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:

ST3GAL1 and βII-spectrin pathways control CAR T cell migration to target tumors

Abstract

Adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells is becoming a promising treatment option for hematological malignancies. However, T cell immunotherapies have mostly failed in individuals with solid tumors. Here, with a CRISPR–Cas9 pooled library, we performed an in vivo targeted loss-of-function screen and identified ST3 β-galactoside α-2,3-sialyltransferase 1 (ST3GAL1) as a negative regulator of the cancer-specific migration of CAR T cells. Analysis of glycosylated proteins revealed that CD18 is a major effector of ST3GAL1 in activated CD8+ T cells. ST3GAL1-mediated glycosylation induces the spontaneous nonspecific tissue sequestration of T cells by altering lymphocyte function-associated antigen-1 (LFA-1) endocytic recycling. Engineered CAR T cells with enhanced expression of βII-spectrin, a central LFA-1-associated cytoskeleton molecule, reversed ST3GAL1-mediated nonspecific T cell migration and reduced tumor growth in mice by improving tumor-specific homing of CAR T cells. These findings identify the ST3GAL1–βII-spectrin axis as a major cell-intrinsic program for cancer-targeting CAR T cell migration and as a promising strategy for effective T cell 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: CAR T cells accumulate in the lungs immediately after adoptive transfer.
Fig. 2: A regulator of CAR T cell migration identified using a CRISPR screen.
Fig. 3: ST3GAL1 regulates LFA-1-mediated CAR T cell migration.
Fig. 4: ST3GAL1–βII-spectrin axis controls LFA-1-mediated T cell migration.
Fig. 5: βII-spectrin expression predicts response to CAR T cell therapy.

Similar content being viewed by others

Data availability

The Protein Data Bank accession code is 5E6U. Signaling pathway analysis was performed with Enrichr using the NCI-Nature 2016 database for pathway enrichment analysis (http://amp.pharm.mssm.edu/Enrichr). Sequencing datasets have been deposited in the Gene Expression Omnibus database under accession codes GSE227429 (CRISPR screen), GSE227509 (CD8+ T cell bulk RNA sequencing) and GSE227430 (CAR T cell bulk RNA sequencing). All other data are included in the article and Supplementary Information files. Source data are provided with this paper.

References

  1. Aghajanian, H., Rurik, J. G. & Epstein, J. A. CAR-based therapies: opportunities for immuno-medicine beyond cancer. Nat. Metab. 4, 163–169 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Lizee, G. et al. Harnessing the power of the immune system to target cancer. Annu. Rev. Med. 64, 71–90 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Bobisse, S. et al. Reprogramming T lymphocytes for melanoma adoptive immunotherapy by T-cell receptor gene transfer with lentiviral vectors. Cancer Res. 69, 9385–9394 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Junghans, R. P. The challenges of solid tumor for designer CAR-T therapies: a 25-year perspective. Cancer Gene Ther. 24, 89–99 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Hernandez-Lopez, A., Tellez-Gonzalez, M. A., Mondragon-Teran, P. & Meneses-Acosta, A. Chimeric antigen receptor-T cells: a pharmaceutical scope. Front. Pharm. 12, 720692 (2021).

    Article  CAS  Google Scholar 

  6. Fowell, D. J. & Kim, M. The spatio-temporal control of effector T cell migration. Nat. Rev. Immunol. 21, 582–596 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mueller, K. T. et al. Cellular kinetics of CTL019 in relapsed/refractory B-cell acute lymphoblastic leukemia and chronic lymphocytic leukemia. Blood 130, 2317–2325 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lee, D. W. et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 385, 517–528 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Morgan, R. A. et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843–851 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Finkle, D. et al. HER2-targeted therapy reduces incidence and progression of midlife mammary tumors in female murine mammary tumor virus huHER2-transgenic mice. Clin. Cancer Res. 10, 2499–2511 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Amitrano, A. M. et al. Optical control of CD8+ T cell metabolism and effector functions. Front. Immunol. 12, 666231 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Paley, M. A. et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. You, R. et al. Active surveillance characterizes human intratumoral T cell exhaustion. J. Clin. Investig. 131, e144353 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Donnadieu, E., Dupre, L., Pinho, L. G. & Cotta-de-Almeida, V. Surmounting the obstacles that impede effective CAR T cell trafficking to solid tumors. J. Leukoc. Biol. 108, 1067–1079 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Platt, R. J. et al. CRISPR–Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zimmermann, M. et al. CRISPR screens identify genomic ribonucleotides as a source of PARP-trapping lesions. Nature 559, 285–289 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hobbs, S. J. & Nolz, J. C. Regulation of T cell trafficking by enzymatic synthesis of O-glycans. Front. Immunol. 8, 600 (2017).

  18. Pietrobono, S. et al. ST3GAL1 is a target of the SOX2–GLI1 transcriptional complex and promotes melanoma metastasis through AXL. Nat. Commun. 11, 5865 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Priatel, J. J. et al. The ST3Gal-I sialyltransferase controls CD8+ T lymphocyte homeostasis by modulating O-glycan biosynthesis. Immunity 12, 273–283 (2000).

  20. Giovannone, N. et al. Human B cell differentiation is characterized by progressive remodeling of O-linked glycans. Front. Immunol. 9, 2857 (2018).

  21. Chen, S. et al. Increased SPC24 in prostatic diseases and diagnostic value of SPC24 and its interacting partners in prostate cancer. Exp. Ther. Med. 22, 923 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Geisler, C. & Jarvis, D. L. Effective glycoanalysis with Maackia amurensis lectins requires a clear understanding of their binding specificities. Glycobiology 21, 988–993 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sen, M. & Springer, T. A. Leukocyte integrin αLβ2 headpiece structures: the αI domain, the pocket for the internal ligand, and concerted movements of its loops. Proc. Natl Acad. Sci. USA 113, 2940–2945 (2016).

  24. Walling, B. L. & Kim, M. LFA-1 in T cell migration and differentiation. Front. Immunol. 9, 952 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Galkina, E. et al. Preferential migration of effector CD8+ T cells into the interstitium of the normal lung. J. Clin. Investig. 115, 3473–3483 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Walch, J. M. et al. Cognate antigen directs CD8+ T cell migration to vascularized transplants. J. Clin. Investig. 123, 2663–2671 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Shulman, Z. et al. Transendothelial migration of lymphocytes mediated by intraendothelial vesicle stores rather than by extracellular chemokine depots. Nat. Immunol. 13, 67–76 (2011).

    Article  PubMed  Google Scholar 

  28. Cyster, J. G. & Goodnow, C. C. Pertussis toxin inhibits migration of B and T lymphocytes into splenic white pulp cords. J. Exp. Med. 182, 581–586 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Surve, C. R., Lehmann, D. & Smrcka, A. V. A chemical biology approach demonstrates G protein βγ subunits are sufficient to mediate directional neutrophil chemotaxis. J. Biol. Chem. 289, 17791–17801 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kim, M., Carman, C. V. & Springer, T. A. Bidirectional transmembrane signaling by cytoplasmic domain separation in integrins. Science 301, 1720–1725 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Kim, M., Carman, C. V., Yang, W., Salas, A. & Springer, T. A. The primacy of affinity over clustering in regulation of adhesiveness of the integrin αLβ2. J. Cell Biol. 167, 1241–1253 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Feng, Y. et al. Exo1: a new chemical inhibitor of the exocytic pathway. Proc. Natl Acad. Sci. USA 100, 6469–6474 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Nishikimi, A. et al. Rab13 acts downstream of the kinase Mst1 to deliver the integrin LFA-1 to the cell surface for lymphocyte trafficking. Sci. Signal 7, ra72 (2014).

    Article  PubMed  Google Scholar 

  34. De Franceschi, N., Hamidi, H., Alanko, J., Sahgal, P. & Ivaska, J. Integrin traffic—the update. J. Cell Sci. 128, 839–852 (2015).

