Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Rejuvenation of tumour-specific T cells through bispecific antibodies targeting PD-L1 on dendritic cells

Abstract

Bispecific T-cell engagers (BiTEs) preferentially targeting tumour-associated antigens and stimulating CD3-mediated signalling are being used in patients to treat acute B-cell lymphoblastic leukemia. However, the potency of BiTEs in solid tumours is limited by their short half-life and their severe toxicity at relevant therapeutic doses. Here we report the design and in vivo performance of a bispecific antibody that simultaneously targets the murine T-cell co-receptor CD3ε and the murine immune checkpoint programmed-death ligand 1 (PD-L1). In multiple syngeneic tumour models, the bispecific antibody generated higher antitumour immune responses than conventional BiTEs targeting tumour-associated antigens and CD3ε. We found that the durable antigen-specific T-cell responses resulted from the rejuvenation of CD8 T cells, owing to the blockade of PD-L1 on dendritic cells (but not on tumour cells) and co-stimulation by B7-1&2 (a peripheral membrane protein on dendritic cells). Bispecific T-cell engagers targeting dendritic cells rather than tumour cells may represent a general means of T-cell rejuvenation for durable cancer immunotherapy.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: PD-L1×CD3 targets PD-L1 to activate T cells in vitro.
Fig. 2: PD-L1×CD3 generates a greater antitumour effect than combination treatment in vivo.
Fig. 3: Pre-existing CD8 T cells are sufficient for the antitumour effect of PD-L1×CD3.
Fig. 4: PD-L1 on dendritic cells is essential for the antitumour effect of PD-L1×CD3.
Fig. 5: PD-L1×CD3 reshapes a distinct immunophenotypic signature in tumour-bearing mice.
Fig. 6: Co-stimulatory signalling is required for PD-L1×CD3-mediated antitumour effects.
Fig. 7: Schematic of hypothesized working model.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. The raw datasets generated during the study are too large to be publicly shared, but they are available for research purposes from the corresponding authors on reasonable request. Source data for the figures are available from figshare with the identifier https://doi.org/10.6084/m9.figshare.14984793.

References

  1. Staerz, U. D., Kanagawa, O. & Bevan, M. J. Hybrid antibodies can target sites for attack by T cells. Nature 314, 628–631 (1985).

    Article  CAS  PubMed  Google Scholar 

  2. Garber, K. Bispecific antibodies rise again. Nat. Rev. Drug Discov. 13, 799–801 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Baeuerle, P. A. & Reinhardt, C. Bispecific T-cell engaging antibodies for cancer therapy. Cancer Res. 69, 4941–4944 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Trabolsi, A., Arumov, A. & Schatz, J. H. T cell-activating bispecific antibodies in cancer therapy. J. Immunol. 203, 585–592 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Bargou, R. et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321, 974–977 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Topp, M. S. et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 16, 57–66 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Maude, S. L., Barrett, D., Teachey, D. T. & Grupp, S. A. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J. 20, 119–122 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Topp, M. S. et al. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J. Clin. Oncol. 32, 4134–4140 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Li, J. et al. CD3 bispecific antibody-induced cytokine release is dispensable for cytotoxic T cell activity. Sci. Transl. Med. 11, eaax8861 (2019).

    Article  PubMed  CAS  Google Scholar 

  10. Reusch, U. et al. Anti-CD3 × anti-epidermal growth factor receptor (EGFR) bispecific antibody redirects T-cell cytolytic activity to EGFR-positive cancers in vitro and in an animal model. Clin. Cancer Res. 12, 183–190 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Cioffi, M., Dorado, J., Baeuerle, P. A. & Heeschen, C. EpCAM/CD3-bispecific T-cell engaging antibody MT110 eliminates primary human pancreatic cancer stem cells. Clin. Cancer Res. 18, 465–474 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Han, H. et al. Bispecific anti-CD3 × anti-HER2 antibody mediates T cell cytolytic activity to HER2-positive colorectal cancer in vitro and in vivo. Int. J. Oncol. 45, 2446–2454 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Kebenko, M. et al. A multicenter phase 1 study of solitomab (MT110, AMG 110), a bispecific EpCAM/CD3 T-cell engager (BiTE(R)) antibody construct, in patients with refractory solid tumors. Oncoimmunology 7, e1450710 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Lutterbuese, R. et al. T cell-engaging BiTE antibodies specific for EGFR potently eliminate KRAS- and BRAF-mutated colorectal cancer cells. Proc. Natl Acad. Sci. USA 107, 12605–12610 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. van Panhuys, N. TCR signal strength alters T-DC activation and interaction times and directs the outcome of differentiation. Front. Immunol. 7, 6 (2016).

    PubMed  PubMed Central  Google Scholar 

  16. Chai, J. G. & Lechler, R. I. Immobilized anti-CD3 mAb induces anergy in murine naive and memory CD4+ T cells in vitro. Int. Immunol. 9, 935–944 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Harding, F. A., McArthur, J. G., Gross, J. A., Raulet, D. H. & Allison, J. P. CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones. Nature 356, 607–609 (1992).

    Article  CAS  PubMed  Google Scholar 

  18. Green, D. R., Droin, N. & Pinkoski, M. Activation-induced cell death in T cells. Immunol. Rev. 193, 70–81 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Curtsinger, J. M. & Mescher, M. F. Inflammatory cytokines as a third signal for T cell activation. Curr. Opin. Immunol. 22, 333–340 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kalos, M. et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 3, 95ra73 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. MacKay, M. et al. The therapeutic landscape for cells engineered with chimeric antigen receptors. Nat. Biotechnol. 38, 233–244 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Labrijn, A. F., Janmaat, M. L., Reichert, J. M. & Parren, P. Bispecific antibodies: a mechanistic review of the pipeline. Nat. Rev. Drug Discov. 18, 585–608 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Seckinger, A. et al. Target expression, generation, preclinical activity, and pharmacokinetics of the BCMA-T cell bispecific antibody EM801 for multiple myeloma treatment. Cancer Cell 31, 396–410 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Rius Ruiz, I. et al. p95HER2-T cell bispecific antibody for breast cancer treatment. Sci. Transl. Med. 10, eaat1445 (2018).

    Article  PubMed  CAS  Google Scholar 

  25. Boise, L. H. et al. CD28 costimulation can promote T cell survival by enhancing the expression of Bcl-XL. Immunity 3, 87–98 (1995).

    Article  CAS  PubMed  Google Scholar 

  26. Garfall, A. L. & June, C. H. Trispecific antibodies offer a third way forward for anticancer immunotherapy. Nature 575, 450–451 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Skokos, D. et al. A class of costimulatory CD28-bispecific antibodies that enhance the antitumor activity of CD3-bispecific antibodies. Sci. Transl. Med. 12, eaaw7888 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Suntharalingam, G. et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N. Engl. J. Med. 355, 1018–1028 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Wu, L. et al. Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation. Nat. Cancer 1, 86–98 (2020).

    Article  CAS  PubMed  Google Scholar 

  30. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Knutson, K. L. & Disis, M. L. Tumor antigen-specific T helper cells in cancer immunity and immunotherapy. Cancer Immunol. Immunother. 54, 721–728 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Jansen, C. S. et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zou, W., Wolchok, J. D. & Chen, L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Sci. Transl. Med. 8, 328rv324 (2016).

    Article  CAS  Google Scholar 

  34. Lin, H. et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J. Clin. Investig. 128, 805–815 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Tang, H. et al. PD-L1 on host cells is essential for PD-L1 blockade–mediated tumor regression. J. Clin. Investig. 128, 580–588 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Garcia-Diaz, A. et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep. 19, 1189–1201 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kohnke, T., Krupka, C., Tischer, J., Knosel, T. & Subklewe, M. Increase of PD-L1 expressing B-precursor ALL cells in a patient resistant to the CD19/CD3-bispecific T cell engager antibody blinatumomab. J. Hematol. Oncol. 8, 111 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Kobold, S., Pantelyushin, S., Rataj, F. & Vom Berg, J. Rationale for combining bispecific T cell activating antibodies with checkpoint blockade for cancer therapy. Front. Oncol. 8, 285 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Schlothauer, T. et al. Novel human IgG1 and IgG4 Fc-engineered antibodies with completely abolished immune effector functions. Protein Eng. Des. Sel. 29, 457–466 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Wei, H. et al. Structural basis of a novel heterodimeric Fc for bispecific antibody production. Oncotarget 8, 51037–51049 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Qiao, J. et al. Targeting tumors with IL-10 prevents dendritic cell-mediated CD8+ T cell apoptosis. Cancer Cell 35, 901–915.e4 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Benonisson, H. et al. CD3-bispecific antibody therapy turns solid tumors into inflammatory sites but does not install protective memory. Mol. Cancer Ther. 18, 312–322 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Diskin, B. et al. PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat. Immunol. 21, 442–454 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Hui, E. et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355, 1428–1433 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kamphorst, A. O. et al. Rescue of exhausted CD8 T cells by PD-1-targeted therapies is CD28-dependent. Science 355, 1423–1427 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kelly, E., Won, A., Refaeli, Y. & van Parijs, L. IL-2 and related cytokines can promote T cell survival by activating AKT. J. Immunol. 168, 597–603 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Heiss, M. M. et al. The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: results of a prospective randomized phase II/III trial. Int. J. Cancer 127, 2209–2221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kiewe, P. et al. Phase I trial of the trifunctional anti-HER2 × anti-CD3 antibody ertumaxomab in metastatic breast cancer. Clin. Cancer Res. 12, 3085–3091 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Schildberg, F. A., Klein, S. R., Freeman, G. J. & Sharpe, A. H. Coinhibitory pathways in the B7-CD28 ligand-receptor family. Immunity 44, 955–972 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Horn, L. A. et al. CD3×PDL1 bi-specific T cell engager (BiTE) simultaneously activates T cells and NKT cells, kills PDL1+ tumor cells, and extends the survival of tumor-bearing humanized mice. Oncotarget 8, 57964–57980 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Mayoux, M. et al. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy. Sci. Transl. Med. 12, eaav7431 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Zhao, Y. et al. PD-L1:CD80 cis-heterodimer triggers the co-stimulatory receptor CD28 while repressing the inhibitory PD-1 and CTLA-4 pathways. Immunity 51, 1059–1073.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Rafiq, S. et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 36, 847–856 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hill, B. T., Roberts, Z. J., Xue, A., Rossi, J. M. & Smith, M. R. Rapid tumor regression from PD-1 inhibition after anti-CD19 chimeric antigen receptor T-cell therapy in refractory diffuse large B-cell lymphoma. Bone Marrow Transplant. 55, 1184–1187 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Wallberg, M. et al. Anti-CD3 treatment up-regulates programmed cell death protein-1 expression on activated effector T cells and severely impairs their inflammatory capacity. Immunology 151, 248–260 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. del Rio, M. L., Bernhardt, G., Rodriguez-Barbosa, J. I. & Forster, R. Development and functional specialization of CD103+ dendritic cells. Immunol. Rev. 234, 268–281 (2010).

    Article  PubMed  Google Scholar 

  58. Refaeli, Y., van Parijs, L., Alexander, S. I. & Abbas, A. K. Interferon γ is required for activation-induced death of T lymphocytes. J. Exp. Med. 196, 999–1005 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Liu, X. et al. Dual targeting of innate and adaptive checkpoints on tumor cells limits immune evasion. Cell Rep. 24, 2101–2111 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the UT Southwestern Institutional Animal Care & Use Committee (IACUC) and the Animal Resources Center (ARC). Y.-X.F. holds the Mary Nell and Ralph B. Rogers Professorship in Immunology. This work was supported by the Cancer Prevention and Research Institute of Texas (CPRIT) grant RR150072, given to Y.-X.F., and the National Institutes of Health (NIH) grant 1U54CA244719-01, to J.Q. We also thank Y. Liang, X. Cao, Z. Ren, A. Zhang, C. Dong, Z. Liu, C. Lu and B. Moon for providing experiment materials and helpful discussions.

Author information

Authors and Affiliations

Authors

Contributions

L.L. and Y.-X.F. designed the study and analysed the data. L.L. performed the experiments. J.C. and J.B. assisted with tumour experiments. H.L. assisted with tissue staining. Z.S. assisted with bispecific antibody purification. C.H. provided mice and reagents. L.L. and Y.-X.F. wrote the manuscript. C.M., E.H. and J.Q. revised the manuscript. J.Q. assisted with data interpretation. Y.-X.F. supervised the project.

Corresponding authors

Correspondence to Jian Qiao or Yang-Xin Fu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Biomedical Engineering thanks Marion Subklewe and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 PD-L1xCD3 generates superior antitumour effects than TAA-targeting BiTE in vivo.

a-b, C57BL/6 J mice were subcutaneously inoculated with 3 × 105 MC38E5 tumor cells and treated with 0.25 mg kg−1 of bispecific antibodies twice on day 10 and 15. Tumor volume was measured twice per week (a). 60 days post treatment, tumor free mice were re-challenged with 3 × 106 tumor cells (b). c-d, C57BL/6 J mice were subcutaneously inoculated with 1 × 106 TC1E5 tumor cells and treated with 0.25 mg kg−1 of bispecific antibodies twice on day 10 and 15. Tumor volume was measured twice per week (c). 60 days post treatment, tumor free mice were re-challenged with 1 × 107 tumor cells (d). e, C57BL/6 J mice were subcutaneously inoculated with 3 × 105 B16E5 tumor cells and treated with 0.25 mg kg−1 of bispecific antibodies intraperitoneally twice on day 8 and 12. f, BALB/c mice were subcutaneously inoculated with 5 × 105 TuBoE5 tumor cells and treated with 0.25 mg kg−1 of fusion proteins intraperitoneally twice on day 10 and 14. g, C57BL/6 J mice were subcutaneously inoculated with 3 × 105 MC38E5 tumor cells and treated with 0.25 mg kg−1 of fusion proteins either intratumorally or intraperitoneally twice on day 9 and 13. Data were presented as mean ± s.e.m from a representative experiment (n = 5 (a, b, f, g), 4 (c-e) biologically independent animals) of two independent experiments. Statistical analysis was performed by two-way ANOVA with Tukey’s multiple comparisons test. ****P ≤ 0.0001.

Extended Data Fig. 2 PD-L1xCD3 targets pre-existing CD8 T cells in the tumour tissue to initiate the antitumour immune response.

a-d, C57BL/6 J mice were inoculated with 1 × 106 MC38 tumor cells and treated with PD-L1xCD3 (0.25 mg kg−1 on day 10 and 15). 200 μg of anti-CD8, anti-CD4, anti-NK1.1 or 500 μg of anti-CSF1R was administrated respectively one day before treatment initiation and then twice a week for 2 weeks. The percentage of CD8 + cells (a), CD4 + cells (b), NK1.1+ cells (c) and CD11b + F4/80+ cells (d) in the spleen were detected by flow cytometry. e, C57BL/6 J mice (n = 5 biologically independent animals) were subcutaneously inoculated with 1 × 106 MC38 tumor cells and treated with 0.25 mg kg−1 of PD-L1xCD3 either intratumorally or intraperitoneally twice on day 10 and 15. f, C57BL/6 J mice (n = 3 biological replicates) were subcutaneously inoculated with 1 × 106 MC38 tumor cells and intraperitoneally treated with 0.25 mg kg−1 of PD-L1xCD3. Concentration of fusion protein in different tissues were measured by hIgG ELISA at indicated time point. Data were shown as mean ± s.e.m from a representative experiment of two independent experiments. Statistical analysis was performed by two-way ANOVA with Tukey’s multiple comparisons test. ****P ≤ 0.0001.

Extended Data Fig. 3 Co-stimulatory signaling blockade abolished PD-L1xCD3 mediated antitumour effects and the in vitro activation of T cells by TAA-targeting BiTE.

a-b, C57BL/6 J mice were inoculated with 1 × 106 MC38 tumor cells and treated with PD-L1xCD3 (0.25 mg kg−1 on day 10 and 15), 200 μg CTLA4-Ig was administrated on day 10, 13 and 15. Experimental design (a) and tumor growth curve (b) are shown. c-e, CD8 T cells were co-cultured with either tumor cells or dendritic cells in the presence of ErbxCD3. T cell activation (c), apoptotic T cells (d), supernatant IL-2 and IFN-γ (e) were measured by flow cytometry. Data were shown as mean ± s.e.m from a representative experiment of two independent experiments (n = 5 biologically independent animals). Statistical analyses were performed by two-way ANOVA with Dunnett’s multiple comparisons test (b), two-tailed unpaired Student’s t-test (c-e). ***P ≤ 0.001, ****P ≤ 0.0001.

Extended Data Fig. 4 Correlation analysis of CD28 expression with CD80/86 expression, dendritic cell infiltration and patient survival.

TCGA database was analyzed for cumulative survival according to CD28 expression (a), correlation of CD28 level with CD80 and CD86 level (b) and correlation of CD28 level with dendritic cell infiltration (c). Skin cutaneous melanoma (SKCM), cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), lung adenocarcinoma (LUAD), colon adenocarcinoma (COAD), breast invasive carcinoma (BRCA). Statistical analyses were performed by log-rank test (a), and Spearman’s rho correlation test (b-c).

Supplementary information

Supplementary Information

Supplementary figures and tables.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, L., Chen, J., Bae, J. et al. Rejuvenation of tumour-specific T cells through bispecific antibodies targeting PD-L1 on dendritic cells. Nat Biomed Eng 5, 1261–1273 (2021). https://doi.org/10.1038/s41551-021-00800-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-021-00800-2

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer