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:

Single-cell mapping of combinatorial target antigens for CAR switches using logic gates

Abstract

Identification of optimal target antigens that distinguish cancer cells from normal surrounding tissue cells remains a key challenge in chimeric antigen receptor (CAR) cell therapy for tumors with intratumoral heterogeneity. In this study, we dissected tissue complexity to the level of individual cells through the construction of a single-cell expression atlas that integrates ~1.4 million tumor, tumor-infiltrating normal and reference normal cells from 412 tumors and 12 normal organs. We used a two-step screening method using random forest and convolutional neural networks to select gene pairs that contribute most to discrimination between individual malignant and normal cells. Tumor coverage and specificity are evaluated for the AND, OR and NOT logic gates based on the combinatorial expression pattern of the pairing genes across individual single cells. Single-cell transcriptome-coupled epitope profiling validates the AND, OR and NOT switch targets identified in ovarian cancer and colorectal cancer.

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: Construction and characterization of tumor-normal single-cell meta-atlas.
Fig. 2: Characterization of single-antigen identification results.
Fig. 3: Characterization of combinatorial antigen identification results.
Fig. 4: Evaluation of selected combinatorial antigens in comparison with previous targets.
Fig. 5: Single-cell epitope analysis of validation targets in ovarian cancer.
Fig. 6: Immunohistochemical staining of CLDN3 and CLDN4 in colon, ovarian, lung and breast cancer.

Similar content being viewed by others

Data availability

The list of cell surface proteins was obtained from the in silico human surfaceome database at http://wlab.ethz.ch/surfaceome/ (ref. 23). The tumor-normal single-cell meta-atlas is available at https://cellatlas.kaist.ac.kr/cart. Raw and processed datasets used in this study are deposited at the Zenodo repository (https://doi.org/10.5281/zenodo.7416669)50. Ovarian cancer single-cell transcriptome data generated in this work are available on the Gene Expression Omnibus with accession number GSE192898 (ref. 51). Source data are provided with this paper.

Code availability

Source codes for CAR target antigen identification, named PCASA (prioritization of combinatorial cancer-associated surface antigens), are available on GitHub at https://github.com/kaistomics/PCASA (ref. 52).

References

  1. Sterner, R. C. & Sterner, R. M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11, 69 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Marofi, F. et al. CAR T cells in solid tumors: challenges and opportunities. Stem Cell Res. Ther. 12, 81 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Taylor, A. M. et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Fan, J. et al. Linking transcriptional and genetic tumor heterogeneity through allele analysis of single-cell RNA-seq data. Genome Res. 28, 1217–1227 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Patel, A. P. et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 344, 1396–1401 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gao, R. et al. Delineating copy number and clonal substructure in human tumors from single-cell transcriptomes. Nat. Biotechnol. 39, 599–608 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pasquini, G., Rojo Arias, J. E., Schäfer, P. & Busskamp, V. Automated methods for cell type annotation on scRNA-seq data. Comput. Struct. Biotechnol. J. 19, 961–969 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Parker, K. R. et al. Single-cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies. Cell 183, 126–142 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jing, Y. et al. Expression of chimeric antigen receptor therapy targets detected by single-cell sequencing of normal cells may contribute to off-tumor toxicity. Cancer Cell 39, 1558–1559 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. Wilkie, S. et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J. Clin. Immunol. 32, 1059–1070 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Hege, K. M. et al. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J. Immunother. Cancer 5, 22 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Yang, Y. et al. Bispecific CAR T cells against EpCAM and inducible ICAM-1 overcome antigen heterogeneity and generate superior antitumor responses. Cancer Immunol. Res. 9, 1158–1174 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Davies, D. M. & Maher, J. Gated chimeric antigen receptor T-cells: the next logical step in reducing toxicity? Transl. Cancer Res. 5, S61–S65 (2016).

    Article  CAS  Google Scholar 

  14. Cho, J. H., Collins, J. J. & Wong, W. W. Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell 173, 1426–1438 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Han, X., Wang, Y., Wei, J. & Han, W. Multi-antigen-targeted chimeric antigen receptor T cells for cancer therapy. J. Hematol. Oncol. 12, 128 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Hu, Z. et al. The cancer surfaceome atlas integrates genomic, functional and drug response data to identify actionable targets. Nat. Cancer 2, 1406–1422 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Orentas, R. J. et al. Paired expression analysis of tumor cell surface antigens. Front .Oncol. 7, 173 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. MacKay, M. et al. The therapeutic landscape for cells engineered with chimeric antigen receptors. Nat. Biotechnol. 38, 233–244 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Dannenfelser, R. et al. Discriminatory power of combinatorial antigen recognition in cancer T cell therapies. Cell Syst. 11, 215–228 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hie, B., Cho, H., DeMeo, B., Bryson, B. & Berger, B. Geometric sketching compactly summarizes the single-cell transcriptomic landscape. Cell Syst 8, 483–493 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Park, J. E. et al. A cell atlas of human thymic development defines T cell repertoire formation. Science 367, eaay3224 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bausch-Fluck, D. et al. The in silico human surfaceome. Proc. Natl Acad. Sci. USA 115, E10988–E10997 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Selvaraju, R. R. et al. Grad-CAM: visual explanations from deep networks via gradient-based localization. Proc. IEEE International Conference on Computer Vision https://ieeexplore.ieee.org/document/8237336 (2017).

