+ All Categories
Home > Documents > VACCINES MAIT cell activation augments adenovirus vector...VACCINES MAIT cell activation augments...

VACCINES MAIT cell activation augments adenovirus vector...VACCINES MAIT cell activation augments...

Date post: 17-Feb-2021
Category:
Upload: others
View: 8 times
Download: 0 times
Share this document with a friend
7
VACCINES MAIT cell activation augments adenovirus vector vaccine immunogenicity Nicholas M. Provine 1 * , Ali Amini 1 , Lucy C. Garner 1 , Alexandra J. Spencer 2 , Christina Dold 3 , Claire Hutchings 4 , Laura Silva Reyes 3 , Michael E. B. FitzPatrick 1 , Senthil Chinnakannan 4 , Blanche Oguti 3 , Meriel Raymond 3 , Marta Ulaszewska 2 , Fulvia Troise 5,6 , Hannah Sharpe 2 , Sophie B. Morgan 7 , Timothy S. C. Hinks 7 , Teresa Lambe 2 , Stefania Capone 8 , Antonella Folgori 8 , Eleanor Barnes 1,2,4 , Christine S. Rollier 3 , Andrew J. Pollard 3 , Paul Klenerman 1,4 * Mucosal-associated invariant T (MAIT) cells are innate sensors of viruses and can augment early immune responses and contribute to protection. We hypothesized that MAIT cells may have inherent adjuvant activity in vaccine platforms that use replication-incompetent adenovirus vectors. In mice and humans, ChAdOx1 (chimpanzee adenovirus Ox1) immunization robustly activated MAIT cells. Activation required plasmacytoid dendritic cell (pDC)derived interferon (IFN)a and monocyte-derived interleukin-18. IFN-ainduced, monocyte-derived tumor necrosis factor was also identified as a key secondary signal. All three cytokines were required in vitro and in vivo. Activation of MAIT cells positively correlated with vaccine-induced T cell responses in human volunteers and MAIT celldeficient mice displayed impaired CD8 + T cell responses to multiple vaccine-encoded antigens. Thus, MAIT cells contribute to the immunogenicity of adenovirus vectors, with implications for vaccine design. M ucosal-associated invariant T (MAIT) cells are unconventional T cells that recognize microbe-derived metabolites of vitamin B2 biosynthesis such as 5-(2-oxopropylideneamino)-6- D- ribitylaminouracil (5-OP-RU) (1). However, MAIT cells can also be activated by cytokines and thereby respond to viruses, which do not synthesize vitamin B2. In vivo, MAIT cells respond to influenza virus to amplify early local immune responses and protect against lethal infection (24). We hypothesized that the ability of MAIT cells to augment early immune responses may play a key role in viral vector vac- cine immunogenicity. Replication-incompetent adenovirus (Ad) vectors are highly potent vaccine platforms for many human diseases (5). They have recently been licensed for use against the Ebola virus (6) and show promise for severe acute respiratory syndrome coro- navirus 2 (SARS-CoV-2) infection (7, 8). We sought to determine whether such vectors activate MAIT cells and whether this affects vaccine immunogenicity. To determine whether MAIT cells respond to Ad vectors, we stimulated human periph- eral blood mononuclear cells (PBMCs) with Ad5 and chimpanzee adenovirus Ox1 (ChAdOx1), which are leading SARS-CoV-2 candidate vac- cines (7, 8). ChAdOx1 induced dose-dependent up-regulation of CD69, granzyme B, and inter- feron (IFN)g by MAIT cells (Fig. 1, A to C, and fig. S1, A to D), whereas Ad5 only weakly ac- tivated MAIT cells. This activation was con- firmed using the MR15-OP-RU tetramer to identify MAIT cells (fig. S1E). Species Cderived Ad vectors have been shown to poorly stimulate innate immune responses as compared with nonspecies C vectors (911). We tested the relative ability of three species C vectors (Ad5, Ad6, and ChAdN13) and five nonspecies C vectors (Ad24, Ad35, ChAd63, ChAd68, and ChAdOx1) (fig. S1F) to activate MAIT cells. After stimulation, we ob- served greater average activation by nonspecies C vectors as compared with species C vectors (Fig. 1, D and E). We next tested the ability of Ad vectors to activate MAIT cells in vivo. Intramuscular (i.m.) ChAdOx1 immunization of C57BL/6J mice strongly induced up-regulation of CD69 and granzyme B by MAIT cells, whereas Ad5 in- duced significantly weaker activation (Fig. 1, F and G, and fig. S2, A to C). We also observed significant up-regulation of CD69 on MAIT cells 1 day after immunization of human volunteers with a candidate ChAdOx1 vaccine (Fig. 1H and fig. S3, A to C). Plasma IFN-g levels markedly increased after vaccination (fig. S3D), as seen in nonhuman primates (10). This increase correlated with levels of MAIT cell activation (Fig. 1I). To investigate the pathways involved, RNA sequencing (RNA-seq) of MAIT cells was per- formed. Eighty-four genes were significantly up-regulated in human MAIT cells after vac- cination (Fig. 2A and data S1). Gene set en- richment analysis (GSEA) (12) identified the strong induction of type I IFN, interleukin (IL)1 family, IL-12 family, and IL-2 family signaling pathways (Fig. 2B). Changes in post- vaccination plasma IFN-a or CCL2, an IFN- regulated chemokine (13), strongly correlated with MAIT cell activation (Fig. 2C and fig. S3, D and E). Comparison of genes up-regulated in MAIT cells after human vaccination, vacci- nation of mice, or in vitro stimulation showed a high degree of overlap. Ninety-eight percent of vaccineup-regulated genes in humans were up-regulated in at least one of the other two conditions, and 63% were up-regulated in both (Fig. 2D; fig. S4, A and B; and data S2 to S4). GSEA on murine MAIT cells and in vitrostimulated human MAIT cells identified sim- ilar enrichment of these cytokine signaling pathways (fig. S4, C and D). In vitro inhibition of type I IFN signaling reduced MAIT cell IFN-g production by >50% (Fig. 2E). Blockade of IL-18 (an IL-1 family member) or IL-12 also reduced MAIT cell activation. By contrast, blockade of IL-15 (an IL-2 family member) had no effect (Fig. 2F and fig. S5A). MAIT cell activation by Ad vectors was independent of T cell receptor signaling (fig. S5B) (2, 3). To understand the cellular origins of these critical cytokines, we examined the cell popu- lations transduced by Ad5 and ChAdOx1. Mono- cytes or conventional dendritic cells were the major transduced population by both vectors [>80% of green fluorescent proteinexpressing (GFP + ) cells] (fig. S5, C to F). ChAdOx1 also efficiently transduced CD123 + plasmacytoid dendritic cells (pDCs), whereas Ad5 did not (fig. S5F) (11). Notably, depletion of CD123 + pDCs resulted in a significant (67%) reduc- tion in IFN-g production by MAIT cells (Fig. 2G) and reduced IFN-a levels by >99% after ChAdOx1 stimulation (Fig. 2H). Depletion of CD14 + monocytes significantly reduced MAIT cell activation after ChAdOx1 stimulation (Fig. 2I and fig. S5G) and abro- gated the secretion of IL-18 (Fig. 2J). The cathepsin BNLRP3 inflammasome pathway (14) was the source of IL-18 in response to ChAdOx1 (fig. S6). Thus, pDC-derived IFN-a and monocyte-derived IL-18 play critical roles in activating MAIT cells in response to Ad vectors. Ad5 induced negligible amounts of IFN-a (fig. S7, A and B) (10, 11). Despite trans- ducing monocytes, Ad5 did not induce IL-18 or IL-12p70 (fig. S7, C and D). By contrast, ChAdOx1 induced robust production of IFN-a and IL-18. Although IFN-a/b and IL-18 together in- duced production of IFN-g by MAIT cells in PBMC culture, this was not seen using iso- lated CD8 + T cells [~75% of human MAIT cells express CD8 (15)] (Fig. 3A), despite the in- duction of CD69 (fig. S8A). Depletion of mono- cytes reduced MAIT cell IFN-g production after stimulation with IFN-a and IL-18 (fig. RESEARCH Provine et al., Science 371, 521526 (2021) 29 January 2021 1 of 6 1 Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK. 2 Jenner Institute, University of Oxford, Oxford, UK. 3 Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK. 4 Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK. 5 Nouscom, SRL, Rome, Italy. 6 Ceinge Biotechnologie Avanzate, Naples, Italy. 7 Respiratory Medicine Unit, Nuffield Department of Medicine Experimental Medicine, University of Oxford, Oxford, UK. 8 ReiThera, SRL, Rome, Italy. *Corresponding author. Email: [email protected] (N.M.P.); [email protected] (P.K.) on July 7, 2021 http://science.sciencemag.org/ Downloaded from
Transcript
  • VACCINES

