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:

Multiplexed transcriptomic profiling of the fate of human CAR T cells in vivo via genetic barcoding with shielded small nucleotides

Abstract

The design of chimeric antigen receptor (CAR) T cells would benefit from knowledge of the fate of the cells in vivo. This requires the permanent labelling of CAR T cell products and their pooling in the same microenvironment. Here, we report a cell-barcoding method for the multiplexed longitudinal profiling of cells in vivo using single-cell RNA sequencing (scRNA-seq). The method, which we named shielded-small-nucleotide-based scRNA-seq (SSN-seq), is compatible with both 3′ and 5′ single-cell profiling, and enables the recording of cell identity, from cell infusion to isolation, by leveraging the ubiquitous Pol III U6 promoters to robustly express small-RNA barcodes modified with direct-capture sequences. By using SSN-seq to track the dynamics of the states of CAR T cells in a tumour-rechallenge mouse model of leukaemia, we found that a combination of cytokines and small-molecule inhibitors that are used in the ex vivo manufacturing of CAR T cells promotes the in vivo expansion of persistent populations of CD4+ memory T cells. By facilitating the probing of cell-state dynamics in vivo, SSN-seq may aid the development of adoptive cell therapies.

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: SSN group barcodes for multiplexed scRNA-seq.
Fig. 2: SSN-seq accurately demultiplexes cell subtypes, and identifies distinct CAR T exhaustion transcriptional programs.
Fig. 3: SSN-seq enables direct comparison of human CD19 CAR T cells manufactured ex vivo using various culture conditions.
Fig. 4: Longitudinal transcription profiling of CAR T cells in vivo using SSN-seq.
Fig. 5: Benchmarking SSN-seq in CAR T cells among different donors and across individual mice.
Fig. 6: SSN-seq is compatible with single-cell 5′ assays.

Similar content being viewed by others

Data availability

Raw and processed data of the scRNA-seq/SSN-seq experiments are available at the GEO under accession code GSE201647. Other public data used in this study include reference genomes for human (https://useast.ensembl.org, GRCh38) and RNA-seq/scRNA-seq datasets (GSE78220, GSE83978, GSE114631, GSE120575 and GSE150992). The raw and analysed datasets generated during the study are available for research purposes from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Wagner, A., Regev, A. & Yosef, N. Revealing the vectors of cellular identity with single-cell genomics. Nat. Biotechnol. 34, 1145–1160 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gehring, J., Hwee Park, J., Chen, S., Thomson, M. & Pachter, L. Highly multiplexed single-cell RNA-seq by DNA oligonucleotide tagging of cellular proteins. Nat. Biotechnol. 38, 35–38 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. McGinnis, C. S. et al. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices. Nat. Methods 16, 619–626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Stoeckius, M. et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jaitin, D. A. et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell 167, 1883–1896 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Datlinger, P. et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat. Methods 14, 297–301 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Biddy, B. A. et al. Single-cell mapping of lineage and identity in direct reprogramming. Nature 564, 219–224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Weinreb, C., Rodriguez-Fraticelli, A., Camargo, F. D. & Klein, A. M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science 367, eaaw3381 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Oren, Y. et al. Cycling cancer persister cells arise from lineages with distinct programs. Nature 596, 576–582 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yang, S. et al. Development of optimal bicistronic lentiviral vectors facilitates high-level TCR gene expression and robust tumor cell recognition. Gene Ther. 15, 1411–1423 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nowicki, T. S. et al. Epigenetic suppression of transgenic T-cell receptor expression via gamma-retroviral vector methylation in adoptive cell transfer therapy. Cancer Discov. 10, 1645–1653 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kong, W. et al. BET bromodomain protein inhibition reverses chimeric antigen receptor extinction and reinvigorates exhausted T cells in chronic lymphocytic leukemia. J. Clin. Invest. 131, e145459 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Mimitou, E. P. et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat. Methods 16, 409–412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Replogle, J. M. et al. Combinatorial single-cell CRISPR screens by direct guide RNA capture and targeted sequencing. Nat. Biotechnol. 38, 954–961 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wang, Z. et al. SETD5-coordinated chromatin reprogramming regulates adaptive resistance to targeted pancreatic cancer therapy. Cancer Cell 37, 834–849 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lanitis, E. et al. Optimized gene engineering of murine CAR-T cells reveals the beneficial effects of IL-15 coexpression. J. Exp. Med. 218, e20192203 (2021).

    Article  CAS  PubMed  Google Scholar 

  18. Schmidt, R. et al. CRISPR activation and interference screens decode stimulation responses in primary human T cells. Science 375, eabj4008 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Replogle, J. M. et al. Mapping information-rich genotype-phenotype landscapes with genome-scale perturb-seq. Cell 185, 2559–2575 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ma, H. et al. CRISPR-Cas9 nuclear dynamics and target recognition in living cells. J. Cell Biol. 214, 529–537 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. Roth, T. L. et al. Pooled knockin targeting for genome engineering of cellular immunotherapies. Cell 181, 728–744 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chew, W. L. et al. A multifunctional AAV-CRISPR-Cas9 and its host response. Nat. Methods 13, 868–874 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Charlesworth, C. T. et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat. Med. 25, 249–254 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Dubrot, J. et al. In vivo screens using a selective CRISPR antigen removal lentiviral vector system reveal immune dependencies in renal cell carcinoma. Immunity 54, 571–585 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Paul, C. P., Good, P. D., Winer, I. & Engelke, D. R. Effective expression of small interfering RNA in human cells. Nat. Biotechnol. 20, 505–508 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Shu, D., Shu, Y., Haque, F., Abdelmawla, S. & Guo, P. Thermodynamically stable RNA three-way junction for constructing multifunctional nanoparticles for delivery of therapeutics. Nat. Nanotechnol. 6, 658–667 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shu, D., Khisamutdinov, E. F., Zhang, L. & Guo, P. Programmable folding of fusion RNA in vivo and in vitro driven by pRNA 3WJ motif of phi29 DNA packaging motor. Nucleic Acids Res. 42, e10 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Filonov, G. S., Kam, C. W., Song, W. & Jaffrey, S. R. In-gel imaging of RNA processing using broccoli reveals optimal aptamer expression strategies. Chem. Biol. 22, 649–660 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Litke, J. L. & Jaffrey, S. R. Highly efficient expression of circular RNA aptamers in cells using autocatalytic transcripts. Nat. Biotechnol. 37, 667–675 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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 

  33. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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  CAS  PubMed  PubMed Central  Google Scholar 

  35. Blank, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zheng, C. et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Neelapu, S. S. et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 377, 2531–2544 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhao, Z. et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell 28, 415–428 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Majzner, R. G. et al. Tuning the antigen density requirement for CAR T-cell activity. Cancer Discov. 10, 702–723 (2020).

    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. 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  Google Scholar 

  42. Rafiq, S., Hackett, C. S. & Brentjens, R. J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020).

    Article  PubMed  Google Scholar 

  43. Sabatino, M. et al. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood 128, 519–528 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gattinoni, L. et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 15, 808–813 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kagoya, Y. et al. BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models. J. Clin. Invest. 126, 3479–3494 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Xu, Y. et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 123, 3750–3759 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Cieri, N. et al. IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 121, 573–584 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. van der Leun, A. M., Thommen, D. S. & Schumacher, T. N. CD8+ T cell states in human cancer: insights from single-cell analysis. Nat. Rev. Cancer 20, 218–232 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2018).

  50. Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science 370, 1328–1334 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sun, D. et al. Identifying phenotype-associated subpopulations by integrating bulk and single-cell sequencing data. Nat. Biotechnol. 40, 527–538 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Deng, Q. et al. Characteristics of anti-CD19 CAR T cell infusion products associated with efficacy and toxicity in patients with large B cell lymphomas. Nat. Med. 26, 1878–1887 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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 

  54. Good, C. R. et al. An NK-like CAR T cell transition in CAR T cell dysfunction. Cell 184, 6081–6100 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zheng, L. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021).

    Article  PubMed  Google Scholar 

  58. Hugo, W. et al. Genomic and transcriptomic features of response to Anti-PD-1 therapy in metastatic melanoma. Cell 165, 35–44 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1CD8+ tumor-infiltrating T cells. Immunity 50, 181–194 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Xia, Y. et al. BCL6-dependent TCF-1+ progenitor cells maintain effector and helper CD4+ T cell responses to persistent antigen. Immunity 55, 1200–1215 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Oh, D. Y. et al. Intratumoral CD4+ T cells mediate anti-tumor cytotoxicity in human bladder cancer. Cell 181, 1612–1625 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Heaton, H. et al. Souporcell: robust clustering of single-cell RNA-seq data by genotype without reference genotypes. Nat. Methods 17, 615–620 (2020).

    Article  CAS  PubMed  Google Scholar 

  66. Kang, H. M. et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat. Biotechnol. 36, 89–94 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Young, R. M., Engel, N. W., Uslu, U., Wellhausen, N. & June, C. H. Next-generation CAR T-cell therapies. Cancer Discov. 12, 1625–1633 (2022).

