Log in

Map** variant effects on anti-tumor hallmarks of primary human T cells with base-editing screens

  • Article
  • Published:

From Nature Biotechnology

View current issue Submit your manuscript

Abstract

Single-nucleotide variants (SNVs) in key T cell genes can drive clinical pathologies and could be repurposed to improve cellular cancer immunotherapies. Here, we perform massively parallel base-editing screens to generate thousands of variants at gene loci annotated with known or potential clinical relevance. We discover a broad landscape of putative gain-of-function (GOF) and loss-of-function (LOF) mutations, including in PIK3CD and the gene encoding its regulatory subunit, PIK3R1, LCK, SOS1, AKT1 and RHOA. Base editing of PIK3CD and PIK3R1 variants in T cells with an engineered T cell receptor specific to a melanoma epitope or in different generations of CD19 chimeric antigen receptor (CAR) T cells demonstrates that discovered GOF variants, but not LOF or silent mutation controls, enhanced signaling, cytokine production and lysis of cognate melanoma and leukemia cell models, respectively. Additionally, we show that generations of CD19 CAR T cells engineered with PIK3CD GOF mutations demonstrate enhanced antigen-specific signaling, cytokine production and leukemia cell killing, including when benchmarked against other recent strategies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Experimental design and phenotypic readouts for massively parallel base-editor screens in T cells.
Fig. 2: Results of BE screens across donors.
Fig. 3: Structure–function analysis of PIK3CD and other gene variants on T cell signaling and activity.
Fig. 4: Enhanced signaling, polyfunctionality and melanoma cell-lytic capacity of epitope-specific T cells engineered with gene variants identified in BE screens.
Fig. 5: Enhanced signaling, cytokine production and leukemia cell-lytic capacity of CD19 CAR T cells equipped with PIK3CD or PIK3R1 variants identified in BE screens.

Similar content being viewed by others

Data availability

All processed screening data are provided as Supplementary Tables. Source data are provided with this paper.

Code availability

Code for generating in silico predicted structures is deposited here: https://github.com/gnikolenyi/izar_vis (ref. 73).

References

  1. Finck, A. V., Blanchard, T., Roselle, C. P., Golinelli, G. & June, C. H. Engineered cellular immunotherapies in cancer and beyond. Nat. Med. 28, 678–689 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Forget, M.-A. et al. Prospective analysis of adoptive TIL therapy in patients with metastatic melanoma: response, impact of anti-CTLA4, and biomarkers to predict clinical outcome. Clin. Cancer Res. 24, 4416–4428 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Rosenberg, S. A. & Restifo, N. P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ruella, M., Korell, F., Porazzi, P. & Maus, M. V. Mechanisms of resistance to chimeric antigen receptor-T cells in haematological malignancies. Nat. Rev. Drug Discov. 22, 976–995 (2023).

  5. Ho, P. et al. The CD58–CD2 axis is co-regulated with PD-L1 via CMTM6 and shapes anti-tumor immunity. Cancer Cell 41, 1207–1221 (2023).

    Article  CAS  PubMed  Google Scholar 

  6. 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 

  7. 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 

  8. Anderson, N. D. et al. Transcriptional signatures associated with persisting CD19 CAR-T cells in children with leukemia. Nat. Med. 29, 1700–1709 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. McGuirk, J. et al. A phase 1 dose escalation and cohort expansion study of the safety and efficacy of allogeneic CRISPR–Cas9-engineered T cells (CTX110) in patients (Pts) with relapsed or refractory (R/R) B-cell malignancies (CARBON). J. Clin. Oncol. 39, TPS7570 (2021).

  10. 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 

  11. Dai, X. et al. Massively parallel knock-in engineering of human T cells. Nat. Biotechnol. 41, 1239–1255 (2023).

    Article  CAS  PubMed  Google Scholar 

  12. Li, B. et al. Cis interactions between CD2 and its ligands on T cells are required for T cell activation. Sci. Immunol. 7, eabn6373 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. 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 

  14. Nahmad, A. D. et al. Frequent aneuploidy in primary human T cells after CRISPR–Cas9 cleavage. Nat. Biotechnol. 40, 1807–1813 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Patel, C. H. et al. TSC2 S1365A mutation potently regulates CD8+ T cell function and differentiation improving adoptive cellular cancer therapy. JCI Insight 8, e167829 (2023).

