Whole-blood cytokine secretion assay as a high-throughput alternative for assessing the cell-mediated immunity profile after two doses of an adjuvanted SARS-CoV-2 recombinant protein vaccine candidate

Stephen C De Rosa, Kristen W Cohen, Matthew Bonaparte, Bo Fu, Sanjay Garg, Catherine Gerard, Paul A Goepfert, Ying Huang, Daniel Larocque, M Juliana McElrath, Daryl Morris, Robbert Van der Most, Guy de Bruyn, Anke Pagnon, Stephen C De Rosa, Kristen W Cohen, Matthew Bonaparte, Bo Fu, Sanjay Garg, Catherine Gerard, Paul A Goepfert, Ying Huang, Daniel Larocque, M Juliana McElrath, Daryl Morris, Robbert Van der Most, Guy de Bruyn, Anke Pagnon

Abstract

Objectives: We previously described the Phase I-II evaluation of SARS-CoV-2 recombinant protein candidate vaccine, CoV2-PreS-dTM, with AF03- or AS03-adjuvant systems (ClinicalTrials.gov, NCT04537208). Here, we further characterise the cellular immunogenicity profile of this vaccine candidate using a whole-blood secretion assay in parallel to intracellular cytokine staining (ICS) of cryopreserved peripheral blood mononuclear cells (PBMCs).

Methods: A randomly allocated subset of 90 healthy, SARS-CoV-2-seronegative adults aged ≥ 18 years who had received (random allocation) one or two separate injections (on study day [D]1 and D22) of saline placebo or CoV2-PreS-dTM formulated with AS03 or AF03 were included. Cytokine secretion was assessed using a TruCulture® whole-blood stimulation system in combination with multiplex bead array, and intracellular cytokine profiles were evaluated on thawed PBMCs following ex vivo stimulation with recombinant S protein at pre-vaccination (D1), post-dose 1 (D22) and post-dose 2 (D36).

Results: Both methods detected similar vaccine-induced responses after the first and second doses. We observed a Th1 bias (Th1/Th2 ratio > 1.0) for most treatment groups when analysed in whole blood, mainly characterised by increased IFN-γ, IL-2 and TNF-α secretion. Among participants aged ≥ 50 years, the Th1/Th2 ratio was higher for those who received vaccine candidate with AS03 versus AF03 adjuvant. ICS revealed that this higher Th1/Th2 ratio resulted from higher levels of IFN-γ expression and that the vaccine induced polyfunctional CD4+ T cells.

Conclusions: The whole-blood cytokine secretion assay is a high-throughput alternative for assessing the quantity and character of vaccine-induced cellular responses.

Keywords: AF03; AS03; CoV2‐PreS‐dTM; SARS‐CoV‐2 recombinant protein candidate vaccine; adjuvant; cell‐mediated immunity; intracellular cytokine staining; multiplex bead array; whole‐blood cytokine secretion assay.

Conflict of interest statement

SdR, KC, YH, MJM and DM are employees of the Fred Hutchinson Cancer Research Center. The Fred Hutchinson Cancer Research Center received funding from Sanofi Pasteur through a contract to perform the ICS analysis. MIB, BF, SG, DL, GdB and AP are Sanofi Pasteur employees and may hold shares and/or stock options in the company. CG and RVdM are employed by, and hold restricted shares in, the GlaxoSmithKline group of companies. All other authors declare no competing interests.

© 2022 Sanofi Pasteur. Clinical & Translational Immunology published by John Wiley & Sons Australia, Ltd on behalf of Australian and New Zealand Society for Immunology, Inc.

