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:

Screening for CD19-specific chimaeric antigen receptors with enhanced signalling via a barcoded library of intracellular domains

An Author Correction to this article was published on 03 February 2023

This article has been updated

Abstract

The immunostimulatory intracellular domains (ICDs) of chimaeric antigen receptors (CARs) are essential for converting antigen recognition into antitumoural function. Although there are many possible combinations of ICDs, almost all current CARs rely on combinations of CD3𝛇, CD28 and 4-1BB. Here we show that a barcoded library of 700,000 unique CD19-specific CARs with diverse ICDs cloned into lentiviral vectors and transduced into Jurkat T cells can be screened at high throughput via cell sorting and next-generation sequencing to optimize CAR signalling for antitumoural functions. By using this screening approach, we identified CARs with new ICD combinations that, compared with clinically available CARs, endowed human primary T cells with comparable tumour control in mice and with improved proliferation, persistence, exhaustion and cytotoxicity after tumour rechallenge in vitro. The screening strategy can be adapted to other disease models, cell types and selection conditions, and could be used to improve adoptive cell therapies and to expand their utility to new disease indications.

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: Selecting for CD69high expression enriches CARs encompassing ITAM-signalling ICDs.
Fig. 2: CARPOOL selections reveal new functional signalling domain configurations.
Fig. 3: CAR variants show enhanced cytotoxicity and cytokine secretion in response to antigen stimulation.
Fig. 4: CAR variants exhibit distinct proliferation and exhaustion profiles upon repeated antigen exposure.
Fig. 5: CARs identified by CARPOOL differ in transcriptional profile and predicted efficacy correlates.
Fig. 6: CAR variants show tumour control similar to that of a BBζ CAR in vivo.

Similar content being viewed by others

Data availability

The NGS selection datasets have been deposited in the Sequence Read Archive and are available under the accession number PRJNA744269. The scRNA-seq data have been deposited in the Gene Expression Omnibus under accession number GSE179767. All data generated or analysed during the study (including the DomainSeq-processed CARPOOL selection data) are included in the paper or its supplementary information.

Code availability

The code used to analyse the domain composition of selected CARs can be accessed in the DomainSeq repository at https://github.com/birnbaumlab/Gordon-et-al-2022.

Change history

References

  1. Waldman, A. D., Fritz, J. M. & Lenardo, M. J. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 20, 651–668 (2020).

    Article  CAS  Google Scholar 

  2. 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  Google Scholar 

  3. Lesch, S. et al. Determinants of response and resistance to CAR T cell therapy. Semin. Cancer Biol. 65, 80–90 (2020).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. Prinzing, B. et al. MyD88/CD40 signaling retains CAR T cells in a less differentiated state. JCI Insight 5, e136093 (2020).

    Article  Google Scholar 

  6. Kagoya, Y. et al. A novel chimeric antigen receptor containing a JAK–STAT signaling domain mediates superior antitumor effects. Nat. Med. 24, 352–359 (2018).

    Article  CAS  Google Scholar 

  7. Wu, W. et al. Multiple signaling roles of CD3ε and its application in CAR-T cell therapy. Cell 182, 855–871.e23 (2020).

    Article  CAS  Google Scholar 

  8. Feucht, J. et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat. Med. 25, 82–88 (2019).

    Article  CAS  Google Scholar 

  9. Zhong, X.-S., Matsushita, M., Plotkin, J., Riviere, I. & Sadelain, M. Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol. Ther. 18, 413–420 (2010).

    Article  CAS  Google Scholar 

  10. Guedan, S. et al. Enhancing CAR T cell persistence through ICOS and 4-1BB costimulation. JCI Insight 3, e96976 (2018).

    Article  Google Scholar 

  11. 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  Google Scholar 

  12. Testi, R., Phillips, J. H. & Lanier, L. L. Leu 23 induction as an early marker of functional CD3/T cell antigen receptor triggering. Requirement for receptor cross-linking, prolonged elevation of intracellular [Ca++] and stimulation of protein kinase C. J. Immunol. 142, 1854–1860 (1989).

