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:

Naturally occurring T cell mutations enhance engineered T cell therapies

Abstract

Adoptive T cell therapies have produced exceptional responses in a subset of patients with cancer. However, therapeutic efficacy can be hindered by poor T cell persistence and function1. In human T cell cancers, evolution of the disease positively selects for mutations that improve fitness of T cells in challenging situations analogous to those faced by therapeutic T cells. Therefore, we reasoned that these mutations could be co-opted to improve T cell therapies. Here we systematically screened the effects of 71 mutations from T cell neoplasms on T cell signalling, cytokine production and in vivo persistence in tumours. We identify a gene fusion, CARD11PIK3R3, found in a CD4+ cutaneous T cell lymphoma2, that augments CARD11–BCL10–MALT1 complex signalling and anti-tumour efficacy of therapeutic T cells in several immunotherapy-refractory models in an antigen-dependent manner. Underscoring its potential to be deployed safely, CARD11–PIK3R3-expressing cells were followed up to 418 days after T cell transfer in vivo without evidence of malignant transformation. Collectively, our results indicate that exploiting naturally occurring mutations represents a promising approach to explore the extremes of T cell biology and discover how solutions derived from evolution of malignant T cells can improve a broad range of T 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: In vitro and in vivo screening identifies T cell mutations that reprogram CAR signalling and functional outputs.
Fig. 2: CARD11–PIK3R3 enhances CBM complex signalling.
Fig. 3: CARD11–PIK3R3 expression leads to signalling, transcriptional and functional enhancements in primary human CD8+ T cells.
Fig. 4: CARD11–PIK3R3 improves the therapeutic efficacy of human and mouse CAR T cells in vivo.
Fig. 5: CARD11–PIK3R3 enhances the therapeutic efficacy of mouse and human TCR-transgenic T cells in vivo.

Similar content being viewed by others

Data availability

Sequencing reads are deposited in the NCBI Sequence Read Archive (BioProject ID PRJNA1029944). All other data are available in the main text or the Supplementary InformationSource data are provided with this paper.

References

  1. Hou, A. J., Chen, L. C. & Chen, Y. Y. Navigating CAR-T cells through the solid-tumour microenvironment. Nat. Rev. Drug Discov. 20, 531–550 (2021).

    Article  CAS  PubMed  Google Scholar 

  2. Wang, L. et al. Genomic profiling of Sézary syndrome identifies alterations of key T cell signaling and differentiation genes. Nat. Genet. 47, 1426–1434 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. 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 

  4. 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 Central  PubMed  Google Scholar 

  5. Mustjoki, S. & Young, N. S. Somatic mutations in “benign” disease. N. Engl. J. Med. 384, 2039–2052 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Walker, S. et al. Identification of a gain-of-function STAT3 mutation (p.Y640F) in lymphocytic variant hypereosinophilic syndrome. Blood 127, 948–951 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  7. Park, J. et al. Genomic analysis of 220 CTCLs identifies a novel recurrent gain-of-function alteration in RLTPR (p.Q575E). Blood 130, 1430–1440 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Park, J. et al. Integrated genomic analyses of cutaneous T-cell lymphomas reveal the molecular bases for disease heterogeneity. Blood 138, 1225–1236 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Daniels, J. et al. Cellular origins and genetic landscape of cutaneous gamma delta T cell lymphomas. Nat. Commun. 11, 1806–1806 (2020).

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

  10. Stadtmauer Edward, A. et al. CRISPR-engineered T cells in patients with refractory cancer. Science 367, eaba7365 (2020).

    Article  PubMed  Google Scholar 

  11. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558, 307–312 (2018).

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

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

  13. Martinez, G. J. et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Schmitz, R. et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature 490, 116–120 (2012).

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

  15. Liu, Y. et al. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat. Genet. 49, 1211–1218 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

  18. Kahan, S. M. et al. Intrinsic IL-2 production by effector CD8 T cells affects IL-2 signaling and promotes fate decisions, stemness, and protection. Sci. Immunol. 7, eabl6322 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Ruland, J. & Hartjes, L. CARD-BCL-10-MALT1 signalling in protective and pathological immunity. Nat. Rev. Immunol. 19, 118–134 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Jattani, R. P., Tritapoe, J. M. & Pomerantz, J. L. Intramolecular interactions and regulation of cofactor binding by the four repressive elements in the caspase recruitment domain-containing protein 11 (CARD11) inhibitory domain. J. Biol. Chem. 291, 8338–8348 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Burke, J. E. Structural basis for regulation of phosphoinositide kinases and their involvement in human disease. Mol. Cell 71, 653–673 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Kutzner, K. et al. Phosphorylation of serine-893 in CARD11 suppresses the formation and activity of the CARD11-BCL10-MALT1 complex in T and B cells. Sci. Signal. 15, eabk3083.

