: how to improve their persistence? Front Immunol. 2022;13:878209.

[13]

JamesJR. Tuning ITAM multiplicity on T cell receptors can control potency and selectivity to ligand density. Sci Signal. 2018;11:eaan1088.

[14]

FeuchtJ, SunJ, EyquemJ, et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat Med. 2019;25:82-88.

[15]

JayaramanJ, Mellody MP, HouAJ, et al. CAR-T design: elements and their synergistic function. EBioMedicine. 2020;58:102931.

[16]

van der StegenSJC, Hamieh M, SadelainM. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov. 2015;14:499-509.

[17]

ZhaoZG, Condomines M, van der StegenSJC, et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015;28:415-428.

[18]

AjinaA, MaherJ. Strategies to address chimeric antigen receptor tonic signaling. Mol Cancer Ther. 2018;17:1795-1815.

[19]

DagarG, GuptaA, MasoodiT, et al. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med. 2023;21:449.

[20]

MengX, JingR, QianL, Zhou C, SunJ. Engineering cytoplasmic signaling of CD28ζ CARs for improved therapeutic functions. Front Immunol. 2020;11:1046.

[21]

WolfNK, Kissiov DU, RauletDH. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat Rev Immunol. 2023;23:90-105.

[22]

LiuE, MarinD, BanerjeeP, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med. 2020;382:545-553.

[23]

PanK, Farrukh H, ChittepuV, XuH, PanCX, ZhuZ. CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J Exp Clin Cancer Res. 2022;41:119.

[24]

ImaiC, Iwamoto S, CampanaD. Genetic modification of primary natural killer cells overcomes inhibitory signals and induces specific killing of leukemic cells. Blood. 2005;106:376-383.

[25]

BurenLG, GuoC, FanY, et al. Abstract B64:Coexpression of a CD19-OX40-CD3ζ CAR with membrane-bound IL-15 enhances natural killer cell function. Cancer Immunol Res. 2020;8:B64-B64.

[26]

DagherOK, PoseyAD Jr. Forks in the road for CAR T and CAR NK cell cancer therapies. Nat Immunol. 2023;24:1994-2007.

[27]

MengFY, ZhangSQ, XieJ, et al. Leveraging CD16 fusion receptors to remodel the immune response for enhancing anti-tumor immunotherapy in iPSC-derived NK cells. J Hematol Oncol. 2023;16:62.

[28]

RavetchJV, LanierLL. Immune inhibitory receptors. Science. 2000;290:84-89.

[29]

RobbinsY, GreeneS, FriedmanJ, et al. Tumor control via targeting PD-L1 with chimeric antigen receptor modified NK cells. eLife. 2020;9:e54854.

[30]

LiuQ, XuYX, MouJL, et al. Irradiated chimeric antigen receptor engineered NK-92MI cells show effective cytotoxicity against CD19 malignancy in a mouse model. Cytotherapy. 2020;22:552-562.

[31]

RiviereI, BroseK, MulliganRC. Effects of retroviral vector design on expression of human adenosine-deaminase in murine bone-marrow transplant recipients engrafted with genetically-modified cells. Proc Natl Acad Sci USA. 1995;92:6733-6737.

[32]

KellyPF, Vandergriff J, NathwaniA, NienhuisAW, VaninEF. Highly efficient gene transfer into cord blood nonobese diabetic/severe combined immunodeficiency repopulating cells by oncoretroviral vector particles pseudotyped with the feline endogenous retrovirus (RD114) envelope protein. Blood. 2000;96:1206-1214.

[33]

LiuE, TongY, DottiG, et al. Cord blood NK cells engineered to express IL-15 and a CD19-targeted CAR show long-term persistence and potent antitumor activity. Leukemia. 2018;32:520-531.

[34]

NicholsonIC, LentonKA, LittleDJ, et al. Construction and characterisation of a functional CD19 specific single chain Fv fragment for immunotherapy of B lineage leukaemia and lymphoma. Mol Immunol. 1997;34:1157-1165.

[35]

LanierLL, YuG, PhillipsJH. Analysis of Fc gamma RIII (CD16) membrane expression and association with CD3 zeta and Fc epsilon RI-gamma by site-directed mutation. J Immunol. 1991;146:1571-1576.

[36]

AlterG, Malenfant JM, AltfeldM. CD107a as a functional marker for the identification of natural killer cell activity. J Immunol Methods. 2004;294:15-22.

[37]

WangR, JawJJ, StutzmanNC, Zou Z, SunPD. Natural killer cell-produced IFN-gamma and TNF-alpha induce target cell cytolysis through up-regulation of ICAM-1. J Leukoc Biol. 2012;91:299-309.

[38]

QuatriniL, Della Chiesa M, SivoriS, MingariMC, PendeD, MorettaL. Human NK cells, their receptors and function. Eur J Immunol. 2021;51:1566-1579.

[39]

WoanKV, KimH, BjordahlR, et al. Harnessing features of adaptive NK cells to generate iPSC-derived NK cells for enhanced immunotherapy. Cell Stem Cell. 2021;28:2062-2075.e5.

[40]

JiaH, YangH, XiongH, Luo KQ. NK cell exhaustion in the tumor microenvironment. Front Immunol. 2023;14:1303605.

[41]

VankayalapatiR, GargA, PorgadorA, et al. Role of NK cell-activating receptors and their ligands in the lysis of mononuclear phagocytes infected with an intracellular bacterium. J Immunol. 2005;175:4611-4617.

[42]

SadelainM, Brentjens R, RivièreI. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol. 2009;21:215-223.

[43]

LanierLL. Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol. 2008;9:495-502.

[44]

HombachAA, AbkenH. Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling. Int J Cancer. 2011;129:2935-2944.

[45]

ZhangHH, LiFL, CaoJ, et al. A chimeric antigen receptor with antigen-independent OX40 signaling mediates potent antitumor activity. Sci Transl Med. 2021;13:eaba7308.

[46]

DaveyAS, CallME, CallMJ. The influence of chimeric antigen receptor structural domains on clinical outcomes and associated toxicities. Cancer. 2021;13:38.

[47]

FujiwaraK, TsuneiA, KusabukaH, Ogaki E, TachibanaM, OkadaN. Hinge and transmembrane domains of chimeric antigen receptor regulate receptor expression and signaling threshold. Cells. 2020;9:1182.

[48]

LiW, QiuS, ChenJ, et al. Chimeric antigen receptor designed to prevent ubiquitination and downregulation showed durable antitumor efficacy. Immunity. 2020;53:456-470.e456.

[49]

DavenportAJ, Jenkins MR, CrossRS, et al. CAR-T cells inflict sequential killing of multiple tumor target cells. Cancer Immunol Res. 2015;3:483-494.

[50]

WalkerAJ, Majzner RG, ZhangL, et al. Tumor antigen and receptor densities regulate efficacy of a chimeric antigen receptor targeting anaplastic lymphoma kinase. Mol Ther. 2017;25:2189-2201.

[51]

RoeK. NK-cell exhaustion, B-cell exhaustion and T-cell exhaustion-the differences and similarities. Immunology. 2022;166:155-168.

[52]

YangK, ZhaoY, SunG, et al. Clinical application and prospect of immune checkpoint inhibitors for CAR-NK cell in tumor immunotherapy. Front Immunol. 2022;13:1081546.

RIGHTS & PERMISSIONS

2024 The Author(s). Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

196

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/