Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2023, Vol. 17 Issue (6) : 1170-1185    https://doi.org/10.1007/s11684-023-0996-8
BGB-A445, a novel non-ligand-blocking agonistic anti-OX40 antibody, exhibits superior immune activation and antitumor effects in preclinical models
Beibei Jiang1, Tong Zhang1, Minjuan Deng2, Wei Jin2, Yuan Hong1, Xiaotong Chen1, Xin Chen1, Jing Wang1, Hongjia Hou1, Yajuan Gao1, Wenfeng Gong1, Xing Wang1, Haiying Li1, Xiaosui Zhou1, Yingcai Feng1, Bo Zhang1, Bin Jiang2, Xueping Lu2, Lijie Zhang2, Yang Li2, Weiwei Song2, Hanzi Sun1, Zuobai Wang3, Xiaomin Song1, Zhirong Shen2, Xuesong Liu1, Kang Li4, Lai Wang1, Ye Liu1()
1. Department of Biology
2. Department of Discovery Biomarkers
3. Department of Clinic Development
4. Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing 102206, China
 Download: PDF(6159 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

OX40 is a costimulatory receptor that is expressed primarily on activated CD4+, CD8+, and regulatory T cells. The ligation of OX40 to its sole ligand OX40L potentiates T cell expansion, differentiation, and activation and also promotes dendritic cells to mature to enhance their cytokine production. Therefore, the use of agonistic anti-OX40 antibodies for cancer immunotherapy has gained great interest. However, most of the agonistic anti-OX40 antibodies in the clinic are OX40L-competitive and show limited efficacy. Here, we discovered that BGB-A445, a non-ligand-competitive agonistic anti-OX40 antibody currently under clinical investigation, induced optimal T cell activation without impairing dendritic cell function. In addition, BGB-A445 dose-dependently and significantly depleted regulatory T cells in vitro and in vivo via antibody-dependent cellular cytotoxicity. In the MC38 syngeneic model established in humanized OX40 knock-in mice, BGB-A445 demonstrated robust and dose-dependent antitumor efficacy, whereas the ligand-competitive anti-OX40 antibody showed antitumor efficacy characterized by a hook effect. Furthermore, BGB-A445 demonstrated a strong combination antitumor effect with an anti-PD-1 antibody. Taken together, our findings show that BGB-A445, which does not block OX40–OX40L interaction in contrast to clinical-stage anti-OX40 antibodies, shows superior immune-stimulating effects and antitumor efficacy and thus warrants further clinical investigation.

Keywords BGB-A445      OX40      agonistic antibody      OX40L noncompetitive     
Corresponding Author(s): Ye Liu   
Just Accepted Date: 28 July 2023   Online First Date: 26 September 2023    Issue Date: 06 February 2024
 Cite this article:   
Beibei Jiang,Tong Zhang,Minjuan Deng, et al. BGB-A445, a novel non-ligand-blocking agonistic anti-OX40 antibody, exhibits superior immune activation and antitumor effects in preclinical models[J]. Front. Med., 2023, 17(6): 1170-1185.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-023-0996-8
https://academic.hep.com.cn/fmd/EN/Y2023/V17/I6/1170
Fig.1  BGB-A445 binds to OX40 potently without interfering with OX40–OX40L interaction. (A) Schematic overview of the discovery of BGB-A445. First, antibody clones were generated via hybridoma fusion with splenocytes from OX40 extracellular domain-immunized BALB/c mice. Then, OX40 binding, T cell costimulation, and OX40L-competition assays were performed by using ELISA, SPR, or cell-based assays, respectively. Finally, hit 445 was humanized and optimized to reduce immunogenicity and improve stability. (B) Binding of BGB-A445 to cell-surface-overexpressed OX40 on HuT78 cells determined via FACS. MOXR0916 was used as a reference antibody. (C) Binding of BGB-A445 to cell-surface-expressed OX40 on human primary CD4+ T cells from six different donors. In consideration of donor variation, the binding signal was further normalized to the receptor occupancy rate of the OX40 receptor. (D) Non-OX40L-competitive property of BGB-A445 in the cell-based assay and comparison with the properties of other clinical-stage agonistic anti-OX40 antibodies. In this assay, the control was set as OX40 alone, and the experimental groups included OX40 in the presence of huIgG, BGB-A445, or reference antibodies. In (B–D) the data are presented as the mean value ± SD from two (B, C) or three (D) replicates.