  23. Li, S., Yang, X., Shao, J. & Shen, Y. Structural insights into the assembly of CARMA1 and BCL10. PLoS ONE 7, e42775 (2012).

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

  24. Grossmann, A. et al. Phospho-tyrosine dependent protein-protein interaction network. Mol. Syst. Biol. 11, 794 (2015).

    Article  PubMed  Google Scholar 

  25. Fan, X., Quezada, S. A., Sepulveda, M. A., Sharma, P. & Allison, J. P. Engagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J. Exp. Med. 211, 715–725 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Massarelli, E. et al. High OX-40 expression in the tumor immune infiltrate is a favorable prognostic factor of overall survival in non-small cell lung cancer. J. Immunother. Cancer 7, 351 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  27. Bardet, M. et al. The T-cell fingerprint of MALT1 paracaspase revealed by selective inhibition. Immunol. Cell Biol. 96, 81–99 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Jiang, T., Zhou, C. & Ren, S. Role of IL-2 in cancer immunotherapy. Oncoimmunology 5, e1163462 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  29. Guedan, S. et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight https://doi.org/10.1172/jci.insight.96976 (2018).

  30. King, M. A. et al. Human peripheral blood leucocyte non-obese diabetic-severe combined immunodeficiency interleukin-2 receptor gamma chain gene mouse model of xenogeneic graft-versus-host-like disease and the role of host major histocompatibility complex. Clin. Exp. Immunol. 157, 104–118 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. Bidlingmaier, S. et al. Identification of MCAM/CD146 as the target antigen of a human monoclonal antibody that recognizes both epithelioid and sarcomatoid types of mesothelioma. Cancer Res. 69, 1570–1577 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  32. Li, Q.-X., Feuer, G., Ouyang, X. & An, X. Experimental animal modeling for immuno-oncology. Pharmacol. Ther. 173, 34–46 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Kalbasi, A. et al. Potentiating adoptive cell therapy using synthetic IL-9 receptors. Nature 607, 360–365 (2022).

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

  34. Overwijk, W. W. & Restifo, N. P. B16 as a mouse model for human melanoma. Curr. Protoc. Immunol. https://doi.org/10.1002/0471142735.im2001s39 (2001).

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

    Article  CAS  PubMed Central  ADS  PubMed  Google Scholar 

  36. Leidner, R. et al. Neoantigen T-cell receptor gene therapy in pancreatic cancer. New Engl. J. Med. 386, 2112–2119 (2022).

    Article  CAS  PubMed  Google Scholar 

  37. Zhao, H. et al. Genome-wide fitness gene identification reveals Roquin as a potent suppressor of CD8 T cell expansion and anti-tumor immunity. Cell Rep. 37, 110083 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. FDA investigating serious risk of T-cell malignancy following BCMA-directed or CD19-directed autologous chimeric antigen receptor (CAR) T cell immunotherapies. FDA https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/fda-investigating-serious-risk-t-cell-malignancy-following-bcma-directed-or-cd19-directed-autologous (28 November 2023).

  39. Cappell, K. M. & Kochenderfer, J. N. Long-term outcomes following CAR T cell therapy: what we know so far. Nat. Rev. Clin. Oncol. 20, 359–371 (2023).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Tward, J. D., Wendland, M. M., Shrieve, D. C., Szabo, A. & Gaffney, D. K. The risk of secondary malignancies over 30 years after the treatment of non-Hodgkin lymphoma. Cancer 107, 108–115 (2006).

    Article  PubMed  Google Scholar 

  41. Chihara, D., Dores, G. M., Flowers, C. R. & Morton, L. M. The bidirectional increased risk of B-cell lymphoma and T-cell lymphoma. Blood 138, 785–789 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Harrison, S. J. et al. CAR+ T-cell lymphoma post ciltacabtagene autoleucel therapy for relapsed refractory multiple myeloma. Blood 142, 6939 (2023).

    Article  Google Scholar 

  43. Bowcock, S. J. et al. High incidence of therapy-related myelodysplasia and acute leukaemia in general haematology clinic patients treated with fludarabine and cyclophosphamide for indolent lymphoproliferative disorders. Br. J. Haematol. 134, 242–243 (2006).

    Article  PubMed  Google Scholar 

  44. Zhu, I. et al. Modular design of synthetic receptors for programmed gene regulation in cell therapies. Cell 185, 1431–1443 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Roybal, K. T. et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell 164, 770–779 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  47. Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. H. Chimeric antigen receptor–modified T cells in chronic lymphoid leukemia. New Engl. J. Med. 365, 725–733 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Jutz, S. et al. Assessment of costimulation and coinhibition in a triple parameter T cell reporter line: simultaneous measurement of NF-κB, NFAT and AP-1. J. Immunol. Methods 430, 10–20 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Northwestern University Flow Cytometry Core, the University of California, San Francisco Flow Cytometry Core, the Northwestern University Research Computing Services, the Northwestern University Skin Biology and Disease Resource-Based Center and Admera Health for their contributions; and J. Eyquem for providing the Nalm6-Luc-expressing cell line. The indicated graphics in Figs. 1a and 5a and Extended Data Fig. 9a were created with BioRender.com and adapted as required. J.D. was supported by NIH NCI grants F30 CA265107 and T32 CA009560. J.G. was supported by NIH NCI grant F31 CA260790. J.C. was supported by NIH grant 1DP2AI136599-01, the Bakewell Foundation and the Mark Foundation Emerging Leader Award. K.T.R. was supported by the Parker Institute for Cancer Immunotherapy, the NIH-NCI Cancer Moonshot Immuno-Oncology Translational Network Center Grant (U54 CA244438), the UCSF Helen Diller Family Comprehensive Cancer Center and the NIH Director’s New Innovator Award (DP2 CA239143).

Author information

Authors and Affiliations

Authors

Contributions

J.G., J.D., K.T.R. and J.C. designed the study. J.G. carried out in vivo screening, primary T cell in vitro experiments and in vivo CAR T cell experiments. J.D. carried out in vitro screening, biochemical experiments, in vivo TCR-transgenic experiments and bioinformatics analysis. J.D. and J.C. identified library mutations. J.G., I.Z. and D.G. designed the lentiviral library; J.G. and I.Z. cloned the library. J.G. produced the lentivirus of each construct. C.T., C.B., J.A. and C.A assisted with in vivo CAR T cell experiments. Y.L., Q.L., K.C. and C.L. assisted with biochemical and in vivo TCR-transgenic experiments. K.C., Q.L. and C.L. carried out and analysed the necropsies. G.M. assisted with primary T cell isolations from human donors. J.G., J.D., K.T.R. and J.C. wrote the manuscript with input from all authors. K.T.R. and J.C. supervised the study.

Corresponding authors

Correspondence to Kole T. Roybal or Jaehyuk Choi.

Ethics declarations

Competing interests

K.T.R. is a co-founder, consultant, SAB member and stockholder of Arsenal Biosciences and Dispatch Therapeutics. He was a founding scientist and consultant and stockholder in Cell Design Labs, now a Gilead Company. K.T.R. holds stock in Gilead. K.T.R. is an exclusive adviser to Venrock. J.G. and J.D. are founding scientists and stockholders of Moonlight Bio. K.T.R. and J.C. are co-founders and stockholders of Moonlight Bio. K.T.R., J.C., J.G., J.D. and I.Z. are inventors on a University of California, San Francisco and Northwestern University provisional patent for enhancing adoptive T cell therapeutics; 63/412,300.

Peer review

Peer review information

Nature thanks Hans Stauss 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 figures and tables

Extended Data Fig. 1 In vitro screening of 28z-CAR and BBz-CAR triple reporter Jurkat cells.

(a) CAR and CD19 expression in cell lines utilized for screening. (b) Percent positive for the indicated reporter construct in 28z-CAR or BBZ-CAR transduced Jurkat cells co-cultured with the indicated cell line (n = 2). (c) Gating strategy to identify library construct transduced Jurkat cells in co-culture. (d) Heatmaps depicting Z scores in vitro screens. Z scores represent mean of two experimental replicates. Z scores are calculated on a row-by-row basis and thus represent different percentage positive values across different experimental conditions, such as with or without CD19 antigen. (e-f) Reproducibility of screening in 28z-CAR Jurkat cells (e) or BBz-CAR Jurkat cells (f). Each replicate indicates an independent lentiviral transduction of Jurkat cells with T cell mutation screening constructs, statistics determined by simple linear regression. (g) Scatterplot indicating CD19-28z CAR (x-axis) and CD19-BBz CAR (y-axis) percent positive for reporter constructs and IL-2 production.

Source Data

Extended Data Fig. 2 Point mutations with statistically significant differences compared to wild-type constructs and antigen specificity of mutations.

(a-c) NFAT, NF-κB, and AP-1 reporter activity. Mutations with a statistically significant difference from the wild-type construct are shown. Each row indicates the mutation construct, co-culture condition and CAR construct. Gain of function indicates mutations which increased reporter activity relative to wild-type, while loss of function refers to mutations with lower reporter activity than the wild-type counterpart. Each dot indicates one biological replicate. (d) Percent of constructs with significant effects in the indicated CAR and co-culture condition. ** indicates P value < 0.01, *** indicates P < 0.001, **** indicates P < 0.0001, Chi-squared test.

Extended Data Fig. 3 Pooled human CAR T cell in vivo persistence screening of T cell mutation library.

(a) Tumor growth curve for mice bearing CD19-K562 subcutaneous tumors treated with vehicle (PBS, control) or 1 x 106 CAR + T cells. (b) Histogram of normalized read count distribution of pre-injection constructs. Constructs with a pre-injection normalized read count of <102 were excluded from analysis. (c) Log2 fold change of CARD11-PIK3R3 normalized reads at each indicated time point in the pooled in vivo experiment (week 1 n = 5, week 2 n = 3, week 3 n = 5). (d) In vitro screen Z scores of mutation constructs identified as positive hits from in vivo screening. Z score represents the mean Z score of two experimental replicates.

Source Data

Extended Data Fig. 4 In Vitro Expansion of CD19-BBz-CAR T cells with and without CARD11-PIK3R3.

(a,b) CAR or CAR + CARD11-PIK3R3 were sorted for purity, then expanded with IL-2 for 12 days. On Day 12, cultures were split, reseeding each group without IL-2 (a) and with IL-2 (b). Cells were counted and split from day 12 to day 30. (c) On Day 30, CD19-BBz-CAR + CARD11-PIK3R3 T cells that were cultured with IL-2 were reseeded without IL-2 and counted and split for an additional 10 days.

Source Data

Extended Data Fig. 5 In vitro analysis of CARD11-PIK3R3 expressing T cells.

(a) Principal component analysis of transcriptomes of human BBz-CAR T cells. (b) Representative histogram of ICOS expression on CD19-BBz-CAR T cells 24 h after co-culture with CD19-K562s. (c) Activation markers expressed on CD8+ CD19-28z-CAR T cells after 24 h co-cultured with CD19-K562s. Ratio of MFI (CAR + CARD11-PIK3R3/CAR) shown. P values determined by two-tailed unpaired T test. (d) Activation markers expressed on untransduced and CARD11-PIK3R3 CD4+ and CD8 + T cells after 24 h co-cultured with CD19-K562s. P values determined by two-tailed ratio paired T test with Holm-Sidak correction for multiple comparisons. (e) Cytokine secretion of CD8 + CD19-CD28z-CAR T cells 48 h post 1:1 co-culture with CD19-K562s. P values determined by two-tailed ratio paired T test, represented as fold change relative to control. (f) Cytokine secretion of control, CARD11-PIK3R3, CD19-BBz-CAR and CD19-BBz-CAR + CARD11-PIK3R3 CD8 + T cells 48 h post 1:1 co-culture with CD19-K562s. (g) Bar graph summarizing CD19-K562 population percentages after 14-day co-culture of CD19-CD28z-CAR T cells with and without CARD11-PIK3R3, and supplemental IL-2. P values determined by multiple paired T test followed by Bonferroni correction. (h-i) Cell counts of CD19-A549 mKate2+ targets co-cultured with CD8+ (h) CD19-BBz-CAR T cells or (i) CD19-CD28z-CAR T cells over 108 h period. Bar graph indicates target cell counts at hour 108, standardized to control. P values calculated by one-way ANOVA followed by Tukey’s multiple comparison test, performed on cell count at 108 h. (j) Cell counts of CD8 + CD19-BBz-CAR T cells or CD19-CD28z-CAR T cells on Day 13 after two stimulations with CD19-K562 targets (n = 2 or 3). P values determined by two-tailed unpaired T test. (a,c-j) Each data point indicates an independent donor (n = 3), with the exception of j (n = 2 or 3). ns indicates not significant, * indicates P value < 0.05, ** indicates P value < 0.01.

Source Data

Extended Data Fig. 6 Weight loss of CD19-BBz-CAR + CARD11-PIK3R3 treated animals ameliorated with TCR Knockout.

(a) Flow cytometry plots indicating CD3 expression of TCR replete or TCR knockout groups after electroporation with TRAC targeted RNPs. (b) Flow cytometry plots indicating CAR (FLAG) and CARD11-PIK3R3 or control blank construct (mCherry) expression in human CD3 + T cells. (c) Radiance of Nalm6-Luc-GFP tumor bearing animals treated with control (n = 5), CD19-BBz-CAR (n = 5), CD19-BBz-CAR + CARD11-PIK3R3 (n = 5) TCR replete T cells dosed at 1e6 CAR + T cells, control cells dosed equivalent to highest total T cell dose in other treatment groups. Dotted lines indicate individual mice, bold line indicates median. Arrow indicates date T cells were injected. (d) Percent weight change from baseline of TCR Knockout or TCR replete CD19-BBz-CAR + CARD11-PIK3R3 treated Nalm6 bearing animals.

Source Data

Extended Data Fig. 7 CD19-BBz-CAR + CARD11-PIK3R3 is well tolerated and effective at high doses.

(a) Flow cytometry plots indicating CAR (FLAG) and CARD11-PIK3R3 (mCherry) expression in human CD3 + T cells. (b-c) Percent weight change from baseline of (b) tumor bearing or (c) non-tumor bearing animals treated with control, CD19-BBz-CAR, CD19-BBz-CAR + CARD11-PIK3R3 or CARD11-PIK3R3 (d) Survival analysis of Fig. 4b surviving CD19-BBz-CAR (n = 3), CD19-BBz-CAR + CARD11-PIK3R3 (n = 4) animals or naïve mice (n = 4) rechallenged with 5e5 Nalm6-Luc-GFP tumors. P values determined by Log-rank Mantel-Cox followed by Bonferroni correction. ** indicates P value < 0.01.

Source Data

Extended Data Fig. 8 In vivo analysis of CARD11-PIK3R3 expressing CAR T cells.

(a,c) Flow cytometry plots indicating FLAG (CAR) and mCherry (CARD11-PIK3R3) expression in human CD3 + T cells. (b) Percent weight change from baseline of control, CD19-CD28z-CAR or BBz-CAR + CARD11-PIK3R3 treated Nalm6 tumor bearing animals. (d) Percent weight change from baseline of control, MCAM-CD28z-CAR or MCAM-CD28z-CAR + CARD11-PIK3R3 treated M28 tumor bearing animals. (e) Flow cytometry plots indicating GFP (CAR) and CARD11-PIK3R3 (mCherry) expression in mouse OT-I T cells. (f) Percent weight change from baseline of control, CD19-BBz-CAR or CD19-BBz-CAR + CARD11-PIK3R3 treated hCD19-B16 tumor bearing animals. (g) Accumulation of CD19-BBz-CAR T cells in spleen of hCD19-B16 tumor bearing animals 5 days post adoptive cell transfer. Each data point indicates one mouse (n = 5), mean + s.e.m. depicted. P values determined using two-tailed unpaired t test. (h) Histograms indicating human CD19 ligand expression on hCD19-B16 tumors, CD19-BBz-CAR or CD19-BBz-CAR + CARD11-PIK3R3 treated that had reached euthanasia endpoint, compared to known CD19 positive B16 tumor sample. * indicates P value < 0.05.

Source Data

Extended Data Fig. 9 In vivo analysis of CARD11-PIK3R3 expressing OT-I T cells.

(a) Cell percentage in total CD8 + T cells of control or CARD11-PIK3R3 OT-I T cells in dual transfer experiment (n = 5). P values determined by two-tailed unpaired T test. The images of the mouse and the cells were created with BioRender.com and adapted as required. (b) Tumor growth curve for mice bearing B16-OVA tumors treated with 1 x 106 OT-I T cells (10% CARD11-PIK3R3 positive). (c) Schematic of competition experiments with pmel-1 CD8 T cells. (d) Tumor growth curves of mice described in (c) (n = 5). (e) Fold-enrichment in the tumor of wild-type CARD11-PIK3R3 or CARD11-PIK3R3 (p.R28A) expressing pmel-1 CD8 T cells compared to vector control 7 days after adoptive transfer (n = 5). Mean + s.e.m. depicted. All P values determined by two-tailed ratio paired T test. (f) Percent change in tumor volume and proportion of transferred cells in control OT-I (n = 8) and CARD11-PIK3R3 OT-I (n = 9) mice bearing B16-OVA subcutaneous tumors. Each data point indicates one mouse, mean + s.e.m. depicted. (g) Frequency of TCF-1 + OT-I cells in spleen (control OT-1 n = 5, CARD11-PIK3R3 OT-1 n = 7) and tumor draining lymph node (LN) (control OT-1 n = 3, CARD11-PIK3R3 OT-1 n = 5) of mice bearing B16-OVA subcutaneous tumors. P values determined by two-tailed unpaired T test. (h) Ex vivo cytokine production with PMA/Ionomycin stimulation of control OT-I (n = 7) or CARD11-PIK3R3 OT-I (n = 7) TILs 7 days post T cell transfer. P values determined by two-tailed unpaired T test. (i) Tumor volume and survival analysis in B16-OVA melanoma bearing mice treated with PBS (n = 5), control OT-I cells (2 × 106) (n = 5), or CARD11-PIK3R3 OT-I cells (2 × 106) (n = 5) day 12 after tumor inoculation. Complete response was defined as an absence of a detectable tumor. P values determined by one-way ANOVA followed by Tukey’s multiple comparisons test (tumor volume) or Log-rank Mantel-Cox (survival). (j) Percent change in body weight in PBS, control OT-I, or CARD11-PIK3R3 OT-I treated, B16-OVA tumor bearing mice. Each data point indicates one mouse, mean + s.e.m. depicted. ns indicates not significant, *** indicates P values < 0.001, **** indicates P value < 0.0001.

Source Data

Extended Data Fig. 10 Long-term evaluation of B6 mice treated with CARD11-PIK3R3 expressing OT-I T cells.

(a) Mice that cleared B16-F10-OVA from Fig. 5e were monitored for up to 240 days after adoptive T cell transfer. Necropsies were performed in as outlined in this schematic. (b) Body weights of all CARD11-PIK3R3 infused mice from Fig. 5e and Extended Data Fig. 9i were measured weekly and compared to expected weight curves published by The Jackson Laboratory Research Institute. P value determined by two-way ANOVA. (c) Spleen weight for three animals that underwent necropsy on day 240 post adoptive transfer. This was calculated as percent of body weight and compared to expected spleen weight published by The Jackson Laboratory Research Institute. P value determined by two-tailed unpaired T test. (d) Necropsy of one representative animal. None of the three animals had gross pathology. (e) Percentage of CD8 T cells in spleen and blood that express CARD11-PIK3R3 240 days post adoptive transfer. (f) Representative hematoxylin and eosin-stained tissue sections from select organs where nodal or extranodal lymphomas can occur. Animals were subject to full-body necropsies (n = 3). Tissues did not show evidence of nuclear atypia, changes in cellular architecture, or presence of neoplastic disease. Representative images at low (left) and high (right) -power magnification are shown with size bars. White box reflects the site of high-magnification image. (g,i) Schematic of blood sampling of mice presented in Fig. 5e 330 days after adoptive transfer (g) or of 2e6 OT-I CARD11-PIK3R3 treated mice presented in Extended Data Fig. 9i 418 days after adoptive transfer (i). (h,j) Percentage of CD8 T cells in blood that express CARD11-PIK3R3 in the two cohorts described in Fig. 5e or Extended Data Fig. 9i. Each data point indicates one mouse, mean + s.e.m. depicted.

Source Data

Supplementary information

Supplementary Fig. 1

Original immunoblotting images for Fig. 2d.

Reporting Summary

Supplementary Fig. 2

Additional flow cytometry gating strategy information.

Supplementary Table 1

Constructs tested in T cell lymphoma library and their associated bar-codes.

Supplementary Table 2

RNA-sequencing results. log2[fold change] (l2fc) and adjusted P values (padj) as determined by DESEQ2 for stimulated CD4+ and CD8+ BBz-CAR T cells. Positive log2[fold change] indicates higher expression in CARD11–PIK3R3-expressing CAR T cells compared to controls.

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

Garcia, J., Daniels, J., Lee, Y. et al. Naturally occurring T cell mutations enhance engineered T cell therapies. Nature 626, 626–634 (2024). https://doi.org/10.1038/s41586-024-07018-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07018-7

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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