  25. Chattopadhay, A., Sarkar, A., Howlader, P. & Balasubramanian, V. N. Grad-CAM++: generalized gradient-based visual explanations for deep convolutional networks. in: 2018 IEEE Winter Conference on Applications of Computer Vision (WACV). https://doi.org/10.1109/WACV.2018.00097 (2018).

  26. Cancer Genome Atlas Research Networket al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 45, 1113–1120 (2013).

    Article  PubMed Central  Google Scholar 

  27. Lonsdale, J. et al. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

    Article  CAS  Google Scholar 

  28. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nakamura, K. et al. CD24 expression is a marker for predicting clinical outcome and regulates the epithelial-mesenchymal transition in ovarian cancer via both the Akt and ERK pathways. Oncol. Rep. 37, 3189–3200 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yang, S. et al. Decontamination of ambient RNA in single-cell RNA-seq with DecontX. Genome Biol. 21, 57 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Tiernan, J. P. et al. Carcinoembryonic antigen is the preferred biomarker for in vivo colorectal cancer targeting. Br. J. Cancer 108, 662–667 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, J. et al. Carbonic anhydrase IV inhibits colon cancer development by inhibiting the Wnt signalling pathway through targeting the WTAP–WT1–TBL1 axis. Gut 65, 1482–1493 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Kharchenko, P. V., Silberstein, L. & Scadden, D. T. Bayesian approach to single-cell differential expression analysis. Nat. Methods 11, 740–742 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Qiu, P. Embracing the dropouts in single-cell RNA-seq analysis. Nat. Commun. 11, 1169 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. T, S. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  Google Scholar 

  37. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Martens, J. H. A. & Stunnenberg, H. G. BLUEPRINT: mapping human blood cell epigenomes. Haematologica 98, 1487–1489 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Davis, C. A. et al. The Encyclopedia of DNA Elements (ENCODE): data portal update. Nucleic Acids Res. 46, D794–D801 (2018).

    Article  CAS  PubMed  Google Scholar 

  40. Mabbott, N. A., Baillie, J. K., Brown, H., Freeman, T. C. & Hume, D. A. An expression atlas of human primary cells: inference of gene function from coexpression networks. BMC Genomics 14, 632 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Karlsson, M. et al. A single-cell type transcriptomics map of human tissues. Sci. Adv. 7, eabh2169 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Polański, K. et al. BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics 36, 964–965 (2020).

    Article  PubMed  Google Scholar 

  43. Tweedie, S. et al. Genenames.org: the HGNC and VGNC resources in 2021. Nucleic Acids Res. 49, D939–D946 (2021).

    Article  CAS  PubMed  Google Scholar 

  44. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580 (2013).

    Article  Google Scholar 

  45. Jiang, L. et al. A quantitative proteome map of the human body. Cell 183, 269–283 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Breiman, L. Random forests. Machine Learning 45, 5–32 (2001).

    Article  Google Scholar 

  47. Sing, T., Sander, O., Beerenwinkel, N. & Lengauer, T. ROCR: visualizing classifier performance in R. Bioinformatics 21, 3940–3941 (2005).

    Article  CAS  PubMed  Google Scholar 

  48. Abadi, M. et al. TensorFlow: a system for large-scale machine learning. Proc. 12th USENIX Symposium on Operating Systems Design and Implementation. https://www.usenix.org/system/files/conference/osdi16/osdi16-abadi.pdf (2016).

  49. Kingma, D. P. & Ba, J. Adam: a method for stochastic optimization. Proc. 3rd International Conference on Learning Representations. https://doi.org/10.48550/arXiv.1412.6980 (2014).

  50. Kwon, J. Integrative single-cell mapping of combinatorial target antigens for logical CAR switches. https://doi.org/10.5281/zenodo.7416669 (2022).

  51. An, H. J. Single cell 5′ RNA sequencing of 9 Korean ovarian cancer patients. Gene Expression Omnibus. https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?&acc=gse192898 (2023).