Fig.2  Crystal structure of the OX40/BGB-A445 Fab complex further confirms the noncompetitiveness of BGB-A445 with OX40L. (A) Crystal structure of OX40 in complex with BGB-A445 Fab. BGB-A445 is shown as a ribbon (heavy chain, cyan; light chain, light cyan). OX40 is shown as a surface representation. HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 of BGB-A445 are highlighted in red, yellow, light golden, pink, blue, and magenta, respectively, and CRD4 of OX40 is colored dark gray. (B) Detailed interactions between BGB-A445 and its key epitopes. BGB-A445 is shown as a gray ribbon and surface, and OX40 is shown as a green ribbon. Key residues are represented as sticks. VDW, van der Waals interaction. (C) Structural alignment of OX40/BGB-A445 Fab with the reported OX40/MOXR0916 (PDB: 6OKN) and OX40/OX40L (PDB: 2HEV) complexes. BGB-A445, MOXR0916, and OX40L are shown as cyan, magenta, and orange ribbons, respectively. Meanwhile, the CRD1–CRD4 regions of OX40 are differentiated with light and dark gray surface representations. (D) Comparison of binding interfaces among BGB-A445, MOXR0916, and OX40L on the OX40 surface. The surfaces colored cyan, magenta, and orange are the binding surfaces of BGB-A445, MOXR0916, and the OX40L trimer, respectively, and the surface colored red represents the overlapping interface of MOXR0916 and the OX40L trimer.
Fig.3  BGB-A445 induces optimal T cell activation and DC maturation. (A) BGB-A445 enhances IL-2 production in conjunction with T cell receptor signaling. HuT78/OX40 cells were cocultured overnight with HEK293/ FcγRI/anti-CD3 cells in the presence of serially diluted OX40 mAbs (BGB-A445 or MOXR0916). IL-2 production in the supernatant was determined by ELISA. (B) Schematic of the CD4+ T/DC coculture system. Immature DCs were cocultured with autologous CD4+ T cells with SEB and OX40 mAbs for 48 h. The secretion of IL-2 by T cells (C, D) and the expression of costimulatory molecules (CD86 and CD83) on DCs (E) were determined by ELISA and flow cytometry, respectively. (D) Indicated OX40 mAbs (10 μg/mL) were added to the coculture system alone or in combination with OX40L-blocking antibodies (10 μg/mL). The data shown in panels (C) and (D) are representative of at least three independent experiments from multiple donors. The data are presented as mean ± SD (*, P < 0.05).
Fig.4  BGB-A445 dose-dependently increased proliferating and activated Teffs in hOX40 knock-in mice bearing MC38 tumors. hOX40 knock-in mice were inoculated with MC38 cells. Single cells isolated from the spleens were stained and analyzed through flow cytometry at 24 h after the second injection of BGB-A445 or MOXR0916. (A) Number of proliferating (Ki67+) CD4 Teffs. (B) Number of proliferating (Ki67+) CD8 T cells. (C) Number of proliferating (Ki67+) memory CD4 (CD44-high) T cells. (D) Number of proliferating (Ki67+) memory CD8 (CD44-high) T cells. All data are presented as mean ± SEM, and each dot represents one mouse sample. Statistically significant differences were analyzed by conducting one-way ANOVA on log-transformed data by using Holm–Sidak’s multiple comparisons test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001).
Fig.5  BGB-A445 preferentially depletes Tregs and increases the CD8+/Treg ratio in vitro and in vivo. (A) In an ADCC assay, NK cells were cocultured with PHA-stimulated PBMCs in the presence of OX40 mAbs (BGB-A445, MOXR0916, or Fc effectorless BGB-A445 mf) overnight. The percentage of Tregs among CD4+ cells was determined by flow cytometry, as described in the Methods and Materials section. CD4+ effector (B) and CD8+ T cell (C) percentages and the CD8+/Treg ratio (D) were also determined. (E–G) hOX40 knock-in mice were inoculated with MC38 cells. Tumors were isolated 24 h after the second injection of BGB-A445 or MOXR0916. TILs (CD45+ cells) isolated from tumor-dissociated single-cell suspensions were stained and analyzed by flow cytometry. (E) Percentage of Tregs among TILs. (F) Percentage of CD8+ T cells among TILs. (G) CD8+/Treg ratio. All data are presented as mean ± SEM, and each dot represents one animal. Statistical differences were analyzed by conducting one-way ANOVA on log-transformed data by using Holm–Sidak’s multiple comparisons test (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001).