    PubMed  PubMed Central  Google Scholar 

  35. Meissner, J. M. et al. αII-Spectrin in T cells is involved in the regulation of cell–cell contact leading to immunological synapse formation? PLoS ONE 12, e0189545 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Capece, T. et al. A novel intracellular pool of LFA-1 is critical for asymmetric CD8+ T cell activation and differentiation. J. Cell Biol. 216, 3817–3829 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Thomsen, P., Roepstorff, K., Stahlhut, M. & van Deurs, B. Caveolae are highly immobile plasma membrane microdomains, which are not involved in constitutive endocytic trafficking. Mol. Biol. Cell 13, 238–250 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Fritzsche, M. & Charras, G. Dissecting protein reaction dynamics in living cells by fluorescence recovery after photobleaching. Nat. Protoc. 10, 660–680 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Lingel, H. et al. CTLA-4-mediated posttranslational modifications direct cytotoxic T-lymphocyte differentiation. Cell Death Differ. 24, 1739–1749 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Heslop, H. E. Safer CARS. Mol. Ther. 18, 661–662 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Beaty, B. T. et al. Talin regulates moesin-NHE-1 recruitment to invadopodia and promotes mammary tumor metastasis. J. Cell Biol. 205, 737–751 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Medina, E. et al. Crumbs interacts with moesin and βHeavy-spectrin in the apical membrane skeleton of Drosophila. J. Cell Biol. 158, 941–951 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gossez, M. et al. Evaluation of a novel automated volumetric flow cytometer for absolute CD4+ T lymphocyte quantitation. Cytometry B 92, 456–464 (2017).

  45. Potrzebowska, K., Lehtonen, J., Samuelsson, M. & Svensson, L. Flow cytometry assay for recycling of LFA-1 in T-lymphocytes. Bio Protoc. 8, e3104 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Morin, N. A. et al. Nonmuscle myosin heavy chain IIA mediates integrin LFA-1 de-adhesion during T lymphocyte migration. J. Exp. Med. 205, 195–205 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bern, M., Kil, Y. J. & Becker, C. Byonic: advanced peptide and protein identification software. Curr. Protoc. Bioinformatics 40, 13.20.11–13.20.14 (2012).

  48. Lim, K. et al. In situ neutrophil efferocytosis shapes T cell immunity to influenza infection. Nat. Immunol. 21, 1046–1057 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Looney, M. R. et al. Stabilized imaging of immune surveillance in the mouse lung. Nat. Methods 8, 91–96 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Susaki, E. A. et al. Advanced CUBIC protocols for whole-brain and whole-body clearing and imaging. Nat. Protoc. 10, 1709–1727 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Shifrut, E. et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell 175, 1958–1971 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Li, W. et al. Quality control, modeling, and visualization of CRISPR screens with MAGeCK-VISPR. Genome Biol. 16, 281 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the High Content Image Core (University of Rochester) for help with fluorescence imaging and use of the Dragonfly spinning-disc confocal. We thank Y. Gao and P. Rock for their technical assistance on the manuscript. We especially thank M. Jin (Weill Cornell) for the anti-HER2 CAR DNA construct and for his help with micro-PET/CT imaging, L. Gan (Augusta University) for help with βII-spectrin transgenic mouse generation, R. Burack and J. Moore for human sample collections and data review, J. Ashton, D. Ghoneim and J. Malik for help with CRISPR transcriptomic analyses and depositing RNA-sequencing data and all members of the Kim Laboratory for their comments during the course of these studies and input during preparation of the manuscript. We obtained hHER2 transgenic mice from Genentech. This study was funded by National Institutes of Health grant AI102851 (M.K. and P.W.O.), National Institutes of Health grant AI147362 (M.K. and R.E.W.), National Institutes of Health grant T32AI007285 (A.M.A. and C.J.S.), National Science and Technology Council in Taiwan MOST 111-2314-B-182A-069 and MOST 111-2321-B-182-002 (T.-C.F.) and K-GRC GO! KRICT project BSF23-113 (K.-D.K.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Y.H., H.-R.K., B.L.W. and M.K. methodology: Y.H., H.-R.K., B.L.W., W.S.S., M.K., J.R.L., A.M.A., C.J.S., K.L., R.K.M., K.-D.K., T.C., E.B.L., N.S.R., K.V., T.-C.F., A.L.-T.Y., P.W.O., R.E.W., C.-D.J., P.M.R. and S.A.G. investigation: Y.H., H.-R.K., B.L.W., W.S.S., J.R.L., A.M.A., C.J.S., K.L., K.-D.K., T.C., E.B.L., N.S.R., K.V. and T.-C.F. funding acquisition: A.M.A., P.W.O., M.K., R.E.W. and K.-D.K. supervision: M.K. writing, original draft: Y.H., H.-R.K., B.L.W. and M.K.

