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.

  • Resource
  • Published:

Repeated mRNA vaccination sequentially boosts SARS-CoV-2-specific CD8+ T cells in persons with previous COVID-19

Abstract

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hybrid immunity is more protective than vaccination or previous infection alone. To investigate the kinetics of spike-reactive T (TS) cells from SARS-CoV-2 infection through messenger RNA vaccination in persons with hybrid immunity, we identified the T cell receptor (TCR) sequences of thousands of index TS cells and tracked their frequency in bulk TCRβ repertoires sampled longitudinally from the peripheral blood of persons who had recovered from coronavirus disease 2019 (COVID-19). Vaccinations led to large expansions in memory TS cell clonotypes, most of which were CD8+ T cells, while also eliciting diverse TS cell clonotypes not observed before vaccination. TCR sequence similarity clustering identified public CD8+ and CD4+ TCR motifs associated with spike (S) specificity. Synthesis of longitudinal bulk ex vivo single-chain TCRβ repertoires and paired-chain TCRɑβ sequences from droplet sequencing of TS cells provides a roadmap for the rapid assessment of T cell responses to vaccines and emerging pathogens.

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: Vaccines expand previously detected and low-abundance clonotypes.
Fig. 2: Longitudinal kinetics of S-reactive clonotypes defined by AIM-scTCRαβ-seq from after infection to before vaccination.
Fig. 3: Trajectories of S-reactive clonotypes defined by AIM-scTCRαβ-seq differentiate CD4+ and CD8+ T cells.
Fig. 4: TCRαβ sequence similarity network shows public CD8+ T cell responses among sequences recovered by AIM-scTCRαβ-seq.
Fig. 5: Transgenic CD8+ T cell-origin TCRs from five public clusters are activated in the context of HLA-A*03:01 and the KCY epitope.
Fig. 6: TRB sequence-defined metrics and nAbs are associated with disease severity before and after mRNA vaccination.

Similar content being viewed by others

Data availability

The whole PBMC and nasal TRB repertoires are available at https://doi.org/10.5281/zenodo.7698787. Sorted naive and memory PBMC T cell subset TRB repertoires from the time point E00, sorted total CD4+ T cells and AIM-sorted CD4+ T cell subsets from the time point E03 are available at https://doi.org/10.5281/zenodo.7686500. Processed single-cell CD69+CD137+ AIM-scTCRɑβ-seq and feature barcode oligonucleotide-labeled mAb data are available at https://zenodo.org/record/6909380. The flow cytometry results from the intracellular cytokine staining of CD4+ and CD8+ S-reactive T cells are available at https://doi.org/10.5281/zenodo.8088178. The sequences of CD8+ and CD4+ T cell-origin TCRs expressed in reporter cells are available from GenBank (OP245920-OP245935 and OR239787-OR239798, respectively). The reference dataset for Cell Ranger used was GRCh-Alts-ensembl-5.0.0 and is available at 10xgenomics.com/support/software/cell-ranger/downloads. Source data are provided with this paper.

Code availability

The code used to analyze and present the data is based on Python v.3.8 or R v.4.1.2 and is available at https://github.com/kmayerb/NIA34780B. The availability of the tool used to classify PBMC TRB repertoires for evidence of CMV infection is discussed at https://www.immunoseq.com/cmv-classifier/.

References

  1. Goldberg, Y. et al. Protection and waning of natural and hybrid immunity to SARS-CoV-2. N. Engl. J. Med. 386, 2201–2212 (2022).

    CAS  PubMed  Google Scholar 

  2. Suarez Castillo, M., Khaoua, H. & Courtejoie, N. Vaccine-induced and naturally-acquired protection against Omicron and Delta symptomatic infection and severe COVID-19 outcomes, France, December 2021 to January 2022. Euro Surveill. 27, 2200250 (2022).

    PubMed  PubMed Central  Google Scholar 

  3. Qu, P. et al. Durability of booster mRNA vaccine against SARS-CoV-2 BA.2.12.1, BA.4, and BA.5 subvariants. N. Engl. J. Med. 387, 1329–1331 (2022).