    Article  CAS  Google Scholar 

  13. Ziegler, S. F., Ramsdell, F. & Alderson, M. R. The activation antigen CD69. Stem Cells 12, 456–465 (1994).

    Article  CAS  Google Scholar 

  14. Underhill, D. M. & Goodridge, H. S. The many faces of ITAMs. Trends Immunol. 28, 66–73 (2007).

    Article  CAS  Google Scholar 

  15. Lindner, S. E., Johnson, S. M., Brown, C. E. & Wang, L. D. Chimeric antigen receptor signaling: functional consequences and design implications. Sci. Adv. 6, eaaz3223 (2020).

    Article  CAS  Google Scholar 

  16. Hombach, A. A., Heiders, J., Foppe, M., Chmielewski, M. & Abken, H. OX40 costimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirected CD4+ T cells. Oncoimmunology 1, 458–466 (2012).

    Article  Google Scholar 

  17. Zhang, H. et al. A chimeric antigen receptor with antigen-independent OX40 signaling mediates potent antitumor activity. Sci. Transl. Med. 13, eaba7308 (2021).

    Article  CAS  Google Scholar 

  18. Wang, E. et al. Generation of potent T-cell immunotherapy for cancer using DAP12-based, multichain, chimeric immunoreceptors. Cancer Immunol. Res 3, 815–826 (2015).

    Article  CAS  Google Scholar 

  19. Ng, Y.-Y. et al. T cells expressing NKG2D CAR with a DAP12 signaling domain stimulate lower cytokine production while effective in tumor eradication. Mol. Ther. 29, 75–85 (2021).

    Article  CAS  Google Scholar 

  20. Töpfer, K. et al. DAP12-based activating chimeric antigen receptor for NK cell tumor immunotherapy. J. Immunol. 194, 3201–3212 (2015).

    Article  Google Scholar 

  21. Long, A. H. et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat. Med. 21, 581–590 (2015).

    Article  CAS  Google Scholar 

  22. Ajina, A. & Maher, J. Strategies to address chimeric antigen receptor tonic signaling. Mol. Cancer Ther. 17, 1795–1815 (2018).

    Article  CAS  Google Scholar 

  23. Shimabukuro-Vornhagen, A. et al. Cytokine release syndrome. J. Immunother. Cancer 6, 56 (2018).

    Article  Google Scholar 

  24. Alizadeh, D. et al. IFNγ is critical for CAR T cell-mediated myeloid activation and induction of endogenous immunity. Cancer Discov. 11, 2248–2265 (2021).

    Article  CAS  Google Scholar 

  25. Lynch, D. H. et al. Flt3 ligand induces tumor regression and antitumor immune responses in vivo. Nat. Med. 3, 625–631 (1997).

    Article  CAS  Google Scholar 

  26. Salmon, H. et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 44, 924–938 (2016).

    Article  CAS  Google Scholar 

  27. Trifilo, M. J., Bergmann, C. C., Kuziel, W. A. & Lane, T. E. CC chemokine ligand 3 (CCL3) regulates CD8+-T-cell effector function and migration following viral infection. J. Virol. 77, 4004–4014 (2003).

    Article  CAS  Google Scholar 

  28. Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).

    Article  CAS  Google Scholar 

  29. Philipson, B. I. et al. 4-1BB costimulation promotes CAR T cell survival through noncanonical NF-κB signaling. Sci. Signal. 13, eaay8248 (2020).

    Article  CAS  Google Scholar 

  30. Rowe, A. M. et al. A cell-intrinsic requirement for NF-κB-inducing kinase in CD4 and CD8 T cell memory. J. Immunol. 191, 3663–3672 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. Zhou, X. et al. Differentiation and persistence of memory CD8+ T cells depend on T cell factor 1. Immunity 33, 229–240 (2010).

    Article  CAS  Google Scholar 

  33. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  Google Scholar 

  34. Boroughs, A. C. et al. A distinct transcriptional program in human CAR T cells bearing the 4-1BB signaling domain revealed by scRNA-Seq. Mol. Ther. 28, 2577–2592 (2020).

    Article  CAS  Google Scholar 

  35. Macian, F. NFAT proteins: key regulators of T-cell development and function. Nat. Rev. Immunol. 5, 472–484 (2005).

    Article  CAS  Google Scholar 

  36. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789.e18 (2019).

    Article  CAS  Google Scholar 

  37. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).

    Article  CAS  Google Scholar 

  38. 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. https://doi.org/10.1038/s41591-020-1061-7 (2020).

  39. Duong, C. P. M. et al. Engineering T cell function using chimeric antigen receptors identified using a DNA library approach. PLoS ONE 8, e63037 (2013).

    Article  CAS  Google Scholar 

  40. Hartl, F. A. et al. Noncanonical binding of Lck to CD3ε promotes TCR signaling and CAR function. Nat. Immunol. 21, 902–913 (2020).

    Article  CAS  Google Scholar 

  41. Eshhar, Z., Waks, T., Bendavid, A. & Schindler, D. G. Functional expression of chimeric receptor genes in human T cells. J. Immunol. Methods 248, 67–76 (2001).

    Article  CAS  Google Scholar 

  42. Hwu, P. et al. Lysis of ovarian cancer cells by human lymphocytes redirected with a chimeric gene composed of an antibody variable region and the Fc receptor gamma chain. J. Exp. Med. 178, 361–366 (1993).

    Article  CAS  Google Scholar 

  43. Pulè, M. A. et al. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol. Ther. 12, 933–941 (2005).

    Article  Google Scholar 

  44. Hombach, A. A. & Abken, H. Costimulation by chimeric antigen receptors revisited the T cell antitumor response benefits from combined CD28-OX40 signalling. Int. J. Cancer 129, 2935–2944 (2011).

    Article  CAS  Google Scholar 

  45. Bartelt, R. R., Cruz-Orcutt, N., Collins, M. & Houtman, J. C. D. Comparison of T cell receptor-induced proximal signaling and downstream functions in immortalized and primary T cells. PLoS ONE 4, e5430 (2009).

    Article  Google Scholar 

  46. Abraham, R. T. & Weiss, A. Jurkat T cells and development of the T-cell receptor signalling paradigm. Nat. Rev. Immunol. 4, 301–308 (2004).

    Article  CAS  Google Scholar 

  47. Zhao, R., Cui, Y., Li, S., Qin, L. & Li, P. Current status and hurdles for CAR-T cell immune therapy. Blood Sci. 1, 148–155 (2019).

    Article  Google Scholar 

  48. Siegler, E. L. & Wang, P. Preclinical models in chimeric antigen receptor-engineered T-cell therapy. Hum. Gene Ther. 29, 534–546 (2018).

    Article  CAS  Google Scholar 

  49. Kato, D. et al. GPC1 specific CAR-T cells eradicate established solid tumor without adverse effects and synergize with anti-PD-1 Ab. eLife 9, e49392 (2020).

    Article  CAS  Google Scholar 

  50. D’Aloia, M. M., Zizzari, I. G., Sacchetti, B., Pierelli, L. & Alimandi, M. CAR-T cells: the long and winding road to solid tumors. Cell Death Dis. 9, 282 (2018).

    Article  Google Scholar 

  51. Newick, K., Moon, E. & Albelda, S. M. Chimeric antigen receptor T-cell therapy for solid tumors. Mol. Ther. Oncolytics 3, 16006 (2016).

    Article  CAS  Google Scholar 

  52. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24, 541–550 (2018).

    Article  CAS  Google Scholar 

  53. Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 38, 947–953 (2020).

    Article  CAS  Google Scholar 

  54. Basar, R., Daher, M. & Rezvani, K. Next-generation cell therapies: the emerging role of CAR-NK cells. Hematol. Am. Soc. Hematol. Educ. Program 2020, 570–578 (2020).

    Article  Google Scholar 

  55. Stoeckius, M. et al. Cell hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    Article  CAS  Google Scholar 

  56. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Koch Institute’s Robert A. Swanson (1969) Biotechnology Center for their technical support, especially the Flow Cytometry Facility, Preclinical Modeling, Imaging and Testing Core, MIT BioMicro Center and High Throughput Sciences Core. We thank G. Paradis, P. Chamberlain, H. Holcombe, V. Spanoudaki and S. Levine for many helpful discussions and suggestions. We also thank Y. Chen for providing the sequence for the 19BB𝛇 CAR. M.E.B. was supported by a Packard Fellowship, a Pew-Stewart Scholarship and a grant from the Deshpande Center. D.J.I. was supported by the Mark Foundation, the National Institutes of Health (R01-CA247632) and the Bridge Project, a partnership between the Koch Institute for Integrative Cancer Research at MIT and the Dana-Farber/Harvard Cancer Center. D.J.I. is an investigator of the Howard Hughes Medical Institute. D.A.L. was supported by US Army Research Office Cooperative Agreement W911NF-19-2-0026 Institute for Collaborative Biotechnologies. This work was supported in part by the Koch Institute Frontier Research Program (to M.E.B. and M.T.H.), and the Koch Institute Support (core) Grant P30-CA14051 from the National Cancer Institute. This work was additionally supported by a fellowship from Human Frontier Science Program to T.K., National Science Foundation Graduate Research Fellowships to K.S.G., C.R.P. and P.V.H., a Paul and Daisy Soros Fellowship to A.R., and support from the National Institute of General Medical Sciences (T32-GM007753) to A.Q.Z. and C.K. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute of General Medical Sciences or the National Institutes of Health. This research is supported in part by the National Research Foundation, Prime Minister’s Office, Singapore under its Campus for Research Excellence and Technological Enterprise (CREATE) programme, through Singapore MIT Alliance for Research and Technology (SMART): Critical Analytics for Manufacturing Personalised-Medicine (CAMP) Inter-Disciplinary Research Group.

Author information

Authors and Affiliations

Authors

Contributions

T.K. and M.E.B. conceived the project. K.S.G., T.K., C.R.P., A.R., A.Q.Z., Y.A., Y.L., C.K. and A.S. conducted experiments. K.S.G., T.K., C.R.P, P.V.H, A.R. and B.J. performed data analyses. D.A.L., M.T.H., D.J.I. and M.E.B. supervised the work. K.S.G., T.K., C.R.P. and M.E.B. wrote the manuscript. All authors contributed to the editing of the manuscript.

Corresponding author

Correspondence to Michael E. Birnbaum.

Ethics declarations

Competing interests

The library approach described in this paper is the subject of a US patent application (PCT/US2020/017794) with T.K. and M.E.B. as inventors. M.E.B. is a founder, consultant and equity holder of Kelonia Therapeutics and Abata Therapeutics. T.K. is presently an employee of Gingko Bioworks. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks the anonymous reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

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 ICD frequency post-selection for 1st and 2nd generation CARs.

Related to Fig. 2b. Heatmap representing log2(fold-change) of individual ICDs, oriented by ICD position within the CAR relative to the plasma membrane. Fold-changes were normalized to frequencies in the EGFP+ sorted population prior to selection.

Extended Data Fig. 2 Characterization of CAR variant expression and function in Jurkat T cells.

a, Design of CD19 targeted CAR candidates. b, CAR surface expression (n = 1), (c) internalization (n = 1), and (d) CD69 upregulation upon antigen stimulation with recombinant human CD19 (n = 6 technical replicates representative of 2 biological replicates). e, Basal CD69 (activation) and (f) PD-1 expression of unstimulated CAR-T cells (n = 7 technical replicates representative of 2 biological replicates). P values shown in panel (e) are 0.002 for Var1 vs. BB𝛇 and 0.0149 for Var2 vs. BB𝛇. P values shown in panel (f) are 0.0117 for Var1 vs. BB𝛇, 0.0110 for Var2 vs. BB𝛇, and 0.0192 for Var3 vs. BB𝛇, as determined by two-tailed unpaired student’s t tests (df = 12). Data shown are individual values in panel (c), while panel (d) shows means ± s.e.m. Panels (e,f) show individual values along with means ± s.e.m.

Source data

Extended Data Fig. 3 Memory and exhaustion phenotypes following tumour rechallenge.

Related to Fig. 4d, e. a, FACS plots displaying changes in CAR-T cell memory differentiation following repeated tumour challenge (days 4 and 18 shown). b, LAG-3 expression following repeated tumour challenge on day 22 (n = 3 technical replicates representative of 2 biological replicates). P values are 0.0465 for CD4+ Var3 vs. BB𝛇, and 0.0358 for CD8+ Var1 vs. BB𝛇, and 0.0112 for CD8+ Var3 vs. BB𝛇 (df = 4 for all). P values in panel (b) were determined using an unpaired student’s t test. Data shown are means ± s.e.m.

Source data

Extended Data Fig. 4 Expression of CD4 and CD8A across different phenotypic clusters.

Differential of CD4 (left) and CD8A (right) expression as determined by single cell sequencing as shown in Fig. 5. Data points shown represent individual cells.

Extended Data Fig. 5 Differential gene expression between Var1 and 19BBζ-expressing CAR-T cells.

Volcano plot showing differentially expressed genes between Var1 (n = 863 cells) and 19BB𝛇-expressing (n = 637 cells) CAR-T cells 48 hours after a third NALM6 rechallenge. P values for each gene were determined using a two-sided Wilcoxon Rank-Sum test. Genes with both a Bonferroni-corrected P value less than 0.05 and an average log2FC of > 1 are highlighted.

Extended Data Fig. 6 High dose Var1, Var3, and BBζ achieve sustained remission.

a, Experimental design. NSG mice were infused intravenously with 5 × 105 FLuc+ CD19+ NALM6 cells, then treated i.v. with 1 × 106 of a 1:1 mixture of human CD8+ and CD4+ CAR-T cells or untransduced control T cells (n = 5 mice for all groups). b, Kaplan-Meier curve for overall survival. Tumour burden was assessed by measuring luminescent activity. c, Quantification of total photon counts are shown. d, Weight loss following ACT was monitored routinely. Data shown in panels (c,d) are individual values.

Source data

Extended Data Fig. 7 High dose BBζ and Var1 confer partial protection against B-ALL recurrence.

a, Experimental design. NSG mice were infused intravenously with 5 × 105 FLuc+ CD19+ NALM6 cells, then treated i.v. with 1 × 106 of a 1:1 mixture of human CD8+ and CD4+ CAR-T cells or untransduced control T cells (n = 5 mice for all groups). Mice were then rechallenged with 5 × 105 FLuc+ CD19+ NALM6 cells on days 44 and 58 post ACT (indicated with arrows). b, Kaplan-Meier curve for overall survival. Tumour burden was assessed by measuring luminescent activity. c, Quantification of total photon counts is shown. d, Weight loss following ACT was monitored routinely. Data shown in panels (c,d) are individual values. Arrows in panels (b–d) indicate dates of NALM6 rechallenge. e, CD19+ NALM6 cells and (f) EGFP+ CAR-T cells were harvested from the peripheral blood (PB), spleen (SP), and bone marrow (BM) of a separate cohort of mice that received the same treatment regimen at day 14 post ACT and quantified via FACS analysis (n = 3). Data shown are individuals with means ± s.e.m.

Source data

Supplementary information

Supplementary Information

Supplementary figures.

Reporting Summary

Peer Review File

Supplementary Table 1

Amino acid compositions of intracellular signalling domains incorporated into the CAR library.

Supplementary Table 2

Barcode frequencies and metadata from NGS of selected CAR-library-expressing Jurkats.

Supplementary Table 3

List of significantly differentially overexpressed genes in each cluster following scRNA-seq.

Supplementary Table 4

Differentially expressed genes between all Var1-expressing cells and all 19BBz-expressing cells.

Supplementary Table 5

List of T-cell-specific curated gene sets used for scGSVA analysis of transcriptional clusters.

Source data

Source Data for Fig. 1

Source data for Fig. 1.

Source Data for Fig. 2

Source data for Fig. 2.

Source Data for Fig. 3

Source data for Fig. 3.

Source Data for Fig. 4

Source data for Fig. 4.

Source Data for Fig. 5

Source data for Fig. 5.

Source Data for Fig. 6

Source data for Fig. 6.

Source Data for ED Fig. 2

Source data for Extended Fig. 2.

Source Data for ED Fig. 3

Source data for Extended Fig. 3.

Source Data for ED Fig. 6

Source data for Extended Fig. 6.

Source Data for ED Fig. 7

Source data for Extended Fig. 7.

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

Gordon, K.S., Kyung, T., Perez, C.R. et al. Screening for CD19-specific chimaeric antigen receptors with enhanced signalling via a barcoded library of intracellular domains. Nat. Biomed. Eng 6, 855–866 (2022). https://doi.org/10.1038/s41551-022-00896-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-022-00896-0

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research