  16. Vang, T. et al. Autoimmune-associated lymphoid tyrosine phosphatase is a gain-of-function variant. Nat. Genet. 37, 1317–1319 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Pawlak-Adamska, E. et al. CD28/CTLA-4/ICOS haplotypes confers susceptibility to Graves’ disease and modulates clinical phenotype of disease. Endocrine 55, 186–199 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Webber, B. R. et al. Highly efficient multiplex human T cell engineering without double-strand breaks using Cas9 base editors. Nat. Commun. 10, 5222 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kluesner, M. G. et al. CRISPR–Cas9 cytidine and adenosine base editing of splice-sites mediates highly-efficient disruption of proteins in primary and immortalized cells. Nat. Commun. 12, 2437 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gaudelli, N. M. et al. Directed evolution of adenine base editors with increased activity and therapeutic application. Nat. Biotechnol. 38, 892–900 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Diorio, C. et al. Cytosine base editing enables quadruple-edited allogeneic CART cells for T-ALL. Blood 140, 619–629 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Glaser, V. et al. Combining different CRISPR nucleases for simultaneous knock-in and base editing prevents translocations in multiplex-edited CAR T cells. Genome Biol. 24, 89 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Levy, J. M. et al. Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses. Nat. Biomed. Eng. 4, 97–110 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Richter, M. F. et al. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat. Biotechnol. 38, 883–891 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Woodruff, R. et al. Large-scale manufacturing of base-edited chimeric antigen receptor T cells. Mol. Ther. Methods Clin. Dev. 31, 101123 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Martin-Rufino, J. D. et al. Massively parallel base editing to map variant effects in human hematopoiesis. Cell 186, 2456–2474 (2023).

    Article  CAS  PubMed  Google Scholar 

  28. Kim, E. & Hart, T. Improved analysis of CRISPR fitness screens and reduced off-target effects with the BAGEL2 gene essentiality classifier. Genome Med. 13, 2 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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 

  30. Schmidt, R. et al. Base-editing mutagenesis maps alleles to tune human T cell functions. Nature 625, 805–812 (2024).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ramezani, M. et al. A genome-wide atlas of human cell morphology. Preprint at bioRxiv https://doi.org/10.1101/2023.08.06.552164 (2023).

  33. Legut, M. et al. A genome-scale screen for synthetic drivers of T cell proliferation. Nature 603, 728–735 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. Han, K. et al. CRISPR screens in cancer spheroids identify 3D growth-specific vulnerabilities. Nature 580, 136–141 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cooper, S. E. et al. scSNV-seq: high-throughput phenoty** of single nucleotide variants by coupled single-cell genoty** and transcriptomics. Genome Biol. 25, 20 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Coelho, M. A. et al. Base editing screens map mutations affecting interferon-γ signaling in cancer. Cancer Cell 41, 288–303 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Carnevale, J. et al. RASA2 ablation in T cells boosts antigen sensitivity and long-term function. Nature 609, 174–182 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. 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 

  40. Puck, J. M. et al. Mutation analysis of IL2RG in human X-linked severe combined immunodeficiency. Blood 89, 1968–1977 (1997).

    CAS  PubMed  Google Scholar 

  41. Gureasko, J. et al. Role of the histone domain in the autoinhibition and activation of the Ras activator Son of Sevenless. Proc. Natl Acad. Sci. USA 107, 3430–3435 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yi, K. H. & Lauring, J. Recurrent AKT mutations in human cancers: functional consequences and effects on drug sensitivity. Oncotarget 7, 4241–4251 (2015).

    Article  PubMed Central  Google Scholar 

  43. Shimizu, H., Toma-Fukai, S., Kontani, K., Katada, T. & Shimizu, T. GEF mechanism revealed by the structure of SmgGDS-558 and farnesylated RhoA complex and its implication for a chaperone mechanism. Proc. Natl Acad. Sci. USA 115, 9563–9568 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Andersson, E. et al. Activating somatic mutations outside the SH2-domain of STAT3 in LGL leukemia. Leukemia 30, 1204–1208 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Vanhaesebroeck, B., Stephens, L. & Hawkins, P. PI3K signalling: the path to discovery and understanding. Nat. Rev. Mol. Cell Biol. 13, 195–203 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Cheung, L. W. et al. Regulation of the PI3K pathway through a p85α monomer–homodimer equilibrium. eLife 4, e06866 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Dornan, G. L. & Burke, J. E. Molecular mechanisms of human disease mediated by oncogenic and primary immunodeficiency mutations in class IA phosphoinositide 3-kinases. Front. Immunol. 9, 575 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Zhao, P. et al. Activated phosphoinositide 3-kinase delta syndrome caused by PIK3CD mutations: expanding the phenotype. Pediatr. Rheumatol. Online J. 22, 24 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Courtney, A. H. et al. A phosphosite within the SH2 domain of Lck regulates its activation by CD45. Mol. Cell 67, 498–511 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yamaguchi, H. & Hendrickson, W. A. Structural basis for activation of human lymphocyte kinase Lck upon tyrosine phosphorylation. Nature 384, 484–489 (1996).

    Article  CAS  PubMed  Google Scholar 

  51. Agarwal, S. et al. Deletion of the inhibitory co-receptor CTLA-4 enhances and invigorates chimeric antigen receptor T cells. Immunity 56, 2388–2407 (2023).

    Article  CAS  PubMed  Google Scholar 

  52. Zhang, X. et al. Activated phosphoinositide 3-kinase delta syndrome misdiagnosed as anti-neutrophil cytoplasmic antibody-associated vasculitis: a case report. J. Int. Med. Res. 49, 03000605211013222 (2021).

    PubMed  PubMed Central  Google Scholar 

  53. Neugebauer, M. E. et al. Evolution of an adenine base editor into a small, efficient cytosine base editor with low off-target activity. Nat. Biotechnol. 41, 673–685 (2023).

    Article  CAS  PubMed  Google Scholar 

  54. Cuella-Martin, R. et al. Functional interrogation of DNA damage response variants with base editing screens. Cell 184, 1081–1097 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wang, Y., Huang, H., Rudin, C. & Shaposhnik, Y. Understanding how dimension reduction tools work: an empirical approach to deciphering t-SNE, UMAP, TriMap, and PaCMAP for data visualization. J. Mach. Learn. Res. 22, 1–73 (2021).

    Google Scholar 

  56. Bae, S., Park, J. & Kim, J.-S. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 30, 1473–1475 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Hanna, R. E. et al. Massively parallel assessment of human variants with base editor screens. Cell 184, 1064–1080 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Szklarczyk, D. et al. The STRING database in 2023: protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 51, D638–D646 (2023).

    Article  CAS  PubMed  Google Scholar 

  60. Frangieh, C. J. et al. Multimodal pooled Perturb-CITE-seq screens in patient models define mechanisms of cancer immune evasion. Nat. Genet. 53, 332–341 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang, T., Lander, E. S. & Sabatini, D. M. Single guide RNA library design and construction. Cold Spring Harb. Protoc. 2016, pdb.prot090803 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  62. McKinney, W. Data structures for statistical computing in Python. In Proceedings of the 9th Python in Science Conference (eds van der Walt, S. & Millman, J.) 56–61 (SciPy, 2010).

  63. Virtanen, P. et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat. Methods 17, 261–272 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Waskom, M. L. seaborn: statistical data visualization. J. Open Source Softw. 6, 3021 (2021).

    Article  Google Scholar 

  65. Hunter, J. D. Matplotlib: a 2D graphics environment. Comput. Sci. Eng. 9, 90–95 (2007).

    Article  Google Scholar 

  66. 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 

  67. Zhang, J., Pei, J., Durham, J., Bos, T. & Cong, Q. Computed cancer interactome explains the effects of somatic mutations in cancers. Protein Sci. 31, 1–19 (2022).

    Article  Google Scholar 

  68. The UniProt Consortium. UniProt: the universal protein knowledgebase. Nucleic Acids Res. 45, D158–D169 (2017).

    Article  Google Scholar 

  69. Kluesner, M. G. et al. EditR: a method to quantify base editing from Sanger sequencing. CRISPR J. 1, 239–250 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nuñez Pedrozo, C. N. et al. In silico performance analysis of web tools for CRISPRa sgRNA design in human genes. Comput. Struct. Biotechnol. J. 20, 3779–3782 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Nguyen, D. N. et al. Polymer-stabilized Cas9 nanoparticles and modified repair templates increase genome editing efficiency. Nat. Biotechnol. 38, 44–49 (2020).

    Article  CAS  PubMed  Google Scholar 

  72. Roth, T. L. et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 559, 405–409 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Walsh, Z. H. et al. Map** variant effects on anti-tumor hallmarks of primary human T cells with base editing screens. Source code. GitHub github.com/gnikolenyi/izar_vis (2024).

Download references

Acknowledgements

N.K. and S.B.S. are equally contributing second authors. B.I. is supported by National Institute of Health grants (R37CA258829, R01CA280414, R01CA266446, U54CA274506); and additionally by the Pershing Square Sohn Cancer Research Alliance Award; the Burroughs Wellcome Fund Career Award for Medical Scientists; a Tara Miller Melanoma Research Alliance Young Investigator Award; the Louis V. Gerstner, Jr. Scholars Program; and the V Foundation Scholars Award. This work was supported by a Herbert Irving Comprehensive Cancer Center (HICCC) Velocity Grant (to B.I.), the HICCC Human Tissue Immunology and Immunotherapy Initiative and NIH Grant P30CA013696. Medical illustrations were prepared by U. Mackensen. The illustration in Extended Data Fig. 9a was created with https://www.biorender.com.

Author information

Authors and Affiliations

Authors

Contributions

B.I. and Z.H.W. conceived the study. B.I. provided overall supervision with support from J.C.M. Z.H.W., P.S. and J.C.M. planned, designed and executed all key experiments. S.B.S., M.M., P.H., M.R. and S.A. performed experiments. N.K. performed computational analyses of screens with support from Z.H.W. and D.Z.B. G.N. performed structural modeling and visualizations. N.V., M.A., J.D.M., A.C. and G.L. provided additional guidance for the design, execution and interpretation of screens. Z.H.W., P.S., J.C.M. and B.I. wrote the manuscript with input and approval from all authors.

Corresponding author

Correspondence to Benjamin Izar.

Ethics declarations

Competing interests

B.I. is a consultant for or received honoraria from Volastra Therapeutics, Johnson & Johnson (Janssen), Novartis, Eisai, AstraZeneca and Merck and has received research funding to Columbia University from Agenus, Alkermes, Arcus Biosciences, Checkmate Pharmaceuticals, Compugen, Immunocore, Regeneron and Synthekine. Z.H.W. and B.I. filed a patent application based on this work. The other authors do not have competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Dimitrios Wagner 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 Optimization of workflows for base editing in primary human T cells.

a, Overview of approach for targeted base editing in primary human T cells. b-d, Target sites of sgRNAs against CD2, B2M, and TRBC1/2 sites predicted to generate gene knockout through several mechanisms (SPLd = splice donor site mutation, SPLa = splice acceptor site mutation, SM = start codon mutation, ES = conversion to early stop codon). e, Representative flow cytometry histograms from one human donor showing ABE-mediated knockout of CD2 and B2M using sgRNAs indicated in (b-c), and f, CBE-mediated knockout of CD2, TRBC1/2, and B2M using sgRNAs indicated in (b-d). g, Quantification of base editing efficiency in (e) (n = 3 independent human donors). h, Quantification of base editing efficiency in (f), (n = independent human 4 donors for B2M_ES and TRBC1/2_ES; n = 2 independent human donors for B2M_SPLd and CD2_SM). i, Representative flow cytometry dotplots and histograms demonstrating CBE-mediated knockout of TCRab. For histograms, red indicates gated mTurquoise-negative cells, and blue indicates gated mTurquoise-positive cells. j, Quantification of ABE-mediated knockout of B2M with lentiviral integration of B2M_SM_1 sgRNA and electroporation of ABE mRNA in CD4 and CD8 T cell subsets (n = 2 independent human donors). k, Editing efficiency (measured by % B2M loss on flow cytometry) and viability of T cells transduced with B2M_SM_1 sgRNA and electroporated with varying doses of ABE. Vertical dotted line represents ABE dose selected (per 1e6 T cells) for screens. Error bars represent mean +/− SD (panels g, h, j).

Source data

Extended Data Fig. 2 Tiling screen targets, library transduction, and pooled base editing of T cells.

a, Classification of sgRNAs in the ClinVar library based on mutation subtype. b, Schematic of gene targets for the 12-Gene tiling screen and their function in T cells. c, Classification of sgRNAs in the 12-Gene tiling library based on mutation subtype. d, Schematic for generation of library base-edited T cells. e, Transduction efficiency of ClinVar base editor library in n = 2 independent human donors.

Extended Data Fig. 3 Metrics for rigor and reproducibility of large-scale base editing screens.

a, Density plots showing LFC values of different categories of guides from the ClinVar library at Day 35 post-electroporation of the long-term expansion screen arm. Dashed line represents the bottom 5% of the distribution of combined empty window and silent mutation controls. Indicated are the percentages of guides in each category falling below this threshold. sgRNAs generating variants in CD3D, CD3E, CD3G, or CD3Z were binned into the ‘CD3 complex’ category. The second donor from the screen is shown (in companion to Fig. 2a). b, Scatter plot showing LFC values of negative control sgRNAs (including both empty window and silent mutations) in both donors from the ClinVar Library at Day 28 post-electroporation in the long-term expansion screen arm. c-d, Distribution of robust rank aggregation (RRA) scores for gene-wise dropout analysis in the c, CD25 hi vs lo (activation) sort and d, CFSE lo vs hi (short-term proliferation) sort arms of the ClinVar library across both donors. The top 5 negatively selected genes in CD25 hi vs lo and in CFSE lo vs hi are listed. e, Shared positive control sgRNA (n = 600) were identified between the ClinVar and 12-gene tiling screens and sgRNA LFCs from matched long-term proliferation arm timepoints (Day 28 of ClinVar Screen, Day 26 of 12-gene Tiling Screen) are plotted. For each screen, the average LFC of each sgRNA across both donors is plotted. Simple linear regression with two-sided Pearson test (panel e).

Extended Data Fig. 4 Analysis of ClinVar screen across readouts.

a, Scatterplot showing LFC of selected sgRNAs generating mutations in LCK, SOS1, and PTPRC. Timepoint shown is Day 28 post-electroporation in the ClinVar long-term expansion screen arm. b, Volcano plot showing enriched and depleted guides in the CFSE lo vs hi proliferation sort. For visualization purposes, one mutation for each labeled sgRNA is shown. One representative donor is shown. False discovery rate (FDR) cutoff <0.05. c, Volcano plot showing enriched and depleted guides in the CD25 hi vs lo proliferation sort. For visualization purposes, one mutation for each labeled sgRNA is shown. FDR cutoff <0.05. One representative donor is shown.

Extended Data Fig. 5 Characterization of variant effects by ClinVar classification.

a, (Top) distribution of negative control sgRNAs in the ClinVar library at day 28 of the long-term proliferation screen arm. (Bottom) sgRNA LFC distributions for selected genes targeted in the ClinVar library. Red lines indicate sgRNAs generating amino acid mutations which are identical to ClinVar-annotated pathogenic variants. b-j, Scatterplots of sgRNAs targeting selected genes in the ClinVar library at day 28 of the long-term proliferation screen arm. Dotted lines represent top and bottom 5% cutoffs of negative control sgRNA distribution. sgRNAs are binned into four distinct categories: predicted to generate an identical mutation to a ClinVar ‘VUS’ (‘Same VUS’; dark blue), predicted to generate a different mutation at an amino acid with a ClinVar ‘VUS’ (‘Diff VUS’; light blue), predicted to generate an identical mutation to a ClinVar ‘pathogenic’, ‘pathogenic/likely pathogenic’, or ‘likely pathogenic’ variant (‘Same P’; red), and sgRNAs predicted to generate a different mutation at an amino acid with a ClinVar ‘pathogenic’, ‘pathogenic/likely pathogenic’, or ‘likely pathogenic’ variant (‘Diff P’; yellow). Selected sgRNAs are annotated. Simple linear regression (panels b-j).

Extended Data Fig. 6 Independent analysis of 12-gene tiling screen and integration of results with ClinVar screen.

a, sgRNA LFCs across both donors at Day 26 of the long-term proliferation arm of the 12-gene tiling screen are plotted. Dotted lines represent top and bottom 10% cutoffs of the distribution of negative control sgRNAs (empty window and silent only sgRNAs) for each donor. Selected sgRNAs, with predicted target gene and mutation, are shown. b, sgRNA LFCs as in a, with blue overlay filtered by target gene. c, Shared sgRNAs (n = 325) were identified between the ClinVar and 12-gene tiling screens and sgRNA LFCs from matched long-term proliferation arm timepoints (Day 28 of ClinVar Screen, Day 26 of 12-gene Tiling Screen) are plotted. For each screen, the average LFC of each sgRNA across both donors is plotted. Selected sgRNAs with shared enrichment/depletion patterns across donors and screens are annotated. Simple linear regression (panel c).

Extended Data Fig. 7 Enrichment and structure-function relationship of variants promoting T cell proliferation.

a, Lollipop plot showing LFC of sgRNAs targeting PIK3CD at Day 15 post-electroporation in the long-term proliferation arm of the ClinVar screen. sgRNAs are mapped to the targeted region of the canonical isoform of PIK3CD (p110δ) and functional domains of the protein are annotated. Selected variants and their predicted mutational consequences are annotated. b,c, Timecourse line graphs of LFC of sgRNAs targeting PIK3CD in both donors in the long-term proliferation arm of the ClinVar screen. d, Lollipop plot for sgRNAs targeting AKT1 at Day 35 post-electroporation in the long-term proliferation arm of the ClinVar screen, mapped to the canonical AKT1 isoform. e, Timecourse line graphs of LFC of sgRNAs targeting AKT1 in the long-term expansion arm of the ClinVar screen. f, Structure and position of mutations in AKT1. (right) Overall predicted structure of AKT1 (blue) and mutated residues (red). (left) Wild-type (WT) and mutated (mut) residues (red). D323G is predicted to localize next to L14 (dark blue). g, Lollipop plots for sgRNAs targeting LCK at day 26 post-electroporation in the long-term proliferation arm of the 12-gene tiling screen, mapped to the canonical LCK isoform. h, Structure and position of mutations in LCK. (top) Overall predicted structure of LCK (blue) and mutated residues (red). (bottom) Wild-type (WT) and mutated (mut) residues (red).

Extended Data Fig. 8 Signaling and impact of subtle differences in editing efficiency and phenotypic readouts.

a, Quantification of S6 phosphorylation (pS235/S236) and b, AKT phosphorylation (pS473) measured by flow cytometry in T cells with indicated genotypes (x axis) after 10 minutes of stimulation with anti-CD3/CD28 antibodies. c, For all validated sgRNAs targeting PIK3CD (that is, Cys416Arg, Tyr524Cys, Glu525Gly_His526Arg, and Glu527Gly_Lys528Glu), sgRNA editing efficiency and effect size on AKT phosphorylation (pS473), d, TNFα MFI, and e, IL2 expression are plotted for each of the 3 donors used in initial validation experiments in Fig. 3. In cases where sgRNAs generated multiple edits within the editing window (for example, PIK3CD Glu525Gly_His526Arg), the average editing efficiency across all targeted bases in the editing window was used. Data in (a-b) was generated from n = 3 independent human donors. Within each donor this data was normalized to the silent control condition. One-way ANOVA with Dunnett’s test for multiple comparisons (panels a,b). Simple linear regression (panels c-e). Error bars represent mean +/− SD (panels a, b).

Source data

Extended Data Fig. 9 Experimental design, functional assays, and melanoma co-culture experiments with NY-ESO-1 TCR T cells engineered with variants identified in base editing screens.

a, Schematic for engineering and expanding NY-ESO-1 specific T cells. b, Representative flow cytometry dotplot of NY-ESO-1 specific T cells prior to sorting. c, Viable A375-dsRed cells relative to time t0 after culture with NY-ESO-1 specific T cells for 48 hours at varying effector to target ratios. aMHCI = MHC class I-blocking antibody (n = 3 independent biological replicates). d, Representative flow cytometry histograms of ABE-mediated knockout of B2M or CD2 in NY-ESO-1 specific T cells. e, Representative contour plots of single or multiplexed base editing of B2M and CD2. f, AKT phosphorylation (pS473), in NY-ESO-1 specific T cells after either 15 minutes of co-culture with A375 cells (+) or media alone (-) (n = 3 independent biological replicates). g, MFI of TNFα and h, GrzB in NY-ESO-1 specific T cells with indicated base edits after 8-hour co-culture with A375-dsRed cells at a 1:1 effector to target ratio (n = 3 independent biological replicates). i, Frequency of NY-ESO-1 specific T cells with indicated genotypes co-expressing TNFα, IL2, and GrzB after 8 hours of co-culture with A375 cells at a 1:1 effector to target ratio (n = 3 independent biological replicates.). NT = non-targeting control sgRNA j, Viable wild-type (WT) or CD58-KO A375 cells relative to time t0 after 48 hours of co-culture with NY-ESO-1 specific T cells at a 1:1 effector to target ratio (n = 3 independent biological replicates.) k, Viable B2M-KO A375 cells relative to time t0 after 48 hours of co-culture with NY-ESO-1 specific T cells with indicated genotypes. Dotted lines in (f-i) represent mean of the control. Dotted lines in (j-k) represent relative viable cell count at time t0. One-way ANOVA with Tukey’s test for multiple comparisons (panel c). One-way ANOVA with Dunnett’s test for multiple comparisons (panels f-i, k). Student’s t test (panel j). Error bars represent mean +/− SD (panels c, f-k).

Source data

Extended Data Fig. 10 Design and results of leukemia co-culture with CD19 CAR-T cells equipped with variants identified in base editing screens.

a, Representative histograms of GFP expression, indicating transduction efficiency of first- and second-generation CD19-CAR constructs (CD19-CD3z or CD19-BBz, respectively) in primary human T cells. Blue histograms represent untransduced control T cells. b, Expression of CTLA4 on CD19-CAR T cells, edited with a control non-targeting sgRNA (NT) or CTLA4-KO sgRNA, following 48-hour co-culture with Nalm6 leukemia cells at an 0.5:1 effector to target ratio. c, Relative cell numbers of Nalm6 cells 48 hours after co-culture with CD19-BBz CAR T variants at several E:T ratios, compared to time t0. d, Relative number of CD19-KO Nalm6 cells 48 hours after co-culture with CD19-BBz CAR T cells at an 0.25:1 effector to target ratio, compared to time t0. e, Quantification of CD19-CD3z CAR T cell AKT phosphorylation (pS473) by flow cytometry after 15 minutes of culture with either Nalm6 leukemia (+) or media alone (−) with representative flow histograms. f, Quantification of CD19-CD3z CAR T cell intracellular expression of TNFα and g, IL2 after 8-hour culture with Nalm6 leukemia (+) or in media only (-). h, Relative cell numbers of wild-type and i, CD19-KO Nalm6 cells 48 hours after co-culture with CD19-CD3z CAR T cells at an 0.5:1 effector to target ratio, compared to time t0. Dotted lines in (e-g) represent mean of the control population. Dotted lines in (panel c, d, h, i) represent relative viable cell count at time t0. One-way ANOVA with Tukey’s test for multiple comparisons (panel b), one-way ANOVA with Dunnett’s test for multiple comparisons (panels d-i). Error bars represent mean +/- SD (panels b-i).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–3.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–8. Each table is in a separate, labeled tab.

Supplementary Data

Raw sequencing reads and library map** percentages for all sequencing runs for screens in this study.

Source data

Source Data Figs. 1 and 3–5 and Extended Data Figs. 1 and 8–10

Statistical source data (contains labeled tabs for all relevant main and Extended Data figures).

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

Walsh, Z.H., Shah, P., Kothapalli, N. et al. Map** variant effects on anti-tumor hallmarks of primary human T cells with base-editing screens. Nat Biotechnol (2024). https://doi.org/10.1038/s41587-024-02235-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-024-02235-x

  • Springer Nature America, Inc.

Navigation