    PubMed  Google Scholar 

  4. Lim, J. M. E. et al. SARS-CoV-2 breakthrough infection in vaccinees induces virus-specific nasal-resident CD8+ and CD4+ T cells of broad specificity. J. Exp. Med. 219, e2020780 (2022).

    Google Scholar 

  5. Peng, Y. et al. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat. Immunol. 21, 1336–1345 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Tavukcuoglu, E., Horzum, U., Cagkan Inkaya, A., Unal, S. & Esendagli, G. Functional responsiveness of memory T cells from COVID-19 patients. Cell. Immunol. 365, 104363 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Rodda, L. B. et al. Functional SARS-CoV-2-specific immune memory persists after mild COVID-19. Cell 184, 169–183 (2021).

    CAS  PubMed  Google Scholar 

  8. Dykema, A. G. et al. SARS-CoV-2 vaccination diversifies the CD4+ spike-reactive T cell repertoire in patients with prior SARS-CoV-2 infection. eBioMedicine 80, 104048 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Minervina, A. A. et al. SARS-CoV-2 antigen exposure history shapes phenotypes and specificity of memory CD8+ T cells. Nat. Immunol. 23, 781–790 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Kared, H. et al. Immune responses in Omicron SARS-CoV-2 breakthrough infection in vaccinated adults. Nat. Commun. 13, 4165 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Altarawneh, H. N. et al. Effects of previous infection and vaccination on symptomatic Omicron infections. N. Engl. J. Med. 387, 21–34 (2022).

    CAS  PubMed  Google Scholar 

  12. Keeton, R. et al. T cell responses to SARS-CoV-2 spike cross-recognize Omicron. Nature 603, 488–492 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Gao, Y. et al. Ancestral SARS-CoV-2-specific T cells cross-recognize the Omicron variant. Nat. Med. 28, 472–476 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Dolton, G. et al. Emergence of immune escape at dominant SARS-CoV-2 killer T cell epitope. Cell 185, 2936–2951 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Naranbhai, V. et al. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all individuals. Cell 185, 1041–1051 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Müller, N. F. et al. Viral genomes reveal patterns of the SARS-CoV-2 outbreak in Washington State. Sci. Transl. Med. 13, eabf0202 (2021).

    PubMed  PubMed Central  Google Scholar 

  17. Mueller, Y. M. et al. Stratification of hospitalized COVID-19 patients into clinical severity progression groups by immuno-phenotyping and machine learning. Nat. Commun. 13, 915 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Elyanow, R. et al. T cell receptor sequencing identifies prior SARS-CoV-2 infection and correlates with neutralizing antibodies and disease severity. JCI Insight 7, e150070 (2022).

    PubMed  PubMed Central  Google Scholar 

  19. Zhang, J. Z. et al. Thermodynamically coupled biosensors for detecting neutralizing antibodies against SARS-CoV-2 variants. Nat. Biotechnol. 40, 1336–1340 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Johansson, A. M. et al. Cross-reactive and mono-reactive SARS-CoV-2 CD4+ T cells in prepandemic and COVID-19 convalescent individuals. PLoS Pathog. 17, e1010203 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Boonyaratanakornkit, J. et al. Clinical, laboratory, and temporal predictors of neutralizing antibodies against SARS-CoV-2 among COVID-19 convalescent plasma donor candidates. J. Clin. Invest. 131, e144930 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Reiss, S. et al. Comparative analysis of activation induced marker (AIM) assays for sensitive identification of antigen-specific CD4 T cells. PLoS ONE 12, e0186998 (2017).

    PubMed  PubMed Central  Google Scholar 

  23. Glanville, J. et al. Identifying specificity groups in the T cell receptor repertoire. Nature 547, 94–98 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Dash, P. et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature 547, 89–93 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Oberhardt, V. et al. Rapid and stable mobilization of CD8+ T cells by SARS-CoV-2 mRNA vaccine. Nature 597, 268–273 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Francis, J. M. et al. Allelic variation in class I HLA determines CD8+ T cell repertoire shape and cross-reactive memory responses to SARS-CoV-2. Sci. Immunol. 7, eabk3070 (2022).

    CAS  PubMed  Google Scholar 

  27. Shomuradova, A. S. et al. SARS-CoV-2 epitopes are recognized by a public and diverse repertoire of human T cell receptors. Immunity 53, 1245–1257 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Szeto, C. et al. Molecular basis of a dominant SARS-CoV-2 spike-derived epitope presented by HLA-A*02:01 recognised by a public TCR. Cells 10, 2646 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Ferretti, A. P. et al. Unbiased screens show CD8+ T cells of COVID-19 patients recognize shared epitopes in SARS-CoV-2 that largely reside outside the spike protein. Immunity 53, 1095–1107 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Robins, H. S. et al. Overlap and effective size of the human CD8+ T cell receptor repertoire. Immunology 2, 47ra64 (2010).

    Google Scholar 

  31. Sethna, Z. et al. OLGA: fast computation of generation probabilities of B- and T-cell receptor amino acid sequences and motifs. Bioinformatics 35, 2974–2981 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Flament, H. et al. Outcome of SARS-CoV-2 infection is linked to MAIT cell activation and cytotoxicity. Nat. Immunol. 22, 322–335 (2021).

    CAS  PubMed  Google Scholar 

  33. Parrot, T. et al. MAIT cell activation and dynamics associated with COVID-19 disease severity. Sci. Immunol. 5, eabe1670 (2020).

    PubMed  PubMed Central  Google Scholar 

  34. Boulouis, C. et al. MAIT cell compartment characteristics are associated with the immune response magnitude to the BNT162b2 mRNA anti-SARS-CoV-2 vaccine. Mol. Med. 28, 54 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Le Gall, S., Stamegna, P. & Walker, B. D. Portable flanking sequences modulate CTL epitope processing. J. Clin. Invest. 117, 3563–3575 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Saggau, C. et al. The pre-exposure SARS-CoV-2-specific T cell repertoire determines the quality of the immune response to vaccination. Immunity 55, 1924–1939 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Mudd, P. A. et al. SARS-CoV-2 mRNA vaccination elicits a robust and persistent T follicular helper cell response in humans. Cell 185, 603–613 (2022).

    CAS  PubMed  Google Scholar 

  38. Pogorelyy, M. V. et al. Resolving SARS-CoV-2 CD4+ T cell specificity via reverse epitope discovery. Cell Rep. Med. 3, 100697 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Gittelman, R. M. et al. Longitudinal analysis of T cell receptor repertoires reveals shared patterns of antigen-specific response to SARS-CoV-2 infection. JCI Insight 7, e151849 (2022).

    PubMed  PubMed Central  Google Scholar 

  40. Carlson, C. S. et al. Using synthetic templates to design an unbiased multiplex PCR assay. Nat. Commun. 4, 2680 (2013).

    PubMed  Google Scholar 

  41. Robins, H. S. et al. Comprehensive assessment of T-cell receptor β-chain diversity in α-β T cells. Blood 114, 4099–4107 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Xu, A. M. et al. Differences in SARS-CoV-2 vaccine response dynamics between class-I- and class-II-specific T-cell receptors in inflammatory bowel disease. Front. Immunol. 13, 880190 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Robins, H. et al. Ultra-sensitive detection of rare T cell clones. J. Immunol. Methods 375, 14–19 (2012).

    CAS  PubMed  Google Scholar 

  44. Xu, J. et al. T cell receptor β repertoires in patients with COVID-19 reveal disease severity signatures. Front. Immunol. 14, 1190844 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Pothast, C. R. et al. SARS-CoV-2-specific CD4+ and CD8+ T cell responses can originate from cross-reactive CMV-specific T cells. eLife 11, e82050 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Jo, N. et al. Aging and CMV infection affect pre-existing SARS-CoV-2-reactive CD8+ T cells in unexposed individuals. Front. Aging 2, 719342 (2021).

    PubMed  PubMed Central  Google Scholar 

  47. Emerson, R. O. et al. Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire. Nat. Genet. 49, 659–665 (2017).

    CAS  PubMed  Google Scholar 

  48. Boppana, S. et al. SARS-CoV-2-specific circulating T follicular helper cells correlate with neutralizing antibodies and increase during early convalescence. PLoS Pathog. 17, e1009761 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Painter, M. M. et al. Rapid induction of antigen-specific CD4+ T cells is associated with coordinated humoral and cellular immunity to SARS-CoV-2 mRNA vaccination. Immunity 54, 2133–2142 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Ngalamika, O., Kawimbe, M. & Mukasine, M. C. Expression of CD40L on CD4+ T cells distinguishes active versus inactive HIV-associated Kaposi’s sarcoma. Cancer Treat. Res. Commun. 27, 100361 (2021).

    PubMed  PubMed Central  Google Scholar 

  51. Curato, C. et al. Frequencies and TCR repertoires of human 2,4,6-trinitrobenzenesulfonic acid-specific T cells. Front. Toxicol. 4, 827109 (2022).

    PubMed  PubMed Central  Google Scholar 

  52. Sette, A. & Crotty, S. Adaptive immunity to SARS-CoV-2 and COVID-19. Cell 184, 861–880 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. DeWitt, W. S. et al. Dynamics of the cytotoxic T cell response to a model of acute viral infection. J. Virol. 89, 4517–4526 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Pogorelyy, M. V. et al. Precise tracking of vaccine-responding T cell clones reveals convergent and personalized response in identical twins. PNAS 115, 12704–12709 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Minervina, A. A. et al. Longitudinal high-throughput TCR repertoire profiling reveals the dynamics of T-cell memory formation after mild COVID-19 infection. eLife 10, e63502 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Meckiff, B. J. et al. Imbalance of regulatory and cytotoxic SARS-CoV-2-reactive CD4+ T cells in COVID-19. Cell 183, 1340–1353 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Goncharov, M. et al. VDJdb in the pandemic era: a compendium of T cell receptors specific for SARS-CoV-2. Nat. Methods 19, 1017–1019 (2022).

    CAS  PubMed  Google Scholar 

  58. Alter, G. et al. Immunogenicity of Ad26.COV2.S vaccine against SARS-CoV-2 variants in humans. Nature 596, 268–272 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Swanson, P. A.2nd et al. AZD1222/ChAdOx1 nCoV-19 vaccination induces a polyfunctional spike protein-specific TH1 response with a diverse TCR repertoire. Sci. Transl. Med. 13, eabj7211 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Harris, P. A. et al. Research electronic data capture (REDCap)—a metadata-driven methodology and workflow process for providing translational research informatics support. J. Biomed. Inform. 42, 377–381 (2009).

    PubMed  Google Scholar 

  61. Bennett, R. S. et al. Scalable, micro-neutralization assay for assessment of SARS-CoV-2 (COVID-19) virus-neutralizing antibodies in human clinical samples. Viruses 13, 893 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Sholukh, A. M. et al. Evaluation of cell-based and surrogate SARS-CoV-2 neutralization assays. J. Clin. Microbiol. 59, e0052721 (2021).

    PubMed  Google Scholar 

  63. Koelle, D. M. Expression cloning for the discovery of viral antigens and epitopes recognized by T cells. Methods 29, 213–226 (2003).

    CAS  PubMed  Google Scholar 

  64. Mayer-Blackwell, K. et al. TCR meta-clonotypes for biomarker discovery with tcrdist3 enabled identification of public, HLA-restricted clusters of SARS-CoV-2 TCRs. eLife 10, e68605 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Hagberg, A. A., Schult, D. A. & Swart, P. J. Exploring network structure, dynamics, and function using NetworkX. In Proc. 7th Python in Science Conference (SciPy2008) (eds Varoquaux, G. et al.) 11–15 (SciPy, 2008).

  66. Britanova, O. V. et al. Age-related decrease in TCR repertoire diversity measured with deep and normalized sequence profiling. J. Immunol. 192, 2689–2698 (2014).

    CAS  PubMed  Google Scholar 

  67. Wagih, O. ggseqlogo: A versatile R package for drawing sequence logos. Bioinformatics 33, 3645–3647 (2017).

    CAS  PubMed  Google Scholar 

  68. Snyder, T. M. et al. Magnitude and dynamics of the T-cell response to SARS-CoV-2 infection at both individual and population levels. Preprint at medRxiv https://doi.org/10.1101/2020.07.31.20165647 (2020).

  69. Nolan, S. et al. A large-scale database of T-cell receptor beta (TCRβ) sequences and binding associations from natural and synthetic exposure to SARS-CoV-2. Preprint at Research Square https://doi.org/10.21203/rs.3.rs-51964/v1 (2020).

  70. Klinger, M. et al. Multiplex identification of antigen-specific T cell receptors using a combination of immune assays and immune receptor sequencing. PLoS ONE 10, e0141561 (2015).

    PubMed  PubMed Central  Google Scholar 

  71. Li, D. et al. The T-cell response to SARS-CoV-2 vaccination in inflammatory bowel disease patients is augmented with anti-TNF therapy. Inflamm. Bowel Dis. 28, 1130–1133 (2022).

    PubMed  PubMed Central  Google Scholar 

  72. Lefranc, M.-P. et al. IMGT, the international ImMunoGeneTics information system 25 years on. Nucleic Acids Res. 43, D413–D422 (2015).

    CAS  PubMed  Google Scholar 

  73. Schmitt, T. M. et al. Generation of higher affinity T cell receptors by antigen-driven differentiation of progenitor T cells in vitro. Nat. Biotechnol. 35, 1188–1195 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Linnemann, C. et al. High-throughput identification of antigen-specific TCRs by TCR gene capture. Nat. Med. 19, 1534–1541 (2013).

    CAS  PubMed  Google Scholar 

  75. Jing, L. et al. Extensive CD4 and CD8 T cell cross-reactivity between alphaherpesviruses. J. Immunol. 196, 2205–2218 (2016).

    CAS  PubMed  Google Scholar 

  76. Ford, E. S. et al. Expansion of the HSV-2-specific T cell repertoire in skin after immunotherapeutic HSV-2 vaccine. Preprint at medRxiv https://doi.org/10.1101/2022.02.04.22270210 (2022).

  77. Jing, L. et al. Cross-presentation and genome-wide screening reveal candidate T cells antigens for a herpes simplex virus type 1 vaccine. J. Clin. Invest. 122, 654–673 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. van Velzen, M. et al. Local CD4 and CD8 T-cell reactivity to HSV-1 antigens documents broad viral protein expression and immune competence in latently infected human trigeminal ganglia. PLoS Pathog. 9, e1003547 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Jing, L. et al. Prevalent and diverse intratumoral oncoprotein-specific CD8+ T cells within polyomavirus-driven Merkel cell carcinomas. Cancer Immunol. Res. 8, 648–659 (2020).

    PubMed  PubMed Central  Google Scholar 

  80. Tigges, M. A. et al. Human CD8+ herpes simplex virus-specific cytotoxic T-lymphocyte clones recognize diverse virion protein antigens. J. Virol. 66, 1622–1634 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank: the participants; the Virology Research Clinic, University of Washington for collecting the specimens and data; D. Geraghty and C.-W. Pyo at the Fred Hutchinson Cancer Center (FHCC) for HLA typing; L. Stamatatos, FHCC, for the SARS-CoV-2 S protein with stabilizing proline substitutions used in the antibody assays; J. Bloom, FHCC, for the expression plasmids encoding the SARS-CoV-2 S protein from strain Wu-1, or Omicron BA.1, BA.2 and BA.4, with 21-amino-acid C-terminal deletions. D.M.K. received support from a National Institutes of Health (NIH) National Institute of Allergy and Infectious Diseases (NIAID) contract no. 75N93019C00063 (D.M.K.). The study has received support from NIH grant nos. AI163999 (D.M.K.), K08 AI148588 (E.S.F.), R01 AI136514 (K.M.-B. and A.F.-G.), F30 CA254168 (T.H.P.), T32 CA080416 (S.J.), P01 CA225517 (A.G.C., D.M.K., L.J., T.H.P. and S.J.), R01 AI134878 (A.M.S., E.L.B. and R.S.G.) and UM1 AI068614 (A.M.S., E.L.B. and R.S.G.). The scientific computing infrastructure at the FHCC was funded by an NIH Office of Research Infrastructure Programs grant no. S10 OD028685 (K.M.-B., A.F.-G. and E.S.F.). The Bill and Melinda Gates Foundation provided support via grant no. INV-027499 (A.F.-G. and K.M.-B.). This study was funded in part with Federal funds from the NIAID, NIH and Department of Health and Human Services under NIH contract no. HHSN272201800013C.

Author information

Authors and Affiliations

Authors

Contributions

D.M.K., E.S.F., K.M.-B., A.F.-G., L.J. and K.J.L. conceptualized the study. A.M.S., E.L.B., R.S.G., M.R.H., B.E., E.E., M.M. and E.P. designed and performed the serological assays. L.J., C.J.B., H.X., T.H.P., K.J.L., H.S.R., R.M.G., R.E. and A.L.G. designed and performed the cellular immunity and sequencing assays. A.G.C., E.W. and M.E. provided the specialty reagents enabling the TCR functional assays. S.S. and C.L.M. processed and managed the specimen and demographic data. A.W. organized the clinical cohort. K.M.-B., E.S.F., K.J.L, L.J., S.J. and A.F.-G. carried out the bioinformatic and statistical analyses. E.S.F., K.M.-B., K.J.L., A.F.-G., L.J. and D.M.K. wrote the manuscript. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Corresponding author

Correspondence to David M. Koelle.

Ethics declarations

Competing interests

H.S.R. and R.E. are employees of Adaptive Biotechnologies. B.E., E.E. and M.R.H. performed this work as employees of Laulima Government Solutions. M.M. and E.P. are subcontractors to Laulima Government Solutions; they performed this work as employees of Tunnell Government Services. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Tao Dong and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Ioana Visan in collaboration with the Nature Immunology editorial team.

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 Schedule of infection, vaccination, and sample collection.

Fifty-three study participants with prior SARS-CoV-2 infection as documented by seropositivity to S and N proteins and participant P845 who was seronegative prior to vaccination received either BNT162 or mRNA-1273 1st dose (closed circle), 2nd dose (closed triangle), and booster (3rd) dose (closed square) on the days after symptom onset as shown. Persons with mild or moderate COVID-19 are shown in magenta, persons with severe COVID-19 in black. Duration of hospitalization in persons with severe COVID-19 is shown in purple. PBMC were obtained at exam visits convalescence (E00, n = 54), late convalescence (E00.5, n = 16), pre-dose 1 (E01, n = 34), post-dose 1 (E02, n = 52), post-dose 2 (E03, n = 53), pre-boost (E04, n = 7), and post-boost (E05, n = 44). 33 persons had samples at E00, E01 and E03, 31 had samples at E00, E01, E02, and E03, and 26 had samples at all of E00, E01, E02, E03, and E05. Three participants were observed to have breakthrough COVID-19 infection based on boosting of anti-nucleocapsid antibody levels at visit E05 (P545, P664, P669), indicated in orange. All participants received primary vaccination but not all received a booster dose.

Source data

Extended Data Fig. 2 Primary vaccination led to expansion in specific TCR clonotypes in vaccinated persons.

Frequency (% of bulk TRB repertoire) of individual clonotypes in E01 vs. E03 in 32 persons with both samples. Expanded (red) (or contracted, purple) clonotypes were defined as log2(fold change) > 2 (or < 0.5) and Fisher’s exact test FDR-adjusted p value < 0.05. Dotted line indicates y = x. Participant ID at top of each graph. ND = not detected. Serologically-naive Participant P845 is not shown.

Source data

Extended Data Fig. 3 Frequency of E01-E03 expanded clonotypes from E00 through E05.

(a) E01-E03 expanded clonotype frequency (abundance) over the course of the study. Each line is an individual clonotype. TRB-PI are shown in black, TRB-PV are in orange (n = 30). ND = not detected. (b) Boxplot at lower right shows the percent of TRB-PI and TRB-PV for each participant that were detectable after a 3rd vaccine dose (E05) (n = 26). Median, IQR and whiskers (1.5*IQR) are noted. Comparison between groups is by two-sided Wilcoxon rank sum test, p = 0.0023. Participants P742 and P758 were not sampled at E02 and trajectories are not shown. Participant P665 had no expanded clonotypes and is not shown. Participant P845 was serologically naive at E00. Participants P545 and P669 experienced breakthrough infection between the E03 and the E05 timepoints and so repertoires at E05 represent both repeat natural infection as well as mRNA booster vaccine dose.

Source data

Extended Data Fig. 4 AIM-scTCRαβ-seq enriches a complex set of clonotypes from PBMC after mRNA vaccination of previously SARS-CoV-2 infected persons.

(a) Frequency of clonotypes detected by CD69+CD137+ AIM-scTCRαβ-seq plotted against the productive frequency of TRB-matched templates in bulk TRB sequencing at E03. Cell types defined by oligonucleotide-labeled mAbs are shown as CD4+ (green), CD8+ (blue), or phenotype not defined (purple) T cells. Clonotype enrichment in CD69+CD137+ AIM-scTCRαβ-seq was determined by cumulative distribution function (CDF) with false discovery rate (FDR) correction (Methods). Clonotypes that were detected, but not enriched, in CD69+CD137+ AIM-scTCRɑβ-seq are shown in red (n = 180) and were omitted from CDR3 motif discovery analysis. ND indicates clonotypes that could not be assigned a TRB unambiguously. (b,c) Productive frequency of CD69+CD137+ AIM-scTCRɑβ-seq detected clonotypes in relation to change in productive frequency from E01 to E03 in bulk TCR-β-seq is shown for 2 representative participants, including one with a lower proportion of representation in CD69+CD137+ AIM-scTCRαβ-seq of their significantly expanded clonotypes (P525, b) and another with a higher proportion of significantly expanded clonotypes also seen by CD69+CD137+ AIM-scTCRαβ-seq (P581, c). (d, e) Density plots show proportion of unique, expanded clonotypes by frequency at E03 (d) or fold change from E01 to E03 (primary vaccination) (e) by detection in CD69+CD137+ AIM-scTCRαβ-seq at E03 amongst 12 participants with both paired E01/E03 bulk TCR-β-seq and CD69+CD137+ AIM-scTCRαβ-seq.

Source data

Extended Data Fig. 5 TS from CD69+CD137+ AIM-scTCRαβ-seq matching TRB clonotypes from nasal swabs in 14 participants at E05.

(a) Rank abundance plots of TRB clonotypes in nasal samples in 14 participants, where blue (CD8+) and green (CD4+) dashes indicate rank of clones identified in the same participant’s blood by CD69+CD137+ AIM-scTCRαβ-seq at E03. (b) In participant P673, rank abundance plot of TRB clonotypes in nasal samples collected at E05. Clones labeled TCR1, TCR2, TCR3, TCR4, TCR8.1 and TCR8.2 indicate TRB clonotypes with exact sequence match to experimentally-confirmed receptors shown to recognize HLA-A*03:01 S epitopes.

Source data

Extended Data Fig. 6 TS and vaccine-expanded clonotypes.

Overlay of TRB sequences from CD69+CD137+ AIM-scTCRɑβ-seq of TS cells onto bulk TRB clonotype frequency at E01 and E03 in two representative participants. E01-to-E03 expanded (Ex) or contracted (Con), TRB clonotypes are shown, with TRB matching a CD69+CD137+ AIM-scTCRɑβ-seq (TS) shown in color and unmatched TRB are shown in gray (Und). Among clonotypes that neither expanded nor contracted (Non-Ex), only CD69+CD137+ AIM-scTCRɑβ-seq TRB-matched clonotypes are shown between red dashed lines. Frequencies of TRB clonotypes pre- and post-vaccine are as resolved by bulk TCR-β-seq. ND = not detected.

Source data

Extended Data Fig. 7 Abundance of TS clonotypes by TCRβ-seq over time.

Longitudinal tracking of abundance of CD69+CD137+ AIM-scTCRαβ-seq-identified CD4+ and CD8+ TS TRB clonotypes in PBMC by TCRβ-seq in all participants with AIM-scTCRαβ-seq. Numbers in top rows indicate the number of unique CD69+CD137+ AIM-scTCRαβ-seq TRB clonotypes from E03 detected at each time point. Percentages refer to the fraction of CD69+CD137+ AIM-scTCRαβ-seq clonotypes detected at the E02 and E03 timepoints, respectively, detected only post-vaccination. Percentages in gray are the fraction of unique clones detected at E03 that are below the level of detection at E02. Not all participants had samples at each time point, indicated by absence of dot symbols at those samples. Participant P669 had SARS-CoV-2 infection between E03 and E05 timepoints and so this E05 repertoire reflects repeat SARS-Co-2 infection and mRNA booster vaccine.

Source data

Extended Data Fig. 8 Selection of AIM-scTCRαβ-seq T cells using CD69, CD137, and CD134/ CD154 marker sets compared to bulk TCRβ-seq and sorted CD4 TCRβ-seq from PBMCs over primary vaccination.

Frequency (% of bulk TRB repertoire) of individual clonotypes in E01 vs. E03 in 7 persons studied by CD69+CD137+ AIM-scTCRαβ-seq and (CD69/CD137)+(CD134/CD154)+ CD4+ AIM-TCRβ-seq. Dotted line indicates y = x. Participant ID at top of each pair of graphs. ND = not detected. All PBMC are shown. Clonotypes found in the total CD4+ sorted fraction are shown in black. Clonotypes present in the total CD4+ sorted fraction and also enriched in sequential sorting of CD4+CD69+CD137+ (green) cells are overlaid with CD4+CD69+(CD134/CD154)+ and CD4+CD137+(CD134/CD154)+ (pink) cells in the left and right panels, respectively, for seven participants. Clonotypes in all three fractions (total CD4+, CD4+CD69+CD137+, and CD4+CD69+(CD134/CD154)+ and CD4+CD137+(CD134/CD154)+) are shown in orange. Gray shaded clonotypes were not identified as CD4+ by any of these methods.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–12.

Reporting Summary

Peer Review File

Supplementary Tables 1–17

All supplementary tables (combined into a single workbook with one tab per table). The first row of each table is the table title.

Supplementary Data

Statistical/bar plot data for Supplementary Figs. 3a,b, 5b,c, 9 and 11.

Source data

Source Data Fig. 1

Source data for Fig. 1.

Source Data Fig. 2

Source data for Fig. 2.

Source Data Fig. 3

Source data for Fig. 3

Source Data Fig. 4

Source data for Fig. 4.

Source Data Fig. 5

Source data for Fig. 5.

Source Data Fig. 6

Source data for Fig. 6.

Source Data Extended Data Fig. 1

Source data for Extended Fig. 1.

Source Data Extended Data Fig. 2

Source data for Extended Fig. 2.

Source Data Extended Data Fig. 3

Source data for Extended Fig. 3.

Source Data Extended Data Fig. 4

Source data for Extended Fig. 4.

Source Data Extended Data Fig. 5

Source data for Extended Fig. 5.

Source Data Extended Data Fig. 6

Source data for Extended Fig. 6.

Source Data Extended Data Fig. 7

Source data for Extended Fig. 7.

Source Data Extended Data Fig. 8

Source data for Extended Fig. 8.

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

Ford, E.S., Mayer-Blackwell, K., Jing, L. et al. Repeated mRNA vaccination sequentially boosts SARS-CoV-2-specific CD8+ T cells in persons with previous COVID-19. Nat Immunol 25, 166–177 (2024). https://doi.org/10.1038/s41590-023-01692-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01692-x

This article is cited by

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