Figures

Figure 1
Figure 1
Vaccine‐induced cytokine concentrations in whole‐blood supernatant after S protein stimulation: whole‐blood cytokine secretion assay. Data show background‐subtracted specific cytokine concentrations in pg mL−1 (log scale) in participants receiving the placebo, the non‐adjuvanted SARS‐CoV2 recombinant protein candidate vaccine (None) or SARS‐CoV2 recombinant protein candidate vaccine adjuvanted with AF03 (red dots) or AS03 (blue dots), by age groups (18–49 or ≥ 50 years) at baseline, 21 days after dose 1 (Post 1st) and 14 days after dose 2 (Post 2nd). For the placebo group, data for all three time points are combined. Box and whisker plots represent median (mid‐line of the box) and 25th and 75th percentiles (bottom and top lines, respectively); the whiskers that extend from the top and bottom of the box extend to the most extreme data points that are no more than 1.5 times the interquartile range or if no value meets this criterion, to the data extremes. Wilcoxon rank P‐values < 0.05 are shown (not adjusted for multiplicity) (exploratory analyses).
Figure 2
Figure 2
Ratios of Th1 (IFN‐γ) to Th2 (IL‐4, IL‐5, IL‐13) responses after one or two doses of SARS‐CoV2 protein adjuvanted with AF03 or AS03 (whole‐blood cytokine secretion assay or intracellular staining of PBMCs). Whole‐blood cell assay: ratios based on background‐adjusted concentrations (values below LLOQ set to LLOQ/2); Intracellular staining (ICS): ratios based on background‐adjusted percentage of CD4+ T cells expressing IFN‐γ to those expressing Th2 cytokines. Data points where the response for IFN‐γ was negative are identified as triangles. Wilcoxon rank P‐values < 0.05 are shown (not adjusted for multiplicity) (exploratory analyses).
Figure 3
Figure 3
Frequencies of CD4+ T cells expressing different cytokines after stimulation with recombinant full‐length S protein: ICS assay. Data show background‐subtracted CD4+ T‐cell frequencies (percentage of CD4+ T cells expressing the indicated cytokine; log scale) for participants receiving the placebo, the non‐adjuvanted SARS‐ CoV2 recombinant protein candidate vaccine (None) or SARS‐CoV2 recombinant protein candidate vaccine adjuvanted with AF03 (red dots) or AS03 (blue dots), by age groups (18–49 or ≥ 50 years) at baseline, 21 days after dose 1 (Post 1st) and 14 days after dose 2 (Post 2nd). For the placebo group, data for all three time points are combined. The y‐axis is truncated at 0.001% and any values below this level are censored. The mid‐line of the box denotes the median, and the ends of the box denote the 25th and 75th percentiles. The whiskers that extend from the top and bottom of the box extend to the most extreme data points that are no more than 1.5 times the interquartile range or if no value meets this criterion, to the data extremes. The Th2 cytokines include co‐expression of CD40L since this reduces the background. Reagents for IL‐5 and IL‐13 were conjugated to the same fluorochrome and were therefore measured as IL‐5 and/or IL‐13. Wilcoxon rank P‐values < 0.05 are shown (not adjusted for multiplicity).
Figure 4
Figure 4
Correlation between cytokine profiles evaluated in the supernatants of whole blood or intracellular staining of PBMCs after the second vaccination (full‐length S protein stimulation). ICS data (frequencies of CD4+ T cells producing each cytokine for the recombinant full‐length S protein stimulation) are expressed as percentage of lymphocytes rather than percentage of CD4+ T cells in order to be more comparable to the measurement of concentrations in whole blood. Since IL‐5 and IL‐13 were not analysed separately by ICS, the data for those two cytokines were summed for the whole‐blood cytokine secretion assay. Spearman’s rank correlation coefficients (rho) are shown.
Figure 5
Figure 5
Polyfunctionality of CD4+ T‐cell responses measured by ICS after stimulation with recombinant full‐length S protein after one or two doses of SARS‐CoV2 protein adjuvanted with AF03 or AS03. Data show background‐subtracted CD4+ T‐cell frequencies (percentage of CD4+ T cells expressing the indicated cytokine combination; log scale) for participants receiving the SARS‐CoV2 recombinant protein candidate vaccine adjuvanted with AF03 (red dots) or AS03 (blue dots), by age groups (18–49 or ≥ 50 years) at baseline, 21 days after dose 1 (Post 1st) and 14 days after dose 2 (Post 2nd). The y‐axis is truncated at 0.0001%, and any values below this level are censored. Each functional profile is shown in the key on the x‐axis as determined by gating as positive or negative for each marker (see Supplementary figure 5 for the gating strategy and representative scatter plots). Because the focus of this figure was polyfunctional T cells, data for cells expressing only one cytokine are not shown. Also, to limit the size of the graph, cellular profiles for which the frequency of cells was very low are not included. Cells co‐expressing IL‐4 or IL‐17 were either not detected or only present at low frequency; all profiles shown are negative for those cytokines and they are not included in the key on the x‐axis. The mid‐line of the box denotes the median and the ends of the box denote the 25th and 75th percentiles. The whiskers that extend from the top and bottom of the box extend to the most extreme data points that are no more than 1.5 times the interquartile range or if no value meets this criterion, to the data extremes. Wilcoxon rank P‐values < 0.05 are shown (not adjusted for multiplicity).

