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:

A single-domain bispecific antibody targeting CD1d and the NKT T-cell receptor induces a potent antitumor response

Abstract

Antibody-mediated modulation of major histocompatibility complex (MHC) molecules, or MHC class I-like molecules, could constitute an effective immunotherapeutic approach. We describe how single-domain antibodies (VHH), specific for the human MHC class I-like molecule CD1d, can modulate the function of CD1d-restricted T cells and how one VHH (1D12) specifically induced strong type I natural killer T (NKT) cell activation. The crystal structure of the VHH1D12-CD1d(α-GalCer)-NKT T-cell receptor (TCR) complex revealed that VHH1D12 simultaneously contacted CD1d and the type I NKT TCR, thereby stabilizing this interaction through intrinsic bispecificity. This led to greatly enhanced type I NKT cell-mediated antitumor activity in in vitro, including multiple myeloma and acute myeloid leukemia patient-derived bone marrow samples, and in vivo models. Our findings underscore the versatility of VHH molecules in targeting composite epitopes, in this case consisting of a complexed monomorphic antigen-presenting molecule and an invariant TCR, and represent a generalizable antitumor approach.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: CD1d-specific VHH1D12 activates type I NKT cells.
Fig. 2: VHH1D12 selectively activates type I NKT cells and enhances their avidity toward (weakly)agonistic antigens presented by CD1d.
Fig. 3: Differential docking of VHH1D5, VHH1D22 and VHH1D12 over CD1d.
Fig. 4: VHH1D5 and VHH1D22 dock over the A′-pocket and flank of the F′-pocket of CD1d, respectively.
Fig. 5: The bispecific properties of VHH1D12 stabilize the CD1d-type I NKT TCR interface.
Fig. 6: VHH1D12 triggers type I NKT cell proliferation and cytokine production and lysis of CD1d-expressing tumor cells in vitro.
Fig. 7: VHH1D12 induces type I NKT cell activation and cytotoxicity toward patient tumor cells that correlates with CD1d expression.
Fig. 8: VHH1D12 induces type I NKT cell antitumor activity in vivo and increases survival in an MM model.

Similar content being viewed by others

Data availability

The crystal structures have been deposited in the PDB: VHH1D5-hCD1d(α-GalCer) under PDB accession code 6V7Y; VHH1D22-hCD1d(α-GalCer) under PDB accession code 6V7Z; and VHH1D12-hCD1d(α-GalCer)-NKT12-TCR under PDB accession code 6V80. The data that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Rossjohn, J. et al. T cell antigen receptor recognition of antigen-presenting molecules. Annu. Rev. Immunol. 33, 169–200 (2015).

    CAS  PubMed  Google Scholar 

  2. Schumacher, T. N., Scheper, W. & Kvistborg, P. Cancer neoantigens. Annu. Rev. Immunol. 37, 173–200 (2019).

    CAS  PubMed  Google Scholar 

  3. Johansen, L. L., Lock-Andersen, J. & Hviid, T. V. F. The pathophysiological impact of HLA class Ia and HLA-G expression and regulatory T cells in malignant melanoma: a review. J. Immunol. Res. 2016, 6829283 (2016).

    PubMed  PubMed Central  Google Scholar 

  4. Exley, M., Garcia, J., Balk, S. P. & Porcelli, S. Requirements for CD1d recognition by human invariant Vα24+ CD4CD8 T cells. J. Exp. Med. 186, 109–120 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Abrahimi, P. et al. Blocking MHC class II on human endothelium mitigates acute rejection. JCI Insight 1, e85293 (2016).

    PubMed  PubMed Central  Google Scholar 

  6. Fugger, L., Jensen, L. T. & Rossjohn, J. Challenges, progress, and prospects of developing therapies to treat autoimmune diseases. Cell 181, 63–80 (2020).

    CAS  PubMed  Google Scholar 

  7. Stone, J. D., Harris, D. T. & Kranz, D. M. TCR affinity for p/MHC formed by tumor antigens that are self-proteins: impact on efficacy and toxicity. Curr. Opin. Immunol. 33, 16–22 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rossjohn, J., Pellicci, D. G., Patel, O., Gapin, L. & Godfrey, D. I. Recognition of CD1d-restricted antigens by natural killer T cells. Nat. Rev. Immunol. 12, 845–857 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Godfrey, D. I., Le Nours, J., Andrews, D. M., Uldrich, A. P. & Rossjohn, J. Unconventional T cell targets for cancer immunotherapy. Immunity 48, 453–473 (2018).