  68. Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Fraietta, J. A. et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 24, 563–571 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Belk, J. A. et al. Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Cancer Cell 40, 768–786 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Chang, M. T. et al. Identifying transcriptional programs underlying cancer drug response with TraCe-seq. Nat. Biotechnol. 40, 86–93 (2022).

    Article  CAS  PubMed  Google Scholar 

  72. Jin, X. et al. In vivo perturb-seq reveals neuronal and glial abnormalities associated with autism risk genes. Science 370, eaaz6063 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Dixit, A. et al. Perturb-Seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Bertrand, E. et al. The expression cassette determines the functional activity of ribozymes in mammalian cells by controlling their intracellular localization. RNA 3, 75–88 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Ma, S. et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Chen, S., Lake, B. B. & Zhang, K. High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell. Nat. Biotechnol. 37, 1452–1457 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Rubin, A. J. et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell 176, 361–376 (2019).

    Article  CAS  PubMed  Google Scholar 

  78. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Yeo, N. C. et al. An enhanced CRISPR repressor for targeted mammalian gene regulation. Nat. Methods 15, 611–616 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Lu, X. et al. Fine-tuned and cell-cycle-restricted expression of fusogenic protein syncytin-2 maintains functional placental syncytia. Cell Rep. 21, 1150–1159 (2017).

    Article  CAS  PubMed  Google Scholar 

  81. Morsut, L. et al. Engineering customized cell sensing and response behaviors using synthetic notch receptors. Cell 164, 780–791 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Marsh, S., Salmon, M. & Hoffman, P. samuel-marsh/scCustomize: version 1.1.1. Zenodo https://doi.org/10.5281/zenodo.7534950 (2023).

  86. McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell rna sequencing data using artificial nearest neighbors. Cell Syst. 8, 329–337 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Steinhart, Z. Code repository for CRISPR activation and interference screens decode stimulation responses in primary human T cells. Zenodo https://doi.org/10.5281/zenodo.5784651 (2022).

  88. 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 

  89. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res 9, 47 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank N. E. Navin for discussions; D. P. Pollock for technical assistance with scRNA-seq; and E. J. Thompson, Y. Zhu and Y. Chen for their help with scRNA-seq. scRNA-seq was performed at the University of Texas MD Anderson Cancer Center Advanced Technology Genomics Core (ATGC) Facility, which is supported in part by a Core Grant CA016672 (ATGC) and a NIH (1S10OD024977-01) award. Part of the figures were generated using BioRender. This work was supported in part by grants from the NIH to P.K.M. (R01CA236949, R01CA236118, R01CA278940, R01CA272843 and R01CA272844), CPRIT Individual Investigator Research Award (RP220391) and CPRIT Scholar in Cancer Research (RR160078), and the DoD PRCRP Career Development Award (CA181486). X.L. was supported by the CPRIT Training Award (RP210028). Y.Z. was supported by the Prostate Cancer Foundation Young Investigator Award, the CPRIT Training Award (RP210028), Odyssey fellowship by the Theodore N. Law Scientific Achievement Fund and Odyssey Program at The University of Texas MD Anderson Cancer Center.

Author information

Authors and Affiliations

Authors

Contributions

X.L. and P.K.M. conceived the project with input from S.M.L. X.L. designed and validated the SSNs and related constructs. X.L. performed all of the experiments, including proof-of-concept scRNA-seq, SSN optimization, CAR T generation and the corresponding SSN-seq. X.L. performed most of the computational data processing and bioinformatic analysis. S.M.L and Y.Z. assisted with data analysis. X.L. and P.K.M. wrote the manuscript, with input from all of the authors.

Corresponding author

Correspondence to Pawel K. Mazur.

Ethics declarations

Competing interests

X.L., S.M.L. and P.K.M. have filed a patent application related to SSN-seq. P.K.M. is a scientific consultant and stockholder of Amplified Medicines, Ikena Oncology and Alternative Bio. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Nicholas Haradhvala, Theodore Roth 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 Expression of standard direct capture sgRNA (STD.guide) as genetic barcodes yield low deconvolution rates across multiple cell lines.

a, Schematic overview of the multiplexed scRNA-seq experiment using standard direct-capture compatible sgRNA (STD.guide) as group barcode carrier. Cells in each experimental group were lentivirally transduced to express unique sgRNAs (denoted as sgRNA-1 to -5) and a cell surface marker NGFR. b, Quantification of indicated parameters in the multiplexed scRNA-seq experiments (as in (a)). sgRNA assignment rates for each cell type are shown. Lentivirus transduction efficiency for each group was determined by a cell surface marker NGFR+ expression using flow cytometry. c, Cell doublets prediction analysis (by DoubletFinder) of sgRNA-1 (human primary T cells) group indicates that the majority of misassigned cells are formed by transcriptionally distinct (heterotypic) cell doublets. d, Violin plots of mouse PDL1 expression levels in control KPC cells (KPC_WT) and PDL1-overexpressing KPC cells (KPC_PDL1). e, DEGs in control KPC (KPC_WT) and PDL1-overexpressing KPC cells (KPC_PDL1). Adjusted P values were calculated by two-sided Wilcoxon rank-sum test with Bonferroni’s correction. Red dots indicate significant genes with adjusted P-values < 0.05 and Log2(fold change) > 0.25.