References

    1. Regulatory Affairs Professionals Society (RAPS) . COVID‐19 vaccine tracker. 2021. Available at . Accessed on 27 April 2021.
    1. Gavi The Vaccine Alliance . Four reasons why we need multiple vaccines for Covid‐19. Available at . Accessed 06 July 2021.
    1. European Medicines Agency . Committee on human medicinal products: Guideline on clinical evaluation of vaccines (draft). 2018. Available at . Accessed 19 July 2021.
    1. World Health Organization . Guidelines on clinical evaluation of vaccines: regulatory expectations: Revision of WHO TRS 924, Annex 1. 2016. Available at . Accessed 19 July 2021.
    1. Tseng CT, Sbrana E, Iwata‐Yoshikawa N et al. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS One 2012; 7: e35421.
    1. Perlman S, Dandekar AA. Immunopathogenesis of coronavirus infections: implications for SARS. Nat Rev Immunol 2005; 5: 917–927.
    1. Roberts A, Lamirande EW, Vogel L et al. Immunogenicity and protective efficacy in mice and hamsters of a β‐propiolactone inactivated whole virus SARS‐CoV vaccine. Viral Immunol 2010; 23: 509–519.
    1. de Alwis R, Chen S, Gan ES, Ooi EE. Impact of immune enhancement on Covid‐19 polyclonal hyperimmune globulin therapy and vaccine development. EBioMedicine 2020; 55: 102768.
    1. Lambert PH, Ambrosino DM, Andersen SR et al. Consensus summary report for CEPI/BC March 12–13, 2020 meeting: Assessment of risk of disease enhancement with COVID‐19 vaccines. Vaccine 2020; 38: 4783–4791.
    1. Del Valle DM, Kim‐Schulze S, Huang HH et al. An inflammatory cytokine signature predicts COVID‐19 severity and survival. Nat Med 2020; 26: 1636–1643.
    1. Peng Y, Mentzer AJ, Liu G 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 2020; 21: 1336–1345.
    1. Sekine T, Perez‐Potti A, Rivera‐Ballesteros O et al. Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID‐19. Cell 2020; 183: 158–168.e114.
    1. Wyllie D, Jones HE, Mulchandani R et al. SARS‐CoV‐2 responsive T cell numbers and anti‐Spike IgG levels are both associated with protection from COVID‐19: a prospective cohort study in keyworkers medRxiv 2021: 2020.2011.2002.20222778.
    1. Cohen KW, Linderman SL, Moodie Z et al. Longitudinal analysis shows durable and broad immune memory after SARS‐CoV‐2 infection with persisting antibody responses and memory B and T cells. Cell Rep Med 2021; 2: 100354.
    1. Ng O‐W, Chia A, Tan AT et al. Memory T cell responses targeting the SARS coronavirus persist up to 11 years post‐infection. Vaccine 2016; 34: 2008–2014.
    1. Flaxman A, Ewer KJ. Methods for measuring T‐cell memory to vaccination: from mouse to man. Vaccines (Basel) 2018; 6: 43.
    1. Bull M, Lee D, Stucky J et al. Defining blood processing parameters for optimal detection of cryopreserved antigen‐specific responses for HIV vaccine trials. J Immunol Methods 2007; 322: 57–69.
    1. Kierstead LS, Dubey S, Meyer B et al. Enhanced rates and magnitude of immune responses detected against an HIV vaccine: effect of using an optimized process for isolating PBMC. AIDS Res Hum Retroviruses 2007; 23: 86–92.
    1. Duffy D, Rouilly V, Braudeau C et al. Standardized whole blood stimulation improves immunomonitoring of induced immune responses in multi‐center study. Clin Immunol (Orlando, Fla) 2017; 183: 325–335.
    1. Converse PJ, Jones SL, Astemborski J, Vlahov D, Graham NM. Comparison of a tuberculin interferon‐gamma assay with the tuberculin skin test in high‐risk adults: effect of human immunodeficiency virus infection. J Infect Dis 1997; 176: 144–150.
    1. Rothel JS, Jones SL, Corner LA, Cox JC, Wood PR. A sandwich enzyme immunoassay for bovine interferon‐gamma and its use for the detection of tuberculosis in cattle. Aust Vet J 1990; 67: 134–137.
    1. Duffy D, Nemes E, Llibre A et al. Immune profiling enables stratification of patients with active TB disease or M. tuberculosis infection. Clin Infect Dis 2021; 73: e3398–e3408.
    1. Goepfert PA, Fu B, Chabanon AL et al. Safety and immunogenicity of SARS‐CoV‐2 recombinant protein vaccine formulations in healthy adults: interim results of a randomised, placebo‐controlled, phase 1–2, dose‐ranging study. Lancet Infect Dis 2021; 21: 1257–1270.
    1. Klucker MF, Dalencon F, Probeck P, Haensler J. AF03, an alternative squalene emulsion‐based vaccine adjuvant prepared by a phase inversion temperature method. J Pharm Sci 2012; 101: 4490–4500.
    1. Garçon N, Vaughn DW, Didierlaurent AM. Development and evaluation of AS03, an Adjuvant System containing α‐tocopherol and squalene in an oil‐in‐water emulsion. Expert Rev Vaccines 2012; 11: 349–366.
    1. Defawe OD, Fong Y, Vasilyeva E et al. Optimization and qualification of a multiplex bead array to assess cytokine and chemokine production by vaccine‐specific cells. J Immunol Methods 2012; 382: 117–128.
    1. Sridhar S, Joaquin A, Bonaparte M, et al. Safety and immunogenicity of a SARS‐CoV‐2 recombinant protein vaccine with AS03 adjuvant in healthy adults: interim findings from a phase 2, randomised, dose‐finding, multi‐centre study. The Lancet Infectious Diseases. 2021;In press.
    1. Bourguignon P, Clément F, Renaud F et al. Processing of blood samples influences PBMC viability and outcome of cell‐mediated immune responses in antiretroviral therapy‐naïve HIV‐1‐infected patients. J Immunol Methods 2014; 414: 1–10.
    1. Ewer KJ, Barrett JR, Belij‐Rammerstorfer S et al. T cell and antibody responses induced by a single dose of ChAdOx1 nCoV‐19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nat Med 2021; 27: 270–278.
    1. Jackson LA, Anderson EJ, Rouphael NG et al. An mRNA vaccine against SARS‐CoV‐2 ‐ preliminary report. N Engl J Med 2020; 383: 1920–1931.
    1. Sadoff J, Gars ML, Shukarev G et al. Safety and immunogenicity of the Ad26.COV2.S COVID‐19 vaccine candidate: interim results of a phase 1/2a, double‐blind, randomized, placebo‐controlled trial. medRxiv 2020: 2020.2009.2023.20199604.
    1. Sahin U, Muik A, Derhovanessian E et al. Concurrent human antibody and TH1 type T‐cell responses elicited by a COVID‐19 RNA vaccine. medRxiv 2020: 2020.2007.2017.20140533.
    1. Moris P, van der Most R, Leroux‐Roels I et al. H5N1 influenza vaccine formulated with AS03 A induces strong cross‐reactive and polyfunctional CD4 T‐cell responses. J Clin Immunol 2011; 31: 443–454.
    1. Richmond P, Hatchuel L, Dong M et al. Safety and immunogenicity of S‐Trimer (SCB‐2019), a protein subunit vaccine candidate for COVID‐19 in healthy adults: a phase 1, randomised, double‐blind, placebo‐controlled trial. Lancet (London, England) 2021; 397: 682–694.
    1. Ward BJ, Gobeil P, Séguin A et al. Phase 1 trial of a candidate recombinant virus‐like particle vaccine for Covid‐19 disease produced in plants. medRxiv 2020: 2020.2011.2004.20226282.
    1. Arunachalam PS, Walls AC, Golden N et al. Adjuvanting a subunit COVID‐19 vaccine to induce protective immunity. Nature 2021; 594: 253–258.
    1. Copaescu A, Smibert O, Gibson A, Phillips EJ, Trubiano JA. The role of IL‐6 and other mediators in the cytokine storm associated with SARS‐CoV‐2 infection. J Allergy Clin Immunol 2020; 146: 518–534. e511.
    1. Lang FM, Lee KMC, Teijaro JR, Becher B, Hamilton JA. GM‐CSF‐based treatments in COVID‐19: reconciling opposing therapeutic approaches. Nat Rev Immunol 2020; 20: 507–514.
    1. Thwaites RS, Sanchez Sevilla Uruchurtu A, Siggins MK et al. Inflammatory profiles across the spectrum of disease reveal a distinct role for GM‐CSF in severe COVID‐19. Sci Immunol 2021; 6: eabg9873.
    1. Prince GA, Curtis SJ, Yim KC, Porter DD. Vaccine‐enhanced respiratory syncytial virus disease in cotton rats following immunization with Lot 100 or a newly prepared reference vaccine. J Gen Virol 2001; 82: 2881–2888.
    1. Castilow EM, Olson MR, Varga SM. Understanding respiratory syncytial virus (RSV) vaccine‐enhanced disease. Immunol Res 2007; 39: 225–239.
    1. Lineburg KE, Neller MA, Ambalathingal GR et al. Rapid whole‐blood assay to detect SARS‐CoV‐2‐specific memory T‐cell immunity following a single dose of AstraZeneca ChAdOx1‐S COVID‐19 vaccine. Clin Transl Immunology 2021; 10: e1326.
    1. Lineburg KE, Srihari S, Altaf M et al. Rapid detection of SARS‐CoV‐2‐specific memory T‐cell immunity in recovered COVID‐19 cases. Clin Transl Immunol 2020; 9: e1219.
    1. De Rosa SC, Carter DK, McElrath MJ. OMIP‐014: validated multifunctional characterization of antigen‐specific human T cells by intracellular cytokine staining. Cytometry Part A 2012; 81: 1019–1021.
    1. Horton H, Thomas EP, Stucky JA et al. Optimization and validation of an 8‐color intracellular cytokine staining (ICS) assay to quantify antigen‐specific T cells induced by vaccination. J Immunol Methods 2007; 323: 39–54.
    1. Sadoff J, Le Gars M, Shukarev G et al. Interim results of a phase 1–2a trial of Ad26.COV2.S Covid‐19 vaccine. N Engl J Med 2021; 384: 1824–1835.
    1. Stamatatos L, Czartoski J, Wan YH et al. mRNA vaccination boosts cross‐variant neutralizing antibodies elicited by SARS‐CoV‐2 infection. Science 2021; Online ahead of print.
    1. Agresti A, Coull BA. Approximate is better than “exact” for interval estimation of binomial proportions. Am Stat 1998; 52: 119–126.

Source: PubMed

3
订阅