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:

TET2 guards against unchecked BATF3-induced CAR T cell expansion

Abstract

Further advances in cell engineering are needed to increase the efficacy of chimeric antigen receptor (CAR) and other T cell-based therapies1,2,3,4,5. As T cell differentiation and functional states are associated with distinct epigenetic profiles6,7, we hypothesized that epigenetic programming may provide a means to improve CAR T cell performance. Targeting the gene that encodes the epigenetic regulator ten–eleven translocation 2 (TET2)8 presents an interesting opportunity as its loss may enhance T cell memory9,10, albeit not cause malignancy9,11,12. Here we show that disruption of TET2 enhances T cell-mediated tumour rejection in leukaemia and prostate cancer models. However, loss of TET2 also enables antigen-independent CAR T cell clonal expansions that may eventually result in prominent systemic tissue infiltration. These clonal proliferations require biallelic TET2 disruption and sustained expression of the AP-1 factor BATF3 to drive a MYC-dependent proliferative program. This proliferative state is associated with reduced effector function that differs from both canonical T cell memory13,14 and exhaustion15,16 states, and is prone to the acquisition of secondary somatic mutations, establishing TET2 as a guardian against BATF3-induced CAR T cell proliferation and ensuing genomic instability. Our findings illustrate the potential of epigenetic programming to enhance T cell immunity but highlight the risk of unleashing unchecked proliferative responses.

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: Effect of TET2 disruption on CAR T cell therapeutic efficacy is dependent on CAR design.
Fig. 2: Effect of CAR design on long-term T cell accumulation upon CRISPR–Cas9 editing of the TET2 locus.
Fig. 3: Hyperproliferative TET2-edited CAR T populations are oligoclonal and biallelically edited for TET2.
Fig. 4: Loss of effector function in hyperproliferative TET2bed CAR T cells.
Fig. 5: The BATF3–MYC axis drives hyperproliferation of TET2bed CAR T cells.

Similar content being viewed by others

Data availability

Data generated from RNA-seq and ATAC-seq have been deposited in the Gene Expression Omnibus with the accession number GSE220259. The publicly available datasets used in this study are GSE23321 for central memory and effector memory phenotype comparison, AKL_HTLV1_UP (M7705), AKL_HTLV1_DN (M9815), the angioimmunoblastic T cell lymphoma dataset (GSE6338) and HALLMARK_MYC_V1 (M5926). Source data are provided with this paper.

References

  1. Kakarla, S. & Gottschalk, S. CAR T cells for solid tumors: armed and ready to go? Cancer J. 20, 151–155 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sadelain, M., Riviere, I. & Riddell, S. Therapeutic T cell engineering. Nature 545, 423–431 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. June, C. H. & Sadelain, M. Chimeric antigen receptor therapy. N. Engl. J. Med. 379, 64–73 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Guedan, S., Calderon, H., Posey, A. D. Jr & Maus, M. V. Engineering and design of chimeric antigen receptors. Mol. Ther. Methods Clin. Dev. 12, 145–156 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Globerson Levin, A., Riviere, I., Eshhar, Z. & Sadelain, M. CAR T cells: building on the CD19 paradigm. Eur. J. Immunol. 51, 2151–2163 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Philip, M. et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545, 452–456 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Khan, O. et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 571, 211–218 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Pastor, W. A., Aravind, L. & Rao, A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat. Rev. Mol. Cell Biol. 14, 341–356 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Carty, S. A. et al. The loss of TET2 promotes CD8+ T cell memory differentiation. J. Immunol. 200, 82–91 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bowman, R. L. & Levine, R. L. TET2 in normal and malignant hematopoiesis. Cold Spring Harb. Perspect. Med. 7, a026518 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Chiba, S. Dysregulation of TET2 in hematologic malignancies. Int. J. Hematol. 105, 17–22 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Kaech, S. M., Wherry, E. J. & Ahmed, R. Effector and memory T-cell differentiation: implications for vaccine development. Nat. Rev. Immunol. 2, 251–262 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Farber, D. L., Yudanin, N. A. & Restifo, N. P. Human memory T cells: generation, compartmentalization and homeostasis. Nat. Rev. Immunol. 14, 24–35 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Wherry, E. J. T cell exhaustion. Nat. Immunol. 12, 492–499 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Majzner, R. G. & Mackall, C. L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 25, 1341–1355 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. 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 

  19. Tahiliani, M. et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Delhommeau, F. et al. Mutation in TET2 in myeloid cancers. N. Engl. J. Med. 360, 2289–2301 (2009).

    Article  PubMed  Google Scholar 

  21. Tefferi, A., Lim, K. H. & Levine, R. Mutation in TET2 in myeloid cancers. N. Engl. J. Med. 361, 1117–1118 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Watatani, Y. et al. Molecular heterogeneity in peripheral T-cell lymphoma, not otherwise specified revealed by comprehensive genetic profiling. Leukemia 33, 2867–2883 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. 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 

  24. Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Lollies, A. et al. An oncogenic axis of STAT-mediated BATF3 upregulation causing MYC activity in classical Hodgkin lymphoma and anaplastic large cell lymphoma. Leukemia 32, 92–101 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Nakagawa, M. et al. Targeting the HTLV-I-regulated BATF3/IRF4 transcriptional network in adult T cell leukemia/lymphoma. Cancer Cell 34, 286–297.e10 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Liang, H. C. et al. Super-enhancer-based identification of a BATF3/IL-2R-module reveals vulnerabilities in anaplastic large cell lymphoma. Nat. Commun. 12, 5577 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gonzalez, M. V. et al. Glucocorticoids antagonize AP-1 by inhibiting the activation/phosphorylation of JNK without affecting its subcellular distribution. J. Cell Biol. 150, 1199–1208 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Patil, R. H. et al. Dexamethasone inhibits inflammatory response via down regulation of AP-1 transcription factor in human lung epithelial cells. Gene 645, 85–94 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Kafer, G. R. et al. 5-Hydroxymethylcytosine marks sites of DNA damage and promotes genome stability. Cell Rep. 14, 1283–1292 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Chen, L. L. et al. SNIP1 recruits TET2 to regulate c-MYC target genes and cellular DNA damage response. Cell Rep. 25, 1485–1500.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Seo, H. et al. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat. Immunol. 22, 983–995 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Man, K. et al. The transcription factor IRF4 is essential for TCR affinity-mediated metabolic programming and clonal expansion of T cells. Nat. Immunol. 14, 1155–1165 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Man, K. et al. Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity 47, 1129–1141.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. McCutcheon, S. et al. CRISPR-based epigenome editing screens in primary human T cells. Mol. Ther. 30, 165–166 (2022).

  37. Ataide, M. A. et al. BATF3 programs CD8+ T cell memory. Nat. Immunol. 21, 1397–1407 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Eferl, R. & Wagner, E. F. AP-1: a double-edged sword in tumorigenesis. Nat. Rev. Cancer 3, 859–868 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. Schreiber, M. et al. Control of cell cycle progression by c-Jun is p53 dependent. Genes Dev. 13, 607–619 (1999).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Logan, M. R., Jordan-Williams, K. L., Poston, S., Liao, J. & Taparowsky, E. J. Overexpression of Batf induces an apoptotic defect and an associated lymphoproliferative disorder in mice. Cell Death Dis. 3, e310 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Quivoron, C. et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell 20, 25–38 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. Couronne, L., Bastard, C. & Bernard, O. A. TET2 and DNMT3A mutations in human T-cell lymphoma. N. Engl. J. Med. 366, 95–96 (2012).

    Article  CAS  PubMed  Google Scholar 

  43. Zhang, X. et al. DNMT3A and TET2 compete and cooperate to repress lineage-specific transcription factors in hematopoietic stem cells. Nat. Genet. 48, 1014–1023 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Riviere, I., Brose, K. & Mulligan, R. C. Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells. Proc. Natl Acad. Sci. USA 92, 6733–6737 (1995).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gallardo, H. F., Tan, C., Ory, D. & Sadelain, M. Recombinant retroviruses pseudotyped with the vesicular stomatitis virus G glycoprotein mediate both stable gene transfer and pseudotransduction in human peripheral blood lymphocytes. Blood 90, 952–957 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Brentjens, R. J. et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat. Med. 9, 279–286 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Brentjens, R. J. et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin. Cancer Res. 13, 5426–5435 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Gong, M. C. et al. Cancer patient T cells genetically targeted to prostate-specific membrane antigen specifically lyse prostate cancer cells and release cytokines in response to prostate-specific membrane antigen. Neoplasia 1, 123–127 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Brentjens, R. J. et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci. Transl Med. 5, 177ra138 (2013).

    Article  Google Scholar 

  51. Stephan, M. T. et al. T cell-encoded CD80 and 4-1BBL induce auto- and transcostimulation, resulting in potent tumor rejection. Nat. Med. 13, 1440–1449 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Stoklasek, T. A., Schluns, K. S. & Lefrancois, L. Combined IL-15/IL-15Rα immunotherapy maximizes IL-15 activity in vivo. J. Immunol. 177, 6072–6080 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Schmidt, M. et al. Detection and direct genomic sequencing of multiple rare unknown flanking DNA in highly complex samples. Hum. Gene Ther. 12, 743–749 (2001).

    Article  CAS  PubMed  Google Scholar 

  55. Gabriel, R. et al. Comprehensive genomic access to vector integration in clinical gene therapy. Nat. Med. 15, 1431–1436 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Paruzynski, A. et al. Genome-wide high-throughput integrome analyses by nrLAM-PCR and next-generation sequencing. Nat. Protoc. 5, 1379–1395 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Afzal, S., Wilkening, S., von Kalle, C., Schmidt, M. & Fronza, R. GENE-IS: time-efficient and accurate analysis of viral integration events in large-scale gene therapy data. Mol. Ther. Nucleic Acids 6, 133–139 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Altschul, S. F., Gish, W., Miller, W., Myers, E. W. & Lipman, D. J. Basic local alignment search tool. J. Mol. Biol. 215, 403–410 (1990).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Sadelain laboratory for helpful discussion and feedback; C. Zebley, B. Youngblood, K. Helin and the Sloan Kettering Institute Centre of Epigenetics Research for advice on epigenetic analysis; J. Boyer for western blot support; N. Socci for advice on exome analysis; M. Schmidt for retroviral integration site analysis; S. Monette and A. Michel from the SKI/CUMC laboratory of Comparative Pathology for conducting pathology analysis; and the following SKI core facilities for their support: Flow Cytometry, Centre of Comparative Medicine and Pathology, Anti-tumour Assessment, Molecular Cytology, Bioinformatics, Integrated Genomics Operation and Cell Therapy and Cell Engineering. Illustrations in Figs. 1a, 2c and 5h and Extended Data Figs. 4a, 7a and 10a were generated using Servier Medical Art. This work was supported by the Pasteur–Weizmann/Servier award, the Leopold Griffuel award, the Leukemia and Lymphoma society (LLS ID: 7014-17) and the MSKCC core grant (P30 CA008748).

Author information

Authors and Affiliations

Authors

Contributions

N.J. and Z.Z. designed the study, performed the experiments, analysed and interpreted data and wrote the manuscript. A.I. and M.L. contributed to RNA-seq and exome analysis. R.K., J.Y. and Y.Z. contributed to ATAC-seq analysis. J.F. and A.D. contributed to animal studies. J.M.-S. contributed to gene targeting. G.G. contributed to vector construction, T cell transduction and animal studies. M.S. designed the study, analysed and interpreted data and wrote the manuscript.

Corresponding author

Correspondence to Michel Sadelain.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Stephen Gottschalk 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 Rv-1928z and Rv-19BBz pre-infusion and in vivo CAR T cell phenotyping.

a,b, Pre-infusion transduction efficiency and phenotyping by flow cytometry of Rv-1928z (a) and Rv-19BBz (b) CAR T cells. c,d, Tumour monitoring of NALM6 bearing mice treated with Rv-1928z (c) and Rv-19BBz (d) CAR T cells. e,f, Bone marrow (e) and Splenic (f) CAR T cell quantification at 3 weeks post infusion. Data is represented as mean±SE [n = 5 (Rv-1928z), n = 6 (Rv-19BBz)]. g,h Differentiation phenotyping of pooled bone marrow CAR T cells at week 3 post infusion. Data from another experiment included in supplementary information. i, CAR T cell inhibitory receptor expression at week 3 post infusion from mouse bone marrow (n = 3). p values were determined by two-sided Mann–Whitney test (e,f) and two-sided χ2 test (h). p < 0.05 was considered statistically significant. p values are denoted: p > 0.05, not significant, NS; *, p < 0.05. Replicate information for g,i are available in Supplementary Table 3. Exact p values are available in Supplementary Table 4.

Source data

Extended Data Fig. 2 Rv-1928z+41BBL and TRAC-1928z pre-infusion and in vivo CAR T cell phenotyping.

a,b, Pre-infusion transduction efficiency and phenotyping by flow cytometry of Rv-1928z+ 41BBL (a) and TRAC-1928z (b) CAR T cells. c,d, Tumour monitoring of NALM6 bearing mice treated with Rv-1928z+41BBL (c) and TRAC-1928z (d) CAR T cells. e,f, Differentiation phenotyping of pooled bone marrow CAR T cells at week 3 post infusion. Data from another experiment included in supplementary information. g, CAR T cell inhibitory receptor expression at week 3 post infusion from mouse bone marrow (n = 3). p values were determined by two-sided χ2 test (f). p < 0.05 was considered statistically significant. p values are denoted: p > 0.05, not significant, NS; *, p < 0.05. Replicate information for e,g are available in Supplementary Table 3.

Source data

Extended Data Fig. 3 Long-term CAR T cell phenotypes upon CRISPR/Cas9 editing of TET2 locus.

a,b, Differentiation phenotyping of retrovirally encoded CAR T cells (day 90) and TRAC-1928z CAR T cells (day 75) isolated from the bone marrow. c, Inhibitory receptor expression of bone marrow Rv-1928z, Rv-19BBz, Rv-1928z+41BBL (day 90) and TRAC-1928z (day 75) CAR T cells. p values were determined by two-sided χ2 test (b). p < 0.05 was considered statistically significant. p values are denoted: p > 0.05, not significant, NS; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001. Replicate information for a,c are available in Supplementary Table 3.

Extended Data Fig. 4 Effect of TET2 editing on CAR T cell accumulation in a prostate cancer model.

a, Schematics of the prostate cancer experimental design. TET2 was edited with the previously discussed gRNA (g1) and an alternative gRNA (g2). PSMA28z+41BBL (PSMA targeted, CD28 costimulated CAR that expresses 41BBL ligand) was used in this study (Dose: 2e5). b, CAR T cell counts in the peripheral blood 30 days post infusion of T cells. Bars show median values. c, Mice with the top 4 CAR T cell peripheral counts at day 30 across both TET2 targeting gRNA (g1, n = 2. g2, n = 2) were euthanized at day 45 along with 5 scrambled gRNA treated PSMA28z+41BBL mice and their splenic CAR T cell numbers were quantified. p values were determined by two-sided Mann-Whitney (b, c) [n = 5 (WT PSMA28z+41BBL), n = 8 (TET2etdg1 PSMA29z+41BBL), n = 11 (TET2etdg2 PSMA29z+41BBL)]. p < 0.05 was considered statistically significant. p values are denoted: *, p < 0.05; **, p < 0.01. Exact p values are available in Supplementary Table 4. The mouse illustration in part a was generated using Servier Medical Art, CC BY 3.0.

Source data

Extended Data Fig. 5 Clonal expansion in all 4 hyper-proliferative CAR T cell populations.

a, Gel image of PCR product for WT CAR T cells and hyper-proliferative TET2-edited CAR T cells. The PCR is designed to amplify the site of gRNA editing. b, Enrichment of TET2-editing from pre-infusion (day 0) in mice to day 21 in Rv-1928z and Rv-1928z+41BBL CAR T cells. p values were determined by two-sided χ2 test. c,d, TCRvβ sequencing reveals hyper-proliferative populations that are dominant for a single clone in TET2bed Rv-1928z (c) and Rv-1928z+41BBL (d). Part of the retroviral vector that was inserted in the TET2 alleles of these clones is highlighted in the figures. eg, Examples of hyper-proliferative Rv-1928z+41BBL CAR T cell populations that are oligoclonal (left panel) with biallelic TET2 editing (right panel). h, Western blot showing total loss of TET2 at protein level in different hyper-proliferative populations. i,j, Examples of oligoclonality in TET2bed TRAC-1928z (i) and Rv-19BBz (j). p < 0.05 was considered statistically significant. p values are denoted: p > 0.05, not significant, NS; *, p < 0.05; **, p < 0.01.

Source data

Extended Data Fig. 6 TCR is dispensable for emergence of hyper-proliferative phenotype in TET2-edited Rv-1928z+41BBL CAR T cells.

a,b, Differentiation phenotyping of TCR+TET2etd RV-1928z+41BBL (a) and TCRTET2etd RV-1928z+41BBL (b) CAR T cells. c, Summary of emergence of hyper-proliferative phenotype post CAR T cell infusion in mice for different donors. Mice were monitored for 90 days. 2e5 CAR T cells were used for both the groups.

Extended Data Fig. 7 Properties of the chimeric antigen receptor design determine composition of TET2bed hyper-proliferative populations.

a, Rv-1928z or Rv-1928z+41BBL CAR T cells were generated from the same donor to assess the effect of CAR design on clonal persistence. 5 Mice were euthanized at day 21 to assess clonal diversity post tumour clearance. 15 mice were followed for emergence of a hyper-proliferative phenotype. b,c, Pair-wise analysis of Rv-1928z (b) and Rv-1928z+41BBL (c) at day 0 and day 21. d, Top 100 Rv-1928z clones at infusion were mapped in the Rv-1928z+41BBL infusion product. These clones were then assessed at day 21 for both the CAR receptors. p values were determined by two-tailed Mann-Whitney test. e,f, Pair-wise analysis (day 0 vs day 90) of the lone hyper-proliferative population found at day 90 for Rv-1928z CAR receptor (e). Representative pair-wise analysis (day 0 vs day 90) of a Rv-1928z+41BBL hyper-proliferative population (f). g, Changes in clonality index over time in Rv-1928z and Rv-1928z+41BBL CAR T cells. h,i, Tracking the fate of the 100 most abundant pre-infusion clones in the hyper-proliferative populations of Rv-1928z (h) and Rv-1928z+41BBL (i). (j) Retro-tracking late-stage dominant clones in the infusion product (Day 0). All dominant clones were isolated at day 90 except for 2-00 which was isolated at day 200. p < 0.05 was considered statistically significant. p values are denoted: p > 0.05, not significant, NS; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001. The human, mouse and lipid bilayer illustrations in part a were generated using Servier Medical Art, CC BY 3.0.

Source data

Extended Data Fig. 8 In vitro and in vivo effector function assessment of TET2-edited and hyper-proliferative TET2bed CAR T cells.

a,b, In vitro cytolytic activity assessment upon co-culture with NALM6 for 16-h as determined by luciferase activity for pre-infusion TET2-edited Rv-1928z (n = 3) and hyper-proliferative TET2bed Rv-1928z (2-2) (n = 3) (a) and pre-infusion TET2-edited Rv-1928z+41BBL (n = 3) and hyper-proliferative TET2bed Rv-1928z+41BBL (17-1) (n = 3) (b). Data is represented as mean±SD. c, NALM6 bearing NSG mice were treated with 2e6 hyper-proliferative TET2bed Rv-1928z (n = 7) or TET2bed Rv-1928z+41BBL (n = 7) CAR T cells to assess their in vivo anti-tumour efficacy. d, Normalized transcript counts of WT Rv-1928z+41BBL and TET2bed Rv-1928z+41BBLCAR T cells isolated from mice at day 90. R=Rest (Transcript counts at isolation). S= Stimulated (Transcript counts 24 h post CD3/28 stimulation). Data is represented as mean±SD (n = 3). e, Schematic of in vitro repeated rechallenge assay for effector function analysis. f,g, Day 1 in vitro cytolytic activity assessment (f) and effector cytokine assessment (g). h,i, Day 8 in vitro cytolytic activity assessment (h) and effector cytokine assessment (i). j,k, Day 15 in vitro cytolytic activity assessment (j) and effector cytokine assessment (k). Data in fk is represented as mean±SD (n = 3). l, TCF1 staining of WT Rv-1928z+41BBL and TET2bed Rv-1928z+41BBL CAR T cells isolated from mice at day 90. WT samples were a pool of 5 mice. TCF1 staining of other hyper-proliferative TET2bed CAR T cells in Supplementary Table 3. p values in a,b,f,h,j were determined by two-sided Student’s unpaired t-test corrected by BKY method. p values in c were determined by two-sided Mann-Whitney test. p values in d,g,i,k were determined by two-sided unpaired t-test. p < 0.05 was considered statistically significant. p values are denoted: p > 0.1, not significant, ns. p < 0.1 are indicated. *, p < 0.05. **, p < 0.01. ***, p < 0.001. ****, p < 0.0001. Exact p values are available in Supplementary Table 4.

Source data

Extended Data Fig. 9 No conserved secondary genetic mutation between different hyper-proliferative TET2bed CAR T populations dominant for a single clone.

a, (Right panel) Copy number changes in TET2bed Rv-1928z+41BBL (17-1). The top panel displays log (ratio) denoted by “(logR)” with chromosomes alternating in the blue and gray. The middle panel displays log (odds-ratio) denoted by “(logOR)”. Segment means are plotted in red lines. In the bottom panel total (black) and minor (red) copy number are plotted for each segment. The bottom bar shows the associated cellular fraction (cf). Dark blue indicates high cf. Light blue indicates low cf. Beige indicates a normal segment (total=2, minor=1). The table shows genetic events occurring at >0.1 cf. (Left panel) CAR T cell clonality as determined by vβ sequencing in TET2bed Rv-1928z+41BBL (17-1). b, Nonsynonymous acquired point mutations in TET2bed Rv-1928z+41BBL (17-1). Mutations that occur at a frequency > ((dominant TCRvβ frequency/2) -0.1) or >0.3 whichever is lower is annotated. c, (Right panel) Copy number changes in TET2bed Rv-1928z (2-2). (Left panel) CAR T cell clonality as determined by vβ sequencing in TET2bed Rv-1928z (2-2). d, Nonsynonymous acquired point mutations in TET2bed Rv-1928z (2-2). e, (Right panel) Copy number changes in TET2bed TRAC-1928z (4-1). (Left panel) CAR T cell clonality as determined by vβ sequencing in TET2bed TRAC-1928z (4-1). f, Nonsynonymous acquired point mutations in TET2bed TRAC-1928z (4-1).

Extended Data Fig. 10 Hyper-proliferative TET2bed Rv-1928z+41BBL do not achieve uncontrolled proliferative state upon secondary transplant.

a, Schematics of secondary transplant of hyper-proliferative TET2bed Rv-1928z+41BBL cells. The exogenous cytokine supplement had to be stopped at day 60 due to deteriorating mice condition in response to frequent injections. b, CAR T cell quantification in peripheral blood under different exogenous supplementation at day 30, day 60 and day 75. Each dot represents a mouse. UD: undetected. Data is represented as mean±SD (n = 5). c, CAR T cell quantification in bone marrow and spleen at day 150 post CAR T cell infusion. Data is represented as mean±SD (n = 5 for no supplement, and IL2. n = 4 for IL7/15). p values were determined by two-sided Mann–Whitney test (b). p < 0.05 was considered statistically significant. p values are denoted: p > 0.05, not significant, NS; *, p < 0.05; **, p < 0.01. (b). Exact p values are available in Supplementary Table 4. The mouse illustration in part a was generated using Servier Medical Art, CC BY 3.0.

Source data

Supplementary information

Supplementary Figures

This file contains Supplementary Figs. 1–4.

Reporting Summary

Supplementary Table 1

Exome analysis of hyper-proliferative TET2bed CAR T cells. 1a, Translocation analysis in TET2bed hyper-proliferative CAR T cells. 1b, Mutation analysis in TET2bed hyper-proliferative CAR T cells. 1c, Copy number analysis in TET2bed hyper-proliferative CAR T cells.

Supplementary Table 2

Retroviral integration site analysis in hyper-proliferative TET2bed CAR T cells. 2a, Retrovirus integration site analysis for TET2bed Rv-1928z+41BBL (17-1). 2b, Retrovirus integration site analysis for TET2bed Rv-1928z (2-2).

Supplementary Table 3

Replicate information on representative figures. 3a-d, Replicate information for selected panels in Extended Data Fig. 1 (a), Extended Data Fig. 2 (b), Extended Data Fig. 3 (c), and Extended Data Fig. 8 (d).

Supplementary Table 4

Exact p values for figures. 4a-i, Exact p values for selected panels in Fig. 2 (a), Fig. 4 (b), Fig. 5 (c), Extended Data Fig. 1 (d), Extended Data Fig. 4 (e), Extended Data Fig. 8 (f), Extended Data Fig. 10 (g), Supplementary Fig. 1 (h), Supplementary Fig. 2 (i).

Supplementary Table 5

List of antibodies used in the flow cytometry.

Supplementary Data

Source Data for Supplementary Fig. 2.

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

Jain, N., Zhao, Z., Feucht, J. et al. TET2 guards against unchecked BATF3-induced CAR T cell expansion. Nature 615, 315–322 (2023). https://doi.org/10.1038/s41586-022-05692-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05692-z

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

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