Corresponding author

Correspondence to Minsoo Kim.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Michael Sixt, Joy Burchell, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: N. Bernard, in collaboration with the Nature Immunology team.

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 hHER2 CAR-T cell assays and in vivo distribution.

a. Gating strategy for detection of hHER2-CAR-T cells. A median of > 90% of mouse CD8 T cells transduced with the hHER2-CAR retrovirus were positive for the transgene as judged by flow cytometry analysis. b. B16F10 cells were transfected with mammalian human HER2 construct with lipofectamine. Cells were grown in the presence of G418 and sorted twice to generate single cell clones. c. CAR-T-mediated B16-HER2 cell killing assay. Apoptotic cells were stained for Annexin V and analyzed by flow cytometry. d. Whole mouse lung before and after CUBIC clearing.

Extended Data Fig. 2 hHER2 CAR-T cell distribution.

A. Deep 3D imaging (1 mm thick) of CAR-T cell accumulation in the cleared mouse lung after 72 hr after i.v. injection (red; CD11c or MHCII, green; CAR-T cell). Graph shows colocalization of red signal versus GFP (CAR-T cells). Pearson coefficient was generated as Green (CAR-T cell) /Red (anti-CD11c or anti-MHCII). Data are presented as mean ± SEM; n = 3 mice/group. B. Flow cytometric analysis of OT-I T cells in the lung, blood, and tumor after i.v. injection (4, 24, 48, and 72 hr; mean ± SEM, n = 3 mice per group). C. PCA of differentially expressed genes in CAR-T cells (T72 vs. T0). D. Gating strategy for detection of hHER2-CAR-T cells from organs (lungs) E. Expression of Ki67+ in CAR-T cells isolated from the lung after i.v. injection. F. Relative expression of IFN-γ and TNF in CAR-T cells isolated from lung at indicated times after i.v. injection. Data are presented as mean ± SEM; n = 3 mice/group. G. Representative flow cytometry results show cell surface PD-1 and Tim-3 expression levels in OT-I naïve T cells, freshly activated in vitro (0 hr), or isolated from the lung after 72 hr post-injection. H. hHER2-CAR-T cells isolated from the lung or dLN after 72 hr i.v. injection were cocultured with B16-hHER2 for 24 hr. Dead B16 cells were stained with 7-AAD. The data reflect 3 independent experiments (the mean ± SEM, n = 3). The data were analyzed by two-tailed Student’s t test (*P = 0.002). I. Representative flow cytometry results showing cell surface PD-1 and Tim-3 expression levels in naïve and CAR-T cells isolated from the lung at the indicated times post-injection.

Source data

Extended Data Fig. 3 In vivo T cell CRISPR screen.

a. Volcano plot shows comparison of differentially expressed genes in naïve vs. in vitro activated CD8 T cells. Red denotes genes increased and blue denotes genes decreased in T cells. P values were adjusted using the Benjamini–Hochberg method. b. Heatmap of RNA-sequencing data from naïve vs. in vitro activated CD8 T cell samples (n = 3). Heatmap represents top 1,316 genes chosen from highest 0.25% p-value summary. c. Retroviral construct used for sgRNA delivery. d. A schematic of the experimental design showing timeline of T cell stimulation, transduction, enrichment, and flow analysis. T cells were purified by FACS sorting at day 7 and i.v injected into recipient tumor bearing mice. e. Scatter plot of the enrichment of candidates (n = 5 sgRNAs per gene) showing that sgRNAs targeting essential genes (left) were significantly reduced in the samples after two representative in vivo screens, but not the non-targeting guides (right). f. Genes with positive beta score (Tmigratory) and negative beta score (TSequestered) (>2 standard deviations from the mean (red)) are highlighted in yellow and blue, respectively.