  52. omics lab @ KAIST. kaistomics/PCASA: PCASA. https://doi.org/10.5281/zenodo.7514043 (2023).

Download references

Acknowledgements

This work was supported by the Bio & Medical Technology Development Program of the National Research Foundation, funded by the Ministry of Science & ICT (NRF-2017M3A9A7050612, NRF-2019M3A9B6064688, NRF-2019M3A9B6064691 and NRF-2021M3A9I402444711), and also supported by the Korea Health Technology R&D Project of the Korea Health Industry Development Institute, funded by the Ministry of Health & Welfare (HI16C1559).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: J. Kwon, J. Kang, J.E.P. and J.K.C. Methodology: J. Kwon, J. Kang, A.J., K.S., D.A., M.Y.B., J.H.L., N.K., H.H.E., J.M.L. and W.Y.P. Investigation: J. Kwon, J. Kang, A.J., K.S., D.A., N.K. and H.H.E. Visualization: J. Kwon, J. Kang and M.Y.B. Funding acquisition: J.K.C. Project administration: H.J.A., J.E.P. and J.K.C. Supervision: S.H., H.J.A., H.O.L., J.E.P. and J.K.C. Writing—original draft: J. Kwon, J. Kang and J.H.L. Writing—review and editing: J. Kwon, J. Kang, J.E.P. and J.K.C.

Corresponding authors

Correspondence to Hee Jung An, Hae-Ock Lee, Jong-Eun Park or Jung Kyoon Choi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Raphael Gottardo, Peter Linsley and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 UMAP representation of the normal cell atlas overlaid with tumor cells and tumor-infiltrating normal cells from selected tumor datasets.

. a, Same plot as Fig. 1e but with cell-type annotation instead of tissue origin information. b, Same plot as Fig. 1e but showing representative cells selected during tumor cellpreserving subsampling by geometric sketching.

Extended Data Fig. 2 Tumor versus normal differences in ECFs and expression levels.

Differences of ECFs (a) and expression levels (b) between tumor and normal cellsfor surfaceome genes ordered by the FI values from the RF model in selected cancer types.

Extended Data Fig. 3 Comparison of cancer types with expression level and FI for surfaceome genes.

a, Clustering heatmap displaying the expression level of surface antigens according to the cancer type. Five major clusters (C1 ~ C5) of genes were denoted. b, Comparison of the different cancer types based on the correlation of the FI values from the cancer type-specific RF model.

Extended Data Fig. 4 Tumor-specific ECF values of subtype marker genes in BRCA samples for which clinical subtype information was available.

The boxplot shows the ECF status of three markers, containing ERBB2, ESR1, and PGR, for all subtypes (ERpositive: n = 11, HER2-positive: n = 8, HER2 and ER double positive: n = 4, TNBC: n = 12): TNBC, Triple-negative breast cancer; ‘+‘ means ‘positive’. Average values of ECFs for ERBB2, ESR1, and PGR, were 0.33 ± 0.15 (s.d.), 0.68 ± 0.15 (s.d), and 0.29 ± 0.18 (s.d.) in ER-positive subtype while HER2-positive subtype showed 0.86 ± 0.19 (s.d.), 0.18 ± 0.3 (s.d.), and 0.01 ± 0.01 (s.d.), respectively. In the case of HER2+ ER+, average values were 0.47 ± 0.19 (s.d.), 0.25 ± 0.23 (s.d.), and 0.06 ± 0.05 (s.d.), and the TNBC subtype showed 0.24 ± 0.14 (s.d), 0.05 ± 0.12 (s.d.), and 0.05 ± 0.15 (s.d.), for ERBB2, ESR1, and PGR, respectively. Data in the box plot represent the first quartile (25%), median (center), and third quartile (75%) with minimum and maximum values. Black points indicate the outliers.

Source data

Extended Data Fig. 5 Correlation between the CNN weight and ECF of each antigen combination in PAAD, LIHC, NSCLC, CRC, and BRCA.

For each of the pairs, the GradCAM weight returned by the CNN model was mapped to the ECF among tumor cells (blue) and normal cells (red). The Pearson correlation coefficient and two-sided P values are shown at the top. Translucent bands around the regression lines represent the confidence interval (CI) of 95%.

Source data

Extended Data Fig. 6 Comparison of tumor versus normal ECFs according to the CNN weight in PAAD, LIHC, NSCLC, CRC, and BRCA.

Tumor and normal ECFs of the combinations were ordered by their CNN weight. The combinations meeting the ECF criteria (> 70% tumor and < 10% normal) were highlighted in color.

Extended Data Fig. 7 Sample-wise co-expression of selected combinations in OV.