    CAS  PubMed  Google Scholar 

  10. Nair, S. & Dhodapkar, M. V. Natural killer T cells in cancer immunotherapy. Front. Immunol. 8, 1178 (2017).

    PubMed  PubMed Central  Google Scholar 

  11. Metelitsa, L. S., Weinberg, K. I., Emanuel, P. D. & Seeger, R. C. Expression of CD1d by myelomonocytic leukemias provides a target for cytotoxic NKT cells. Leukemia 17, 1068–1077 (2003).

    CAS  PubMed  Google Scholar 

  12. Exley, M. A. et al. A major fraction of human bone marrow lymphocytes are Th2-like CD1d-reactive T cells that can suppress mixed lymphocyte responses. J. Immunol. 167, 5531–5534 (2001).

    CAS  PubMed  Google Scholar 

  13. Lameris, R. et al. Generation and characterization of CD1d-specific single-domain antibodies with distinct functional features. Immunology 149, 111–121 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Kjer-Nielsen, L. et al. A structural basis for selection and cross-species reactivity of the semi-invariant NKT cell receptor in CD1d/glycolipid recognition. J. Exp. Med. 203, 661–673 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Uldrich, A. P. et al. CD1d-lipid antigen recognition by the γδ TCR. Nat. Immunol. 14, 1137–1145 (2013).

    CAS  PubMed  Google Scholar 

  16. Le Nours, J. et al. Atypical natural killer T-cell receptor recognition of CD1d-lipid antigens. Nat. Commun. 7, 10570 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Luoma, A. M. et al. Crystal structure of Vδ1 T cell receptor in complex with CD1d-sulfatide shows MHC-like recognition of a self-lipid by human γδ T cells. Immunity 39, 1032–1042 (2013).

    CAS  PubMed  Google Scholar 

  18. Borg, N. A. et al. CD1d-lipid-antigen recognition by the semi-invariant NKT T-cell receptor. Nature 448, 44–49 (2007).

    CAS  PubMed  Google Scholar 

  19. Patel, O. et al. Recognition of CD1d-sulfatide mediated by a type II natural killer T cell antigen receptor. Nat. Immunol. 13, 857–863 (2012).

    CAS  PubMed  Google Scholar 

  20. Girardi, E. et al. Type II natural killer T cells use features of both innate-like and conventional T cells to recognize sulfatide self antigens. Nat. Immunol. 13, 851–856 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Pellicci, D. G. et al. Recognition of β-linked self glycolipids mediated by natural killer T cell antigen receptors. Nat. Immunol. 12, 827–833 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Miyamoto, K., Miyake, S. & Yamamura, T. A synthetic glycolipid prevents autoimmune encephalomyelitis by inducing TH2 bias of natural killer T cells. Nature 413, 531–534 (2001).

    CAS  PubMed  Google Scholar 

  23. Raju, R. et al. Synthesis and evaluation of 3″- and 4″-deoxy and -fluoro analogs of the immunostimulatory glycolipid, KRN7000. Bioorg. Med. Chem. Lett. 19, 4122–4125 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Wun, K. S. et al. A molecular basis for the exquisite CD1d-restricted antigen specificity and functional responses of natural killer T cells. Immunity 34, 327–339 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wun, K. S. et al. A minimal binding footprint on CD1d-glycolipid is a basis for selection of the unique human NKT TCR. J. Exp. Med. 205, 939–949 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, J. et al. Lipid binding orientation within CD1d affects recognition of Borrelia burgorferi antigens by NKT cells. Proc. Natl Acad. Sci. USA 107, 1535–1540 (2010).

    CAS  PubMed  Google Scholar 

  27. Kanamori, M., Tasumi, Y., Iyoda, T., Ushida, M. & Inaba, K. Sulfatide inhibits α-galactosylceramide presentation by dendritic cells. Int. Immunol. 24, 129–136 (2012).

    CAS  PubMed  Google Scholar 

  28. Bai, L. et al. Lysosomal recycling terminates CD1d-mediated presentation of short and polyunsaturated variants of the NKT cell lipid antigen αGalCer. Proc. Natl Acad. Sci. USA 106, 10254–10259 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Im, J. S. et al. Kinetics and cellular site of glycolipid loading control the outcome of natural killer T cell activation. Immunity 30, 888–898 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Godfrey, D. I. & Kronenberg, M. Going both ways: immune regulation via CD1d-dependent NKT cells. J. Clin. Invest. 114, 1379–1388 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Dmitriev, O. Y., Lutsenko, S. & Muyldermans, S. Nanobodies as probes for protein dynamics in vitro and in cells. J. Biol. Chem. 291, 3767–3775 (2016).

    CAS  PubMed  Google Scholar 

  32. McMillin, D. W. et al. Compartment-specific bioluminescence imaging platform for the high-throughput evaluation of antitumor immune function. Blood 119, e131–e138 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kimbrel, E. A., Davis, T. N., Bradner, J. E. & Kung, A. L. In vivo pharmacodynamic imaging of proteasome inhibition. Mol. Imaging 8, 140–147 (2009).

    CAS  PubMed  Google Scholar 

  34. Im, J. S. et al. Direct measurement of antigen binding properties of CD1 proteins using fluorescent lipid probes. J. Biol. Chem. 279, 299–310 (2004).

    CAS  PubMed  Google Scholar 

  35. Brossay, L. et al. CD1d-mediated recognition of an α-galactosylceramide by natural killer T cells is highly conserved through mammalian evolution. J. Exp. Med. 188, 1521–1528 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Reantragoon, R. et al. Structural insight into MR1-mediated recognition of the mucosal associated invariant T cell receptor. J. Exp. Med. 209, 761–774 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Heemskerk, M. H. M. et al. Redirection of antileukemic reactivity of peripheral T lymphocytes using gene transfer of minor histocompatibility antigen HA-2-specific T-cell receptor complexes expressing a conserved alpha joining region. Blood 102, 3530–3540 (2003).

    CAS  PubMed  Google Scholar 

  38. Holst, J., Vignali, K. M., Burton, A. R. & Vignali, D. A. A. Rapid analysis of T-cell selection in vivo using T cell-receptor retrogenic mice. Nat. Methods 3, 191–197 (2006).

    CAS  PubMed  Google Scholar 

  39. Lameris, R., Schneiders, F. L., de Gruijl, T. D. & van der Vliet, H. J. Exploiting the CD1d-iNKT cell axis for potentiation of DC-based cancer vaccines. Methods Mol. Biol. 1139, 155–165 (2014).

    CAS  PubMed  Google Scholar 

  40. Aricescu, A. R., Lu, W. & Jones, E. Y. A time- and cost-efficient system for high-level protein production in mammalian cells. Acta Crystallogr. D Biol. Crystallogr. 62, 1243–1250 (2006).

    PubMed  Google Scholar 

  41. Aragão, D. et al. MX2: a high-flux undulator microfocus beamline serving both the chemical and macromolecular crystallography communities at the Australian Synchrotron. J. Synchrotron Radiat. 25, 885–891 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Winn, M. D. et al. Overview of the CCP4 suite and current developments. Acta Crystallogr. D Biol. Crystallogr. 67, 235–242 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Bricogne, G. et al. NBUSTER v.2.10 (Global Phasing Ltd, 2017).

  47. Lefranc, M.-P. et al. IMGT unique numbering for immunoglobulin and T cell receptor variable domains and Ig superfamily V-like domains. Dev. Comp. Immunol. 27, 55–77 (2003).

    CAS  PubMed  Google Scholar 

  48. Geuze, R. E. et al. Luciferase labeling for multipotent stromal cell tracking in spinal fusion versus ectopic bone tissue engineering in mice and rats. Tissue Eng. Part A 16, 3343–3351 (2010).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by CCA-VICI grant no. 2000483 from the VU University Medical Center, grant no. 140343 from Worldwide Cancer Research, funding from LAVA Therapeutics (R.L., J.V. and H.J.V.), the National Health and Medical Research Council of Australia (NHMRC; grant nos. 1113293 and 1140126), the Australian Research Council (ARC; grant no. CE140100011) and the Cancer Council of Victoria (J.R., D.I.G.). S.G. is supported by an NHMRC Senior Research Fellowship (no. GNT#1159272). A.R.H. is supported by National Institutes of Health (grant no. R01 GM111849). D.I.G. is supported by an NHMRC Senior Principal Research Fellowship (no. 1117766). A.P.U. is supported by an ARC Future Fellowship (no. FT140100278). J.L.N is supported by an ARC Future Fellowship (no. FT160100074). J.R. is supported by an ARC Laureate Fellowship (no. FL160100049). We thank the Monash Macromolecular Crystallization Facility staff for assistance with crystallization, the Australian Synchrotron for assistance with data collection, the University of Melbourne, Department of Microbiology and Immunology Flow Cytometry facility for flow cytometry support, L. Smit and K. K. Sivaramanand for assistance and P.W.H.I. Parren for comments.

Author information

Authors and Affiliations

Authors

Contributions

R.L., A.S., J.V., S.J.J.R. and S.M.Q-P. performed the research. R.L. and A.S. analyzed the data. R.L., A.S., S.Z., D.I.G., T.D.G., J.R. and H.J.V. designed the research and interpreted the data. S.G., D.G.P., A.P.U., J.L.N., S.K.R., A.R.H. and R.W.J.G. provided unique reagents. R.L., A.S., D.I.G., T.D.G., J.R. and H.J.V. wrote the manuscript.

Corresponding authors

Correspondence to Jamie Rossjohn or Hans J. van der Vliet.

Ethics declarations

Competing interests

H.J.V. is Chief Scientific Officer of LAVA Therapeutics, a company working on bispecific antibodies. H.J.V. and T.D.G. are shareholders of LAVA Therapeutics. D.I.G. is a member of the scientific advisory board and a shareholder of Avalia Immunotherapies, a company working on NKT cell-based vaccines.

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 CD1d-specific VHH1D12 activates type I NKT cells.

a-f, Expression (%) of CD25 or CD107a on type I NKT cells after 24 h or 4 h co-culture with either HeLa wild type (WT), HeLa.CD1d (a,c,f), MM.1 s.WT, MM.1 s.CD1d cells (b), A431 (d) or CCRF-CEM (e), plus NC, α-GalCer (100 ng/ml), VHH1D control (100 nM or concentration range (e)) and/or VHH1D12 (100 nM or concentration range (e)) (n = 3 (a-d, f) or n = 4 (e) independent experiments with type I NKT cells obtained from individual donors), analysed by flow cytometry. Transwell; type I NKT cells were placed in the upper chamber. g, Representative histogram depicting expression of CD1d (by means of binding of biotinylated VHH1D22) to MM.1S.CD1d after 24 h incubation with negative control (NC) or VHH1D12 (100 nM). h, Exemplifying gating strategy used for type I NKT cell CD107a expression analysis (here after co-culture with CCRF-CEM cells). a-d,f, The bars indicate the mean +s.d.; e, nonlinear regresssion with 95% confidence bands (dotted lines). a,d, One-way analysis of variance (ANOVA) with Tukey multiple comparisons test; b,c, two-way ANOVA with Šídák; f, Tukey multiple comparisons test.

Source data

Extended Data Fig. 2 VHH1D12 selectively activates type I NKT cells.

a, Fold change in CD69 expression on gated Jurkat-76 (J76) or SKW3 cell lines transduced with the CD1d(α-GalCer)-restricted NKT15-, 9B3- or 9C1-TCRs after overnight co-culture with either C1R.WT (CD1d negative) or C1R.GFP.CD1d plus NC, α-GalCer (100 ng/ml), VHH1D5 (1 µM), VHH1D12 (1 µM), VHH1D22 (1 µM) or a combination thereof (n = 4 (NKT15) or n = 3 (9B3-, and 9C1 TCR) independent experiments), analysed by flow cytometry. MF, median fluorescence. b, Fold change in CD69 expression on gated Jurkat J.RT3-T3.5 (JRT3) cell line transduced with the CD1d(sulfatide)-restricted DP10.7 TCR after overnight co-culture with either C1R.WT (CD1d negative) or C1R.CD1d plus NC, sulfatide (25 µg/ml), VHH1D22 (1 µM) or a combination thereof (n = 3 independent experiments), analysed by flow cytometry. c, Histograms depicting binding of lipid loaded CD1d-tetramers, pre-incubated with (red histograms) or without (dark grey histograms) VHH1D12 (~5 times molar excess), to JRT3.DP10.7 cell line. Number in each histogram indicates MF. Data are representative of 3 separate experiments. d, Type I NKT cell binding of lipid loaded CD1d tetramers, simultaneously (+) or after 60 minutes (>) incubated with or without VHH1D12 and/or 1D22 (~5 times molar excess) as indicated (n = 3 independent experiments with type I NKT cells obtained from individual donors). a,b,d, The bars indicate the mean +s.d. a,b,d, One-way analysis of variance (ANOVA) with Tukey multiple comparisons test.

Source data

Extended Data Fig. 3 VHH1D5 and VHH1D22 dock over the A’-pocket and flank of the F′-pocket of CD1d respectively.

a, Binding of VHH1D5 to CD1d wild-type (WT) and alanine-substitution mutants expressed on C1R cells (normalized for GFP expression and made relative to CD1d WT) (n = 5 independent experiments). b, Footprint of NKT12 TCR (yellow), VHH1D22 (dark-red) and overlapping area (orange). c, Binding of VHH1D22 to CD1d WT and alanine-substitution mutants expressed on C1R cells (normalized for GFP expression and made relative to CD1d WT) (n = 5 independent experiments). a,c, The bars indicate mean +s.d.

Source data

Extended Data Fig. 4 Bispecific properties of VHH1D12 stabilize the CD1d-type I NKT-TCR interface.

a, Enlarged view of VHH1D12 CDR1 (green), CDR3 (blue), and CDR1 and CDR3 loop interactions with CD1d(α-GalCer). Residues involved in contacts are represented as sticks, with hydrogen bonds represented as red dashed lines. Nitrogen, oxygen, and sulphate are coloured in blue, red, and yellow respectively. VHH1D12 residues are labelled in purple, CD1d and β2m residues in black. b, Sensorgrams and saturation plot of concentrations series for VHH1D12 passed over CD1d(endogenous). Sensorgram data are representative of 2 independent experiments performed in duplicate, saturation plot indicates data points from both independent experiments.. RU, response unit, KD, equilibrium dissociation constant, Ka, association rate constant, Kd, dissociation rate constant. c, Footprint of NKT12 TCR CDR regions on CD1d(α-GalCer) molecular surface and enlarged view of CDR2β (orange), CDR3β (yellow), CDR1α (teal), CDR2α (green) and CDR3α (purple) loop interactions with CD1d(α-GalCer) (grey). Residues and bonds are represented as above. d, Expression (%) of CD107a on type I NKT cells after 4 h co-culture with MM.1S.mCherry/luc.CD1d cells plus 10 nM VHH1D12 wild type (WT) or indicated alanine substitution mutants (n = 4 independent experiments), and e, histogram depicting binding of biotinylated VHH1D12 and alanine substitution mutants to C1R.GFP.CD1d, analysed by flow cytometry. f, Histogram depicting binding of lipid loaded CD1d-tetramer, whether or not pre-incubated with VHH1D12 (red histogram) or VHH1D12 R96A (blue histogram) (~5 times molar excess), to type I NKT cells. Data are representative of 3 separate experiments with type I NKT cells obtained from individual donors. g,h, Sensorgrams and saturation plots of concentrations series for NKT12 TCR passed over VHH1D12 R45A-CD1d(α-GalCer) (g) or VHH1D12 R96A (i). Sensorgram data are representative of 2 independent experiments performed in duplicate, saturation plot indicates data points from both independent experiments. d, The bars indicate mean +s.d.

Source data

Extended Data Fig. 5 VHH1D12 triggers type I NKT cell cytokine production, anti-tumor cytotoxicity and expansion.

a, Cytokine secretion by type I NKT cells after 24 h co-culture with HeLa.CD1d cells plus negative control (NC), α-GalCer (100 ng/ml), OCH (100 ng/ml), VHH1D12 (100 nM) or a combination thereof, analysed by CBA (n = 4 independent experiments with type I NKT cells obtained from individual donors). b, Living (annexin V and 7-AAD) CCRF-CEM cells after 16 h co-culture with type I NKT cells (effector:target ratio 1:2) plus NC or VHH1D12 (100 nM), quantified by flow cytometric counting beads and made relative to CCRF-CEM cells only (n = 5 independent experiments with type I NKT cells obtained from individual donors). c, Cytotoxicity of type I NKT cells plus NC, α-GalCer (100 ng/ml), VHH1D12 (100 nM) or a combination thereof towards CCRF-CEM cells as determined by annexin V+ and 7-AAD+ after 18 h co-culture (effector:target as indicated) analysed by flow cytometry (n = 4 independent experiments with type I NKT cells obtained from individual donors). d, Exemplifying gating strategy used to analyse type I NKT cell cytotoxicity towards tumor cells (here CCRF-CEM cells). e, Mean percentage CD4+, CD8+, and CD4CD8 type I NKT cell (fresh isolated) after 7-day co-culture with MM.1S.mCherry/luc.CD1d cells plus NC, α-GalCer (100 ng/ml) or VHH1D12 (100 nM) (effector:target ratio of 1:2) (n = 3 independent experiments with type I NKT cells obtained from individual donors). a, The box and whisker plots indicate the median and 25th to 75th percentiles, and minimun to maximum; b,c, the bars indicate the mean +s.d. a, One-way analysis of variance (ANOVA) with Tukey multiple comparisons test; b, two-tailed paired t-test; c, two-way ANOVA with Šídák multiple comparisons test.

Source data

Extended Data Fig. 6 Type I NKT cell cytokine secretion induced by VHH1D12 and CD1d expression and type I NKT cell frequency in MM and AML patient tumor samples.

a, Cytokine secretion by type I NKT cells after 16 h co-culture with fresh bone marrow (BM) derived MM patient samples plus NC, α-GalCer (100 ng/ml) or VHH1D12 (100 nM) (n = 7 individual patients), analysed by CBA. BM, indicates bone marrow cells without added effector cells. b, Exemplary histograms depicting CD1d expression on MM and AML cells. c,d, Exemplifying gating strategy used to analyse BM derived MM and AML patient samples (here after co-culture with type I NKT cells). e, type I NKT cell frequency (% of CD3pos cells) in BM samples of MM and AML patients. a,e, The box and whisker plots indicate the median and 25th to 75th percentiles, and minimum to maximum. a, One-way analysis of variance (ANOVA) with Tukey multiple comparisons test.

Source data

Extended Data Fig. 7 CD1d expression on tumor cells in mice treated with VHH1D12 and type I NKT cells.

a, Histogram depicting CD1d expression on pre-infusion MM.1S.mCherry/luc.CD1d and intraperitoneal plasmacytomas found in 2 mice treated with type I NKT cells plus VHH1D12 (from left to right mouse 2 and 4 in Fig. 5b). b, Exemplifying gating strategy used to analyse tumor-infiltrating type I NKT cells and tumor CD1d expression after tumor dissociation of mouse 2.

Extended Data Fig. 8 Surface expression of CD1a, CD1b, CD1c or CD1d and lipid antigen structures.

a,b, Histograms depicting expression of CD1a, CD1b, CD1c or CD1d on C1R transfectants, K562 transfectant, HeLa wild type (WT), HeLa transfectant, A431, MM.1S.WT, MM.1S transfectant, CCRF-CEM, MOLM-13 and NOMO-1. FMO, fluorescence minus one control. MF, median fluorescence. c, Structures of lipid antigens used in CD1d tetramers. PC, phosphatidylcholine.

Supplementary information

Supplementary Tables

Supplementary Tables 1–6.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lameris, R., Shahine, A., Pellicci, D.G. et al. A single-domain bispecific antibody targeting CD1d and the NKT T-cell receptor induces a potent antitumor response. Nat Cancer 1, 1054–1065 (2020). https://doi.org/10.1038/s43018-020-00111-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-020-00111-6

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