Source data

Extended Data Fig. 4 Identification of CD18.

a. The spectrum of one of the CD18 peptides obtained by nanospray-ion trap tandem mass spectrometry. b. The deconvoluted MS/MS spectrum of tryptic peptide NVTR with 3 HexNAc. M: the parent ion of NVTR-HexNAc(3) at m/z 549.760 Da. M-18: loss one H2O in NVTR-HexNA(3). a2: the observed m/z 186.1218 Da matched to the a2 ion. y1: the observed m/z 175.1261 Da matched to the y1 ion. c. The crystal structure of LFA-1 headpiece (5E6U.pbd). The region encircled by dots from the left view is enlarged (right), representing the O-glycosylation on Thr256 in βI-domain.

Extended Data Fig. 5 Activated T cell migration in vitro.

a. Recruitment of CAR-T cells into dLN was blocked by LFA-1-blocking Ab and PTx treatments. Percentages of total transferred CD8 + T cells are shown. b & c. Activated CD8+ T cell migration on ICAM-1-coated plates ± CXCL12 (B) or CXCL10 (C) with varying concentrations as indicated. Data were collected from 3 independent experiments (n = 3, 11–20 individual cells per mouse). Data represent mean ± SEM. Statistical analyses were performed using one-way ANOVA with Bonferroni post-test. d. Representative Transwell assay of in vitro activated CD8+ T cell migration in response CXCL12 (2 µg/ml). Data were collected from 3 independent experiments (1 × 106 cells/well). Data represent mean ± SEM. Statistical analyses were performed using two-sided, unpaired Student’s t test. e. Schematic depicting the flow chamber assay. f & g. Activated CD8+ T cell migration on ICAM-1 coated plates with flow. Cells were treated with PTx (6 h) (F) and/or gallein (30 min) (G) ± CXCL12 (G) where indicated. Cells were allowed to settle for 10 minutes prior to 1 dyne/cm2 of fresh media being run across the surface of the plate. Data were collected from 3 independent experiments (n = 3, 25–45 individual cells per mouse). Data represent mean ± SEM. Statistical analyses were performed using one-way ANOVA with Bonferroni post-test. h. Activated CD8+ T cell migration on bEND.3 coated plates. bEND.3 cells were allowed to grow to a monolayer for at least 12 h prior to imaging. CD8+ were treated with PTx (6 h) where indicated. T cells were allowed to settle for 10 minutes prior to 1 dyne/cm2 of fresh media being run across the surface of the plate. Data were collected from 3 independent experiments (n = 3, 20–39 individual cells per mouse). Data represent mean ± SEM. Statistical analyses were performed using two-sided, unpaired Student’s t test.

Source data

Extended Data Fig. 6 TIRFM measurements of LFA-1 distribution.

a. Schematic of TIRFM measurements. Molecules near the surface fluoresce more brightly than molecules between microvilli and those farther from the substrate. b. Human CD8 T cells labeled for LFA-1 spreading on glass, BSA, or ICAM-1 coated substrate. Contrast and brightness have been adjusted for visibility, but the original gray values are indicated in the scale bars to the right of each image. All images in the same row are at the same magnification. Scale bars, 5 μm. Representative images from > five independent experiments are shown.

Extended Data Fig. 7 βII-spectrin expression in T cells.

a. Generation of GFP-ROSAβII-spectrin mouse model (Rosa26tm(CAG-LSL-Sptbn1-IRES-GFP)). Mouse Sptbn1 cDNA was inserted into the CAG-STOP-GFP-Rosa targeting vector, CTV, between a floxed Stop cassette and the internal ribosome entry site (IRES) followed by the enhanced Green Fluorescent Protein gene (eGFP). Transcription is under control of the CAG promoter. The targeting vector contained Rosa26 homology arms (1 kb 5′ and 3.8 kb 3′), so that the entire loxP-stop-loxP-Tmc2-IRES-GFP transcriptional cassette was inserted into the first intron of Rosa26 gene on chromosome 6. b & c. Flow cytometry analysis of IFNγ and TNF expression in hHER2-CAR transfected T cells from GFP-ROSAβII-spectrin mouse (TβII-spectrin) after treated with PBS or Tat–Cre Recombinase. Cells were co-cultured with B16-HER2 cells. Data represent mean ± SEM. n = 9. d. CAR-mediated B16-HER2 cell killing assay with TβII-spectrin cells after treated with PBS or Tat-Cre Recombinase. Cell death was stained for NucSpot. Data represent mean ± SEM. n = 9. e. Activated CD4+ T cell (OT-II) migration on ICAM-1 coated plates ± CXCL12. Cells were treated with PTx (6 hr) where indicated. Data were collected from 2 independent experiments (n = 2, 17–34 individual cells per mouse). Data represent mean ± SEM Statistical analyses were performed using one-way ANOVA with Bonferroni post-test. *P = 0.007. f. The pie charts depict the proportion of CD4 T cells distributed in the tumour, blood, LN/spleen, or lung/liver 72 h post-injection. g. Expression levels of βII-spectrin and St3gal1 in human CD4 and CD8 T cells (before and after activation). Loading control: β-actin. Representative western blot images from three independent experiments are shown. h. Expression levels of βII-spectrin in human CD8 memory T cells (CD8+CD45RO+CD45RACD56CD57). Loading control: β-actin. Representative western blot images from three independent experiments are shown.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

MS data.

Supplementary Table 2

LC–MS/MS analysis data.

Supplementary Table 3

CD19 CAR T cell participant information.

Supplementary Video 1

In vitro CAR T cell killing assay. WT CD8+ T cells or hHER2 CAR T cells were co-incubated with B16-hHER2 cells (green) in the presence of propidium iodide (red); scale bar, 20 μm.

Supplementary Video 2

In vivo CAR T cell migration. hHER2 CAR T cells (green) were intravenously injected by the tail vein of the B12-hHER2-bearing mouse, and intravital imaging was simultaneously performed to visualize T cells in the lung, LNs and tumor. The blood vessels were labeled with anti-CD31 (red). Time is marked as min:s; scale bars, 50 μm (parts 1, 3 and 4) and 20 μm (part 2).

Supplementary Video 3

CAR T cell accumulation. hHER2 CAR T cells (green) were intravenously injected by the tail vein of the B12-hHER2-bearing mouse, and intravital imaging was performed 48 and 72 h after infusion to visualize T cells in the lung. The blood vessels were labeled with anti-CD31 (red). Time is marked as min:s; scale bar, 50 μm.

Supplementary Video 4

CAR T cell accumulation. Deep 3D imaging (1-mm thick) of CAR T cell accumulation in a cleared mouse lung 72 h after intravenous injection (red, blood vessels (CD31); green, CAR T cells (GFP)).

Supplementary Video 5

In vitro CAR T cell migration to a tumor spheroid. Long-term time-lapse imaging (9 h) of hHER2 CAR T cells (red) embedded in 3D Matrigel around an hHER2-B16 (blue) spheroid ± PTx pretreatment; scale bars, 100 µm. Time is represented in h:min.

Supplementary Video 6

In vitro CD8+ T cell migration. Migration of naive and activated (day 5) mouse CD8+ T cells on ICAM-1 ± CXCL12-coated plates. Time is marked as min:s; scale bar, 50 μm.

Supplementary Video 7

In vitro CD8+ T cell migration. Migration of activated (day 5) mouse CD8+ T cells on ICAM-1-coated plates with (1 dyn cm–2) and without (0 dyn cm–2) flow; scale bar, 50 μm.

Supplementary Video 8

In vitro CD8+ T cell migration with βII-spectrin expression. Activated CD8+ T cells were transfected with GFP only (mock) or βII-spectrin cDNA and migrated on ICAM-1 ± CXCL12-coated plates. Time is marked as min:s; scale bar, 20 μm.

Supplementary Video 9

LFA-1-mediated CD8+ T cell spreading. TIRF microscopy images of activated CD11a–mYFP CD8+ T cell adhesion on ICAM-1 ± CXCL12-coated plates with and without βII-spectrin expression. LFA-1 intensity (YFP) is shown on a pseudocolor scale (from low (blue) to high (red)). Each frame is 15 s; scale bar, 10 μm.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

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. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data

Unprocessed western blots and gels.

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

Hong, Y., Walling, B.L., Kim, HR. et al. ST3GAL1 and βII-spectrin pathways control CAR T cell migration to target tumors. Nat Immunol 24, 1007–1019 (2023). https://doi.org/10.1038/s41590-023-01498-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01498-x

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