Extended Data Fig. 2 Benchmarking performance of sgRNA- and F30-based SSN barcode designs across multiple cell types.

a, Schematics of all sgRNA- and F30-based SSN barcode designs tested in K562 cells. The control groups G01_STD.guide_mock and G02_STD.guide_Cas9 were transduced with lentiviral vectors expressing standard (without SSN) sgRNA (STD.guide) ± dCas9, respectively. G03_SSN.guide_A group expresses sgRNA-based SSN (SSN.guide) with 20 nt group barcode (GBC) whereas G04-G07 groups have 8 nt GBC. Cells in groups G08-G09 express SSN.guide cassette in the reverse orientation. G10_SSN.F30_A, cells express F30-based SSN barcode (SSN.F30) where the capture sequence is inserted into the Arm1 and in G11-G12 groups the capture sequence is inserted into the Arm2. G04-G07, G08-G09 or G11-G12 express GBC with different nucleotide sequences to assess the effect of barcode sequence on performance within the same barcode groups. b, The Pearson correlation analysis of transcription profiles between the indicated SSN-barcoded groups indicates that within the same cell type there are no signs of transcriptome perturbation due to the expression of different SSN barcodes. Of note, human HA.GD2.28z CAR T cells group (G20) exhibits a lower correlation with CD19.28z CAR T cells (G19) or primary human T cell groups (G17-G18) consistent with the observed enhanced exhaustion phenotype (see Fig. 2b). Group combining all identified cell multiples exhibits high transcriptome correlation with K562 groups suggesting that K562 cells are main contributors. The ‘not assigned’ group is highly correlated with human primary T cells congruent with the observed overall lower SSN-assignment rate (see Fig. 2e). c, Quantification of RNA content in each cell type as determined by the number of total RNA UMI counts per cell. Analysis for mouse KPC, human AsPC-1, mouse primary T and human primary T cells (as in Fig. 1c) and human K562, human AsPC-1, mouse EL4 and human primary T cells (as in Fig. 2d). The number above each violin plot indicates the median number of total RNA UMI counts. Note that primary T cells have significantly lower total RNA UMI counts per cell.

Extended Data Fig. 3 Quality control of SSN barcode sequencing library preparation using standard and optimized (enrichSSN) protocol.

a, Comparison of two strategies for SSN library preparation (the same cDNA input of SSN.guide-labelled human primary T cells). Schematics of the standard and optimized (enrichSSN) approach for library preparation with 10x Genomics Feature PCR or optimized enrichSSN amplification and the final library construction. The final SSN-seq libraries quality control was performed using Agilent Bioanalyzer. DNA electropherograms of the final libraries with indicated (arrows) expected product sizes: ~310 bp with 10x Feature PCR or ~280 bp with enrichSSN amplification (due to the omission of the TSO sequence). CBC, cell barcode. UMI, unique molecular identifier. CS1, 10x Capture Sequence 1. GBC, group barcode. TSO, template switch oligo. The TSO adds a common 5′ sequence to full-length cDNA after reverse transcription. b, Quantification of SSN UMI counts per cell and SSN assignment rates using standard Feature PCR or enrichSSN amplification for the final library preparation. Total sequencing reads and sequencing saturation were shown. Higher sequencing saturation indicates that a larger fraction of the library complexity has been captured. To enable fair comparison, only cells bearing the same cell barcodes were included. The enrichSSN strategies generated higher SSN UMI counts per cell. The center line indicates the median and the box marks the 25th and 75th percentiles. The whiskers extend to the smallest/largest value no further than 1.5 times the interquartile range. Outliers beyond the whisker ends are not plotted. The number to the right of each violin plot indicates the median number of UMIs counts. The enrichSSN amplification led to a higher barcode assignment rate compared to the standard PCR protocol.