Expression correlations of the OV gene pairs were computed by using pan-cancer TCGA samples (n = 11,768) and GTEx normal samples (n = 17,382). The Pearson correlation coefficient was used.

Source data

Extended Data Fig. 8 Across-sample expression patterns of selected logical CAR switches in PAAD, LIHC, NSCLC, CRC, and BRCA.

Shown are the top 10 gene combinations with the largest CNN weight for each cancer type and each logic switch (AND, OR, and NOT). The heatmap intensity scales with the combinatorial tumor (left) and normal (right) ECF values.

Source data

Extended Data Fig. 9 Additional single-cell epitope analysis results for EPCAM and CD24 in ovarian cancer.

a, Distribution of EPCAM and CD24 protein expression levels broken down by annotated cell type. Log2 fold changes comparing CITE-seq ADT signals from targeting antibodies versus isotype controls were computed and are displayed in each cell type. b, Clustering and cell-type mapping of tumor and tumor-resident normal cells based on the CITE-seq transcriptome. c, Single-cell maps, as in (b), overlaid with mRNA expression levels retrieved from CITEseq. d, Single-cell maps, as in (b), overlaid with protein expression levels retrieved from CITE-seq. e, Single-cell maps, as in (b), overlaid with the mRNA expression status indicating cells with both EPCAM and CD24 (brown), only EPCAM (light blue), only CD24 (light red), or none (gray). f, Single-cell maps, as in (b), overlaid with the epitope expression status indicating cells with both EPCAM and CD24 (brown), only EPCAM (light blue), only CD24 (light red), or none (gray).

Extended Data Fig. 10 Single-cell epitope analysis of validation targets in colorectal cancer.

a, Clustering and cell-type mapping of the tumor and tumor-infiltrating normal cells based on the CITE-seq transcriptome of our pooled CRC samples. b, Single-cell maps, as in (a), overlaid with mRNA expression levels retrieved from the CITE-seq experiments. c, Single-cell maps, as in (b), overlaid with protein expression levels retrieved from the CITE-seq experiments. d, Distribution of protein expression levels broken down by annotated cell type. Log2 fold changes comparing CITE-seq ADT signals from targeting antibodies versus isotype controls were computed and are displayed for each protein in each cell type. e, Cell-by-cell protein expression patterns in the tumor and tumor-resident normal cells for CEACAM5 versus EPCAM, CEACAM5 versus CA4, CEACAM5 versus CPM, and CEACAM5 versus VSIG2. Cells enclosed in the marked zones indicate the expression of both proteins for the AND gating and only one protein (CEACAM5) for the NOT gating.

Supplementary information

Reporting Summary

Supplementary Tables 1–9

Supplementary Table 1: Summary of our cancer cell atlas. Supplementary Table 2: Status of cell types for tumor datasets integrated with normal cell atlas. Supplementary Table 3: Summary of selected tumor datasets of six cancer types. Supplementary Table 4: Performance of RF cell classifier for each cancer type. Supplementary Table 5: RF FI for top 100 genes for each cancer type. Supplementary Table 6: CNN weights calculated by Grad-CAM for each cancer type. Supplementary Table 7: Performance of CNN cell classifier for each cancer type. Supplementary Table 8: Sample-wise co-expression for OV target combinations. Supplementary Table 9: Normal ECF distribution by cell types for OV target combinations.

Source data

Source Data Fig. 2

Fig 2a: Sample-wise ECF and pseudo bulk expression level. Fig 2c,e and Extended Data Figs 2 and 3: ECF and expression level for each gene. Fig 2d: Sample-wise differentially expressed gene analysis result.

Source Data Fig. 3

Fig 3e,f: ECF of gene combinations for OV.

Source Data Fig. 4

Fig 4c: ECF for each gene combination of AND and NOT gates depending on the cell type in OV. Fig 4e: Sample-wise ECF status for each gene combination in OV.

Source Data Extended Data Fig./Table 4

ECF status of BRCA subtype-specific markers.

Source Data Extended Data Fig./Table 5

ECF of gene combinations for PAAD, LIHC, NSCLC, CRC and BRCA.

Source Data Extended Data Fig./Table 7

Co-expression for each gene combination for OV in GTEx and TCGA data.

Source Data Extended Data Fig./Table 8

Extended Data Fig 8: Sample-wise ECF status for each gene combination in PAAD, LIHC, NSCLC, CRC and BRCA.

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

Kwon, J., Kang, J., Jo, A. et al. Single-cell mapping of combinatorial target antigens for CAR switches using logic gates. Nat Biotechnol 41, 1593–1605 (2023). https://doi.org/10.1038/s41587-023-01686-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-023-01686-y

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer