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:

Clonal expansion of T memory stem cells determines early anti-leukemic responses and long-term CAR T cell persistence in patients

Abstract

Low-affinity CD19 chimeric antigen receptor (CAR) T cells display enhanced expansion and persistence, enabling fate tracking through integration site analysis. Here we show that integration sites from early (1 month) and late (>3 yr) timepoints cluster separately, suggesting different clonal contribution to early responses and prolonged anti-leukemic surveillance. CAR T central and effector memory cells in patients with long-term persistence remained highly polyclonal, whereas diversity dropped rapidly in patients with limited CAR T persistence. Analysis of shared integrants between the CAR T cell product and post-infusion demonstrated that, despite their low frequency, T memory stem cell clones in the product contributed substantially to the circulating CAR T cell pools, during both early expansion and long-term persistence. Our data may help identify patients at risk of early loss of CAR T cells and highlight the critical role of T memory stem cells both in mediating early anti-leukemic responses and in long-term surveillance by CAR T cells.

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: Immunophenotypic characterization of patients with long-lasting CAR T cells in the product and after infusion over time.
Fig. 2: Immunophenotypic characterization of patients with short-living CAR T cells in the product and after infusion over time.
Fig. 3: Distribution and abundance of ISs collected in the product and after infusion over time in Pt4 and Pt6.
Fig. 4: Diversity of ISs in different T cell subtypes over time from Pt4.
Fig. 5: Diversity of ISs in different T cell subtypes over time from Pt6.
Fig. 6: Tracking of IS in different T cell subtypes over time in Pt4 and Pt6.
Fig. 7: Diversity of ISs in different T cell subtypes over time from Pt10 and Pt17.
Fig. 8: Comparison of IS distribution pre- and post-infusion.

Similar content being viewed by others

Data availability

The fastq files relative to sequencing of IS amplicons generated for this study are available through the NCBI repository (https://submit.ncbi.nlm.nih.gov/), BioProject accession number PRJNA718947. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

No new code was generated or used in this manuscript.

References

  1. Sallusto, F., Lenig, D., Förster, R., Lipp, M. & Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature https://doi.org/10.1038/35005534 (1999).

  2. Gattinoni, L. et al. A human memory T cell subset with stem cell-like properties. Nat. Med. 17, 1290–1297 (2011).

    Article  CAS  Google Scholar 

  3. Heslop, H. E. et al. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood https://doi.org/10.1182/blood-2009-08-239186 (2010).

  4. Berger, C. et al. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J. Clin. Invest. https://doi.org/10.1172/JCI32103 (2008).

  5. Biasco, L. et al. In vivo tracking of T cells in humans unveils decade-long survival and activity of genetically modified T memory stem cells. Sci. Transl. Med. 7, 273ra13 (2015).

    Article  Google Scholar 

  6. Oliveira, G. et al. Tracking genetically engineered lymphocytes long-term reveals the dynamics of T cell immunological memory. Sci. Transl. Med. 7, 317ra198 (2015).

    Article  Google Scholar 

  7. Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. https://doi.org/10.1056/NEJMoa1407222 (2014).

  8. Lee, D. W. et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet https://doi.org/10.1016/S0140-6736(14)61403-3 (2015).

  9. Gardner, R. A. et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood https://doi.org/10.1182/blood-2017-02-769208 (2017).

  10. Maude, S. L. et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N. Engl. J. Med. https://doi.org/10.1056/NEJMoa1709866 (2018).

  11. Mueller, K. T. et al. Cellular kinetics of CTL019 in relapsed/refractory B-cell acute lymphoblastic leukemia and chronic lymphocytic leukemia. Blood https://doi.org/10.1182/blood-2017-06-786129 (2017).

  12. Sommermeyer, D. et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia https://doi.org/10.1038/leu.2015.247 (2016).

  13. Turtle, C. J. et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. https://doi.org/10.1172/JCI85309 (2016).

  14. Xu, Y. et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood https://doi.org/10.1182/blood-2014-01-552174 (2014).

  15. Sheih, A. et al. Clonal kinetics and single-cell transcriptional profiling of CAR-T cells in patients undergoing CD19 CAR-T immunotherapy. Nat. Commun. 11, 219 (2020).

    Article  CAS  Google Scholar 

  16. Ghorashian, S. et al. Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat. Med. https://doi.org/10.1038/s41591-019-0549-5 (2019).

  17. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature https://doi.org/10.1038/s41586-018-0178-z (2018).

  18. Aiuti, A. et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science 341, 6148 (2013).

    Article  Google Scholar 

  19. Scala, S. et al. Dynamics of genetically engineered hematopoietic stem and progenitor cells after autologous transplantation in humans. Nat. Med. 24, 1683–1690 (2018).

    Article  CAS  Google Scholar 

  20. Biasco, L. et al. In vivo tracking of human hematopoiesis reveals patterns of clonal dynamics during early and steady-state reconstitution phases. Cell Stem Cell 19, 107–119 (2016).

    Article  CAS  Google Scholar 

  21. Milone, M. C. et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol. Ther. https://doi.org/10.1038/mt.2009.83 (2009).

  22. Long, A. H. et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. https://doi.org/10.1038/nm.3838 (2015).

  23. Biasco, L. Integration site analysis in gene therapy patients: expectations and reality. Hum. Gene Ther. 28, 1122–1129 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was funded the JP Moulton Foundation and supported by the National Institute for Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children NHS Foundation Trust and University College London. P.J.A. is a recipient of an NIHR Research Professorship which also supported S.G. M.P. is supported by the National Institute of Health Research University College London Hospital Biomedical Research Centre. A.J.T. is a recipient of a Wellcome Trust Senior Fellowship. The work of L.B. was supported by the Wellcome Trust (grant no. 104807/Z/14/Z, Principal Research Fellowship awarded to A.J.T.) and the National Institute for Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK. L.B. is also currently Director of R&D for the gene therapy company AVROBIO located in Cambridge, MA, USA (none of what is presented in this work is supported by or relates to AVROBIO). The work of L.B. was also in part performed using the resources of the Gene Therapy Program of the Dana Farber/Boston Children’s Cancer and Blood Disorders Center.

Author information

Authors and Affiliations

Authors

Contributions

L.B. designed and co-supervised the study, performed computational analyses and wrote the manuscript. N.I. and C.R. performed cell isolation and molecular insertion sites retrieval. S.G. and R.R. collected and provided clinical sample material for analysis. A.G. cryopreserved study samples and performed flow cytometric staining. R.H. and R.W. were Principal Investigators for the clinical study. B.P. wrote study documentation and provided trial management. A.L. provided statistical analysis for the study. M.P. generated the CAR construct and participated in its preclinical characterization. A.J.T. conceived the idea and participated in the experimental design and data analysis. P.J.A. supervised the study as PI and wrote the manuscript.

Corresponding author

Correspondence to Persis J. Amrolia.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Justin Eyquem, Alena Gros and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.' after the Supplementary information section.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Immunophenotyping of CD3+ cells in Pt4 and Pt6.

Additional FACS plot on CD62L+CD45RA+CD95 expression in CAR+ T cells, subtype frequencies within total CD3+ cells and relative summary of data on the CD3+ T cell composition overtime for Pt4 (top panels) and Pt6 (bottom panels). Percentages of CD62L/CD45RA compartments are showed inside each gate. Longitudinal contribution of each subpopulations to the CD3+ cell compartment is shown below for each patient according to the color code legend on the right.

Source data

Extended Data Fig. 2 Integration sites collected from Pt4 and Pt6.

Summary of number of IS (grey bars) and relative sequencing reads (white bars) in Pt4 (top panel) and Pt6 (bottom panel) (d = days after treatment).

Source data

Extended Data Fig. 3 Distribution of integration sites collected from Pt4 and Pt6.

Distribution of IS with respect to TSS (left plots) or gene content of the loci (right plots) in the cell product (red) or after infusion at early or late timepoints after treatment (blue) for Pt4 (top panels) and Pt6 (bottom panels).

Extended Data Fig. 4 Gene categories relative to integration sites collected from Pt4 and Pt6.

Plots on the left show word clouds of hit genes in the cell product (in red) and after infusion at early or late timepoints after treatment (in blue) for Pt4 (top panels) and Pt6 (bottom panels). Relative gene enrichment analysis for top biological processes of hit genes relative to each word cloud in the product (red bars) or after infusion (blue bars) is shown on the right plots (significance reported as -log10 binomial p-value from one-tailed tests decreasing from top to bottom).

Extended Data Fig. 5 Relative abundance and diversity of integration sites collected from Pt4 and Pt6.

Scattered dot plots showing relative abundance of IS in the product (a) and at early (b) or late (c) timepoints after treatment in Pt4 (plots on the left) and Pt6 (plots on the right) (d = days after treatment, m = months after treatment). Mean percent abundance is shown as a dotted line for each sample. Number of events in each dataset is equal to what reported in Extended Data Fig. 2. d) Longitudinal plots showing Gini/Simpson Diversity Index (left y-axis) of IS overtime in TSCM (orange lines) and in TCM/TEM (dark blue) and in all T cells (green lines). The grey lines show the percentage of CAR cells overtime (right y-axis).

Source data

Extended Data Fig. 6 Correlation of integration sites and number of cells collected from Pt4 and Pt6.

Longitudinal plots showing number of IS collected (a) and number of cells collected (b) in TSCM (orange lines) and in TCM/TEM (dark blue) at early timepoints for Pt4 (left panels) and Pt6 (right panels). The plot in (c) shows the correlation observed between number of cells collected (y-axis) and clonal diversity for all samples and both patients (x-axis, Shannon Diversity Index) shown as blue dots. Interpolation with best fit curve and R squared value are shown in black.

Source data

Extended Data Fig. 7 Integration sites collected from Pt10 and Pt17.

Summary of number of IS (grey bars) and relative sequencing reads (white bars) in Pt10 (top panel) and Pt17 (bottom panel) (d = days after treatment).

Source data

Extended Data Fig. 8 Sharing of integration sites in the product with the CAR T populations in Pt10 and Pt17 after infusion.

Ring plots showing relative contribution from each subtype (coloured section of the ring) to the pool of IS detected in the product and at early timepoints in Pt10 (left) and Pt6 (right). The total number of IS captured both in the product and after infusion is shown inside each plot. The relative percentage of IS belonging to each T cell subtype of the product that were shared with samples after infusion is shown in white inside each section of each ring plot.

Supplementary information

Source data

Source Data Fig. 1

Raw datasets relative to figure panels.

Source Data Fig. 2

Raw datasets relative to figure panels.

Source Data Fig. 4

Raw datasets relative to figure panels.

Source Data Fig. 5

Raw datasets relative to figure panels.

Source Data Fig. 6

Raw datasets relative to figure panels.

Source Data Fig. 7

Raw datasets relative to figure panels.

Source Data Fig. 8

Raw datasets relative to figure panels.

Source Data Extended Data Fig. 1

Raw datasets relative to figure panels.

Source Data Extended Data Fig. 2

Raw datasets relative to figure panels.

Source Data Extended Data Fig. 5

Raw datasets relative to figure panels.

Source Data Extended Data Fig. 6

Raw datasets relative to figure panels.

Source Data Extended Data Fig. 7

Raw datasets relative to figure panels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Biasco, L., Izotova, N., Rivat, C. et al. Clonal expansion of T memory stem cells determines early anti-leukemic responses and long-term CAR T cell persistence in patients. Nat Cancer 2, 629–642 (2021). https://doi.org/10.1038/s43018-021-00207-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00207-7

This article is cited by

Search

Quick links

Nature Briefing: Cancer

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

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