Extended Data Fig. 4 The treatment with specific pathway inhibitors during the ex vivo manufacturing process modulates transcriptional profiles of the final CAR T cell infusion product.

a, Number of CAR T cells identified by SSN.guide and/or SSN.F30 barcoding (88% and 84%, respectively) indicate similar performances of both barcoding strategies. b, Expression analysis of selected markers of T cell effector (GZMB, GZMK, NKG7) and memory (CCR7, SELL, TCF7) functions for each CAR T cell treatment group (as in Fig. 3f). c, Distribution of CAR T cells from each treatment group in the identified gene clusters as in Fig. 3g based on SSN-seq analyses of pooled CAR T infusion product. The dot plots on the right indicate the corresponding expression of CD8A or CD4 in each cluster. The four CD8+ clusters (C01-C04) and the four CD4+ clusters (C07-C10) include approximately 91% of analysed cells. Of note, clusters C01+C07 contain mainly TWS119-treated, C03+C09 JQ1-treated, C02+C08 are dominated by Combo-treated and C04+C10 are mainly contributed by Mock control groups. d, Identification of active gene signatures in in vivo CAR T cell clusters using AUCell algorithm. AUCell analysis of curated gene sets from the MSigDB Immunologic Signatures database: genes down-regulated in naïve vs. effector CD8 T cells (GSE10239) and naïve vs. activated CD4 T cells (GSE28726) identified significant downregulation of naïve and elevation of activation/effector T cell signatures in clusters C04 and C10 containing mostly the mock-treated cells.

Extended Data Fig. 5 Conditions of ex vivo manufacturing impact long-term in vivo CAR T cells expansion and the ratio of CD4+ and CD8+ CAR T cells in NALM6 leukaemia mouse model.

a, Quantification of the in vivo expansion advantage of each CAR T cell treatment group normalized to Mock_IL2. The fold change is corrected by their input cell proportion (calculated by the distribution in the infusion product). b-c, Enrichment analysis (Fisher′s exact test) of each treatment group within the indicated CD4+ or CD8+ populations of CAR T cells in vivo (b) or the infusion product (c). The odds ratio indicates the ratio of two sets of odds: the odds of a treatment group presenting in one cluster shown in Fig. 4e versus the odds of the treatment group presenting in the remaining cells of all other clusters. Colour of the dots indicates the Log2-transformed odds ratio (blue: <0, underrepresented; red: >0, overrepresented). Dot size indicates the –Log10-transformed false discover rate (FDR) calculated using Benjamini-Hochberg correction. d, The CD8+/CD4+ composition changes in the CAR T infusion product (Day 0) versus persistent CAR T cells retrieved after in vivo tumour rechallenge (Day 42).

Extended Data Fig. 6 Culture conditions of ex vivo expansion impact long-term CAR T cells transcription profiles associated with cell lineage, exhaustion and anti-tumour activity in vivo.

a, Expression analysis of selected markers of T cell lineage (CD4, CD8A), memory (TCF7, CCR7) and effector (GZMB, GZMK, NKG7, PRF1) functions, chemokines (CXCL13), cytokines (IFNG), immune checkpoints (PDCD1, LAG3, HAVCR2, ENTPD1, TIGIT, CTLA4) and transcription factors (TOX, TOX2, EOMES, TBX21) for each cell cluster from the in vivo 8-plex CAR T cells experiment. b, Enrichment analysis (Fisher′ss exact test) of in vivo CAR T cells from each treatment group in the identified 12 phenotypic clusters. Colour of the dots indicates the Log2-transformed odds ratio (blue: <0, underrepresented; red: >0, overrepresented). Dot size indicates the –Log10-transformed false discover rate (FDR) calculated using Benjamini-Hochberg correction. c, The Scissor predicted anti-PD1 responses with inputs from four datasets56,58,59,60 are overlaid on UMAP visualization as in Fig. 4e. The dashed eclipse indicates cluster C01.

Supplementary information

Supplementary Information

Supplementary Figures and Notes, and descriptions of Supplementary Datasets 1–5.

Reporting Summary

Peer Review File

Supplementary Dataset 1

DEGs within each cluster (from Figs. 2h, 3f, 4f, 5d,f and 6e).

Supplementary Dataset 2

Summary of CAR T cell characteristics of the in vivo SSN-seq study in individual animals (from Fig. 5).

Supplementary Dataset 3

Annotated SSN-seq plasmid sequences.

Supplementary Dataset 4

Summary of quality metrics in all SSN-seq experiments.

Supplementary Dataset 5

PCR primers for SSN amplification.

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

Lu, X., Lofgren, S.M., Zhao, Y. et al. Multiplexed transcriptomic profiling of the fate of human CAR T cells in vivo via genetic barcoding with shielded small nucleotides. Nat. Biomed. Eng 7, 1170–1187 (2023). https://doi.org/10.1038/s41551-023-01085-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-023-01085-3

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