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.

  • Letter
  • Published:

Inefficient CAR-proximal signaling blunts antigen sensitivity

Abstract

Rational design of chimeric antigen receptors (CARs) with optimized anticancer performance mandates detailed knowledge of how CARs engage tumor antigens and how antigen engagement triggers activation. We analyzed CAR-mediated antigen recognition via quantitative, single-molecule, live-cell imaging and found the sensitivity of CAR T cells toward antigen approximately 1,000-times reduced as compared to T cell antigen-receptor-mediated recognition of nominal peptide–major histocompatibility complexes. While CARs outperformed T cell antigen receptors with regard to antigen binding within the immunological synapse, proximal signaling was significantly attenuated due to inefficient recruitment of the tyrosine-protein kinase ZAP-70 to ligated CARs and its reduced concomitant activation and subsequent release. Our study exposes signaling deficiencies of state-of-the-art CAR designs, which presently limit the efficacy of CAR T cell therapies to target tumors with diminished antigen expression.

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: Protein-functionalized planar glass SLB system allows for highly resolved spatiotemporal imaging of CAR- or TCR-mediated antigen recognition.
Fig. 2: CMV-specific T cells detect antigen more efficiently than CAR T cells by 2–3 orders of magnitude.
Fig. 3: CAR T cells engage antigen efficiently yet require a high number of antigen-ligated CARs for ITAM phosphorylation.
Fig. 4: After synapse recruitment, ZAP-70 is activated less efficiently and is more stably antigen- receptor-associated in CAR T cells.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the authors upon reasonable request; see the author contributions for specific datasets.

Code availability

The custom code employed for the analysis of calcium signaling can be made accessible at the reader’s request.

References

  1. Maher, J., Brentjens, R. J., Gunset, G., Rivière, I. & Sadelain, M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ /CD28 receptor. Nat. Biotechnol. 20, 70–75 (2002).

    Article  PubMed  CAS  Google Scholar 

  2. Imai, C. et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18, 676–684 (2004).

    Article  PubMed  CAS  Google Scholar 

  3. Finney, H. M., Lawson, A. D. G., Bebbington, C. R. & Weir, A. N. C. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J. Immunol. 161, 2791–2797 (1998).

    PubMed  CAS  Google Scholar 

  4. Srivastava, S. & Riddell, S. R. Engineering CAR-T cells: design concepts. Trends Immunol. 36, 494–502 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Huang, J. et al. A single peptide-major histocompatibility complex ligand triggers digital cytokine secretion in CD4+ T cells. Immunity 39, 846–857 (2013).

    Article  PubMed  CAS  Google Scholar 

  6. Irvine, D. J., Purbhoo, M. A., Krogsgaard, M. & Davis, M. M. Direct observation of ligand recognition by T cells. Nature 419, 845–849 (2002).

    Article  PubMed  CAS  Google Scholar 

  7. Purbhoo, M. A., Irvine, D. J., Huppa, J. B. & Davis, M. M. T cell killing does not require the formation of a stable mature immunological synapse. Nat. Immunol. 5, 524–530 (2004).

    Article  PubMed  CAS  Google Scholar 

  8. Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Turtle, C. J. et al. CD19 CAR–T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 126, 2123–2138 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. 378, 439–448 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Fry, T. J. et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat. Med. 24, 20–28 (2018).

    PubMed  CAS  Google Scholar 

  12. Huppa, J. B. et al. TCR–peptide–MHC interactions in situ show accelerated kinetics and increased affinity. Nature 463, 963–967 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Douglass, A. D. & Vale, R. D. Single-molecule microscopy reveals plasma membrane microdomains created by protein–protein networks that exclude or trap signaling molecules in T cells. Cell 121, 937–950 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Hudecek, M. et al. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 19, 3153–3164 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Srivastava, S. et al. Logic-gated ROR1 chimeric antigen receptor expression rescues T cell-mediated toxicity to normal tissues and enables selective tumor targeting. Cancer Cell 35, 489–503.e8 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Alberola-Ila, J., Takaki, S., Kerner, J. D. & Perlmutter, R. M. Differential signaling by lymphocyte antigen receptors. Annu. Rev. Immunol. 15, 125–154 (1997).

    Article  PubMed  CAS  Google Scholar 

  17. Chan, A. C., Iwashima, M., Turck, C. W. & Weiss, A. ZAP-70: a 70 kd protein-tyrosine kinase that associates with the TCR ζ chain. Cell 71, 649–662 (1992).

    Article  PubMed  CAS  Google Scholar 

  18. Bunnell, S. C. et al. T cell receptor ligation induces the formation of dynamically regulated signaling assemblies. J. Cell Biol. 158, 1263–1275 (2002).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Pores-Fernando, A. T. & Zweifach, A. Calcium influx and signaling in cytotoxic T-lymphocyte lytic granule exocytosis. Immunol. Rev. 231, 160–173 (2009).

    Article  PubMed  CAS  Google Scholar 

  20. Watanabe, K. et al. Target antigen density governs the efficacy of anti-CD20-CD28-CD3 ζ chimeric antigen receptor-modified effector CD8+ T cells. J. Immunol. 194, 911–920 (2015).

    Article  PubMed  CAS  Google Scholar 

  21. Walker, A. J. et al. Tumor antigen and receptor densities regulate efficacy of a chimeric antigen receptor targeting anaplastic lymphoma kinase. Mol. Ther. 25, 2189–2201 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Potter, T. A., Bluestone, J. A. & Rajan, T. V. A single amino acid substitution in the α3 domain of an H-2 class I molecule abrogates reactivity with CTL. J. Exp. Med. 166, 956–966 (1987).

    Article  PubMed  CAS  Google Scholar 

  23. Baskar, S., Wiestner, A., Wilson, W. H., Pastan, I. & Rader, C. Targeting malignant B cells with an immunotoxin against ROR1. MAbs 4, 349–361 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Yang, J. et al. Therapeutic potential and challenges of targeting receptor tyrosine kinase ROR1 with monoclonal antibodies in B-cell malignancies. PLoS ONE 6, e21018 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Hudecek, M. et al. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol. Res. 3, 125–135 (2015).

    Article  PubMed  CAS  Google Scholar 

  26. Choudhuri, K., Wiseman, D., Brown, M. H., Gould, K. & van der Merwe, P. A. T-cell receptor triggering is critically dependent on the dimensions of its peptide-MHC ligand. Nature 436, 578–582 (2005).

    Article  PubMed  CAS  Google Scholar 

  27. Stewart-Jones, G. et al. Rational development of high-affinity T-cell receptor-like antibodies. Proc. Natl Acad. Sci. USA 106, 5784–5788 (2009).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  28. Maus, M. V. et al. An MHC-restricted antibody-based chimeric antigen receptor requires TCR-like affinity to maintain antigen specificity. Mol. Ther. Oncolytics 3, 16023 (2016).

    Article  Google Scholar 

  29. Maus, M. V. et al. Adoptive immunotherapy for cancer or viruses. Annu. Rev. Immunol. 32, 189–225 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Long, A. H. et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 21, 581–590 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Chmielewski, M., Hombach, A., Heuser, C., Adams, G. P. & Abken, H. T cell activation by antibody-like immunoreceptors: increase in affinity of the single-chain fragment domain above threshold does not increase T cell activation against antigen-positive target cells but decreases selectivity. J. Immunol. 173, 7647–7653 (2004).

    Article  PubMed  CAS  Google Scholar 

  32. Chmielewski, M., Hombach, A. A. & Abken, H. CD28 cosignalling does not affect the activation threshold in a chimeric antigen receptor-redirected T-cell attack. Gene Ther. 18, 62–72 (2011).

    Article  PubMed  CAS  Google Scholar 

  33. Iwashima, M., Irving, B. A., van Oers, N. S., Chan, A. C. & Weiss, A. Sequential interactions of the TCR with two distinct cytoplasmic tyrosine kinases. Science 263, 1136–1139 (1994).

    Article  PubMed  CAS  Google Scholar 

  34. Pelosi, M. et al. Tyrosine 319 in the interdomain B of ZAP-70 is a binding site for the Src homology 2 domain of Lck. J. Biol. Chem. 274, 14229–14237 (1999).

    Article  PubMed  CAS  Google Scholar 

  35. Katz, Z. B., Novotná, L., Blount, A. & Lillemeier, B. F. A cycle of Zap70 kinase activation and release from the TCR amplifies and disperses antigenic stimuli. Nat. Immunol. 18, 86–95 (2017).

    Article  PubMed  CAS  Google Scholar 

  36. Varma, R., Campi, G., Yokosuka, T., Saito, T. & Dustin, M. L. T cell receptor-proximal signals are sustained in peripheral microclusters and terminated in the central supramolecular activation cluster. Immunity 25, 117–127 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Ruella, M. & Maus, M. V. Catch me if you can: leukemia escape after CD19-directed T cell immunotherapies. Comput. Struct. Biotechnol. J. 14, 357–362 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Evans, A. G. et al. Evolution to plasmablastic lymphoma evades CD19-directed chimeric antigen receptor T cells. Br. J. Haematol. 171, 205–209 (2015).

    Article  PubMed  CAS  Google Scholar 

  39. Orlando, E. J. et al. Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat. Med. 24, 1504–1506 (2018).

    PubMed  CAS  Google Scholar 

  40. Wang, X. et al. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood 118, 1255–1263 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Zacharias, D. A., Violin, J. D., Newton, A. C. & Tsien, R. Y. Partitioning of lipid-modified monomeric GFPs into membrane microdomains of live cells. Science 296, 913–916 (2002).

    Article  PubMed  CAS  Google Scholar 

  42. Hudecek, M. et al. The B-cell tumor-associated antigen ROR1 can be targeted with T cells modified to express a ROR1-specific chimeric antigen receptor. Blood 116, 4532–4541 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Brown, C. E. et al. Biophotonic cytotoxicity assay for high-throughput screening of cytolytic killing. J. Immunol. Methods 297, 39–52 (2005).

    Article  PubMed  CAS  Google Scholar 

  44. Klesmith, J. R., Wu, L., Lobb, R. R., Rennert, P. D. & Hackel, B. J. Fine epitope mapping of the CD19 extracellular domain promotes design. Biochemistry 58, 4869–4881 (2019).

    Article  PubMed  CAS  Google Scholar 

  45. Garboczi, D. N., Hung, D. T. & Wiley, D. C. HLA-A2-peptide complexes: refolding and crystallization of molecules expressed in Escherichia coli and complexed with single antigenic peptides. Proc. Natl Acad. Sci. USA 89, 3429–3433 (1992).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  46. Clements, C. S. et al. The production, purification and crystallization of a soluble heterodimeric form of a highly selected T-cell receptor in its unliganded and liganded state. Acta Crystallogr. D Biol. Crystallogr. 58, 2131–2134 (2002).

    Article  CAS  Google Scholar 

  47. Axmann, M., Schütz, G. J. & Huppa, J. B. Single molecule fluorescence microscopy on planar supported bilayers. J. Vis. Exp. e53158 (2015).

  48. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  PubMed  CAS  Google Scholar 

  49. Schindelin, J., Rueden, C. T., Hiner, M. C. & Eliceiri, K. W. The ImageJ ecosystem: an open platform for biomedical image analysis. Mol. Reprod. Dev. 82, 518–529 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Gao, Y. & Kilfoil, M. L. Accurate detection and complete tracking of large populations of features in three dimensions. Opt. Express 17, 4685–4704 (2009).

    Article  PubMed  Google Scholar 

  51. Wieser, S., Moertelmaier, M., Fuertbauer, E., Stockinger, H. & Schütz, G. J. (Un)confined diffusion of CD59 in the plasma membrane determined by high-resolution single molecule microscopy. Biophys. J. 92, 3719–3728 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Roe, M. W., Lemasters, J. J. & Herman, B. Assessment of Fura-2 for measurements of cytosolic free calcium. Cell Calcium 11, 63–73 (1990).

    Article  PubMed  CAS  Google Scholar 

  53. Lowe, D. G. Distinctive image features from scale-invariant keypoints. Int. J. Comput. Vis. 60, 91–110 (2004).

    Article  Google Scholar 

  54. Jaqaman, K. et al. Robust single-particle tracking in live-cell time-lapse sequences. Nat. Methods 5, 695–702 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Tinevez, J.-Y. et al. TrackMate: an open and extensible platform for single-particle tracking. Methods 115, 80–90 (2017).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Austrian Science Fund through project no. P 25775-B2 (V.G., S.K., L.S. and J.H.), the Vienna Science and Technology Fund (no. LS14-031 to V.G. and J.H.), the Platform for Advanced Cellular Therapies (J.H., R.K.), the Doctoral Program Biomolecular Technology of Proteins supported by the Austrian Science Fund (no. W1224 to E.L. and R.K.) and the Innovative Training Network EN-ACTI2NG (European Network on Anti-Cancer Immuno-Therapy Improvement by modification of CAR and TCR Interactions and Nanoscale Geometry) supported by the European Commission. M.H. is a member of the Young Scholar Program (Junges Kolleg) and an extraordinary member of the Bavarian Academy of Sciences. This work was supported by a grant from German Cancer Aid (Max Eder Program, grant no. 70110313 to M.H.) and the German Research Foundation (project no. 324392634, TRR 221 to M.H.). We thank V. Mühlgrabner (Medical University of Vienna) for help with fluorescence-activated cell sorting; R. Platzer (Medical University of Vienna) for help with single-molecule tracking; O. Dushek (Oxford University) for sending us the T1-CAR constructs; and U. Jäger and N. Worel (Medical University of Vienna), M. Lehner (St. Anna Children’s Cancer Research) and A. Rehm (Max Delbrück Center for Molecular Medicine) for constructive criticism.

Author information

Authors and Affiliations

Authors

Contributions

V.G., J.R., M.H. and J.B.H. conceived the project and wrote the manuscript. V.G. conducted and analyzed all imaging-related and biochemical experiments. J.R. and V.D.R.G. generated all T cell lines and conducted classical immunological assays. I.D-P. wrote the analysis software used for the single-cell calcium analysis. S.K. carried out the initial experiments. L.S. produced the HLA-A2 and HLA-A2(D227T/K228A) imaging probes. H.E. and H.S. contributed important ideas. E.L. and R.K. provided the CD19 cells for the SLB-related imaging experiments. E.L. contributed to the CD19-related imaging experiments.

Corresponding authors

Correspondence to Michael Hudecek or Johannes B. Huppa.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Jamie D. K.Wilson was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Biochemical and imaging-based analysis of SLB-embedded proteins as well as in vitro analysis of cytotoxicity, IFNγ secretion and proliferation of CMVsp- and ROR1-CAR-T cells.

a, 12.5% SDS PAGE analysis of the recombinant proteins employed for SLB functionalization. Data shown are representative of (n=3) independent experiments. b, The immobile fraction of SLB-anchored proteins was determined via Fluorescence Recovery After Photobleaching (FRAP). Fluorescence intensities were normalized with regard to initial intensity values and plotted versus time. Data shown are representative of (n=5) independent experiments. c, The diffraction-limited fluorescence signal of a single ROR1-AF555 molecule diffusing laterally in an SLB is marked with a green circle. The multi-step single molecule time trajectory is indicated as a green trace (scale bar: 1 µm). Data shown are representative of (n ≥ 2500) single molecule traces. Scale bar = 2 µm. d, SDS-PAGE of whole cell lysates performed under non-reducing/non-boiling and reducing/boiling conditions combined with immunoblot analysis carried out using an affinity-purified polyclonal rabbit anti-CD3ζ antibody confirms dimeric state of CAR-constructs featuring an IgG4-hinge domain, as employed in this study. Data shown are representative of (n=3) independent experiments. e, Schematic outline of CAR-constructs employed in this study. f, Assessment of CAR-T cell effector functions in cell-based in vitro assays. (top panel) Cytotoxicity of R12 4-1BB ζ-CAR-T cells, which were also specific for CMV, co-cultured with K562 or K562-ROR1 or K562-HLA-A2/CMV target cells (10:1 E:T ratio) over time. (middle panel) Proliferation of CFSE-labeled CMVsp-R12 4-1BB ζ-CAR-T cells co-cultured for 72 hours with K562-ROR1 or K562-HLA-A2/CMV target cells (4:1 ratio) or high IL-2 concentrations. As indicated, the decrease in fluorescence was reflective of the number of cell divisions. Data shown are representative of (n=3) independent experiments. (bottom panel) IFNγ secretion of CMVsp-R12 4-1BB ζ-CAR-T cells co-cultured for 24 hours with K562-ROR1 or K562-HLA-A2/CMV target cells (4:1 ratio) or in the presence of PMA and ionomycin. IFNγ concentration in the supernatant was analyzed by ELISA. Data (top and bottom panels) correspond to T cells derived from healthy donors (n=3) and are displayed as mean ± s.d. Data (middle panel) shown is representative of (n=3) independent experiments.

Extended Data Fig. 2 Epitopes targeted by ROR1-specific CARs, CD45 segregation patterns and the influence of the presence of B7-1 on CAR-T cell antigen sensitivity.

a, Schematic representation of ROR1-epitopes targeted by R12, R11 and 2A2 CARs with previously published binding constants 23,24. b, Antigen dose-response of R11 4-1BB ζ-s featuring a size-adjusted and a short linker as is indicated. Each data point comprises data from (n ≥ 230) cells. Data shown are representative of (n=3) independent experiments. c, Montage of representative immunofluorescence images (left panel) of a R12 4-1BB ζ-CAR-T cell confronted with an SLB presenting ROR1 (318 ROR1-AF555 μm²), ICAM-1 and B7-1. The yellow line in the merged channel denotes the position of the fluorescence intensity line scan shown in the upper middle panel. The Pearson coefficient (upper right panel) was derived by plotting anti-CD45-antibody-AF647 intensities against ROR1-AF555 intensities within individually scanned pixels and contributed to the quantitation shown in lower middle panel. Distributions of Pearson coefficients (lower middle and right panels) are shown for indicated SLB densities of ROR-AF555 / HLA-A2/CMV-AF555 and anti-CD45 antibody-AF647within synaptic antigen clusters (lower panels; n ≥ 40 clusters per condition; n = 16 cells per panel). The negative Pearson coefficients indicate anti-correlated distribution of TCR/CAR and CD45. The degree of exclusion of CD45 from areas of bound antigen is indistinguishable for CAR-T cells engaging ROR1 and CMVsp T cells recognizing HLA-A2/CMV. Data shown are representative of (n=3) independent experiments; scale bar = 2 µm. d, Quantitation of the magnitude of the calcium signal (as measured using the ratiometric calcium sensor fura-2) of activated R12 CAR-T cells featuring a 4-1BB-, CD28- or no intracellular costimulatory module in response to 1.9 ROR1 molecules µm−2. Data are representative of (n=3) independent experiments with data points displaying the measured means (n ≥ 57 cells) ± s.d.. e, Relative surface expression levels (upper panels) of ROR1-CARs and CD28. Calcium response (lower panel) of T cells (n ≥ 841 cells per data point) modified with indicated CARs to increasing antigen densities in the presence or absence of B7-1. Data shown all panels are representative of independent experiments (n=3).

Extended Data Fig. 3 TCR/CAR expression levels and Lck-phosphorylation in the course of antigen recognition.

a, Comparison of TCR- and CAR-surface densities. (left panel) CMVsp-ROR1-CAR-T cells were labeled with either recombinant ROR-AF647 or AF647-conjugated anti-TCR mAb (clone IP26) to determine concentrations required for label saturation. (middle panel) Flow cytometric calibration of AF647 intensity values with the use of QuantumTM Alexa Flour® 647 MESF beads. Resulting MFIs were plotted against the number of equivalent AF647 fluorophores. (right panel) Quantitation of CARs or TCRs on the surface of CMVsp-ROR1-CAR-T cells based on label saturation (left panel) and flow cytometric calibration (middle panel). (left panel) Data shown is representative of independent experiments (n=3). (right and middle panel) Data corresponding to (n=4) donors is shown as mean ± s.d. b, Quantification of synaptic Lck activation (1st panel) and inhibition (2nd panel) as quantified via immunofluorescence in the course of antigen recognition by CMV-specific T cells as well as CAR-T cells as is indicated. Each data point shows mean ± s.e.m (n=30 cells). Plotted data are representative of independent experiments (n=3).

Extended Data Fig. 4 Relative ZAP-70 expression levels, synaptic architecture and the influence of ITAM numbers on CAR-T cell antigen sensitivity.

a, Levels of ectopically expressed ZAP-70-GFP as compared via immunoblot analysis to the expression of endogenous ZAP-70. Data shown are representative of independent experiments (n=3). b, Representative synapses of R12 4-1BB ζ-CAR-T cells and CMVsp-T cells contacting SLBs presenting ROR1-AF555 at a density of 11 molecules µm−2 or HLA-A2/CMV-AF555 at a density of 7 molecules µm−2, respectively (n=30 cells per condition). Data shown are representative of at independent experiments (n=5); scale bar = 5µm. c, Antigen dose-response of T cells modified with R12 4-1BB CARs featuring 2 x 1 ITAMs, 2 x 3 ITAMs or 2 x 6 ITAMs. Each data point comprises data from T cells (n ≥ 402). Data shown are representative of independent experiments (n=3).

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gudipati, V., Rydzek, J., Doel-Perez, I. et al. Inefficient CAR-proximal signaling blunts antigen sensitivity. Nat Immunol 21, 848–856 (2020). https://doi.org/10.1038/s41590-020-0719-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0719-0

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