    MAIT cell activation augments adenovirus vectorvaccine immunogenicityNicholas M. Provine1*, Ali Amini1, Lucy C. Garner1, Alexandra J. Spencer2, Christina Dold3,Claire Hutchings4, Laura Silva Reyes3, Michael E. B. FitzPatrick1, Senthil Chinnakannan4,Blanche Oguti3, Meriel Raymond3, Marta Ulaszewska2, Fulvia Troise5,6, Hannah Sharpe2,Sophie B. Morgan7, Timothy S. C. Hinks7, Teresa Lambe2, Stefania Capone8, Antonella Folgori8,Eleanor Barnes1,2,4, Christine S. Rollier3, Andrew J. Pollard3, Paul Klenerman1,4*

    Mucosal-associated invariant T (MAIT) cells are innate sensors of viruses and can augment early immuneresponses and contribute to protection. We hypothesized that MAIT cells may have inherent adjuvantactivity in vaccine platforms that use replication-incompetent adenovirus vectors. In mice and humans,ChAdOx1 (chimpanzee adenovirus Ox1) immunization robustly activated MAIT cells. Activation requiredplasmacytoid dendritic cell (pDC)–derived interferon (IFN)–a and monocyte-derived interleukin-18.IFN-a–induced, monocyte-derived tumor necrosis factor was also identified as a key secondary signal.All three cytokines were required in vitro and in vivo. Activation of MAIT cells positively correlated withvaccine-induced T cell responses in human volunteers and MAIT cell–deficient mice displayed impairedCD8+ T cell responses to multiple vaccine-encoded antigens. Thus, MAIT cells contribute to theimmunogenicity of adenovirus vectors, with implications for vaccine design.

    Mucosal-associated invariant T (MAIT)cells are unconventional T cells thatrecognizemicrobe-derivedmetabolitesof vitamin B2 biosynthesis such as5-(2-oxopropylideneamino)-6-D-

    ribitylaminouracil (5-OP-RU) (1). However,MAIT cells can also be activated by cytokinesand thereby respond to viruses, which do notsynthesize vitamin B2. In vivo, MAIT cellsrespond to influenza virus to amplify earlylocal immune responses and protect againstlethal infection (2–4). We hypothesized that theability of MAIT cells to augment early immuneresponses may play a key role in viral vector vac-cine immunogenicity. Replication-incompetentadenovirus (Ad) vectors are highly potentvaccine platforms for many human diseases(5). They have recently been licensed for useagainst the Ebola virus (6) and show promisefor severe acute respiratory syndrome coro-navirus 2 (SARS-CoV-2) infection (7, 8). Wesought to determine whether such vectorsactivate MAIT cells and whether this affectsvaccine immunogenicity.To determine whether MAIT cells respond

    to Ad vectors, we stimulated human periph-eral bloodmononuclear cells (PBMCs) with Ad5and chimpanzee adenovirus Ox1 (ChAdOx1),

    which are leading SARS-CoV-2 candidate vac-cines (7, 8). ChAdOx1 induced dose-dependentup-regulation of CD69, granzyme B, and inter-feron (IFN)–g by MAIT cells (Fig. 1, A to C, andfig. S1, A to D), whereas Ad5 only weakly ac-tivated MAIT cells. This activation was con-firmed using the MR1–5-OP-RU tetramer toidentify MAIT cells (fig. S1E).Species C–derived Ad vectors have been

    shown to poorly stimulate innate immuneresponses as compared with non–species Cvectors (9–11). We tested the relative ability ofthree species C vectors (Ad5,Ad6, andChAdN13)and five non–species C vectors (Ad24, Ad35,ChAd63, ChAd68, and ChAdOx1) (fig. S1F) toactivate MAIT cells. After stimulation, we ob-served greater average activation by non–species C vectors as compared with speciesC vectors (Fig. 1, D and E).We next tested the ability of Ad vectors to

    activate MAIT cells in vivo. Intramuscular(i.m.) ChAdOx1 immunization of C57BL/6Jmicestrongly induced up-regulation of CD69 andgranzyme B by MAIT cells, whereas Ad5 in-duced significantly weaker activation (Fig. 1, Fand G, and fig. S2, A to C). We also observedsignificant up-regulation of CD69 onMAIT cells1 day after immunization of human volunteerswith a candidate ChAdOx1 vaccine (Fig. 1Hand fig. S3, A to C). Plasma IFN-g levelsmarkedly increased after vaccination (fig.S3D), as seen in nonhuman primates (10).This increase correlated with levels of MAITcell activation (Fig. 1I).To investigate the pathways involved, RNA

    sequencing (RNA-seq) of MAIT cells was per-formed. Eighty-four genes were significantlyup-regulated in human MAIT cells after vac-cination (Fig. 2A and data S1). Gene set en-richment analysis (GSEA) (12) identified the

    strong induction of type I IFN, interleukin(IL)–1 family, IL-12 family, and IL-2 familysignaling pathways (Fig. 2B). Changes in post-vaccination plasma IFN-a or CCL2, an IFN-regulated chemokine (13), strongly correlatedwith MAIT cell activation (Fig. 2C and fig. S3,D and E). Comparison of genes up-regulatedin MAIT cells after human vaccination, vacci-nation of mice, or in vitro stimulation showeda high degree of overlap. Ninety-eight percentof vaccine–up-regulated genes in humans wereup-regulated in at least one of the other twoconditions, and 63%were up-regulated in both(Fig. 2D; fig. S4, A and B; and data S2 to S4).GSEA on murine MAIT cells and in vitro–stimulated human MAIT cells identified sim-ilar enrichment of these cytokine signalingpathways (fig. S4, C and D).In vitro inhibition of type I IFN signaling

    reduced MAIT cell IFN-g production by >50%(Fig. 2E). Blockade of IL-18 (an IL-1 familymember) or IL-12 also reduced MAIT cellactivation. By contrast, blockade of IL-15 (anIL-2 familymember) had no effect (Fig. 2F andfig. S5A). MAIT cell activation by Ad vectorswas independent of T cell receptor signaling(fig. S5B) (2, 3).To understand the cellular origins of these

    critical cytokines, we examined the cell popu-lations transducedbyAd5 andChAdOx1.Mono-cytes or conventional dendritic cells were themajor transduced population by both vectors[>80% of green fluorescent protein–expressing(GFP+) cells] (fig. S5, C to F). ChAdOx1 alsoefficiently transduced CD123+ plasmacytoiddendritic cells (pDCs), whereas Ad5 did not(fig. S5F) (11). Notably, depletion of CD123+

    pDCs resulted in a significant (67%) reduc-tion in IFN-g production by MAIT cells (Fig.2G) and reduced IFN-a levels by >99% afterChAdOx1 stimulation (Fig. 2H).Depletion of CD14+ monocytes significantly

    reduced MAIT cell activation after ChAdOx1stimulation (Fig. 2I and fig. S5G) and abro-gated the secretion of IL-18 (Fig. 2J). Thecathepsin B–NLRP3 inflammasome pathway(14) was the source of IL-18 in response toChAdOx1 (fig. S6). Thus, pDC-derived IFN-aandmonocyte-derived IL-18 play critical rolesin activating MAIT cells in response to Advectors. Ad5 induced negligible amounts ofIFN-a (fig. S7, A and B) (10, 11). Despite trans-ducing monocytes, Ad5 did not induce IL-18or IL-12p70 (fig. S7, C and D). By contrast,ChAdOx1 induced robust production of IFN-aand IL-18.Although IFN-a/b and IL-18 together in-

    duced production of IFN-g by MAIT cells inPBMC culture, this was not seen using iso-lated CD8+ T cells [~75% of humanMAIT cellsexpress CD8 (15)] (Fig. 3A), despite the in-duction of CD69 (fig. S8A). Depletion ofmono-cytes reduced MAIT cell IFN-g productionafter stimulation with IFN-a and IL-18 (fig.

    RESEARCH

    Provine et al., Science 371, 521–526 (2021) 29 January 2021 1 of 6

    1Translational Gastroenterology Unit, Nuffield Department ofMedicine, University of Oxford, Oxford, UK. 2Jenner Institute,University of Oxford, Oxford, UK. 3Oxford Vaccine Group,Department of Paediatrics, University of Oxford, and theNational Institute for Health Research (NIHR) OxfordBiomedical Research Centre, Oxford, UK. 4Peter MedawarBuilding for Pathogen Research, University of Oxford, Oxford,UK. 5Nouscom, SRL, Rome, Italy. 6Ceinge BiotechnologieAvanzate, Naples, Italy. 7Respiratory Medicine Unit, NuffieldDepartment of Medicine – Experimental Medicine, Universityof Oxford, Oxford, UK. 8ReiThera, SRL, Rome, Italy.*Corresponding author. Email: [email protected](N.M.P.); [email protected] (P.K.)

    on July 7, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • S8B). The addition of monocytes rescuedthe response (Fig. 3B). Conditioned super-natant or the provision of PBMCs across atranswell significantly rescued MAIT cellIFN-g production (Fig. 3C and fig. S8C), sug-gesting the presence of a soluble, monocyte-derived, IFN-a–dependent signal. Unexpectedly,IFN-a–treated monocytes secreted tumor ne-crosis factor (TNF) (Fig. 3D and fig. S8D).Additionally, TNFR2 signaling pathwayswere strongly induced in stimulated MAITcells (fig. S8, E to G). Thus, we investigatedwhether TNF was the IFN-a–dependent in-termediary signal. Addition of TNF or ananti-TNFR2 agonist to isolated CD8+ T cellsstimulated with IFN-a and IL-18 enhancedMAIT cell IFN-g production by >300% (Fig.3E and fig. S8H). Addition of an anti-TNFantibody (adalimumab) inhibited MAIT cellIFN-g production in response to IFN-a andIL-18 stimulation or to conditioned super-natant (fig. S8, I and J). TNF blockade usingeither adalimumab or recombinant TNFR2-Fcfusion protein (etanercept), but not a controlantibody (vedolizumab), inhibited IFN-g pro-duction byMAIT cells in response to ChAdOx1(Fig. 3F). Depletion of monocytes reducedChAdOx1-induced TNF production by 94%(Fig. 3G). Ad5 inducedminimal TNF (fig. S8K),consistent with the poor ability to stimulateIFN-a (fig. S7A).These data suggest a model in which pDC-

    derived IFN-a acts directly and indirectly viainduction of TNF bymonocytes (with IL-18) toactivate MAIT cells in response to ChAdOx1(fig. S9). To test thismodel in vivo, wild-type (WT)C57BL/6J, Il18rap−/−, Ifnar−/−, and Tnfrsf1a−/−

    Tnfrsf1b−/−micewere immunizedwithChAdOx1.MAIT cells from these animals were thenanalyzed by RNA-seq (fig. S10, A and B, anddata S5 to S8). Principal components analysisidentified a strong gradient of activation, whereMAIT cells from Ifnar−/− mice were mostsimilar to those from naïve animals, and MAITcells from Tnfrsf1a−/−Tnfrsf1b−/− and Il18rap−/−

    mice had intermediate transcriptional profiles(Fig. 3, H and I). The effector genes Cd69 (atboth the gene and protein level), Cxcl10, Cxcl11,Ccl5, and Gzmb were all regulated along thisgradient (fig. S10, C and D). Other genes (suchas Cxcl9) were only regulated by TNF signaling(fig. S10D). In total, 51% of the genes induced byvaccination were regulated by one or moreof these cytokine pathways, and 11%were co-regulated by two or more of these pathways(Fig. 3J and data S9). Thus, TNF, IL-18, andespecially type I IFN play a critical role in vivoin Ad vector–induced MAIT cell activation.Human volunteers showed a significant in-

    crease in IFN-g–producing T cells after ChAdOx1boosting immunization (Fig. 4A). The degree ofexpansion positively correlated with MAIT cellactivation (Fig. 4B). To determine whether thiswas a causal relationship, WT and Mr1−/− mice,

    Provine et al., Science 371, 521–526 (2021) 29 January 2021 2 of 6

    A B

    D E

    C

    F

    G

    H I

    naïvenaïve

    naïvenaïve

    Fig. 1. Activation of human and murine MAIT cells by adenovirus vectors. (A to C) Human PBMCs (n = 9donors; four experiments) were stimulated with Ad5-GFP or ChAdOx1-GFP [multiplicity of infection (MOI) = 0 to104 vp (viral particles)]. MAIT cell CD69 (A), granzyme B (GzmB) (B), and IFN-g (C) expression was measuredafter 24 hours. (D and E) Human PBMCs (n = 5 donors; two experiments) were stimulated with the indicatedvectors (species in parentheses). MAIT cell GzmB (D) or IFN-g (E) expression was measured after 24 hours.C, species C; non-C, non–species C. (F and G) C57BL/6J mice (n = 6 mice per group; representative of twoexperiments) were immunized intramuscularly (i.m.) with 108 IU (infectious units) of Ad5-GFP or ChAdOx1-GFP.Inguinal lymph node (iLN) and liver MAIT cell CD69 (F) and GzmB (G) expression was measured after24 hours. (H and I) Healthy human volunteers (n = 14) were immunized with a 5 × 1010 vp dose of ChAdOx1MenB.1. (H) MAIT cell CD69 expression 1 day before and 1 day after immunization. (I) Pearson correlation ofchange in plasma IFN-g levels after vaccination with the change in MAIT cell CD69 expression. *P < 0.05; **P <0.01; ***P < 0.001. Unpaired t test [(A) to (C)], two-way analysis of variance (ANOVA) [(D) and (E)], one-wayANOVA with Sidak correction for multiple comparisons [(F) and (G)], or Wilcoxon rank-sum test (H). Symbolsindicate average response [(A) to (C)] or individual mice or volunteers [(D) to (I)]. Mean ± SEM is shown.

    RESEARCH | REPORTon July 7, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • which lack MAIT cells (16), were used (fig. S11, Ato C). After vaccinationwith ChAdOx1 expressinganoptimizedhepatitis C virus (HCV) antigen (17),Mr1−/− mice had significantly reduced frequen-cies of HCV-specific CD8+ T cells compared withWT mice (Fig. 4C and fig. S11D). No significantdefect in HCV-specific CD4+ T cells was observed(fig. S11E). We also observed defects in the CD8+

    T cell responses of Mr1−/− mice vaccinated withthe candidate SARS-CoV-2 vaccine, ChAdOx1-

    nCoV-19 (Fig. 4D and fig. S11F) (7). WT andMr1−/− mice were then given a homologousChAd63-ovalbumin (OVA) prime-boost immu-nization (fig. S11G) (18).Mr1−/−mice displayedreduced OVA-specific CD8+ T cell responsesafter both priming and boosting (Fig. 4, E andF). Differences in the microbiome (19) or gen-eral immunodeficiency ofMr1−/−mice did notexplain these differences in immunogenicity(fig. S12).

    MAIT cells can sense the diversity of theAd vector–induced innate immune activationlandscape (e.g., IFN-a, TNF, IL-18), integrat-ing these signals to augment vaccine-inducedCD8+ T cell immunity. The blend of signalsrequired to maximally trigger MAIT cellsdescribed here includes a critical pathwayvia type I IFN–dependent TNF release, relieson cross-talk between two distinct popula-tions of transduced cells, and varies between

    Provine et al., Science 371, 521–526 (2021) 29 January 2021 3 of 6

    Fig. 2. Activation of MAIT cells by adenovirus vectors requires pDC-derivedIFN-a and monocyte-derived IL-18. (A and B) Gene expression analysis ofMAIT cells isolated from the PBMCs of human volunteers 1 day before and 1 dayafter vaccination with ChAdOx1 MenB.1 (n = 14 vacinees). (A) Volcano plot ofdifferentially expressed genes [log2 fold change (FC) > 1, adjusted P < 0.05]. Thetop 10 up-regulated genes are annotated. (B) Selected cytokine signalingpathways from the Reactome database enriched by GSEA. NES, normalizedenrichment score. (C) Pearson correlation of change in plasma IFN-a level aftervaccination with the change in MAIT cell CD69 expression. (D) Overlap of genesup-regulated in MAIT cells from ChAdOx1-vaccinated volunteers (“human in vivo”),from human PBMCs stimulated with ChAdOx1 (“human in vitro”), and from thedraining inguinal LNs of ChAdOx1-vaccinated mice (“mouse in vivo”). (E and F)Human PBMCs were stimulated with ChAdOx1-GFP, and the following inhibitorswere used: vaccinia virus–derived type I IFN antagonist B18R (1 or 10 mg/ml;

    n = 7 donors; three experiments) or anti-IFNAR2 antibody (10 or 25 mg/ml;n = 5 or 3 donors; two or one experiments, respectively) (E); or anti-IL-12,anti-IL-15, or anti-IL-18 antibodies (10 mg/ml; n = 5 donors; two experiments)(F). MAIT cell IFN-g expression was measured after 24 hours. PBS, phosphate-buffered saline. (G and H) PBMCs were depleted of CD123+ pDCs or leftuntreated and stimulated with ChAdOx1-GFP. MAIT cell IFN-g expression(n = 8 donors; three experiments) (G) or levels of IFN-a in the cell culturesupernatant (n = 4 donors; one experiment) (H) were measured after 24 hours.(I and J) PBMCs were depleted of CD14+ monocytes or left untreated andstimulated with ChAdOx1-GFP. MAIT cell IFN-g expression (n = 4 donors; twoexperiments) (I) or IL-18 levels in the supernatant (n = 4 donors; three experiments)(J) were measured after 24 hours. *P < 0.05; **P < 0.01; ***P < 0.001.Repeated-measures one-way ANOVAwith Dunnett correction [(E) and (F)] or unpairedt test [(G) to (J)]. Symbols indicate individual donors. Mean ± SEM is shown.

    RESEARCH | REPORTon July 7, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • adenovirus serotypes. Our data, coupled withstudies in the lung (4, 20, 21), support amodelthat places MAIT cells in a critical bridgingposition between innate and adaptive immu-

    nity. The mechanism by which MAIT cell ac-tivation promotes antigen-specific CD8+ T cellresponses remains to be defined. However,local production of chemokine CXCL10 repre-

    sents a promising candidate as it can promoteCD8+ T cell priming (22).It is notable that the activation of MAIT

    cells is tightly linked to the immunogenicity

    Provine et al., Science 371, 521–526 (2021) 29 January 2021 4 of 6

    Fig. 3. IFN-a acts directly and indirectly through the induction of TNF toactivate MAIT cells. (A) Human PBMCs or purified CD8+ T cells (n = 3 donors;one experiment) were stimulated with the indicated cytokines (50 ng/ml). MAITcell IFN-g expression was measured after 24 hours. (B) Purified CD8+ T cells withor without CD14+ monocytes (n = 4 donors; one experiment) were stimulatedwith IFN-a and IL-18 (50 ng/ml). MAIT cell IFN-g expression was measured after24 hours. (C) Purified monocytes (n = 3 donors; one experiment) werestimulated with IFN-a (50 ng/ml) or left untreated. After 24 hours, supernatantswere transferred with or without IL-18 (50 ng/ml) to autologous purified CD8+

    T cells. MAIT cell IFN-g expression was measured after 24 hours. (D) TNFproduction by IFN-a–treated CD14-purified monocytes was measured after24 hours (n = 3 donors; one experiment). (E) Purified CD8+ T cells (n = 10 donors;four experiments) were stimulated with IFN-a and IL-18 with or without TNF(50 ng/ml) or anti-TNFR2 agonist antibody (2.5 mg/ml). MAIT cell IFN-g expressionwas measured after 24 hours. (F) PBMCs were stimulated with ChAdOx1, and thefollowing inhibitors were added: vedolizumab (anti-a4b7 integrin antibody, n = 8donors; two experiments), adalimumab (anti-TNF antibody, n = 11 donors; three

    experiments), or etanercept (TNFR2-Fc fusion protein, n = 8 donors; two experiments)(10 mg/ml). MAIT cell IFN-g expression was measured after 24 hours. (G) PBMCswith or without CD14 depletion were stimulated with ChAdOx1. Concentrationof TNF in the supernatant was measured after 24 hours (n = 4 donors; oneexperiment). (H to J) C57BL/6J (n = 4), Il18rap−/− (n = 3), Tnfrsf1a−/−Tnfrsf1b−/−

    (n = 4), or Ifnar−/− (n = 4) mice were immunized i.m. with 108 IU of ChAdOx1-GFP.Naïve C57BL/6J mice (n = 4) were used as a control. After 24 hours, MAITcells were isolated from the iLNs and sorted for RNA-seq (one experiment). (H)Principal components analysis. (I) Heatmap of the up-regulated differentiallyexpressed genes (log2 FC > 1, adjusted P < 0.05) between MAIT cells fromChAdOx1-immunized and naïve C57BL/6J mice, with all other groups shown forcomparison. (J) Overlap of the genes up-regulated (log2 FC > 1, adjusted P < 0.05)in MAIT cells from ChAdOx1-immunized and naïve C57BL/6J mice, and the genesup-regulated in MAIT cells from ChAdOx1-immunized C57BL/6J mice as comparedto each of the ChAdOx1-immunized knockout strains. *P < 0.05; **P < 0.01.Unpaired t test [(B), (C), and (G)], repeated-measures one-way ANOVA with Dunnettcorrection [(E) and (F)]. Symbols indicate individual donors. Mean ± SEM is shown.

    RESEARCH | REPORTon July 7, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • of adenovirus vectors. This technology hasemerged as a potent platform for T cellimmunogenicity in clinical trials for HIV(23) and as vaccines for emerging virusessuch as Ebola (6) and SARS-CoV-2 (7, 8).This knowledge can be harnessed to improvethe design of these vaccines against majorpathogens and cancers.

    REFERENCES AND NOTES

    1. N. M. Provine, P. Klenerman, Annu. Rev. Immunol. 38, 203–228(2020).

    2. L. Loh et al., Proc. Natl. Acad. Sci. U.S.A. 113, 10133–10138(2016).

    3. B. van Wilgenburg et al., Nat. Commun. 7, 11653 (2016).4. B. van Wilgenburg et al., Nat. Commun. 9, 4706 (2018).5. A. Vitelli et al., Expert Rev. Vaccines 16, 1241–1252 (2017).6. European Medicines Agency, New vaccine for prevention of

    Ebola virus disease recommended for approval in theEuropean Union, press release (29 May 2020); www.ema.europa.eu/en/documents/press-release/new-vaccine-prevention-ebola-virus-disease-recommended-approval-european-union_en.pdf.

    7. P. M. Folegatti et al., Lancet 396, 467–478 (2020).8. F.-C. Zhu et al., Lancet 396, 479–488 (2020).9. K. M. Quinn et al., J. Clin. Invest. 125, 1129–1146 (2015).10. J. E. Teigler, M. J. Iampietro, D. H. Barouch, J. Virol. 86,

    9590–9598 (2012).11. M. J. Johnson et al., J. Immunol. 188, 6109–6118 (2012).12. A. Subramanian et al., Proc. Natl. Acad. Sci. U.S.A. 102,

    15545–15550 (2005).

    13. S. A. Samarajiwa, S. Forster, K. Auchettl, P. J. Hertzog,Nucleic Acids Res. 37 (suppl. 1), D852–D857 (2009).

    14. V. Hornung et al., Nat. Immunol. 9, 847–856 (2008).

    15. L. C. Garner, P. Klenerman, N. M. Provine, Front. Immunol. 9,1478 (2018).

    16. E. Treiner et al., Nature 422, 164–169 (2003).

    17. T. Donnison et al., Vaccine 38, 5036–5048 (2020).

    18. A. Gola et al., Sci. Transl. Med. 10, eaap9128 (2018).19. A. Varelias et al., J. Clin. Invest. 128, 1919–1936 (2018).20. A. Meierovics, W.-J. C. Yankelevich, S. C. Cowley,

    Proc. Natl. Acad. Sci. U.S.A. 110, E3119–E3128 (2013).21. A. I. Meierovics, S. C. Cowley, J. Exp. Med. 213, 2793–2809

    (2016).22. V. Peperzak et al., J. Immunol. 191, 3025–3036

    (2013).

    23. D. H. Barouch et al., Lancet 392, 232–243 (2018).

    Provine et al., Science 371, 521–526 (2021) 29 January 2021 5 of 6

    Fig. 4. Impact of MAIT cell deficiency onT cell responses after ChAdOx1 or ChAd63immunization. (A) Frequency of IFN-g–producingPBMCs measured by peptide enzyme-linkedimmunospot (ELISPOT) in ChAdOx1 MenB.1 vacci-nated volunteers before boost (n = 14) orday 14 after boost (n = 13). SFU, spot formingunits. (B) Spearman rank correlation analysis ofthe change in MAIT cell CD69 expression frombefore boost to day 1 after boost versus theincrease in IFN-g–producing PBMCs from beforeboost to day 14 after boost. (C) C57BL/6J (n = 12)or Mr1−/− (n = 9) mice were immunized i.m. with108 IU of ChAdOx1-HCV-GT1-6_D_TM-Ii+L (twoexperiments). On day 16, HCV-specific CD107a+,IFN-g+, TNF+, or IFN-g+TNF+ CD8+ T cell responseswere measured. (D) C57BL/6J (n = 12) or Mr1−/−

    (n = 11) mice were immunized i.m. with 108 IU ofChAdOx1-nCoV-19 (two experiments). On day 13,SARS-CoV-2 spike-specific CD107a+, IFN-g+, TNF+,or IFN-g+TNF+ CD8+ T cell responses weremeasured. (E and F) C57BL/6J (n = 12) orMr1−/− (n = 12, n = 11 after boost) were primed i.m.with 107 IU of ChAd63-OVA and boosted intra-venously on day 28 with 108 IU (squares) or 109 IU(circles) of ChAd63-OVA. SIINFEKL-specificCD107a+, IFN-g+, TNF+, or IFN-g+TNF+ CD8+ T cellresponses were measured either 3 weeks afterprime (E) or 3 weeks after boost (F). *P < 0.05;**P < 0.01; ***P < 0.001. Wilcoxon rank-sum test(A) or two-way ANOVA [(C) to (F)]. Symbolsindicate individual volunteers or mice. Mean ± SEMis shown.

    RESEARCH | REPORTon July 7, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    https://www.ema.europa.eu/en/documents/press-release/new-vaccine-prevention-ebola-virus-disease-recommended-approval-european-union_en.pdfhttps://www.ema.europa.eu/en/documents/press-release/new-vaccine-prevention-ebola-virus-disease-recommended-approval-european-union_en.pdfhttps://www.ema.europa.eu/en/documents/press-release/new-vaccine-prevention-ebola-virus-disease-recommended-approval-european-union_en.pdfhttps://www.ema.europa.eu/en/documents/press-release/new-vaccine-prevention-ebola-virus-disease-recommended-approval-european-union_en.pdfhttp://science.sciencemag.org/

  • ACKNOWLEDGMENTSWe thank H. Ferry and L. Hardy for assistance with cell sorting;S. Slevin, C.-P. Hackstein, and C. Willberg for critical discussions;M. Salio and V. Cerundolo for the Mr1−/− mice; J. Rehwinkelfor the Ifnar−/− mice; M. Esposito, H. Al-Mossawi, L. Ni Lee, andT. Donnison for reagents; the NIH Tetramer Facility for the MR1tetramers; and all of the volunteers for sample donation andparticipation in the trial. Funding: N.M.P. is supported by anOxford-UCB Postdoctoral Fellowship. A.A. is supported by aWellcome Clinical Training Fellowship (216417/Z/19/Z). L.C.G. issupported by a Wellcome PhD Studentship (109028/Z/15/Z).M.E.B.F. is supported by an Oxford-Celgene Doctoral Fellowship.S.B.M. and T.S.C.H. are supported by the Wellcome Trust (211050/Z/18/Z and 211050/Z/18/A). E.B. is supported by the MedicalResearch Council (STOP-HCV and MR/R014485/1), an NIHR SeniorFellowship, the NIHR Biomedical Research Centre (Oxford), andthe UKRI/NIHR through the UK Coronavirus ImmunologyConsortium (UK-CIC). C.S.R. is supported by the NIHR BiomedicalResearch Centre and is a Jenner Institute Investigator. A.J.P. is

    supported by the NIHR Oxford Biomedical Research Centre and isan NIHR Senior Investigator. P.K. is supported by the WellcomeTrust (WT109965MA), the NIHR Biomedical Research Centre(Oxford), the UKRI/NIHR through the UK Coronavirus ImmunologyConsortium (UK-CIC), and an NIHR Senior Fellowship. TheChAdOx1 MenB.1 clinical trial is funded by the Medical ResearchCouncil DPFS (MRM0076931). The views expressed are those ofthe authors and not necessarily those of the NHS, the NIHR, or theDepartment of Health. Author contributions: N.M.P. and P.K.designed the project. N.M.P., A.J.S., C.D., T.S.C.H., E.B., C.S.R.,A.J.P., and P.K. designed the experiments. N.M.P., A.A., L.C.G.,A.J.S., C.D., C.H., L.S.R., M.E.B.F., M.U., H.S., and S.B.M.performed the experiments. S.C., B.O., M.R., F.T., T.L., S.C., A.F.,E.B., C.S.R., and A.J.P. provided samples and reagents. Allauthors contributed to the writing and editing of the manuscript.Competing interests: C.D., C.S.R., and A.J.P. are namedinventors on a patent application in the field of meningococcalvaccines. A.J.P. waives his rights under any patent. P.K. is anamed inventor on a patent application in the field of cancer

    vaccines. Data and materials availability: All gene expressiondata are deposited in the Gene Expression Omnibus underGSE158835. All data are available in the manuscript or thesupplementary materials.

    SUPPLEMENTARY MATERIALS

    science.sciencemag.org/content/371/6528/521/suppl/DC1Materials and MethodsFigs. S1 to S12Tables S1 and S2

    References (24–44)

    MDAR Reproducibility Checklist

    Data S1 to S9

    View/request a protocol for this paper from Bio-protocol.

    1 May 2019; resubmitted 20 July 2020Accepted 19 November 202010.1126/science.aax8819

    Provine et al., Science 371, 521–526 (2021) 29 January 2021 6 of 6

    RESEARCH | REPORTon July 7, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    https://science.sciencemag.org/content/371/6528/521/suppl/DC1https://en.bio-protocol.org/cjrap.aspx?eid=10.1126/science.aax8819http://science.sciencemag.org/

  • MAIT cell activation augments adenovirus vector vaccine immunogenicity

    Christine S. Rollier, Andrew J. Pollard and Paul KlenermanSharpe, Sophie B. Morgan, Timothy S. C. Hinks, Teresa Lambe, Stefania Capone, Antonella Folgori, Eleanor Barnes,Michael E. B. FitzPatrick, Senthil Chinnakannan, Blanche Oguti, Meriel Raymond, Marta Ulaszewska, Fulvia Troise, Hannah Nicholas M. Provine, Ali Amini, Lucy C. Garner, Alexandra J. Spencer, Christina Dold, Claire Hutchings, Laura Silva Reyes,

    DOI: 10.1126/science.aax8819 (6528), 521-526.371Science

    , this issue p. 521; see also p. 460Sciencebe exploited to enhance the efficacy of vaccines.

    T cell immunity to target antigens after vaccination. This work suggests an additional pathway that could+impaired CD8positively correlated with vaccine-mediated T cell responses in human subjects, and mice deficient in MAIT cells showedplasmacytoid dendritic cells as well as monocyte-derived interleukin-18 and tumor necrosis factor. MAIT cell activation

    produced byαimmunized mice (see the Perspective by Juno and O'Connor). This activation required interferon- report that a leading adenoviral vector vaccine, ChAdOx1, activated MAIT cells inet al.context of viral infections. Provine

    recognize derivatives of microbiota-derived vitamin B2 precursors but can also be activated by certain cytokines in the Mucosal-associated invariant T (MAIT) cells are a T cell subset important for mucosal homeostasis. These cells

    Vaccines get a help-MAIT

    ARTICLE TOOLS http://science.sciencemag.org/content/371/6528/521

    MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2021/01/27/371.6528.521.DC1

    CONTENTRELATED

    http://stke.sciencemag.org/content/sigtrans/14/680/eabe1757.fullhttp://stm.sciencemag.org/content/scitransmed/11/519/eaaw1673.fullhttp://stm.sciencemag.org/content/scitransmed/11/519/eaav1800.fullhttp://stm.sciencemag.org/content/scitransmed/12/548/eaaz7715.fullhttp://stm.sciencemag.org/content/scitransmed/12/571/eabc6659.fullhttp://science.sciencemag.org/content/sci/371/6528/460.full

    REFERENCES

    http://science.sciencemag.org/content/371/6528/521#BIBLThis article cites 42 articles, 12 of which you can access for free

    PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

    Terms of ServiceUse of this article is subject to the

    is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

    Science. No claim to original U.S. Government WorksCopyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

    on July 7, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/content/371/6528/521http://science.sciencemag.org/content/suppl/2021/01/27/371.6528.521.DC1http://science.sciencemag.org/content/sci/371/6528/460.fullhttp://stm.sciencemag.org/content/scitransmed/12/571/eabc6659.fullhttp://stm.sciencemag.org/content/scitransmed/12/548/eaaz7715.fullhttp://stm.sciencemag.org/content/scitransmed/11/519/eaav1800.fullhttp://stm.sciencemag.org/content/scitransmed/11/519/eaaw1673.fullhttp://stke.sciencemag.org/content/sigtrans/14/680/eabe1757.fullhttp://science.sciencemag.org/content/371/6528/521#BIBLhttp://www.sciencemag.org/help/reprints-and-permissionshttp://www.sciencemag.org/about/terms-servicehttp://science.sciencemag.org/

Recommended