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:

Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation

Abstract

Despite the significant therapeutic advances provided by immune-checkpoint blockade and chimeric antigen receptor T cell treatments, many malignancies remain unresponsive to immunotherapy. Bispecific antibodies targeting tumor antigens and activating T cell receptor signaling have shown some clinical efficacy; however, providing co-stimulatory signals may improve T cell responses against tumors. Here, we developed a trispecific antibody that interacts with CD38, CD3 and CD28 to enhance both T cell activation and tumor targeting. The engagement of both CD3 and CD28 affords efficient T cell stimulation, whereas the anti-CD38 domain directs T cells to myeloma cells, as well as to certain lymphomas and leukemias. In vivo administration of this antibody suppressed myeloma growth in a humanized mouse model and also stimulated memory/effector T cell proliferation and reduced regulatory T cells in non-human primates at well-tolerated doses. Collectively, trispecific antibodies represent a promising platform for 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: Optimization of α-CD3×CD28 CODV-Fab antibody and assessment of anti-CD28 contribution to T cell stimulation, survival and proliferation.
Fig. 2: CD38 trispecific antibody with minimal FcR binding reduced non-specific cytokine release by human PBMCs in vitro and lysed human multiple myeloma cells.
Fig. 3: Characterization of in vitro T cell subset expansion in response to CD38/CD28×CD3.
Fig. 4: CD28 expression on multiple myeloma cells increases susceptibility to cytolysis.
Fig. 5: In vitro activation of human PBMCs by α-CD28 superagonist requires bivalency of the antibody.
Fig. 6: Structural basis for CD3 and CD28 target recognition.
Fig. 7: Comparison of the effects of subcutaneous versus intravenous routes of administration on maximum concentration, inflammatory cytokine release and CD4/CD8 cell binding in vivo in NHPs.
Fig. 8: Vital microscopy analysis of myeloma cell cytolysis by the CD38 trispecific Ab in vitro in the presence of primary human T cells and protection against disseminated human multiple myeloma cell tumor growth in vivo.

Similar content being viewed by others

Data availability

Atomic coordinates and structure factors of the CD28 CODV-Fab and CD28:CODV-Fab complexes from Fig. 6 have been deposited in the PDB under accession codes 6O89 and 6O8D, respectively. Source data for Figs. 14 and Extended Data Fig. 2 are provided with the paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Bhoj, V. G. et al. Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood 128, 360–370 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hui, R. et al. Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial. Ann. Oncol. 28, 874–881 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 (1996).

    Article  CAS  PubMed  Google Scholar 

  4. Zuniga, E. I., Macal, M., Lewis, G. M. & Harker, J. A. Innate and adaptive immune regulation during chronic viral infections. Annu. Rev. Virol. 2, 573–597 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Vesely, M. D., Kershaw, M. H., Schreiber, R. D. & Smyth, M. J. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 29, 235–271 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Sharma, P. & Allison, J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kochenderfer, J. N. et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 116, 4099–4102 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rosenberg, S. A. & Restifo, N. P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  12. Buie, L. W., Pecoraro, J. J., Horvat, T. Z. & Daley, R. J. Blinatumomab: a first-in-class bispecific T-cell engager for precursor B-cell acute lymphoblastic leukemia. Ann. Pharmacother. 49, 1057–1067 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Xu, L. et al. Trispecific broadly neutralizing HIV antibodies mediate potent SHIV protection in macaques. Science 358, 85–90 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sun, L. L. et al. Anti-CD20/CD3 T cell-dependent bispecific antibody for the treatment of B cell malignancies. Sci. Transl. Med. 7, 287ra270 (2015).

    Google Scholar 

  15. Zugmaier, G. et al. Long-term survival and T-cell kinetics in relapsed/refractory ALL patients who achieved MRD response after blinatumomab treatment. Blood 126, 2578–2584 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Esensten, J. H., Helou, Y. A., Chopra, G., Weiss, A. & Bluestone, J. A. CD28 costimulation: from mechanism to therapy. Immunity 44, 973–988 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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 

  18. 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 

  19. Steinmetz, A. et al. CODV-Ig, a universal bispecific tetravalent and multifunctional immunoglobulin format for medical applications. MAbs 8, 867–878 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Waibler, Z. et al. Signaling signatures and functional properties of anti-human CD28 superagonistic antibodies. PLoS ONE 3, e1708 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. 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 

  22. Alegre, M. L. et al. A non-activating “humanized” anti-CD3 monoclonal antibody retains immunosuppressive properties in vivo. Transplantation 57, 1537–1543 (1994).

    Article  CAS  PubMed  Google Scholar 

  23. McKeage, K. Daratumumab: first global approval. Drugs 76, 275–281 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Gratama, J. W. et al. Tetramer-based quantification of cytomegalovirus (CMV)-specific CD8+ T lymphocytes in T-cell-depleted stem cell grafts and after transplantation may identify patients at risk for progressive CMV infection. Blood 98, 1358–1364 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Halliley, J. L. et al. Long-lived plasma cells are contained within the CD19CD38hiCD138+ subset in human bone marrow. Immunity 43, 132–145 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Robillard, N. et al. CD28, a marker associated with tumoral expansion in multiple myeloma. Clin. Cancer Res. 4, 1521–1526 (1998).

    CAS  PubMed  Google Scholar 

  27. Nair, J. R. et al. CD28 expressed on malignant plasma cells induces a prosurvival and immunosuppressive microenvironment. J. Immunol. 187, 1243–1253 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Hunig, T. The rise and fall of the CD28 superagonist TGN1412 and its return as TAB08: a personal account. FEBS J. 283, 3325–3334 (2016).

    Article  PubMed  CAS  Google Scholar 

  29. Tyrsin, D. et al. From TGN1412 to TAB08: the return of CD28 superagonist therapy to clinical development for the treatment of rheumatoid arthritis. Clin. Exp. Rheumatol. 34, 45–48 (2016).

    PubMed  Google Scholar 

  30. Findlay, L. et al. Improved in vitro methods to predict the in vivo toxicity in man of therapeutic monoclonal antibodies including TGN1412. J. Immunol. Methods 352, 1–12 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Evans, E. J. et al. Crystal structure of a soluble CD28-Fab complex. Nat. Immunol. 6, 271–279 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Kroschinsky, F. et al. New drugs, new toxicities: severe side effects of modern targeted and immunotherapy of cancer and their management. Crit. Care 21, 89 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Saber, H., Del Valle, P., Ricks, T. K. & Leighton, J. K. An FDA oncology analysis of CD3 bispecific constructs and first-in-human dose selection. Regul. Toxicol. Pharmacol. 90, 144–152 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Zuch de Zafra, C. L. et al. Targeting multiple myeloma with AMG 424, a novel anti-CD38/CD3 bispecific T-cell-recruiting antibody optimized for cytotoxicity and cytokine release. Clin. Cancer Res. 25, 3921–3933 (2019).

    Article  PubMed  CAS  Google Scholar 

  35. Brudno, J. N. & Kochenderfer, J. N. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 127, 3321–3330 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ishiguro, T. et al. An anti-glypican 3/CD3 bispecific T cell-redirecting antibody for treatment of solid tumors. Sci. Transl. Med. 9, eaal4291 (2017).

    Article  PubMed  CAS  Google Scholar 

  37. June, C. H., O’Connor, R. S., Kawalekar, O. U., Ghassemi, S. & Milone, M. C. CAR T cell immunotherapy for human cancer. Science 359, 1361–1365 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wolchok, J. D. et al. Development of ipilimumab: a novel immunotherapeutic approach for the treatment of advanced melanoma. Ann. NY Acad. Sci. 1291, 1–13 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Przepiorka, D. et al. FDA approval: blinatumomab. Clin. Cancer Res. 21, 4035–4039 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Malavasi, F. et al. Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol. Rev. 88, 841–886 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Wang, L. et al. CD38 expression predicts poor prognosis and might be a potential therapy target in extranodal NK/T cell lymphoma, nasal type. Ann. Hematol. 94, 1381–1388 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Burgler, S. Role of CD38 expression in diagnosis and pathogenesis of chronic lymphocytic leukemia and its potential as therapeutic target. Crit. Rev. Immunol. 35, 417–432 (2015).

    Article  PubMed  Google Scholar 

  44. Konopleva, M., Rissling, I. & Andreeff, M. CD38 in hematopoietic malignancies. Chem. Immunol. 75, 189–206 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Poret, N. et al. CD38 in hairy cell leukemia is a marker of poor prognosis and a new target for therapy. Cancer Res. 75, 3902–3911 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Bolzoni, M. et al. The link between bone microenvironment and immune cells in multiple myeloma: emerging role of CD38. Immunol. Lett. 205, 65–70 (2019).

    Article  CAS  PubMed  Google Scholar 

  47. Van de Donk, N., Richardson, P. G. & Malavasi, F. CD38 antibodies in multiple myeloma: back to the future. Blood 131, 13–29 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Van de Donk, N. W. et al. Clinical efficacy and management of monoclonal antibodies targeting CD38 and SLAMF7 in multiple myeloma. Blood 127, 681–695 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Lonial, S. et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 387, 1551–1560 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Dimopoulos, M. A. et al. Daratumumab, lenalidomide, and dexamethasone for multiple myeloma. N. Engl. J. Med. 375, 1319–1331 (2016).

    Article  CAS  PubMed  Google Scholar 

  51. Palumbo, A. et al. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N. Engl. J. Med. 375, 754–766 (2016).

    Article  CAS  PubMed  Google Scholar 

  52. Mateos, M. V. et al. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N. Engl. J. Med. 378, 518–528 (2018).

    Article  CAS  PubMed  Google Scholar 

  53. Martin, T. et al. A phase 1b study of isatuximab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma. Blood 129, 3294–3303 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hoffmann, M. et al. Exhaustion of activated CD8 T cells predicts disease progression in primary HIV-1 infection. PLoS Pathog. 12, e1005661 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Rodriguez-Alba, J. C. et al. HIV disease progression: overexpression of the ectoenzyme CD38 as a contributory factor? Bioessays 41, e1800128 (2019).

    PubMed  Google Scholar 

  56. Chen, L., Li, Y., Yi, X. & Gibbons, D. L. Targeting CD38 to improve anti-PD-1/CTLA-4 combination therapy in lung cancer. J. Clin. Oncol. 36, 144 (2018).

    Article  Google Scholar 

  57. Tai, Y. T. & Anderson, K. C. Targeting CD38 alleviates tumor-induced immunosuppression. Oncotarget 8, 112166–112167 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  58. DeFrancesco, L. CAR-T cell therapy seeks strategies to harness cytokine storm. Nat. Biotechnol. 32, 604 (2014).

    Article  PubMed  Google Scholar 

  59. 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 

  60. Penaranda, C., Tang, Q. & Bluestone, J. A. Anti-CD3 therapy promotes tolerance by selectively depleting pathogenic cells while preserving regulatory T cells. J. Immunol. 187, 2015–2022 (2011).

    Article  CAS  PubMed  Google Scholar 

  61. Tabares, P. et al. Human regulatory T cells are selectively activated by low-dose application of the CD28 superagonist TGN1412/TAB08. Eur. J. Immunol. 44, 1225–1236 (2014).

    Article  CAS  PubMed  Google Scholar 

  62. Krejcik, J. et al. Monocytes and granulocytes reduce CD38 expression levels on myeloma cells in patients treated with daratumumab. Clin. Cancer Res. 23, 7498–7511 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Raghu, G. et al. SAR156597 in idiopathic pulmonary fibrosis: a phase 2 placebo-controlled study (DRI11772). Eur. Respir. J. 52, 1801130 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Merchant, A. M. et al. An efficient route to human bispecific IgG. Nat. Biotechnol. 16, 677–681 (1998).

    Article  CAS  PubMed  Google Scholar 

  65. Smith, K. B. & Ellis, S. A. Standardisation of a procedure for quantifying surface antigens by indirect immunofluorescence. J. Immunol. Methods 228, 29–36 (1999).

    Article  CAS  PubMed  Google Scholar 

  66. Cobbold, M. et al. Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers. J. Exp. Med. 202, 379–386 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kabsch, W. XDS. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Evans, P. R. & Murshudov, G. N. How good are my data and what is the resolution? Acta Crystallogr. D Biol. Crystallogr. 69, 1204–1214 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Morin, A. et al. Collaboration gets the most out of software. eLife 2, e01456 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank R. Mashal and H. Van de Velde for critical comments on the manuscript, T. Majid and C. Lawendowski for excellent program management, A. E. Schroeer and B. DelGiudice for graphic arts support, and M. Sanicola-Nadel, T. Schmidt, T. Bouquin, D. Wiederschain, S. Sidhu, B. Thurberg, K. Klinger, J. Darbyshire, C. Dangler, Z. Jayyosi and C. J. Wei for organizational support. We also thank J. Kingsbury, S. Kathuria, L. Chen, N. Maestrali, S. Somarriba, E. Deschamps, N. Couteault and L. Maton for technical support.

Author information

Authors and Affiliations

Authors

Contributions

Z.-y.Y., L.W., E.S., L.X., E.R., R.R.W., V.C.-R., F.S., P.P., N.E.M., S.R., K.R., P.K., J.F., R.V. and G.J.N. designed the research. Z.-y.Y., L.W., E.S., L.X., R.R.W., D.M.L., V.C.-R., B.C., C.P., B.Z., H.Q., P.K., J.F. and R.V. carried out the research. Z.-y.Y., L.W., E.S., L.X., D.M.L., V.P., B.O., G.U., E.F., C.B., T.D., T.B., S.P., C.L., A.P., G.D. and Z.S. performed the experiments. Z.-y.Y., L.W., E.S., L.X., R.R.W., D.M.L., V.C.-R., B.C., C.P., P.P., G.U., N.E.M., P.K., J.F., R.V., A.L. and G.J.N. analyzed the data. Z.-y.Y., L.W., E.S., D.M.L., R.R.W., G.U., P.P., T.B., N.E.M. and G.J.N. wrote the paper. All authors reviewed the paper.

Corresponding authors

Correspondence to Zhi-yong Yang or Gary J. Nabel.

Ethics declarations

Competing interests

All authors are or were employees of Sanofi while engaged in this research project. Sanofi develops and manufactures cancer treatment medicines. G.J.N. is the Chief Scientific Officer of Sanofi. G.J.N., Z.-y.Y., R.R.W., L.X., E.S., L.W., T.D., B.C., C.L. and C.P. are listed on intellectual property (WO2019074973A2) developed and owned by Sanofi related to the development of novel cancer treatments.

Additional information

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

Extended data

Extended Data Fig. 1 Expression of relevant cell surface antigens on an IL-2 promoter luciferase indicator line, as well as Bcl-xL kinetics and annexin V analysis in primary T cells.

(a) Cell surface expression of CD3, CD28, or CD38 respectively on GloResponse™ IL2-luc2P Jurkat Cells (Promega) was determined by flow cytometry as described in Methods in 1 independent experiment. (b) Induction of pro-survival protein Bcl-xL (left) and annexin V (right) after CD38 trispecific Ab, or indicated mutant Ab, treatment of primary CD3+ cells (n = 3 donors). Cells were isolated as described in Methods and analyzed by flow cytometry targeting CD3, Annexin V, and using a fixable viability dye. The data points and error bars were plotted with mean ± SD.

Extended Data Fig. 2 Binding of alternative human IgG4 Fc mutants to human Fc receptors.

(a) Alternative mutations in the Fc region of IgG4 were prepared for analysis in Fc receptor binding assays. (b) Binding in the biacore assay was used to measure the affinity of the specified IgG4 Fc variants to indicated human Fc receptors immobilized on the chip. IgG4 Fc variants were used at 150 nM. The assay was repeated once with similar results. The binding to human FcRn was measured using ELISA by coating the human FcRn antigen on the plate in a single experiment.

Source data

Extended Data Fig. 3 Crystal structure and modeling of the CD38 trispecific Ab.

The crystal structure of CD28: anti-CD28xCD3 CODV-Fab (a) and molecular model of anti-CD38 Fab. (b) were used to model the binary CD28: trispecific antibody model complex (Fig. 6b). The model is compatible with concurrent binding of the antibody with CD28 and CD38. The distance between the CDRH3s for anti-CD28/CD3 CODV-Fab measured ~60 Å, comparable to the anti-IL4/IL13 CODV-Fab and shorter than the anti-HIV CODV-Fab (exceeds 100 Å), possibly because different linkers were used in the CODV Fabs. The structure of CD3 in presence of CD3mid Fv has not been solved and thus not modeled.

Extended Data Fig. 4 Effect of trispecific Ab on immune cell distribution and binding of CD38 trispecific Ab to T cell subsets in the blood of NHPs.

Blood samples from animals treated with a single IV (30 ug/kg) or SC administration (100 ug/kg) were analyzed by flow cytometry (a) to quantitate T lymphocyte, B lymphocyte, NK cells and monocytes as indicated. T lymphocytes were selected using CD2 + /CD16-, B lymphocytes with CD2-/CD20 + /HLA-DR + , and NK cells with CD2 + / CD16 + , and (b) to quantitate cell-bound trispecific Ab levels on CD4+ T cells (CD2 + /CD4 + /CD8) or CD8 cells (CD2 + /CD4-/CD8 + ). CD38 trispecific binding was determined using anti-human IgG4 (Southern Biotech) to determine percentage of cells bound with trispecific Ab on indicated subpopulations of T cells. The data were plotted as average of n = 3 animals ± SD.

Extended Data Fig. 5 Effect of CD38 trispecific Ab on T lymphocyte subset distribution in the blood of NHPs.

Blood samples from animals (n = 3) treated with a single SC administration (100 ug/kg) were analyzed by flow cytometry to quantitate CD4, CD8, CD8 effector memory and T regulatory cells as indicated. CD4+ T cells were selected using cell surface staining for CD2 + /CD4 + /CD8-, CD8 + T cells with CD2 + /CD4-/CD8 + ; CD8 + effector memory by CD2 + /CD4-/CD8 + /CD45RA + /CD197- and regulatory T cells with CD2 + /CD4 + /CD8-/CD127-/CD25hi. The data were plotted as individual animals.

Extended Data Fig. 6 Effect of intra-animal dose escalation on serum levels and inflammatory cytokine release in NHP.

Animals (n = 3) were injected IV with the CD38 trispecific Ab at 10 ug/kg on day 1, 30 ug/kg on day 4, and 100 ug/kg on day 7. Serum samples were measured for trispecific Ab serum concentration using a Gyrolab immunoassay (Gyros Protein Technologies). Plasma samples were analyzed for inflammatory cytokines IL-6 and IFN-γ as described in Methods. The data were plotted as individual animals.

Extended Data Fig. 7 Cytolysis of the CD38 trispecific Ab of CD38+ hematological cancer cell lines.

Cytolytic activity of the CD38/CD28xCD3 trispecific FALA mutant Ab against indicated CD38+CD28- lines, including acute myelocytic leukemia (AML (KG-1)), a B cell lymphoma (OCI-Ly19), acute T lymphocytic leukemia (ALL (KOPN8)), and chronic lymphocytic lymphoma (CLL(Z-138)). The representative nonlinear regression graphs (ordinary fit) ± SD are from 2 PBMC donors performed in two separate experiments.

Supplementary information

Supplementary Information

Supplementary Tables 1–6.

Reporting Summary

Supplementary Video 1

Time-lapse vital microscopy of myeloma cell cytolysis by the CD38 trispecific antibody in vitro in the presence of primary human T cells.

Source data

Source Data Fig. 1

Flow cytometry statistical source data

Source Data Fig. 2

Flow cytometry statistical source data

Source Data Fig. 3

Flow cytometry statistical source data

Source Data Fig. 4

Flow cytometry statistical source data

Source Data Extended Data Fig. 2

SPR and ELISA source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, L., Seung, E., Xu, 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). https://doi.org/10.1038/s43018-019-0004-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-019-0004-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing