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:

Modular chimeric cytokine receptors with leucine zippers enhance the antitumour activity of CAR T cells via JAK/STAT signalling

Abstract

The limited availability of cytokines in solid tumours hinders maintenance of the antitumour activity of chimeric antigen receptor (CAR) T cells. Cytokine receptor signalling pathways in CAR T cells can be activated by transgenic expression or injection of cytokines in the tumour, or by engineering the activation of cognate cytokine receptors. However, these strategies are constrained by toxicity arising from the activation of bystander cells, by the suboptimal biodistribution of the cytokines and by downregulation of the cognate receptor. Here we show that replacement of the extracellular domains of heterodimeric cytokine receptors in T cells with two leucine zipper motifs provides optimal Janus kinase/signal transducer and activator of transcription signalling. Such chimeric cytokine receptors, which can be generated for common γ-chain receptors, interleukin-10 and -12 receptors, enabled T cells to survive cytokine starvation without induction of autonomous cell growth, and augmented the effector function of CAR T cells in vitro in the setting of chronic antigen exposure and in human tumour xenografts in mice. As a modular design, leucine zippers can be used to generate constitutively active cytokine receptors in effector immune cells.

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: Two leucine zipper motifs endow ZipRs with constitutive signal transduction.
Fig. 2: Zip2R and Zip7R activate distinct signalling pathways as determined by multiplexed phosphoproteomics.
Fig. 3: ZipRs improve CAR T cell effector function during chronic antigen exposure in vitro.
Fig. 4: ZipRs augment CAR T cell antitumour activity in vivo.
Fig. 5: Leucine zipper ectodomains enable the generation of ZipRs from diverse cytokine receptor families.
Fig. 6: CAR.ZipR T cells sustain effector function through memory/survival or hybrid memory/effector transcriptional programmes.

Similar content being viewed by others

Data availability

The RNA-seq data files are available through Synapse (https://www.synapse.org/#!Synapse:syn52457643). The raw and analysed datasets generated during the study are available for research purposes from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

The code used to analyse the cluster size of colocalized Zip receptors can be accessed in the AutomatedImageAnalysis repository at https://github.com/Jorge-Ibanez-StJude/AutomatedImageAnalysis.

References

  1. 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  CAS  PubMed  PubMed Central  Google Scholar 

  2. Melenhorst, J. J. et al. Decade-long leukaemia remissions with persistence of CD4(+) CAR T cells. Nature 602, 503–509 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Heczey, A. et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol. Ther. 25, 2214–2224 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lamers, C. H., Klaver, Y., Gratama, J. W., Sleijfer, S. & Debets, R. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells—a completed study overview. Biochem. Soc. Trans. 44, 951–959 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Adusumilli, P. S. et al. A Phase I trial of regional mesothelin-targeted CAR T-cell therapy in patients with malignant pleural disease, in combination with the anti-PD-1 agent pembrolizumab. Cancer Discov. 11, 2748–2763 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Qi, C. et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat. Med. 28, 1189–1198 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Heczey, A. et al. Anti-GD2 CAR-NKT cells in relapsed or refractory neuroblastoma: updated phase 1 trial interim results. Nat. Med. https://doi.org/10.1038/s41591-023-02363-y (2023).

  8. Del Bufalo, F. et al. GD2-CART01 for relapsed or refractory high-risk neuroblastoma. N. Engl. J. Med. 388, 1284–1295 (2023).

    Article  PubMed  Google Scholar 

  9. Markley, J. C. & Sadelain, M. IL-7 and IL-21 are superior to IL-2 and IL-15 in promoting human T cell-mediated rejection of systemic lymphoma in immunodeficient mice. Blood 115, 3508–3519 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Koneru, M., Purdon, T. J., Spriggs, D., Koneru, S. & Brentjens, R. J. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo. Oncoimmunology 4, e994446 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Krenciute, G. et al. Transgenic expression of IL15 improves antiglioma activity of IL13Ralpha2-CAR T cells but results in antigen loss variants. Cancer Immunol. Res. 5, 571–581 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Batra, S. A. et al. Glypican-3-specific CAR T cells coexpressing IL15 and IL21 have superior expansion and antitumor activity against hepatocellular carcinoma. Cancer Immunol. Res. 8, 309–320 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Chmielewski, M. & Abken, H. CAR T cells releasing IL-18 convert to T-Bet(high) FoxO1(low) effectors that exhibit augmented activity against advanced solid tumors. Cell Rep. 21, 3205–3219 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Ma, X. et al. Interleukin-23 engineering improves CAR T cell function in solid tumors. Nat. Biotechnol. 38, 448–459 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stach, M. et al. Inducible secretion of IL-21 augments anti-tumor activity of piggyBac-manufactured chimeric antigen receptor T cells. Cytotherapy 22, 744–754 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Heczey, A. et al. Anti-GD2 CAR-NKT cells in patients with relapsed or refractory neuroblastoma: an interim analysis. Nat. Med. 26, 1686–1690 (2020).

    Article  CAS  PubMed  Google Scholar 

  17. O’Cearbhaill, R. E. et al. A phase I clinical trial of autologous chimeric antigen receptor (CAR) T cells genetically engineered to secrete IL-12 and to target the MUC16ecto antigen in patients (pts) with MUC16ecto+recurrent high-grade serous ovarian cancer (HGSOC). Gynecol. Oncol. 159, 42 (2020).

    Article  Google Scholar 

  18. Zhang, L. et al. Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma. Clin. Cancer Res. 21, 2278–2288 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Shum, T. et al. Constitutive signaling from an engineered IL7 receptor promotes durable tumor elimination by tumor-redirected T cells. Cancer Discov. 7, 1238–1247 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lange, S. et al. A chimeric GM-CSF/IL18 receptor to sustain CAR T-cell function. Cancer Discov. 11, 1661–1671 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hunter, M. R. et al. Chimeric gammac cytokine receptors confer cytokine independent engraftment of human T lymphocytes. Mol. Immunol. 56, 1–11 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Sockolosky, J. T. et al. Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science 359, 1037–1042 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kochenderfer, J. N. et al. Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum interleukin-15 levels. J. Clin. Oncol. 35, 1803–1813 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yeku, O. O., Purdon, T. J., Koneru, M., Spriggs, D. & Brentjens, R. J. Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment. Sci. Rep. 7, 10541 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Vera, J. F. et al. Genetic manipulation of tumor-specific cytotoxic T lymphocytes to restore responsiveness to IL-7. Mol. Ther. 17, 880–888 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Agarwal, Y. et al. Intratumourally injected alum-tethered cytokines elicit potent and safer local and systemic anticancer immunity. Nat. Biomed. Eng. 6, 129–143 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mischnik, M. et al. IKAP: a heuristic framework for inference of kinase activities from Phosphoproteomics data. Bioinformatics 32, 424–431 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Nguyen, P. et al. Route of 41BB/41BBL costimulation determines effector function of B7-H3-CAR.CD28zeta T cells. Mol. Ther. Oncolytics 18, 202–214 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kramer, A., Green, J., Pollard, J. Jr. & Tugendreich, S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics 30, 523–530 (2014).

    Article  PubMed  Google Scholar 

  31. Bruniquel, D., Borie, N., Hannier, S. & Triebel, F. Regulation of expression of the human lymphocyte activation gene-3 (LAG-3) molecule, a ligand for MHC class II. Immunogenetics 48, 116–124 (1998).

    Article  CAS  PubMed  Google Scholar 

  32. Meyaard, L., Hovenkamp, E., Otto, S. A. & Miedema, F. IL-12-induced IL-10 production by human T cells as a negative feedback for IL-12-induced immune responses. J. Immunol. 156, 2776–2782 (1996).

    Article  CAS  PubMed  Google Scholar 

  33. Joshi, N. S. et al. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kanai, T. et al. Identification of STAT5A and STAT5B target genes in human T cells. PLoS ONE 9, e86790 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Pearce, E. L. et al. Control of effector CD8+ T cell function by the transcription factor Eomesodermin. Science 302, 1041–1043 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Wei, J. et al. Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature 576, 471–476 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Omilusik, K. D. et al. Transcriptional repressor ZEB2 promotes terminal differentiation of CD8+ effector and memory T cell populations during infection. J. Exp. Med. 212, 2027–2039 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Stuhlmann-Laeisz, C. et al. Forced dimerization of gp130 leads to constitutive STAT3 activation, cytokine-independent growth, and blockade of differentiation of embryonic stem cells. Mol. Biol. Cell 17, 2986–2995 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Seubert, N. et al. Active and inactive orientations of the transmembrane and cytosolic domains of the erythropoietin receptor dimer. Mol. Cell 12, 1239–1250 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Patel, N., Herrman, J. M., Timans, J. C. & Kastelein, R. A. Functional replacement of cytokine receptor extracellular domains by leucine zippers. J. Biol. Chem. 271, 30386–30391 (1996).

    Article  CAS  PubMed  Google Scholar 

  42. Brooks, A. J. et al. Mechanism of activation of protein kinase JAK2 by the growth hormone receptor. Science 344, 1249783 (2014).

    Article  PubMed  Google Scholar 

  43. Ross, S. H. et al. Phosphoproteomic analyses of interleukin 2 signaling reveal integrated JAK kinase-dependent and -independent networks in CD8(+) T cells. Immunity 45, 685–700 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chinnasamy, D. et al. Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin. Cancer Res. 18, 1672–1683 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chen, Y. et al. Eradication of neuroblastoma by T cells redirected with an optimized GD2-specific chimeric antigen receptor and interleukin-15. Clin. Cancer Res. 25, 2915–2924 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Straathof, K. C. et al. An inducible caspase 9 safety switch for T-cell therapy. Blood 105, 4247–4254 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Prinzing, B. et al. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity. Sci. Transl. Med. 13, eabh0272 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Haydar, D. et al. Cell-surface antigen profiling of pediatric brain tumors: B7-H3 is consistently expressed and can be targeted via local or systemic CAR T-cell delivery. Neuro. Oncol. 23, 999–1011 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Mount, C. W. et al. Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M(+) diffuse midline gliomas. Nat. Med. 24, 572–579 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Freitas, K. A. et al. Enhanced T cell effector activity by targeting the Mediator kinase module. Science 378, eabn5647 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Blokon-Kogan, D. et al. Membrane anchored IL-18 linked to constitutively active TLR4 and CD40 improves human T cell antitumor capacities for adoptive cell therapy. J. Immunother. Cancer 10, e001544 (2022).

    Article  PubMed Central  Google Scholar 

  52. Hurton, L. V. et al. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc. Natl Acad. Sci. USA 113, E7788–E7797 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hu, J. et al. Cell membrane-anchored and tumor-targeted IL-12 (attIL12)-T cell therapy for eliminating large and heterogeneous solid tumors. J. Immunother. Cancer 10, e003633 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Kakarla, S. et al. Antitumor effects of chimeric receptor engineered human T cells directed to tumor stroma. Mol. Ther. 21, 1611–1620 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Yi, Z., Prinzing, B. L., Cao, F., Gottschalk, S. & Krenciute, G. Optimizing EphA2-CAR T cells for the adoptive immunotherapy of glioma. Mol. Ther. Methods Clin. Dev. 9, 70–80 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Li, N. et al. CAR T cells targeting tumor-associated exons of glypican 2 regress neuroblastoma in mice. Cell Rep. Med. 2, 100297 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Moll, J. R., Ruvinov, S. B., Pastan, I. & Vinson, C. Designed heterodimerizing leucine zippers with a ranger of pIs and stabilities up to 10(-15) M. Protein Sci. 10, 649–655 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  59. Bolte, S. & Cordelieres, F. P. A guided tour into subcellular colocalization analysis in light microscopy. J. Microsc. 224, 213–232 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Tan, H. et al. Integrative proteomics and phosphoproteomics profiling reveals dynamic signaling networks and bioenergetics pathways underlying T cell activation. Immunity 46, 488–503 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang, X. et al. JUMP: a tag-based database search tool for peptide identification with high sensitivity and accuracy. Mol. Cell. Proteomics 13, 3663–3673 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Niu, M. et al. Extensive peptide fractionation and y1 ion-based interference detection method for enabling accurate quantification by isobaric labeling and mass spectrometry. Anal. Chem. 89, 2956–2963 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Du, X. et al. Hippo/Mst signalling couples metabolic state and immune function of CD8alpha(+) dendritic cells. Nature 558, 141–145 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ritchie, M. E. et al. limma Powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Hornbeck, P. V. et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 43, D512–D520 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. McCommon and A. George (St. Jude Animal Resource Center) and C. Coleman (St. Jude Center for In Vivo Imaging and Therapeutics) for assistance with in vivo mouse studies. We thank Q. Pan for assistance with bioinformatics and figure design. We also thank K. Nichols and S. Albeituni for helpful discussions on the use of ruxolitinib. Animal imaging was performed by the St. Jude Center for In Vivo Imaging and Therapeutics, which is supported by St. Jude Children’s Research Hospital and the National Cancer Institute (NCI; no. P30 CA021765). We thank the Computational Structural Biology Center in the Department of Structural Biology for support. Cellular images were acquired at the St. Jude Cell and Tissue Imaging Center, which is supported by St. Jude Children’s Research Hospital and NCI (no. P30 CA021765). The schematic shown in Fig. 1a was created with Biorender.com, for which we have a licence. This work was supported by NCI grant no. F31CA250401-01A1 to M.B., grant nos. R01NS121249 and R01NS122859 to G.K. and the American Lebanese Syrian Associated Charities to J.Y., J.P., M.M.B., G.K. and S.G. The content is solely the responsibility of the authors and does not necessarily represent the official views of NIH.

Author information

Authors and Affiliations

Authors

Contributions

This study was conceptualized by M.B., G.K. and S.G. Experimentation and analysis were performed by M.B., S.L., B.I.S., H.Shi., X.S., X.M., J.I., P.N., M.S., J.W., A.K., D.L., S.L.P., H.T., R.V.P., Y.L., Z-F.Y., A.A.A., H.S., and P.V. M.H. provided the CT3 scFv. J.Y., J.P., H.C., M.M.B., G.K. and S.G. supervised the study. M.B., J.Y., J.P., H.C., M.M.B., G.K. and S.G provided funding and resources. M.B and S.G wrote the paper. All authors reviewed and edited the paper.

Corresponding author

Correspondence to Stephen Gottschalk.

Ethics declarations

Competing interests

M.B., B.I.S., A.A.A., M.M.B., G.K. and S.G. are coinventors on a patent application for the developed Zip receptor technology. S.L., A.A.A., G.K. and S.G. are coinventors on patent applications in the fields of cell or gene therapy for cancer. M.H. is an inventor on international patent application no. PCT/US2019/045338 assigned to NIH, ‘High affinity monoclonal antibodies targeting glypican-2 and uses thereof’. H.C. is a consultant of Kumquat Biosciences, Inc. S.G. is a consultant of TESSA Therapeutics, a member of the Data and Safety Monitoring Board of Immatics and has received honoraria from Tidal, Catamaran Bio, Sanofi and Novartis within the past 2 years. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Even Weber and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 Zip2R colocalization analysis by confocal microscopy.

a, Pearson correlation analysis of mRuby and mClover in HEK293T cells transfected with indicated constructs (IL-2Rβ(1x): N = 16, IL-2Rγ(1x): N = 8, Zip2R(1x): N = 37, IL-2Rβ(2x): N = 33, IL-2Rγ(2x): N = 24, Zip2R(2x): N = 28, mean ± SD, ****p < 0.0001, one-way ANOVA with Tukey’s multiple comparisons test). b, Representative images.

Source data

Extended Data Fig. 2 Zip2R(2x) subcellular localization and trafficking determined by confocal microscopy.

a, Schematic of SNAP/CLIP tag system. b, Representative images of Zip2R(2x) with endosomal markers. Scale bar = 10 µm. c-f, Representative images and Zip2R(2x) colocalization analysis of indicated regions of interest (ROI) with (c) cell membrane, (d) Lamp1, (e) Rab11, and (f) Rab5. g, Comparison of Zip2R(2x) and Zip2R(1x) colocalization with indicated subcellular markers (Membrane: N = 14, Lamp1, Rab11, Rab5: N = 4, *p = 0.0224 (membrane), *p = 0.0479 (Lamp1), mean ± SD, two-tailed t-test).

Source data

Extended Data Fig. 3 ZipR signaling is inhibited by ruxolitinib.

a, pSTAT5 expression in Zip2R(2x) transduced T cells treated with increasing concentrations of ruxolitinib. IC50 is indicated with a dashed line (N = 2). b, pSTAT5 expression in Zip2R(2x) transduced T cells at baseline or following 24-hour incubation with 5 µM ruxolitinib (N = 4, mean ± SD, ***p = 0.0005, two-tailed t-test). c, pSTAT5 expression in Zip7R(2x) transduced T cells treated with increasing concentrations of ruxolitinib. IC50 is indicated with a dashed line (N = 3, mean ± SD). d, pSTAT5 expression in Zip7R(2x) transduced T cells at baseline or following 24-hour incubation with 5 µM ruxolitinib (N = 5, mean ± SD, ***p < 0.001, two-tailed t-test). e, Viability of untreated or 5 µM ruxolitinib-treated T cells after 7 days of cytokine starvation as determined by flow cytometry (Viability dye/Annexin V) (N = 3, mean ± SD, ****p < 0.0001, one-way ANOVA with Tukey’s multiple comparisons test).

Source data

Extended Data Fig. 4 ZipRs augment CAR T cell antitumor activity against LM7 osteosarcoma without altering antigen specificity.

a,b, Cytotoxicity assay after 24-hour co-culture of LM7 WT (left) or LM7 B7H3 KO (right) cells with CAR T cells at indicated effector:target cell (E:T) ratios (N = 4 (a), N = 3 (b) biological replicates, mean ± SD, ****p < 0.0001, two-way ANOVA with Tukey’s multiple comparisons test). c, Cytokine production after 24-hour co-culture of LM7 WT (left) or LM7 B7-H3 KO (right) cells with CAR or CAR.Zip2R T cells at a 2:1 E:T measured by multiplex analysis (Left: CAR,CAR.Zip2R: N = 3, mean ± SD, ΔCAR, ΔCAR.Zip2R: N = 2; Right: N = 2, mean, two-way ANOVA with Tukey’s multiple comparisons test). d, Cytokine production after 24-hour co-culture of A549 WT (left) or LM7 WT (right) cells with CAR or CAR.Zip7R T cells at a 2:1 E:T measured by multiplex analysis (N = 2 biological replicates, mean). e, Number of stimulations in 7 day repeat stimulation assay with LM7 WT cells and CAR T cells at 2:1 E:T (N = 4, mean ± SD, *p = 0.0154, two-tailed t-test). f, Fold expansion of three representative donors used in repeat stimulation assays with LM7 WT cells. Data represented in (e). g, Repeat stimulation assay with LM7 B7-H3 KO cells and CAR T cells at 2:1 E:T (N = 4, mean ± SD). h, Stimulations of tumor cell killing in 7-day repeat stimulation assay with LM7 WT cells and CAR T cells at 2:1 E:T (N = 3, mean ± SD, *p = 0.0202, two-tailed t-test). i, Fold expansion of three representative donors used in repeat stimulation assays with LM7 WT cells. Data represented in (h). j, Repeat stimulation assay with LM7 B7-H3 KO cells and CAR T cells at 2:1 E:T (N = 3 biological replicates, mean ± SD).

Source data

Extended Data Fig. 5 Phenotypic analysis of B7-H3-CAR T cells following stimulation with A549 WT.

B7-H3-CAR+, CAR+Zip2R+, or CAR+Zip7R+ T cells were stimulated twice with A549 WT. CAR positive or CAR and ZipR positive T cells were quantified by flow cytometry pre and post 2nd stimulation (stim) and CD4 and CD8 positive T cells post 2nd stim (N = 2 donors). a, Percent CAR+ or CAR+ZipR+ cells pre and post 2nd stim (***p = 0.0005, two-tailed t-test). b, Percent CD4+ (left panel) or CD8+ (right panel) cells post 2nd stimulation (**p = 0.001, ***p = 0.0003, two-tailed t-test).

Source data

Extended Data Fig. 6 Zip2R and Zip7R augment EphA2-CAR T cell antitumor activity in vitro.

a, Transduction efficiency of EphA2-CAR, EphA2-CAR.Zip2R, and EphA2-CAR.Zip7R T cells (N = 3, mean ± SD). b, Frequency of CD4+ and CD8+ T cells transduced with indicated constructs (N = 3, mean ± SD). c, Immunophenotype of CD4+ (left) or CD8+ (right) T cells with indicated constructs (TN-Like: CCR7+ CD45RA+, TEM: CCR7- CD45RA, TCM: CCR7+ CD45RA, TEMRA: CCR7 CD45RA+, N = 3, mean ± SD). d, Fold expansion of ΔCAR (left) or CAR (right) T cells stimulated with A673 cells every 7 days (N = 3, mean ± SD). e, Rounds of stimulation (left) and relative expansion compared to CAR (right) in repeat stimulation assays with A673 cells (N = 3, mean ± SD, **p = 0.0011, ****p < 0.0001, two-way ANOVA with Tukey’s multiple comparisons test).

Source data

Extended Data Fig. 7 Zip2R augments the antitumor activity of EphA2-CAR in the A673 model.

a, Experimental scheme of s.c. A673 model; mice received a single i.v. dose of 1x106 CAR T cells on day 7 post tumor cell injection. b, Tumor volume of mice treated with indicated constructs (N = 4; donor 1). c, Kaplan-Meier survival curve (*p = 0.0438, log-rank test). d, Tumor volume of mice treated with indicated constructs (N = 5; donor 2). e, Tumor volume following rechallenge (dashed line) with A673 WT cells on the contralateral flank (N = 5 (CAR), N = 4 (CAR.Zip2R); tumor rejection: CAR: 0/5; CAR.Zip2R: 2/4). f, Kaplan Meier survival following rechallenge.

Source data

Extended Data Fig. 8 Analysis of B7H3-CAR.Zip7R T cell toxicity in vivo.

a-d, Mice received a single i.v. dose of 3x105 B7-H3.CAR.Zip7R.ffLuc T cells on day 7 post A549 cell injection; non-tumor bearing mice served as a control (N = 5 per group). (a) Serial bioluminescence images. b, Quantification of bioluminescence. c, Kaplan-Meier survival (N = 5). d, Representative IHC of lung in non-tumor bearing mice treated with CAR.Zip7R T cells at day 48 post T cell injection. e, Kaplan-Meier survival of A549-tumor bearing mice post i.v. injection of 3x105 B7-H3-CAR.Zip7R control (AAVS1ko) or T cell receptor (TRACko) KO T cells (N = 5). f, A549 bearing mice received CAR.Zip7R.ffLuc T cells and on day 7 ruxolitinib was started (shaded area) in ½ of the mice (untreated: n = 5; treated: n = 5). Quantification of CAR.Zip7R.ffLuc T cell bioluminescence in untreated or ruxolitinib chow-treated mice. g, Fold expansion on day 7 post start of ruxolitinb treatment (N = 4 (untreated), N = 5 (Ruxolitinib), mean ± SD, *p = 0.0159, two-tailed Mann-Whitney U test). h, Kaplan-Meier survival (N = 4 (untreated), N = 5 (Ruxolitinib), **p = 0.0027, log rank test).

Source data

Extended Data Fig. 9 Immunophenotype and antigen specificity of CAR.Zip21R or CAR.Zip12R T cells.

a, Frequency of CD4+ and CD8+ CAR and CAR.Zip21R T cells (N = 4, mean ± SD). b, Immunophenotype of CD4+ (left) or CD8+ (right) T cells ((TN-Like: CCR7+ CD45RA+, TEM: CCR7- CD45RA, TCM: CCR7+ CD45RA, TEMRA: CCR7 CD45RA+, N = 4, mean ± SD). c, Frequency of CD4+ and CD8+ CAR and CAR.Zip12R T cells (N = 3 (CAR), N = 2 (ΔCAR), mean ± SD). d, Representative flow cytometry plots. e, Immunophenotype of CD4+ (left) and CD8+ (right) T cells (N = 3 (CAR, CAR.Zip12R), N = 2 (ΔCAR, ΔCAR.Zip12R), mean ± SD, **p = 0.0031, *p = 0.0238 (TN-Like), *p = 0.0265 (TEMRA), two-way ANOVA with Tukey’s multiple comparisons test). f, Transduction efficiency of ΔCAR and ΔCAR.Zip12R T cells (N = 2, mean). g, Fold expansion of ΔCAR and ΔCAR.Zip12R T cells stimulated with A549 WT cells every seven days (N = 2, mean).

Source data

Extended Data Fig. 10 Transcriptomic analysis of CD4+ CAR.ZipR T cells by scRNAseq.

a, GSEA of unstimulated or stimulated CD4+ CAR and CAR.ZipR T cell populations. b, Expression of memory, effector/cytotoxicity, inhibition/exhaustion, and activation markers in unstimulated or stimulated CD4+ CAR and CAR.ZipR T cell populations. c, Expression of selected genes in unstimulated or stimulated CD4+ CAR and CAR.ZipR T cell populations (Wilcoxon rank sum test with Bonferroni correction; ****adjusted p value < 0.0001 and log2 FC > 0.5 or < −0.5).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–15.

Reporting Summary

Peer Review File

Supplementary dataset 1

Phosphoproteomics dataset.

Supplementary dataset 2

Zip receptor sequences.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bell, M., Lange, S., Sejdiu, B.I. et al. Modular chimeric cytokine receptors with leucine zippers enhance the antitumour activity of CAR T cells via JAK/STAT signalling. Nat. Biomed. Eng (2023). https://doi.org/10.1038/s41551-023-01143-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-023-01143-w

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research