A Single-Dose Intranasal ChAd Vaccine Protects Upper and Lower Respiratory Tracts against SARS-CoV-2

Ahmed O Hassan, Natasha M Kafai, Igor P Dmitriev, Julie M Fox, Brittany K Smith, Ian B Harvey, Rita E Chen, Emma S Winkler, Alex W Wessel, James Brett Case, Elena Kashentseva, Broc T McCune, Adam L Bailey, Haiyan Zhao, Laura A VanBlargan, Ya-Nan Dai, Meisheng Ma, Lucas J Adams, Swathi Shrihari, Jonathan E Danis, Lisa E Gralinski, Yixuan J Hou, Alexandra Schäfer, Arthur S Kim, Shamus P Keeler, Daniela Weiskopf, Ralph S Baric, Michael J Holtzman, Daved H Fremont, David T Curiel, Michael S Diamond, Ahmed O Hassan, Natasha M Kafai, Igor P Dmitriev, Julie M Fox, Brittany K Smith, Ian B Harvey, Rita E Chen, Emma S Winkler, Alex W Wessel, James Brett Case, Elena Kashentseva, Broc T McCune, Adam L Bailey, Haiyan Zhao, Laura A VanBlargan, Ya-Nan Dai, Meisheng Ma, Lucas J Adams, Swathi Shrihari, Jonathan E Danis, Lisa E Gralinski, Yixuan J Hou, Alexandra Schäfer, Arthur S Kim, Shamus P Keeler, Daniela Weiskopf, Ralph S Baric, Michael J Holtzman, Daved H Fremont, David T Curiel, Michael S Diamond

Abstract

The coronavirus disease 2019 pandemic has made deployment of an effective vaccine a global health priority. We evaluated the protective activity of a chimpanzee adenovirus-vectored vaccine encoding a prefusion stabilized spike protein (ChAd-SARS-CoV-2-S) in challenge studies with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mice expressing the human angiotensin-converting enzyme 2 receptor. Intramuscular dosing of ChAd-SARS-CoV-2-S induces robust systemic humoral and cell-mediated immune responses and protects against lung infection, inflammation, and pathology but does not confer sterilizing immunity, as evidenced by detection of viral RNA and induction of anti-nucleoprotein antibodies after SARS-CoV-2 challenge. In contrast, a single intranasal dose of ChAd-SARS-CoV-2-S induces high levels of neutralizing antibodies, promotes systemic and mucosal immunoglobulin A (IgA) and T cell responses, and almost entirely prevents SARS-CoV-2 infection in both the upper and lower respiratory tracts. Intranasal administration of ChAd-SARS-CoV-2-S is a candidate for preventing SARS-CoV-2 infection and transmission and curtailing pandemic spread.

Keywords: COVID-19; IgA; SARS-CoV-2; T cells; antibody; intranasal; mucosal immunity; pathogenesis; protection; vaccine.

Conflict of interest statement

Declaration of Interests M.S.D. is a consultant for Inbios, Vir Biotechnology, and NGM Biopharmaceuticals and on the Scientific Advisory Board of Moderna. The Diamond laboratory has received unrelated funding support from Moderna, Vir Biotechnology, and Emergent BioSolutions. M.S.D., D.T.C., A.O.H., and I.P.D. have filed a disclosure with Washington University for possible development of ChAd-SARS-CoV-2. M.J.H. is a member of the DSMB for AstraZeneca and founder of NuPeak Therapeutics. The Baric laboratory has received unrelated funding support from Takeda, Pfizer, and Eli Lilly.

Copyright © 2020 Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Immunogenicity of ChAd-SARS-CoV-2-S (A) Diagram of transgene cassettes: ChAd-control has no transgene insert; ChAd-SARS-CoV-2-S encodes for SARS-CoV-2 S protein with the two indicated proline mutations. (B) Binding of ChAd-SARS-CoV-2-S transduced 293 cells with anti-S mAbs. (Left) Summary: +, ++, +++, and - indicate 50%, and no binding, respectively. MFI, mean fluorescence intensity. (Right) Representative flow cytometry histograms of two experiments are shown. (C–F) Four-week-old female BALB/c mice were immunized via intramuscular route with ChAd-control or ChAd-SARS-CoV-2-S and boosted 4 weeks later. Antibody responses in sera of immunized mice at day 21 after priming or boosting were evaluated. An ELISA measured anti-S and RBD IgG and IgA levels ([D] and [E]), and an FRNT determined neutralization activity (F). Data are pooled from two experiments (n = 15–30). (G and H) Cell-mediated responses were analyzed at day 7 post-booster immunization after re-stimulation with an S protein peptide pool (Table S1). Splenocytes were assayed for IFNγ and granzyme B expression in CD8+ T cells and granzyme B only in CD4+ T cells by flow cytometry (G). A summary of frequencies and numbers of positive cell populations is shown in (H) (n = 5). Bars indicate median values, and dotted lines are the limit of detection (LOD) of the assays. (I) Spleens were harvested at 7 days post-boost, and SARS-CoV-2 spike-specific IgG+ antibody-secreting cell (ASC) frequency was measured by ELISPOT (n = 13). (J) CD8+ T cells in the lung were assayed for IFNγ and granzyme B expression by flow cytometry after re-stimulation with an S protein peptide pool (n = 4–5). (K) Lung CD8+ T cells also were assayed for expression of CD69 and CD103. (L) Spleens were harvested at 7 days post-boosting, and SARS-CoV-2 spike-specific IgA+ ASC frequency was measured by ELISPOT (n = 4–5). (M and N) SARS-CoV-2 S- (M) and RBD-specific (N) IgG and IgA levels in BAL fluid were determined by ELISA (n = 4–5). Bars and columns show median values, and dotted lines indicate the limit of detection (LOD) of the assays. For (D), (F), (H), (I), and (J): Mann-Whitney test: ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns, not significant. See Figure S1, Figure S2, and S3 and Table S1.
Figure S1
Figure S1
ChAd-SARS-CoV-2-S Vaccine Induces Neutralizing Antibodies as Measured by FRNT, Related to Figure 1 Four-week old female BALB/c mice were primed or primed and boosted with ChAd-control or ChAd-SARS-CoV-2-S via intramuscular route. A-B. Serum samples from ChAd-control or ChAd-SARS-CoV-2 vaccinated mice were collected at day 21 after priming (A) or boosting (B) and assayed for neutralizing activity by FRNT. Serum neutralization curves corresponding to individual mice are shown for the indicated vaccines (n = 15-30 per group). Each point represents the mean of two technical replicates. (C). An ELISA measured anti-SARS-CoV-2 NP IgG responses in paired sera obtained 5 days before and 8 days after SARS-CoV-2 challenge of ChAd-control or ChAd-SARS-CoV-2-S mice vaccinated by an intramuscular route (n = 5: ∗∗p < 0.01; ∗∗∗p < 0.001; paired t test). Dotted lines represent the mean IgG titers from naive sera.
Figure S2
Figure S2
Gating Strategy for Analyzing T Cell Responses, Related to Figure 1 Four-week old female BALB/c mice were immunized with ChAd-control or ChAd-SARS-CoV-2-S and boosted four weeks later. T cell responses were analyzed in splenocytes at day 7 post-boost. Cells were gated for lymphocytes (FSC-A/SSC-A), singlets (SSC-W/SSC-H), live cells (Aqua-), CD45+, CD19- followed by CD4+ or CD8+ cell populations expressing IFNγ or granzyme B.
Figure S3
Figure S3
Impact of Pre-existing ChAd Immunity on Transduction of Mice with Hu-AdV5-hACE2, Related to Figures 1 and 2 Four-week old female BALB/c mice were primed or primed and boosted. Serum samples were collected one day prior to Hu-AdV5-hACE2 transduction. Neutralizing activity of Hu-AdV5-hACE2 in the sera from the indicated vaccine groups was determined by FRNT after prime only (A) or prime and boost (B). Each symbol represents a single animal; each point represents two technical repeats and bars indicate the range. A positive control (anti-Hu-Adv5 serum) is included as a frame of reference.
Figure 2
Figure 2
Protective Efficacy of Intramuscularly Delivered ChAd-SARS-CoV-2-S against SARS-CoV-2 Infection (A) Scheme of vaccination and challenge. Four-week-old BALB/c female mice were immunized ChAd-control or ChAd-SARS-CoV-2-S. Some mice received a booster dose of the homologous vaccine. On day 35 post-immunization, mice were challenged with SARS-CoV-2 as follows: animals were treated with anti-Ifnar1 mAb and transduced with Hu-AdV5-hACE2 via an intranasal route 1 day later. Five days later, mice were challenged with 4 × 105 focus-forming units (FFUs) of SARS-CoV-2 via the intranasal route. (B and C) Tissues were harvested at 4 and 8 dpi for analysis. Infectious virus in the lung was measured by plaque assay (B), and viral RNA levels were measured in the lung, spleen, and heart at 4 and 8 dpi by qRT-PCR (C; n = 3–7). (D) Viral RNA in situ hybridization using SARS-CoV-2 probe (brown color) in the lungs harvested at 4 dpi. Images show low- (top; scale bars, 100 μm) and medium- (middle; scale bars, 100 μm) power magnification with a high-power magnification inset (representative images from n = 3 per group). (E) Fold change in gene expression of indicated cytokines and chemokines from lung homogenates at 4 dpi was determined by qRT-PCR after normalization to Gapdh levels and comparison with naive unvaccinated, unchallenged controls (n = 7). (F and G) Mice that received a prime-boost immunization were challenged on day 35 post-booster immunization. Tissues were collected at 4 dpi for analysis. Infectious virus in the lung was determined by plaque assay (F), and viral RNA was measured in the lung, spleen, and heart using qRT-PCR (G; n = 6–7). (B, C, and E–G) Columns show median values, and dotted lines indicate the LOD of the assays. For (B), (C), (E), (F), and (G): Mann-Whitney test: ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001. See Figure S3.
Figure 3
Figure 3
Single-Dose Intramuscular Vaccination with ChAd-SARS-CoV-2-S Protects Mice against SARS-CoV-2-Induced Inflammation in the Lung Four-week-old female BALB/c mice were immunized with ChAd-control and ChAd-SARS-CoV-2-S and challenged following the scheme described in Figure 2. Lungs were harvested at 8 dpi. Sections were stained with hematoxylin and eosin and imaged at 40× (left; scale bar, 250 μm), 200× (middle; scale bar, 50 μm), and 400× (right; scale bar, 25 μm) magnifications. Each image is representative of a group of 3 mice.
Figure 4
Figure 4
Immune Responses after Intranasal Immunization of ChAd-SARS-CoV-2-S (A) Scheme of experiments. 5-week-old BALB/c female mice were immunized with ChAd-control or ChAd-SARS-CoV-2-S via an intranasal route. (B–D) Antibody responses in sera of immunized mice at 1 month after priming were evaluated. An ELISA measured SARS-CoV-2 S- and RBD-specific IgG (B) and IgA levels (C), and a FRNT determined neutralization activity (D). Data are pooled from two experiments with n = 10–25 mice per group. (E–J) Mice that received a booster dose were sacrificed 1 week later to evaluate mucosal and cell-mediated immune responses. SARS-CoV-2 S- and RBD-specific IgG (E) and IgA levels (F) in BAL fluid were determined by ELISA. Neutralizing activity of BAL fluid against SARS-CoV-2 was measured by FRNT (G). CD8+ T cells in the lung were assayed for IFNγ and granzyme B expression by flow cytometry after re-stimulation with an S protein peptide pool (H). CD8+ T cells in the lung also were phenotyped for expression of CD103 and CD69 (I). SARS-CoV-2 spike-specific IgG+ and IgA+ ASC frequency in the spleen harvested 1 week post-boost was measured by ELISPOT (J). Data for mucosal and cell-mediated responses are pooled from two experiments (E–I: n = 7–9 per group; J: n = 5 per group). (B–J) Bars and columns show median values, and dotted lines indicate the LOD of the assays. Mann-Whitney test: ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. See Figure S4 and Table S1.
Figure S4
Figure S4
Intranasal Inoculation of ChAd-SARS-CoV-2-S Induces Neutralizing Antibodies as Measured by FRNT and Protects against SARS-CoV-2 Replication, Related to Figures 4 and 5 Five-week old female BALB/c mice were immunized with ChAd-control or ChAd-SARS-CoV-2-S via an intranasal inoculation route. Serum samples collected one month after immunization were assayed for neutralizing activity by FRNT. Mice were boosted at day 30 after priming and were sacrificed one week later to evaluate immune responses. (A) Serum samples from ChAd-control or ChAd-SARS-CoV-2-S vaccinated mice were tested for neutralizing activity with SARS-CoV-2 strain 2019 n-CoV/USA_WA1/2020 (n = 8-10 per group). (B) Serum samples from ChAd-SARS-CoV-2-S vaccinated mice were tested for neutralization of recombinant luciferase-expressing SARS-CoV-2 viruses (wild-type (left) and D614G variant (middle)). (Right) Paired EC50 values are indicated (n = 5; n.s. not significant, paired t test). (C) BAL fluid was collected from ChAd-control or ChAd-SARS-CoV-2-S vaccinated mice, and neutralization of SARS-CoV-2 strain 2019 n-CoV/USA_WA1/2020 was measured using a FRNT assay (n = 8-10 per group). Each point represents the mean of two technical replicates. (D) On day 35 post-immunization, mice were challenged via intranasal route with 4 × 105 FFU of SARS-CoV-2 five days after Hu-AdV5-hACE2 transduction and anti-Ifnar1 mAb treatment as described in Figure 2. Tissues were collected at 4 dpi for viral burden measurements. Genomic RNA (ORF1a) levels were determined in lungs and nasal turbinates (two experiments, n = 6-9; ∗∗∗∗p < 0.0001; Mann-Whitney test). Columns show median values, and dotted lines indicate the LOD of the assays.
Figure 5
Figure 5
Single-Dose Intranasal Immunization with ChAd-SARS-CoV-2-S Protects against SARS-CoV-2 Infection Five-week-old BALB/c female mice were immunized with ChAd-control or ChAd-SARS-CoV-2-S via an intranasal route. On day 35 post-immunization, mice were challenged as follows: animals were treated with anti-Ifnar1 mAb and transduced with Hu-AdV5-hACE2 via the intranasal route 1 day later. Five days later, mice were challenged intranasally with 4 × 105 FFUs of SARS-CoV-2. (A–C) Tissues and nasal washes were collected at 4 and 8 dpi for analysis. Infectious virus in the lung was measured by plaque assay (A). Viral RNA levels in the lung, spleen, heart, nasal turbinates, and nasal washes were measured at 4 and 8 dpi by qRT-PCR (B). Fold change in gene expression of indicated cytokines and chemokines was determined by qRT-PCR, normalized to Gapdh, and compared to naive controls in lung homogenates at 4 dpi (C; 2 experiments, n = 6–9; median values are shown). Columns show median values, and dotted lines indicate the LOD of the assays. (D) Lungs were harvested at 8 dpi. Sections were stained with hematoxylin and eosin and imaged at 40× (left; scale bar, 250 μm), 200× (middle; scale bar, 50 μm), and 400× (right; scale bar, 25 μm) magnifications. Each image is representative of a group of 3 mice. (E) An ELISA measured anti-SARS-CoV-2 NP IgM (left) and IgG (right) antibody responses in paired sera obtained 5 days before and 8 days after SARS-CoV-2 challenge of ChAd-control or ChAd-SARS-CoV-2-S mice vaccinated by an intranasal route (n = 6). Dotted lines represent the LOD of the assay. Dashed lines indicate the mark for a 4-fold increase of pre-boost IgM and IgG levels. For (A)–(C): Mann-Whitney test: ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001; for (E): ∗∗p < 0.01, ∗∗∗∗p < 0.0001; paired t test. See Figures S4 and S5.
Figure S5
Figure S5
SARS-CoV-2 NP-Specific IgM and IgG Antibody Responses following SARS-CoV-2 Challenge, Related to Figure 5 Five-week-old BALB/c female mice were immunized with ChAd-control or ChAd-SARS-CoV-2-S via an intranasal route. One month later, mice were transduced with Hu-AdV5-ACE2 and challenged with SARS-CoV-2 as described in Figure 5. An ELISA measured anti-SARS-CoV-2 NP IgM and IgG responses in paired sera obtained 5 days before and 8 days after SARS-CoV-2 challenge. Serum ELISA curves corresponding to individual mice are shown for the indicated vaccines or control naive BALB/c mice (n = 6 per group).
Figure 6
Figure 6
Immunogenicity and Protective Efficacy after Intranasal Immunization of ChAd-SARS-CoV-2-S in K18-hACE2 Mice (A) Scheme of experiments. Four-week-old K18-hACE2 female mice were immunized with ChAd-control or ChAd-SARS-CoV-2-S via an intranasal route. (B–D) Antibody responses in sera of immunized mice at 4 weeks after priming were evaluated. An ELISA measured SARS-CoV-2 S-and RBD-specific IgG (B) and IgA levels (C), and a FRNT determined neutralization activity (D; n = 7). (E–G) At 1 month post-immunization, mice were challenged with 103 FFUs of SARS-CoV-2. Tissues and nasal washes were harvested at 4 dpi. Infectious virus in lungs was measured using plaque assay (E). Viral RNA levels in the lung, spleen, heart, nasal turbinates, and nasal washes were measured at 4 dpi by qRT-PCR (F). Fold change in gene expression of indicated cytokines and chemokines was determined by qRT-PCR, normalized to Gapdh, and compared to naive controls in lung homogenates at 4 dpi (G; n = 7). Boxes indicate median values, and dotted lines indicate the LOD of the assays. For (B)–(G): Mann-Whitney test: ∗∗p < 0.01; ∗∗∗p < 0.001. See Figure S6.
Figure S6
Figure S6
Intranasal Inoculation of ChAd-SARS-CoV-2-S Induces Neutralizing Antibodies in K18-hACE2 Mice, Related to Figure 6 Four-week old female K18-hACE2 mice were immunized with ChAd-control or ChAd-SARS-CoV-2-S via an intranasal inoculation route. Serum samples collected four weeks after immunization were assayed for neutralizing activity by FRNT (n = 7 per group). Each point represents the mean of two technical replicates.
Figure 7
Figure 7
Comparison of Immunogenicity of Single-Dose Administration of ChAd-SARS-CoV-2-S by Intramuscular and Intranasal Delivery Routes (Upper panel) After intramuscular inoculation, ChAd-SARS-CoV-2 vaccine transduces antigen-presenting cells (APCs) at the site of injection in muscle tissues. APCs migrate to lymphoid tissues where antigen-specific CD8+T cells become activated, proliferate, and produce IFNγ and granzyme B. Antigen-specific B cells proliferate, some of which become plasmablasts and plasma cells that secrete anti-S IgG. After SARS-CoV-2 challenge, activated CD8+ T cells migrate to the lungs to control infection and anti-S IgG neutralizes virus particles. Intramuscular vaccination protects against lower, but not upper, airway infection and does not efficiently induce mucosal immunity. (Lower panel) After intranasal inoculation, ChAd-SARS-CoV-2 transduces APCs in the upper respiratory tract. APCs then migrate to bronchial- or mucosal-associated lymphoid tissues to present antigens to lymphocytes, including B and T cells. After SARS-CoV-2 challenge, activated CD8+ T cells migrate to the lungs, secrete cytokines, and attack virus-infected cells. Some CD8+ T cells likely adopt a tissue-resident memory phenotype (CD103+ CD69+), enabling them to reside in the lung (or upper airway) and respond more rapidly after re-encountering cognate antigen (SARS-CoV-2 S peptides). The activated B cell becomes responsive after intranasal vaccination produces cells that secrete anti-SARS-CoV-2-S IgG or IgA, the latter of which neutralizes virus within the upper and lower respiratory tracts. The mucosal immunity generated by intranasal inoculation of ChAd-SARS-CoV-2 likely controls infection at the point of initiation in the upper respiratory tract.

References

    1. Abbink P., Larocca R.A., De La Barrera R.A., Bricault C.A., Moseley E.T., Boyd M., Kirilova M., Li Z., Ng’ang’a D., Nanayakkara O. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys. Science. 2016;353:1129–1132.
    1. Alharbi N.K., Qasim I., Almasoud A., Aljami H.A., Alenazi M.W., Alhafufi A., Aldibasi O.S., Hashem A.M., Kasem S., Albrahim R. Humoral immunogenicity and efficacy of a single dose of ChAdOx1 MERS vaccine candidate in dromedary camels. Sci. Rep. 2019;9:16292.
    1. Alsoussi W.B., Turner J.S., Case J.B., Zhao H., Schmitz A.J., Zhou J.Q., Chen R.E., Lei T., Rizk A.A., McIntire K.M. A potently neutralizing antibody protects mice against SARS-CoV-2 infection. J. Immunol. 2020;205:915–922.
    1. Amanat F., Krammer F. SARS-CoV-2 vaccines: status report. Immunity. 2020;52:583–589.
    1. Bolles M., Deming D., Long K., Agnihothram S., Whitmore A., Ferris M., Funkhouser W., Gralinski L., Totura A., Heise M., Baric R.S. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J. Virol. 2011;85:12201–12215.
    1. Burton D.R., Walker L.M. Rational vaccine design in the time of COVID-19. Cell Host Microbe. 2020;27:695–698.
    1. Calzas C., Chevalier C. Innovative mucosal vaccine formulations against influenza A virus infections. Front. Immunol. 2019;10:1605.
    1. Cao Y., Su B., Guo X., Sun W., Deng Y., Bao L., Zhu Q., Zhang X., Zheng Y., Geng C. Potent neutralizing antibodies against SARS-CoV-2 identified by high-throughput single-cell sequencing of convalescent patients’ B cells. Cell. 2020;182:73–84.e16.
    1. Case J.B., Rothlauf P.W., Chen R.E., Liu Z., Zhao H., Kim A.S., Bloyet L.M., Zeng Q., Tahan S., Droit L. Neutralizing antibody and soluble ACE2 inhibition of a replication-competent VSV-SARS-CoV-2 and a clinical isolate of SARS-CoV-2. Cell Host Microbe. 2020 doi: 10.1016/j.chom.2020.06.021. Published July 3, 2020.
    1. Chandrashekar A., Liu J., Martinot A.J., McMahan K., Mercado N.B., Peter L., Tostanoski L.H., Yu J., Maliga Z., Nekorchuk M. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science. 2020;369:812–817.
    1. Cheung E.W., Zachariah P., Gorelik M., Boneparth A., Kernie S.G., Orange J.S., Milner J.D. Multisystem inflammatory syndrome related to COVID-19 in previously healthy children and adolescents in New York City. JAMA. 2020 doi: 10.1001/jama.2020.10374. Published online June 8, 2020.
    1. Corbett K.S., Flynn B., Foulds K.E., Francica J.R., Boyoglu-Barnum S., Werner A.P., Flach B., O’Connell S., Bock K.W., Minai M. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in nonhuman primates. N. Engl. J. Med. 2020 doi: 10.1056/NEJMoa2024671. Published online July 28, 2020.
    1. Davis C.W., Jackson K.J.L., McElroy A.K., Halfmann P., Huang J., Chennareddy C., Piper A.E., Leung Y., Albarino C.G., Crozier I. Longitudinal Analysis of the Human B Cell Response to Ebola Virus Infection. Cell. 2019;177:1566–1582.
    1. de Alwis R., Chen S., Gan E.S., Ooi E.E. Impact of immune enhancement on Covid-19 polyclonal hyperimmune globulin therapy and vaccine development. EBioMedicine. 2020;55:102768.
    1. de Wit E., van Doremalen N., Falzarano D., Munster V.J. SARS and MERS: recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 2016;14:523–534.
    1. Diamond M.S., Pierson T.C. The challenges of vaccine development against a new virus during a pandemic. Cell Host Microbe. 2020;27:699–703.
    1. Dicks M.D., Spencer A.J., Edwards N.J., Wadell G., Bojang K., Gilbert S.C., Hill A.V., Cottingham M.G. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity. PLoS ONE. 2012;7:e40385.
    1. Douglas A.D., de Cassan S.C., Dicks M.D., Gilbert S.C., Hill A.V., Draper S.J. Tailoring subunit vaccine immunogenicity: maximizing antibody and T cell responses by using combinations of adenovirus, poxvirus and protein-adjuvant vaccines against Plasmodium falciparum MSP1. Vaccine. 2010;28:7167–7178.
    1. Dutta A., Huang C.T., Lin C.Y., Chen T.C., Lin Y.C., Chang C.S., He Y.C. Sterilizing immunity to influenza virus infection requires local antigen-specific T cell response in the lungs. Sci. Rep. 2016;6:32973.
    1. Folegatti P.M., Bittaye M., Flaxman A., Lopez F.R., Bellamy D., Kupke A., Mair C., Makinson R., Sheridan J., Rohde C. Safety and immunogenicity of a candidate Middle East respiratory syndrome coronavirus viral-vectored vaccine: a dose-escalation, open-label, non-randomised, uncontrolled, phase 1 trial. Lancet Infect. Dis. 2020;20:816–826.
    1. Folegatti P.M., Ewer K.J., Aley P.K., Angus B., Becker S., Belij-Rammerstorfer S., Bellamy D., Bibi S., Bittaye M., Clutterbuck E.A. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet. 2020;396:467–478.
    1. Gao Q., Bao L., Mao H., Wang L., Xu K., Yang M., Li Y., Zhu L., Wang N., Lv Z. Development of an inactivated vaccine candidate for SARS-CoV-2. Science. 2020;369:77–81.
    1. Graham B.S. Rapid COVID-19 vaccine development. Science. 2020;368:945–946.
    1. Grifoni A., Weiskopf D., Ramirez S.I., Mateus J., Dan J.M., Moderbacher C.R., Rawlings S.A., Sutherland A., Premkumar L., Jadi R.S. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020;181:1489–1501.e15.
    1. Guan W.J., Ni Z.Y., Hu Y., Liang W.H., Ou C.Q., He J.X., Liu L., Shan H., Lei C.L., Hui D.S.C., China Medical Treatment Expert Group for Covid-19 Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020;382:1708–1720.
    1. Hashem A.M., Algaissi A., Agrawal A.S., Al-Amri S.S., Alhabbab R.Y., Sohrab S.S., Almasoud A.S., Alharbi N.K., Peng B.-H., Russell M. A highly immunogenic, protective, and safe adenovirus-based vaccine expressing Middle East respiratory syndrome coronavirus S1-CD40L fusion protein in a transgenic human dipeptidyl peptidase 4 mouse model. J. Infect. Dis. 2019;220:1558–1567.
    1. Hassan A.O., Dmitriev I.P., Kashentseva E.A., Zhao H., Brough D.E., Fremont D.H., Curiel D.T., Diamond M.S. A gorilla adenovirus-based vaccine against Zika virus induces durable immunity and confers protection in pregnancy. Cell Rep. 2019;28:2634–2646.e4.
    1. Hassan A.O., Case J.B., Winkler E.S., Thackray L.B., Kafai N.M., Bailey A.L., McCune B.T., Fox J.M., Chen R.E., Alsoussi W.B. A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies. Cell. 2020;182:744–753.e4.
    1. He T.C., Zhou S., da Costa L.T., Yu J., Kinzler K.W., Vogelstein B. A simplified system for generating recombinant adenoviruses. Proc. Natl. Acad. Sci. USA. 1998;95:2509–2514.
    1. Hillen W., Berens C. Mechanisms underlying expression of Tn10 encoded tetracycline resistance. Annu. Rev. Microbiol. 1994;48:345–369.
    1. Hodgson S.H., Ewer K.J., Bliss C.M., Edwards N.J., Rampling T., Anagnostou N.A., de Barra E., Havelock T., Bowyer G., Poulton I.D. Evaluation of the efficacy of ChAd63-MVA vectored vaccines expressing circumsporozoite protein and ME-TRAP against controlled human malaria infection in malaria-naive individuals. J. Infect. Dis. 2015;211:1076–1086.
    1. Hoffmann M., Kleine-Weber H., Schroeder S., Krüger N., Herrler T., Erichsen S., Schiergens T.S., Herrler G., Wu N.H., Nitsche A. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181:271–280.e8.
    1. Kobinger G.P., Figueredo J.M., Rowe T., Zhi Y., Gao G., Sanmiguel J.C., Bell P., Wivel N.A., Zitzow L.A., Flieder D.B. Adenovirus-based vaccine prevents pneumonia in ferrets challenged with the SARS coronavirus and stimulates robust immune responses in macaques. Vaccine. 2007;25:5220–5231.
    1. Korber B., Fischer W.M., Gnanakaran S., Yoon H., Theiler J., Abfalterer W., Hengartner N., Giorgi E.E., Bhattacharya T., Foley B. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell. 2020;182:812–827.e19.
    1. Laurie K.L., Carolan L.A., Middleton D., Lowther S., Kelso A., Barr I.G. Multiple infections with seasonal influenza A virus induce cross-protective immunity against A(H1N1) pandemic influenza virus in a ferret model. J. Infect. Dis. 2010;202:1011–1020.
    1. Letko M., Marzi A., Munster V. Functional assessment of cell entry and receptor usage for SARS-CoV-2 and other lineage B betacoronaviruses. Nat. Microbiol. 2020;5:562–569.
    1. Liu L., Wei Q., Lin Q., Fang J., Wang H., Kwok H., Tang H., Nishiura K., Peng J., Tan Z. Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight. 2019;4:e123158.
    1. López-Camacho C., Abbink P., Larocca R.A., Dejnirattisai W., Boyd M., Badamchi-Zadeh A., Wallace Z.R., Doig J., Velazquez R.S., Neto R.D.L. Rational Zika vaccine design via the modulation of antigen membrane anchors in chimpanzee adenoviral vectors. Nat. Commun. 2018;9:2441.
    1. Maizel J.V., Jr., White D.O., Scharff M.D. The polypeptides of adenovirus. I. Evidence for multiple protein components in the virion and a comparison of types 2, 7A, and 12. Virology. 1968;36:115–125.
    1. Mao R., Qiu Y., He J.-S., Tan J.-Y., Li X.-H., Liang J., Shen J., Zhu L.-R., Chen Y., Iacucci M. Manifestations and prognosis of gastrointestinal and liver involvement in patients with COVID-19: a systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2020;5:667–678.
    1. McCray P.B., Jr., Pewe L., Wohlford-Lenane C., Hickey M., Manzel L., Shi L., Netland J., Jia H.P., Halabi C., Sigmund C.D. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 2007;81:813–821.
    1. Munster V.J., Wells D., Lambe T., Wright D., Fischer R.J., Bushmaker T., Saturday G., van Doremalen N., Gilbert S.C., de Wit E., Warimwe G.M. Protective efficacy of a novel simian adenovirus vaccine against lethal MERS-CoV challenge in a transgenic human DPP4 mouse model. NPJ Vaccines. 2017;2:28.
    1. Munster V.J., Feldmann F., Williamson B.N., van Doremalen N., Pérez-Pérez L., Schulz J., Meade-White K., Okumura A., Callison J., Brumbaugh B. Respiratory disease in rhesus macaques inoculated with SARS-CoV-2. Nature. 2020 doi: 10.1038/s41586-020-2324-7. Published online May 12, 2020.
    1. Neutra M.R., Kozlowski P.A. Mucosal vaccines: the promise and the challenge. Nat. Rev. Immunol. 2006;6:148–158.
    1. Ni L., Ye F., Cheng M.L., Feng Y., Deng Y.Q., Zhao H., Wei P., Ge J., Gou M., Li X. Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity. 2020;52:971–977.e3.
    1. Pallesen J., Wang N., Corbett K.S., Wrapp D., Kirchdoerfer R.N., Turner H.L., Cottrell C.A., Becker M.M., Wang L., Shi W. Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen. Proc. Natl. Acad. Sci. USA. 2017;114:E7348–E7357.
    1. Penaloza-MacMaster P., Provine N.M., Ra J., Borducchi E.N., McNally A., Simmons N.L., Iampietro M.J., Barouch D.H. Alternative serotype adenovirus vaccine vectors elicit memory T cells with enhanced anamnestic capacity compared to Ad5 vectors. J. Virol. 2013;87:1373–1384.
    1. Pinto D., Park Y.J., Beltramello M., Walls A.C., Tortorici M.A., Bianchi S., Jaconi S., Culap K., Zatta F., De Marco A. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature. 2020;583:290–295.
    1. Rathnasinghe R., Strohmeier S., Amanat F., Gillespie V.L., Krammer F., García-Sastre A., Coughlan L., Schotsaert M., Uccellini M. Comparison of transgenic and adenovirus hACE2 mouse models for SARS-CoV-2 infection. bioRxiv. 2020 doi: 10.1101/2020.07.06.190066.
    1. Reyes-Sandoval A., Berthoud T., Alder N., Siani L., Gilbert S.C., Nicosia A., Colloca S., Cortese R., Hill A.V. Prime-boost immunization with adenoviral and modified vaccinia virus Ankara vectors enhances the durability and polyfunctionality of protective malaria CD8+ T-cell responses. Infect. Immun. 2010;78:145–153.
    1. Roy S., Medina-Jaszek A., Wilson M.J., Sandhu A., Calcedo R., Lin J., Wilson J.M. Creation of a panel of vectors based on ape adenovirus isolates. J. Gene Med. 2011;13:17–25.
    1. Sabo M.C., Luca V.C., Prentoe J., Hopcraft S.E., Blight K.J., Yi M., Lemon S.M., Ball J.K., Bukh J., Evans M.J. Neutralizing monoclonal antibodies against hepatitis C virus E2 protein bind discontinuous epitopes and inhibit infection at a postattachment step. J. Virol. 2011;85:7005–7019.
    1. Sheehan K.C., Lai K.S., Dunn G.P., Bruce A.T., Diamond M.S., Heutel J.D., Dungo-Arthur C., Carrero J.A., White J.M., Hertzog P.J., Schreiber R.D. Blocking monoclonal antibodies specific for mouse IFN-alpha/beta receptor subunit 1 (IFNAR-1) from mice immunized by in vivo hydrodynamic transfection. J. Interferon Cytokine Res. 2006;26:804–819.
    1. Slifka M.K., Amanna I. How advances in immunology provide insight into improving vaccine efficacy. Vaccine. 2014;32:2948–2957.
    1. Takamura S. Persistence in temporary lung niches: a survival strategy of lung-resident memory CD8+ T cells. Viral Immunol. 2017;30:438–450.
    1. van Doremalen N., Lambe T., Spencer A., Belij-Rammerstorfer S., Purushotham J.N., Port J.R., Avanzato V.A., Bushmaker T., Flaxman A., Ulaszewska M. ChAdOx1nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques. Nature. 2020;30 doi: 10.1038/s41586-020-2608.
    1. Weingartl H., Czub M., Czub S., Neufeld J., Marszal P., Gren J., Smith G., Jones S., Proulx R., Deschambault Y. Immunization with modified vaccinia virus Ankara-based recombinant vaccine against severe acute respiratory syndrome is associated with enhanced hepatitis in ferrets. J. Virol. 2004;78:12672–12676.
    1. Wichmann D., Sperhake J.-P., Lütgehetmann M., Steurer S., Edler C., Heinemann A., Heinrich F., Mushumba H., Kniep I., Schröder A.S. Autopsy findings and venous thromboembolism in patients with COVID-19: a prospective cohort study. Ann. Intern. Med. 2020 doi: 10.7326/M20-2003. Published online May 6, 2020.
    1. Winkler E.S., Bailey A.L., Kafai N.M., Nair S., McCune B.T., Yu J., Fox J.M., Chen R.E., Earnest J.T., Keeler S.P. SARS-CoV-2 infection in the lungs of human ACE2 transgenic mice causes severe inflammation, immune cell infiltration, and compromised respiratory function. bioRxiv. 2020 doi: 10.1101/2020.07.09.196188.
    1. Wrapp D., Wang N., Corbett K.S., Goldsmith J.A., Hsieh C.-L., Abiona O., Graham B.S., McLellan J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367:1260–1263.
    1. Yu J., Tostanoski L.H., Peter L., Mercado N.B., McMahan K., Mahrokhian S.H., Nkolola J.P., Liu J., Li Z., Chandrashekar A. DNA vaccine protection against SARS-CoV-2 in rhesus macaques. Science. 2020;369:806–811.
    1. Yuan M., Wu N.C., Zhu X., Lee C.D., So R.T.Y., Lv H., Mok C.K.P., Wilson I.A. A highly conserved cryptic epitope in the receptor binding domains of SARS-CoV-2 and SARS-CoV. Science. 2020;368:630–633.
    1. Yusuf H., Kett V. Current prospects and future challenges for nasal vaccine delivery. Hum. Vaccin. Immunother. 2017;13:34–45.
    1. Zhou F., Yu T., Du R., Fan G., Liu Y., Liu Z., Xiang J., Wang Y., Song B., Gu X. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062.
    1. Zhou P., Yang X.L., Wang X.G., Hu B., Zhang L., Zhang W., Si H.R., Zhu Y., Li B., Huang C.L. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273.
    1. Zhu F.C., Li Y.H., Guan X.H., Hou L.H., Wang W.J., Li J.X., Wu S.P., Wang B.S., Wang Z., Wang L. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: a dose-escalation, open-label, non-randomised, first-in-human trial. Lancet. 2020;395:1845–1854.
    1. Zost S.J., Gilchuk P., Chen R.E., Case J.B., Reidy J.X., Trivette A., Nargi R.S., Sutton R.E., Suryadevara N., Chen E.C. Rapid isolation and profiling of a diverse panel of human monoclonal antibodies targeting the SARS-CoV-2 spike protein. Nat. Med. 2020 doi: 10.1038/s41591-020-0998-x. Published online July 10, 2020.

Source: PubMed

3
Iratkozz fel