Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Jan 6:15:1494206.
doi: 10.3389/fimmu.2024.1494206. eCollection 2024.

Effective cancer immunotherapy combining mRNA-encoded bispecific antibodies that induce polyclonal T cell engagement and PD-L1-dependent 4-1BB costimulation

Affiliations

Effective cancer immunotherapy combining mRNA-encoded bispecific antibodies that induce polyclonal T cell engagement and PD-L1-dependent 4-1BB costimulation

Oana Hangiu et al. Front Immunol. .

Abstract

Background: Immune checkpoint inhibitors have revolutionized cancer therapy, but many patients fail to respond or develop resistance, often due to reduced T cell activity. Costimulation via 4-1BB has emerged as a promising approach to enhance the effector function of antigen-primed T cells. Bispecific T cell-engaging (TCE) antibodies are an effective way to provide tumor-specific T cell receptor-mediated signaling to tumor-infiltrating lymphocytes. mRNA-based delivery of bispecific antibodies, offer a novel approach to enhance tumor-specific immune responses while minimizing adverse effects.

Methods: Two bispecific antibodies were generated: the EGFR x CD3 TCE antibody (LiTE) and the PD-L1 x 4-1BB costimulatory antibody (LiTCo), which was further fused to a high FcRn albumin variant (Albu-LiTCo). The mRNA encoding these bispecific antibodies contains an N1-methylpseudouridine modified nucleoside and regulatory sequences to ensure proper expression and stability. A series of in vitro assays and cell-based analyses were performed to characterize both antibodies. The in vivo efficacy of the mRNA-encoded bispecific antibodies was evaluated in xenograft tumor models expressing EGFR.

Results: We investigated the combined effect of two mRNA-encoded Fc-free bispecific antibodies with complementary mechanisms of action: an EGFR-targeting TCE and a half-life extended PD-L1 x 4-1BB costimulatory antibody. The mRNAs encoding both bispecific LiTERNA and Albu-LiTCoRNA, showed similar binding specificity and in vitro function to their protein analogues. Pharmacokinetic studies demonstrated sustained expression of both bispecific antibodies following intravenous administration of the mRNAs formulated using a polymer/lipid-based nanoparticle (LNP) but different pharmacokinetic profiles, shorter for the TCE and longer for the PD-L1 x 4-1BB. When administered as a mRNA-LNP combination (ComboRNA), the growth of EGFR-positive tumors in immunocompetent mice was significantly inhibited, resulting in tumor regression in 20% of cases with no associated toxicity. Histological analysis confirmed increased T cell infiltration in the tumors treated with LITERNA and ComboRNA. Repeated administration resulted in sustained production of bispecific antibodies with different exposure cycles and potent antitumor activity with a favorable safety profile.

Conclusions: These results highlight the potential of combining two mRNA-encoded bispecific antibodies with different mechanisms of action and programmable half-life for cancer immunotherapy.

Keywords: T-cell engager; cancer immunotherapy; combined RNA; costimulatory antibody; mRNA encoded bispecific antibodies.

PubMed Disclaimer

Conflict of interest statement

OH, RN and MC are all employees of Leadartis. SF formerly worked at Leadartis. LA-V and LS are co-founders of Leadartis. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Figures

Figure 1
Figure 1
Functional characterization and mechanisms of action of LiTE and Albu-LiTCo antibodies. Schematic representation of EGFR-targeted LiTE and Albu-LiTCo antibodies (A). LiTE-mediated EGFR-dependent activation of tumor cells (B). CT26 and CT26EGFR were cocultured with mouse splenocytes (E:T ratio 5:1) in the presence of LiTE or anti-CD3 IgG at 3.34 nM equimolar concentration. Mouse CD69 expression was measured by flow cytometry after 24 hours (B). LiTE-mediated EGFR-dependent cytotoxicity of tumor cells (C). Luciferase-expressing CT26 and CT26EGFR cells were cocultured with splenocytes (E:T ratio 5:1) and different concentrations of LiTE. Specific lysis was measured by bioluminescence at 48 hours. Percent specific lysis was calculated relative to control. Data are mean ± SD (n = 3). Significance was calculated by unpaired Student’s t test. Competition ELISA for PD-L1/PD-1 interaction inhibition by anti-PD-L1 IgG or Albu-LiTCo (D). Data are mean ± SD (n = 3). One representative experiment from two independent experiments is shown. LogIC50 is shown. Blocking activity of Albu-LiTCo in a cell-based bioassay (E). JurkatPD-1 cells were cocultured with CHOPD-L1 cells and increasing concentrations of anti-PD-L1 IgG or Albu-LiTCo. Luminescence was measured after 6 hours. Data are expressed as fold induction relative to unstimulated JurkatPD-1 cells. Representative dose-concentration curves are shown as mean ± SD (n = 3). IC50 is shown. Agonistic activity of Albu-LiTCo (F). Wild type CHO cells or CHOPD-L1/EGFR were cocultured with LiTE-activated mouse CD8a+ T cells (3.34 nM) in the presence of Albu-LiTCo, anti-4-1BB IgG or anti-PD-L1 IgG at 6.67 nM equimolar dose. IFNγ secretion was determined after 72 h Negative controls (–) consisted of CD8+ T cells cultured with either CHO or CHOPD-L1/EGFR, and in the absence (- control in the left) or presence (- control in the right) of soluble LiTE. Data are mean ± SD (n = 3). One representative experiment of three independent experiments is shown. Significance was calculated by unpaired Student’s t test.
Figure 2
Figure 2
Functional characterization of LiTERNA and Albu-LiTCoRNA. Western blot detection using a HRP-conjugated anti-VHH mAb in the conditioned media of LiTERNA and Albu-LiTCoRNA transfected HEK293 cells. Molecular mass (kDa) is indicated (A). Specific binding of LiTERNA (B) and Albu-LiTCoRNA (C, D) to plastic immobilized specific antigens (hEGFR and m4-1BB, mPD-L1, respectively) demonstrated by ELISA using an HRP-conjugated anti-VHH mAb cocktail (B, C) or HRP-conjugated anti-human serum albumin (HRP-HSA) mAb (D). Data are represented as a mean ± SD (n = 3). BSA, bovine serum albumin; NFDM, non-fat dry milk. Binding of LiTERNA and Albu-LiTCoRNA to cell surface expressed antigens mCD3 (E), m4-1BB (F), hEGFR (G) and hPDL-1 (H) by flow cytometry. The y-axis shows the relative cell number, and the x-axis represents the R-phycoerythrin (PE)-fluorescence, expressed on a linear scale. The anti-CD3 IgG, anti-4-1BB IgG, anti-EGFR IgG and anti-PD-L1 IgG were used as controls. One representative experiment out of two independent experiments is shown. The number indicates the percentage of positive cells (%).
Figure 3
Figure 3
Effects of LiTERNA and Albu-LiTCoRNA on tumor cell cytotoxicity, agonistic costimulatory activity and tumor growth inhibition. EGFR-dependent cytotoxicity by LiTERNA (A). Luciferase-expressing CT26 and CT26EGFR cells were co-cultured with mouse splenocytes at an E:T ratio of 5:1. Specific tumor cell lysis was measured by bioluminescence. The percentage of specific lysis was calculated relative to the same number of tumor cells cultured with splenocytes. Data are expressed as mean ± SD (n = 3). Significance was calculated by unpaired Student’s t test. CHO or CHOPD-L1/EGFR cells were plated with mouse CD8a+ T cells activated with LiTERNA in the presence of Albu-LiTCo (B) or with LiTE in the presence of Albu-LiTCoRNA (C). IFNγ secretion was determined after 72 hours. Data are expressed as mean ± SD (n = 3). Significance was calculated by unpaired Student’s t test. Anti-4-1BB IgG and anti-PD-L1 IgG (B) or EGFPRNA (C) were used as controls. Pharmacokinetic profile of LiTERNA (solid blue line) and Albu-LiTCoRNA (solid red line) expressed as ng/mL (left Y-axis) after a single intravenous (i.v.) administration of mRNA-LNP in BALB/c mice (D). Ex vivo specific tumor lysis of CT26EGFR cells mediated by LiTE-containing mouse serum (dashed blue line) and Albu-LiTCo-containing mouse serum (dashed red line) expressed as % of Lysis (right Y-axis) (D). Target cells modified for the expression of luciferase were co-cultured with splenocytes at the effector/target (E/T) ratio of 5:1 in the presence of mouse serum obtained at 4, 96 and 168 hours post-mRNA-LNP administration. After 48 hours, the percentage of specific tumor lysis was measured by bioluminescence (right Y-axis). Data are presented as mean ± SD (n = 3). BALB/c mice were subcutaneously (s.c.) inoculated with CT26EGFR cells, randomized into n=5/group with similar mean tumor sizes and SDs and treated with 3 i.v. injections of LiTERNA, Albu-LiTCoRNA (10 µg/mouse) as monotherapy or combined (ComboRNA) (E). Average tumor volume growth of mice in each group is shown. Data are presented as mean ± SD. Significance was determined by one-way ANOVA with Tukey’s correction test for multiple comparisons. Quantitative analysis of intratumoral CD8+ T cells in mouse tumor tissue (n = 4/group) (F). Data were calculated as percentage of CD8+ versus total cell number and presented as mean ± SD. Significance was determined by one-way ANOVA with Tukey’s test correction for multiple comparisons.

References

    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. (2012) 12:252–64. doi: 10.1038/nrc3239 - DOI - PMC - PubMed
    1. Sprooten J, De Wijngaert P, Vanmeerbeerk I, Martin S, Vangheluwe P, Schlenner S, et al. . Necroptosis in immuno-oncology and cancer immunotherapy. Cells. (2020) 9:1823. doi: 10.3390/cells9081823 - DOI - PMC - PubMed
    1. Warmuth S, Gunde T, Snell D, Brock M, Weinert C, Simonin A, et al. . Engineering of a trispecific tumor-targeted immunotherapy incorporating 4-1BB co-stimulation and PD-L1 blockade. Oncoimmunology. (2021) 10:2004661. doi: 10.1080/2162402X.2021.2004661 - DOI - PMC - PubMed
    1. Qiao Y, Qiu Y, Ding J, Luo N, Wang H, Ling X, et al. . Cancer immune therapy with PD-1-dependent CD137 co-stimulation provides localized tumour killing without systemic toxicity. Nat Commun. (2021) 12:6360. doi: 10.1038/s41467-021-26645-6 - DOI - PMC - PubMed
    1. Geuijen C, Tacken P, Wang L-C, Klooster R, van Loo PF, Zhou J, et al. . A human CD137×PD-L1 bispecific antibody promotes anti-tumor immunity via context-dependent T cell costimulation and checkpoint blockade. Nat Commun. (2021) 12:4445. doi: 10.1038/s41467-021-24767-5 - DOI - PMC - PubMed

MeSH terms