Fig.6  BGB-A445 executes dose-dependent antitumor activity in MC38 syngeneic tumor models and presents superior efficacy in the PD-1 resistant model PAN02. (A) MC38 tumor cells were implanted s.c. into hOX40 knock-in mice. On day 7 after cell implantation, the mice were randomly allocated into five groups and treated with the vehicle, BGB-A445, or MOXR0916 (0.4, 2, or 10 mpk) i.p. once a week for 2 weeks. The data are presented as mean bodyweight ± SEM for 10 animals in each group. (B) TGI on day 16 was correlated with the dose used in the MC38 model. (C) Animal body weight in the MC38 model group. Statistical differences were analyzed by using nonparametric Dunn’s multiple comparisons test, where * denotes an adjusted P < 0.05, **** denotes an adjusted P < 0.0001, and ns denotes no statistical significance. (D) PAN02 cells were implanted s.c. into hOX40 knock-in mice. The mice were randomly allocated into four groups and treated with the vehicle, BGB-A445, muCh15mt, or MOXR0916 once a week. The data are presented as the mean tumor volume ± SEM from nine animals in each group. The data of the treatment groups were compared with log-transformed data by using Holm–Sidak’s multiple comparisons test (**, P < 0.01; ***, P < 0.001). (E) Body weights of PAN02 model animals.
Fig.7  BGB-A445 exerts a combination effect when used with an anti-PD-1 antibody. (A) BGB-A445 enhances MLR. In vitro-differentiated DCs were cocultured with allogeneic CD4+ T cells in the presence of BGB-A445 or MOXR0916 (0.1–10 μg/mL) with or without 50 ng/mL BGB-A317 for 2 days. IL-2 in the supernatant was detected by ELISA. All data are presented as the mean ± SD from quadruplicates. Statistical significance: *, P < 0.05; **, P < 0.01. (B) MC38 tumor cells were implanted s.c. in hOX40 knock-in mice. On day 10 after implantation, the mice were randomly allocated into four groups and treated with the vehicle, BGB-A445 monotherapy, muCh15mt monotherapy, or combination treatment i.p. once a week for 2 weeks. The data are presented as the mean tumor volume ± SEM from nine animals in each group. (C) Tumor volumes in individual animals for the MC38 model. (D) Bodyweights of the MC38 model animals.
Fig.8  Non-OX40L-blocking anti-OX40 antibody may induce superior T cell activation to an OX40L-blocking antibody. BGB-A445 can bind to an OX40 epitope that is different from OX40L and stimulate T cells without impairing APC activation, hence promoting T cell proliferation and activation to maximum levels.
1 J Liu, Z Chen, Y Li, W Zhao, J Wu, Z Zhang. PD-1/PD-L1 checkpoint inhibitors in tumor immunotherapy. Front Pharmacol 2021; 12: 731798
https://doi.org/10.3389/fphar.2021.731798
2 S Piconese, B Valzasina, MP Colombo. OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med 2008; 205(4): 825–839
https://doi.org/10.1084/jem.20071341
3 R Duhen, C Ballesteros-Merino, AK Frye, E Tran, V Rajamanickam, SC Chang, Y Koguchi, CB Bifulco, B Bernard, RS Leidner, BD Curti, BA Fox, WJ Urba, RB Bell, AD Weinberg. Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat Commun 2021; 12(1): 1047
https://doi.org/10.1038/s41467-021-21383-1
4 JA Marin-Acevedo, B Dholaria, AE Soyano, KL Knutson, S Chumsri, Y Lou. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol 2018; 11(1): 39
https://doi.org/10.1186/s13045-018-0582-8
5 H Wajant. Principles of antibody-mediated TNF receptor activation. Cell Death Differ 2015; 22(11): 1727–1741
https://doi.org/10.1038/cdd.2015.109
6 E Stüber, M Neurath, D Calderhead, HP Fell, W Strober. Cross-linking of OX40 ligand, a member of the TNF/NGF cytokine family, induces proliferation and differentiation in murine splenic B cells. Immunity 1995; 2(5): 507–521
https://doi.org/10.1016/1074-7613(95)90031-4
7 Y Ohshima, Y Tanaka, H Tozawa, Y Takahashi, C Maliszewski, G Delespesse. Expression and function of OX40 ligand on human dendritic cells. J Immunol 1997; 159(8): 3838–3848
https://doi.org/10.4049/jimmunol.159.8.3838
8 AD Weinberg, KW Wegmann, C Funatake, RH Whitham. Blocking OX-40/OX-40 ligand interaction in vitro and in vivo leads to decreased T cell function and amelioration of experimental allergic encephalomyelitis. J Immunol 1999; 162(3): 1818–1826
https://doi.org/10.4049/jimmunol.162.3.1818
9 T Ito, R Amakawa, M Inaba, T Hori, M Ota, K Nakamura, M Takebayashi, M Miyaji, T Yoshimura, K Inaba, S Fukuhara. Plasmacytoid dendritic cells regulate Th cell responses through OX40 ligand and type I IFNs. J Immunol 2004; 172(7): 4253–4259
https://doi.org/10.4049/jimmunol.172.7.4253
10 M Croft. Control of immunity by the TNFR-related molecule OX40 (CD134). Annu Rev Immunol 2010; 28(1): 57–78
https://doi.org/10.1146/annurev-immunol-030409-101243
11 J Song, T So, M Croft. Activation of NF-κB1 by OX40 contributes to antigen-driven T cell expansion and survival. J Immunol 2008; 180(11): 7240–7248
https://doi.org/10.4049/jimmunol.180.11.7240
12 A Song, X Tang, KM Harms, M Croft. OX40 and Bcl-xL promote the persistence of CD8 T cells to recall tumor-associated antigen. J Immunol 2005; 175(6): 3534–3541
https://doi.org/10.4049/jimmunol.175.6.3534
13 PR Rogers, J Song, I Gramaglia, N Killeen, M Croft. OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells. Immunity 2001; 15(3): 445–455
https://doi.org/10.1016/S1074-7613(01)00191-1
14 J Song, T So, M Cheng, X Tang, M Croft. Sustained survivin expression from OX40 costimulatory signals drives T cell clonal expansion. Immunity 2005; 22(5): 621–631
https://doi.org/10.1016/j.immuni.2005.03.012
15 CA Huddleston, AD Weinberg, DC Parker. OX40 (CD134) engagement drives differentiation of CD4+ T cells to effector cells. Eur J Immunol 2006; 36(5): 1093–1103
https://doi.org/10.1002/eji.200535637
16 CE Ruby, AD Weinberg. OX40-enhanced tumor rejection and effector T cell differentiation decreases with age. J Immunol 2009; 182(3): 1481–1489
https://doi.org/10.4049/jimmunol.182.3.1481
17 I Gramaglia, AD Weinberg, M Lemon, M Croft. Ox-40 ligand: a potent costimulatory molecule for sustaining primary CD4 T cell responses. J Immunol 1998; 161(12): 6510–6517
https://doi.org/10.4049/jimmunol.161.12.6510
18 Y Ohshima, Y Tanaka, H Tozawa, Y Takahashi, C Maliszewski, G Delespesse. Expression and function of OX40 ligand on human dendritic cells. J Immunol 1997; 159(8): 3838–3848
https://doi.org/10.4049/jimmunol.159.8.3838
19 S Piconese, B Valzasina, MP Colombo. OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med 2008; 205(4): 825–839
https://doi.org/10.1084/jem.20071341
20 S Aspeslagh, S Postel-Vinay, S Rusakiewicz, JC Soria, L Zitvogel, A Marabelle. Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer 2016; 52: 50–66
https://doi.org/10.1016/j.ejca.2015.08.021
21 KS Voo, L Bover, ML Harline, LT Vien, V Facchinetti, K Arima, LW Kwak, YJ Liu. Antibodies targeting human OX40 expand effector T cells and block inducible and natural regulatory T cell function. J Immunol 2013; 191(7): 3641–3650
https://doi.org/10.4049/jimmunol.1202752
22 MC St Rose, RA Taylor, S Bandyopadhyay, HZ Qui, AT Hagymasi, AT Vella, AJ Adler. CD134/CD137 dual costimulation-elicited IFN-γ maximizes effector T-cell function but limits Treg expansion. Immunol Cell Biol 2013; 91(2): 173–183
https://doi.org/10.1038/icb.2012.74
23 T Ito, YH Wang, O Duramad, S Hanabuchi, OA Perng, M Gilliet, FX Qin, YJ Liu. OX40 ligand shuts down IL-10-producing regulatory T cells. Proc Natl Acad Sci USA 2006; 103(35): 13138–13143
https://doi.org/10.1073/pnas.0603107103
24 N Kitamura, S Murata, T Ueki, E Mekata, RT Reilly, EM Jaffee, T Tani. OX40 costimulation can abrogate Foxp3+ regulatory T cell-mediated suppression of antitumor immunity. Int J Cancer 2009; 125(3): 630–638
https://doi.org/10.1002/ijc.24435
25 A Burocchi, P Pittoni, A Gorzanelli, MP Colombo, S Piconese. Intratumor OX40 stimulation inhibits IRF1 expression and IL-10 production by Treg cells while enhancing CD40L expression by effector memory T cells. Eur J Immunol 2011; 41(12): 3615–3626
https://doi.org/10.1002/eji.201141700
26 Y Bulliard, R Jolicoeur, J Zhang, G Dranoff, NS Wilson, JL Brogdon. OX40 engagement depletes intratumoral Tregs via activating FcγRs, leading to antitumor efficacy. Immunol Cell Biol 2014; 92(6): 475–480
https://doi.org/10.1038/icb.2014.26
27 AD Weinberg, NP Morris, M Kovacsovics-Bankowski, WJ Urba, BD Curti. Science gone translational: the OX40 agonist story. Immunol Rev 2011; 244(1): 218–231
https://doi.org/10.1111/j.1600-065X.2011.01069.x
28 SN Linch, MJ McNamara, WL Redmond. OX40 agonists and combination immunotherapy: putting the pedal to the metal. Front Oncol 2015; 5: 34
https://doi.org/10.3389/fonc.2015.00034
29 P Zhang, GH Tu, J Wei, P Santiago, LR Larrabee, S Liao-Chan, T Mistry, ML Chu, T Sai, K Lindquist, H Long, J Chaparro-Riggers, S Salek-Ardakani, YA Yeung. Ligand-blocking and membrane-proximal domain targeting anti-OX40 antibodies mediate potent T cell-stimulatory and anti-tumor activity. Cell Rep 2019; 27(11): 3117–3123.e5
https://doi.org/10.1016/j.celrep.2019.05.027
30 AI Chen, AJ McAdam, JE Buhlmann, S Scott, ML Jr Lupher, EA Greenfield, PR Baum, WC Fanslow, DM Calderhead, GJ Freeman, AH Sharpe. Ox40-ligand has a critical costimulatory role in dendritic cell:T cell interactions. Immunity 1999; 11(6): 689–698
https://doi.org/10.1016/S1074-7613(00)80143-0
31 T Zhang, X Song, L Xu, J Ma, Y Zhang, W Gong, Y Zhang, X Zhou, Z Wang, Y Wang, Y Shi, H Bai, N Liu, X Yang, X Cui, Y Cao, Q Liu, J Song, Y Li, Z Tang, M Guo, L Wang, K Li. The binding of an anti-PD-1 antibody to FcγRI has a profound impact on its biological functions. Cancer Immunol Immunother 2018; 67(7): 1079–1090
https://doi.org/10.1007/s00262-018-2160-x
32 W Kabsch. XDS. Acta Crystallogr D Biol Crystallogr 2010; 66(2): 125–132
https://doi.org/10.1107/S0907444909047337
33 AJ McCoy, RW Grosse-Kunstleve, PD Adams, MD Winn, LC Storoni, RJ Read. Phaser crystallographic software. J Appl Cryst 2007; 40(4): 658–674
https://doi.org/10.1107/S0021889807021206
34 DM Compaan, SG Hymowitz. The crystal structure of the costimulatory OX40–OX40L complex. Structure 2006; 14(8): 1321–1330
https://doi.org/10.1016/j.str.2006.06.015
35 P Bernasconi-Elias, T Hu, D Jenkins, B Firestone, S Gans, E Kurth, P Capodieci, J Deplazes-Lauber, K Petropoulos, P Thiel, D Ponsel, S Hee Choi, P LeMotte, A London, M Goetcshkes, E Nolin, MD Jones, K Slocum, MJ Kluk, DM Weinstock, A Christodoulou, O Weinberg, J Jaehrling, SA Ettenberg, A Buckler, SC Blacklow, JC Aster, CJ Fryer. Characterization of activating mutations of NOTCH3 in T-cell acute lymphoblastic leukemia and anti-leukemic activity of NOTCH3 inhibitory antibodies. Oncogene 2016; 35(47): 6077–6086
https://doi.org/10.1038/onc.2016.133
36 P Emsley, B Lohkamp, WG Scott, K Cowtan. Features and development of Coot. Acta Crystallogr D Biol Crystallogr 2010; 66(4): 486–501
https://doi.org/10.1107/S0907444910007493
37 PV Afonine, RW Grosse-Kunstleve, N Echols, JJ Headd, NW Moriarty, M Mustyakimov, TC Terwilliger, A Urzhumtsev, PH Zwart, PD Adams. Towards automated crystallographic structure refinement with phenix. refine. Acta Crystallogr D Biol Crystallogr 2012; 68(4): 352–367
https://doi.org/10.1107/S0907444912001308
38 F Li, N Vijayasankaran, AY Shen, R Kiss, A Amanullah. Cell culture processes for monoclonal antibody production. MAbs 2010; 2(5): 466–479
https://doi.org/10.4161/mabs.2.5.12720
39 IL Tourkova, ZR Yurkovetsky, MR Shurin, GV Shurin. Mechanisms of dendritic cell-induced T cell proliferation in the primary MLR assay. Immunol Lett 2001; 78(2): 75–82
https://doi.org/10.1016/S0165-2478(01)00235-8
40 JL Bodmer, P Schneider, J Tschopp. The molecular architecture of the TNF superfamily. Trends Biochem Sci 2002; 27(1): 19–26
https://doi.org/10.1016/S0968-0004(01)01995-8
41 Y Yang, SH Yeh, S Madireddi, WL Matochko, C Gu, P Pacheco Sanchez, M Ultsch, G De Leon Boenig, SF Harris, B Leonard, SJ Scales, JW Zhu, E Christensen, JQ Hang, RJ Brezski, S Marsters, A Ashkenazi, S Sukumaran, H Chiu, R Cubas, JM Kim, GA Lazar. Tetravalent biepitopic targeting enables intrinsic antibody agonism of tumor necrosis factor receptor superfamily members. MAbs 2019; 11(6): 996–1011
https://doi.org/10.1080/19420862.2019.1625662
42 X Wang, M Mathieu, RJ Brezski. IgG Fc engineering to modulate antibody effector functions. Protein Cell 2018; 9(1): 63–73
https://doi.org/10.1007/s13238-017-0473-8
43 A Lee, SJ Keam. Tislelizumab: first approval. Drugs 2020; 80(6): 617–624
https://doi.org/10.1007/s40265-020-01286-z
44 S Aspeslagh, S Postel-Vinay, S Rusakiewicz, JC Soria, L Zitvogel, A Marabelle. Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer 2016; 52: 50–66
https://doi.org/10.1016/j.ejca.2015.08.021
45 M Cella, D Scheidegger, K Palmer-Lehmann, P Lane, A Lanzavecchia, G Alber. Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation. J Exp Med 1996; 184(2): 747–752
https://doi.org/10.1084/jem.184.2.747
46 M GaudreauC MilburnC GaoA PritskerM FereshtehZ YangB BarnhartA Korman M Quigley. Abstract 2782: examining the dynamic regulation of OX40 following receptor agonism and T-cell activation: Implications for antibody-mediated enhancement of T-cell function. Cancer Res. 2018; 78. 2782–2782
47 C Baniel CHeinzeCM.Hoefges A.Sumiec EG.Hank JA.Carlson PM.Jin WJ.Patel RB. SriramaneniRN.GilliesSD. ErbeAK. Schwarz CN.PieperAA. RakhmilevichAL.SondelPM. Morris ZS. In situ vaccine plus checkpoint blockade induces memory humoral response. Front. Immun 2020; 11, 1610
48 R Montler, RB Bell, C Thalhofer, R Leidner, Z Feng, BA Fox, AC Cheng, TG Bui, C Tucker, H Hoen, A Weinberg. OX40, PD-1 and CTLA-4 are selectively expressed on tumor-infiltrating T cells in head and neck cancer. Clin Transl Immunology 2016; 5(4): e70
https://doi.org/10.1038/cti.2016.16
49 J Li, NJ Stagg, J Johnston, MJ Harris, SA Menzies, D DiCara, V Clark, M Hristopoulos, R Cook, D Slaga, R Nakamura, L McCarty, S Sukumaran, E Luis, Z Ye, TD Wu, T Sumiyoshi, D Danilenko, GY Lee, K Totpal, D Ellerman, I Hötzel, JR James, TT Junttila. Membrane-proximal epitope facilitates efficient T cell synapse formation by anti-FcRH5/CD3 and is a requirement for myeloma cell killing. Cancer Cell 2017; 31(3): 383–395
https://doi.org/10.1016/j.ccell.2017.02.001
50 KLS Cleary, HTC Chan, S James, MJ Glennie, MS Cragg. Antibody distance from the cell membrane regulates antibody effector mechanisms. J Immunol 2017; 198(10): 3999–4011
https://doi.org/10.4049/jimmunol.1601473
[1] FMD-23015-OF-LY_suppl_1 Download
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed