Summary
Recent publications have explored intranasal (i.n.) adenovirus-based (Ad) vaccines as an effective strategy for SARS-CoV-2 in pre-clinical models. However, the effects of prior immunizations and infections have yet to be considered. Here, we investigate the immunomodulatory effects of Mycobacterium bovis BCG pre-immunization followed by vaccination with an S-protein-expressing i.n. Ad, termed Ad(Spike). While i.n. Ad(Spike) retains some protective effect after 6 months, a single administration of BCG-Danish prior to Ad(Spike) potentiates its ability to control viral replication of the B.1.351 SARS-CoV-2 variant within the respiratory tract. Though BCG-Danish did not affect Ad(Spike)-generated humoral immunity, it promoted the generation of cytotoxic/Th1 responses over suppressive FoxP3+ TREG cells in the lungs of infected mice. Thus, this vaccination strategy may prove useful in limiting future pandemics by potentiating the long-term efficacy of mucosal vaccines within the context of the widely distributed BCG vaccine.
Subject areas: Biological sciences, Immunology, Immune response, Microbiology
Graphical abstract
Highlights
-
•
SARS-CoV-2 intranasal adenovirus vaccine immunity was found to wane over six months
-
•
BCG-primed mice had enhanced viral clearance/vaccine immunity after COVID-19 vaccination
-
•
BCG promotes COVID-19 vaccine-specific cytotoxic/TH1 responses over TREG responses
-
•
BCG is safe, widely available, and has the potential to synergize with other vaccines
Biological sciences; Immunology; Immune response; Microbiology
Introduction
The COVID-19 pandemic has resulted in over 766 million cases and over 6.9 million deaths as of May 2023.1 Infection with SARS-CoV-2 can cause a broad spectrum of disease ranging from mild symptoms to severe lung injury and multi-organ failure, potentially leading to death, especially in the elderly and those with comorbidities.2 Additionally, there is evidence that recovered individuals can experience symptoms termed “long COVID,” which can involve multiple organ systems (e.g., lung, heart, kidneys, liver, etc.).3 Consequently, COVID-19 has had severe consequences on global health and the economy and has been the target of novel immunization approaches. Vaccination, in combination with public health measures, has effectively slowed the progression and hospitalization rates associated with SARS-CoV-24; however, these immunization strategies have so far failed to fully prevent viral transmission and infection.5 Moreover, vaccine efficacy at preventing infection declines by six months after full vaccination,6 and breakthrough infections, especially by novel SARS-CoV-2 variants, have been reported in previously vaccinated individuals.7 In addition, there is an urgent need to provide equitable access to affordable and effective vaccines amongst developing nations to prevent the ongoing morbidity and mortality as well as reduce the risk of novel variants emerging.
Among the strategies in preclinical development, a promising approach involves the use of intranasal (i.n.) vaccines, theoretically capable of eliciting local mucosal immune responses within the respiratory tract that can effectively neutralize SARS-CoV-2 entry and prevent viral replication at the site of initial infection.8 However, while some vaccines are undergoing phase I/II clinical trials, the success of this approach has thus far been elusive.9 For example, i.n. administration of a chimpanzee adenovirus-based vaccine carrying the Spike protein (ChAd-SARS-CoV-2-S) successfully generated neutralizing IgA antibodies and T cell responses in the lungs of hACE-2 mice10 and provides at least one month of protection in mice and rhesus macaques.11 However, a recent progress report from a phase I clinical trial of the i.n. administration of ChadOx1 has announced a failure to substantially increase mucosal immunity in humans.12 Nonetheless, the safety results were sufficiently encouraging to pursue approaches that enhance mucosal immunity generated using adenovirus vaccine vectors.
In many countries, the live-attenuated Mycobacterium bovis Bacillus Calmette-Guérin (BCG) vaccine is administered intradermally to newborns soon after birth to protect them from disseminated Mycobacterium tuberculosis infection. In fact, most individuals worldwide are BCG vaccinated; of the approximately 140 million babies born per year, approximately 100 million of them are vaccinated with BCG.13 Curiously, vaccination with BCG has also been reported to reduce child mortality14 from neonatal sepsis and lower respiratory tract infections.15 There is cumulating evidence that BCG acts on the antiviral immune response by boosting the activity of innate immune cells, a concept known as trained immunity,16 as well as by promoting heterologous T cell activation.17 As such, many researchers have speculated on the possibility that BCG could provide immunity against SARS-CoV-2 infection,18 although there are limited experimental data thus far to support this idea.19,20 A multi-country randomized trial with over 3,000 healthcare workers compared BCG vaccination to placebo, but it did not show a lower COVID-19 risk in the BCG group.21 Still, there remains great promise that BCG can be combined with conventional SARS-CoV-2 vaccination strategies to improve their effectiveness.22 As such, we hypothesized that BCG may provide a novel, cost-effective, and safe priming strategy to enhance the long-term efficacy of i.n. immunization with adenovirus-based COVID-19 vaccines.
Here, we sought to evaluate the effect of prior administration of BCG on the immunogenicity and efficacy of an i.n. human adenovirus (AdV) serotype 5 vaccine expressing the SARS-CoV-2 S (Spike)-protein, referred to as Ad(Spike), in a C57BL/6 mouse model of infection with the B.1.351 variant of SARS-CoV-2. Previous work has shown that a single i.n. immunization with adenovirus-based vaccines against SARS-CoV-2 is sufficient to provide effective protection from infection in naive animals.23 In this report, we demonstrate that the protection conferred by our intranasally administered Ad(Spike) vaccine alone declines in mice by 6 months post-immunization. Importantly, a single dose of BCG administered systemically prior to Ad(Spike) vaccination was capable of boosting the Spike-specific cytotoxic T cell response in the lungs and, in doing so, significantly decreased the replication and production of infectious viral particles up to 6 months after i.n. immunization with the Ad(Spike) vaccine. This approach is analogous to previous “prime-pull” vaccination strategies that employ a conventional vaccine to elicit systemic T cell responses (“prime”), followed by the recruitment of these activated T cells and associated cytokines to the desired body site via the administration of a localized, secondary vaccine (“pull”).24 However, in this instance, we attempted to exploit the non-specific or off-target immune enhancing properties of BCG as the priming mechanism. Overall, the results herein provide proof-of-concept data that highlight a novel and innovative approach to the use of heterologous vaccine strategies incorporating the widely approved BCG vaccine to protect against current and future viral pandemics.
Results
The in vivo protection provided by a single dose of intranasal recombinant Ad(Spike) attenuates with time
To investigate the long-term protection provided by i.n. vaccine administration, we developed a replication-deficient (ΔE1-, ΔE3-) human adenovirus serotype 5 expressing the full-length S-protein of the ancestral strain of SARS-Co-V-2 (Ad(Spike)) which was codon optimized for expression in human and mouse cell lines (Figure 1A). Exogenous gene expression was verified in vitro by western blot using S-protein specific antibodies (Figure 1B). Female C57BL/6 mice were vaccinated with either PBS or 109 TCID50 Ad(Spike) i.n. The infectious dose of Ad(Spike) was determined using a pilot study and was chosen based on the induction of Spike-specific IgA antibodies in lung homogenates from vaccinated animals (Figure S1). Study animals were challenged 2 or 6 months later with the Beta variant (B.1.351) of SARS-CoV-2. In each case, the animals were followed for 5 days post-infection (dpi) to determine viral shedding from oral swabs (3 dpi) and the viral load in the lungs at 5 dpi (Figure 1C).
Figure 1.
The protection provided by a single dose of intranasal recombinant Ad(Spike) attenuates with time in C57BL/6 mice
(A) Diagram of the SARS-CoV-2 Spike protein (ancestral strain) transgene cassette expressed in our recombinant ΔE1/E3 human adenovirus serotype 5, termed Ad(Spike).
(B) Western blot analyses of antigen expression from SF-BMAd-R cells infected with Ad(Spike). Samples were run against a molecular weight ladder; lanes 1 and 2. The negative control contains SF-BMAd-R cells infected with a ΔE1/E3 adenovirus with an empty gene cassette, termed Ad(e); lane 3. Ad(Spike) was amplified in 3 batches of cells and western blots were run on cell lysates from each batch; lanes 4–6, before being combined and purified. This western blot was run with an RBD-specific antibody, thereby capturing the S1 portion.
(C) Study design schematic. At time 0, animals were vaccinated intranasally (i.n.) with 109 mean tissue culture infectious dose (TCID50) of Ad(Spike) in 30 μL. In the case of the sham control, at time 0 animals were vaccinated i.n. with 30 μL PBS. Mice were then challenged with 106 TCID50 SARS-CoV-2 South African strain (B.1.351) at month 2 or month 6 post-vaccination. In both challenge models, animals were followed for 5 days with nasal swab collection on day 3 and euthanasia on day 5.
(D–F) Infectious viral load in mice challenged with the B.1.351 variant of SARS-CoV-2, 2 and 6 months post-immunization with Ad(Spike). Viral load (TCID50) in (D) oral swabs at 3 days post-infection (dpi), and (E) lungs at 5 dpi, quantified by the Spearman–Kärber method. Viral RNA in mouse (F) lungs at 5 dpi. N = 6. Data points represent individual mice, means ± SD are shown. For (D)–(F), Kruskal-Wallis test with Dunn’s multiple comparisons: ∗p < 0.05; ∗∗p < 0.01; ns = not significant. Schematics made with BioRender.com.
As expected, Ad(Spike)-immunized mice displayed a significant reduction in the production of infectious virus in oral swabs (Figure 1D). Notably, animals challenged 6 months post-vaccination, showed lower viral titers compared to unvaccinated mice, although this did not reach statistical significance (Figure 1D). Similarly, infectious viral titers (Figure 1E) and viral RNA (Figure 1F) assessed directly in the lungs of infected animals at 6 months post-vaccination were not statistically different from unvaccinated mice as opposed to data from mice challenged 2 months after immunization, where significant differences were observed.
BCG administration prolongs the protective effect of Ad(Spike) in immunized mice
We next investigated the effectiveness of a prime-boost vaccination regimen using BCG. Female C57BL/6 mice were pre-immunized with 106 colony forming units (CFU) of the BCG-Danish strain containing an empty plasmid (BCG(e)) intraperitoneally (i.p.), 1 month prior to i.n. Ad(Spike) vaccination (month 0; Figure 2A). Although BCG is routinely administered intradermally in humans, a range of administration routes have been applied to the mouse model including i.n., i.p., aerosol, subcutaneous (s.c.) and intravenous (i.v.).19,25,26,27,28,29 For this study, we chose to use the i.p. route for ease of handling and to maximize the systemic distribution of BCG upon a single immunization. In addition, the i.p. route is more immunogenic than the s.c. route and is effective at inducing both systemic and mucosal immunity.25,30 Naive controls received the vehicle (PBS) in place of both BCG(e) and Ad(Spike). First, we tested the possibility that BCG alone could provide non-specific protection against infection or severe disease following SARS-CoV-2 challenge in our animal model, since reports from human data are controversial.31 Animals were pre-immunized with BCG(e) and after one month were vaccinated with an adenoviral vector containing an empty gene cassette (Ad(e)) before challenge with SARS-CoV-2 two months later. Mice pre-immunized with BCG-Danish/Ad(e) displayed no significant reduction in infectious viral titers or viral RNA in oral swabs or lungs (Figure S2) compared to PBS controls, confirming the BCG-Danish vaccine did not provide significant non-specific protection to challenged mice.19,32
Figure 2.
Exposure to BCG prolongs the protective effect of Ad(Spike) in immunized C57BL/6 mice
(A) Study design schematic. Animals were primed intraperitoneally (i.p.) with 106 cfu BCG containing an empty gene cassette (BCG(e)) or PBS at time −1 month. At time 0, animals were vaccinated intranasally (i.n.) with 109 mean tissue culture infectious dose (TCID50) of Ad(Spike) in 30 μL. In the case of the sham control, at time 0 animals were vaccinated i.n. with 30 μL PBS. Mice were then challenged with 106 TCID50 SARS-CoV-2 South African strain (B.1.351) 6 months post-vaccination. Animals were followed for 5 days post challenge, with nasal swabs collected on days 1, 3, and 5 post-infection (dpi) and euthanasia on day 5.
(B–E) Viral load quantified in oral swabs at 1, 3, and 5 dpi, and (D) in lungs at 5 dpi, quantified by TCID50. Viral RNA in (C) oral swabs at 5 dpi and (E) lungs at 5 dpi. N = 4–6. Data points represent individual mice, means ± SD are shown. For (B), two-way ANOVA with Tukey’s multiple comparisons: ∗p < 0.05; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001. For (C)–(E), Kruskal-Wallis test with Dunn’s multiple comparisons: ∗p < 0.05; ∗∗p < 0.01; ns = not significant. Schematic made with BioRender.com.
We then investigated the effect of prior BCG-Danish administration on the duration of our Ad(Spike) vaccine 6 months after vaccination (Figure 2A) by quantifying the daily variation in viral replication and infectious particles in oral swabs and the pulmonary tissue of mice infected with SARS-CoV-2. Here, we found that while a single dose of i.n. Ad(Spike) could reduce infectious viral particles in oral swabs at 1 and 3 dpi, mean viral titers in animals that were first pre-immunized with BCG were significantly lower than those that were not (Figure 2B). At 5 dpi, we also assessed the reduction of viral RNA in oral swabs (Figure 2C), infectious virus in the lungs measured by TCID50 (Figure 2D), and viral RNA in the lungs (Figure 2E). We found that by 6 months post-vaccination, although a single dose of Ad(Spike) was able to reduce viral burden compared to controls, this reduction was not statistically significant. However, in animals that first received a single administration of BCG-Danish, we did observe a statistically significant protective effect after 6 months, highlighting the fact that BCG is able to potentiate the ability of the i.n. Ad(spike) vaccine to control viral replication within both the upper and lower respiratory tract.
Ad(Spike)-vaccinated animals are protected from SARS-CoV-2-induced histological changes 6 months post-vaccination
SARS-CoV-2 infection in C57BL/6 mice causes pulmonary inflammation characterized by immune cell infiltration, lung atelectasis, and bronchial constriction. Since we observed a greater reduction of viral particle load in BCG-Danish exposed mice, we next assessed the extent of pulmonary damage at 6 months post-Ad(Spike) vaccination, both with and without BCG pre-immunization. Scoring of lung histology showed an overall significant reduction in cellular and tissue damage (CTL), circulatory/vascular damage (CVL), and inflammatory patterns (RIP) in all Ad(Spike) vaccinated animals that was not observably enhanced through pre-immunization with BCG(e) (Figures 3A–3C and Table S1). Thus, we could not distinguish any protective effect of BCG-Danish, as both groups displayed a significant reduction in histological changes within the lung compared to the controls. Collectively, these results confirm that while a single i.n. dose of Ad(Spike) vaccine fails to prevent infection, it does protect against severe SARS-CoV-2-induced lung alterations as long as 6 months post-vaccination.
Figure 3.
SARS-CoV-2 induced pulmonary pathology is well prevented in Ad(Spike) immunized mice, regardless of BCG exposure
(A and B) Heatmap and (B) graphical summary of lung pathology (5 days post infection (dpi)) scored in categories: Fibrosis, CTD, CVL, RIP, RR. CTD: Cell/tissue damage which is comprised of bronchoepithelial necrosis (scored 1–3), inflammatory cells/debris in bronchi (1–3), intraepithelial neutrophils (1–3) alveolar emphysema (Yes = 1/No = 0). CVL: Circulatory/vascular lesions comprised of alveolar hemorrhage (Y/N), significant alveolar edema (Y/N), endothelial/vasculitis (1–3). RIP: Reaction/inflammatory patterns comprised of necrosis/suppurative bronchitis (Y/N), intra-alveolar macrophages (Y/N), mononuclear inflammation around airways (Y/N), neutrophilic/heterophilic inflammation (1–3), mesothelial reaction (1–3). RR: Regeneration/repair, includes alveolar epithelial cell regeneration/proliferation (1–3) and bronchiolar epithelial cells regeneration/proliferation (1–3).
(C) Lungs of vaccinated animals, infected after 6 months, were harvested at 5 dpi and stained with Masson’s trichrome for fibrosis (top row) and H&E staining for pathology scoring (bottom row). Row one is imaged at 100× magnification (scale bar, 100 μm). Row two is imaged at 200× magnification (scale bar, 100 μm) showing airway mononuclear inflammation in control animals. Each image is representative for the group. N = 4–6. Data points represent individual mice, means ± SD are shown. For (B), two-way ANOVA with Tukey’s multiple comparisons: ∗p < 0.05.
Ad(Spike)-induced antibody profiles are not influenced by pre-immunization with BCG
Vaccine protection against SARS-CoV-2 infection is largely attributed to its ability to generate high quantities of neutralizing antibodies, although the potential role of BCG in promoting antibody production remains to be assessed. To determine if BCG influenced the quantity and affinity of the antibody response generated by Ad(Spike), we bled immunized animals at select timepoints pre- and post-vaccination. Antibody titers against the full Spike protein (Figure 4) and the receptor binding domain (RBD) (Figure S3) were assessed via ELISA. As expected, animals in the PBS and BCG(e)+Ad(e) groups did not produce Spike or RBD-specific serum antibodies throughout the study. Spike-specific IgG was detectable at high levels 1 month after vaccination and continued to rise until 2 months after vaccination when levels slowly declined until the end of the study for both Ad(Spike) and BCG(e)+Ad(Spike) groups (Figure 4A). Although RBD-specific IgG was significantly higher in Ad(Spike) animals than those that received BCG(e)+Ad(Spike) at 1 month post-vaccination, this difference was not observed at later time points (Figure S3A). As such, Spike-specific IgG was similar between vaccinated groups.
Figure 4.
Exposure to BCG does not significantly influence the quantity or quality of Ad(Spike) generated antibodies
Spike-specific antibodies in the (A–G) serum and (H–M) bronchoalveolar lavage fluid (BALF).
(A) IgG titers in mouse sera throughout the study schedule determined by ELISA.
(B–D) IgG1 and (C) IgG2c at 6 months post-vaccination. The ratio of Spike-specific IgG1/IgG2c at 6 months post-vaccination is given in (D).
(E) IgG avidity index at −1, 0, 1, 2, and 6 months post-vaccination.
(F) cPass determined antibody neutralization activity in serum at 6 months post-vaccination.
(G) IgA titers in mouse sera calculated at 0, 3, and 6 months post-vaccination. N = 7–10.
(H–K) Spike-specific IgG, (I) IgG1, and (J) IgG2c with the ratio of IgG1/IgG2c given in (K).
(L) cPass determined antibody neutralization activity in BALF at 6 months post-vaccination.
(M) IgA in BALF at 6 months post-vaccination calculated by ELISA. N = 7. Data points represent individual mice, means ± SD are shown. The included legend applies to both serum and BALF data. For (A), (G), two-way ANOVA with Tukey’s multiple comparisons: ∗∗p < 0.01; ∗∗∗∗p < 0.0001. For (B), (F), (K), one-way ANOVA with Tukey’s multiple comparisons: ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns = not significant. For (C), (D), (H)–(J), (L), (M), Kruskal-Wallis test with Dunn’s multiple comparisons: ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001.
Since BCG is known to induce IFNγ,33 a promoter of IgG2c production,34 we then addressed if BCG influenced the isotype of antibodies produced upon Ad(Spike) vaccination in the animals prior to challenge. Both groups of vaccinated animals similarly expressed Spike-specific IgG1 (Figure 4B) and IgG2c (Figure 4C) and displayed the same ratio of IgG1/IgG2c (Figure 4D). Correspondingly, both groups of vaccinated animals expressed similar levels of RBD-specific IgG1 (Figure S3D) and IgG2c (Figure S3E) in similar ratios (Figure S3F), confirming that BCG-Danish pre-immunization did not influence isotype-switching upon Ad(Spike) vaccination.
We then assessed IgG avidity by ELISA. As expected, Spike- and RBD-specific IgG avidity rose from 0 to 2 months (Figures 4E and S3B, respectively) and Spike-specific avidity was maintained in both vaccinated groups until 6 months, demonstrating that BCG did not influence the avidity of IgG. Interestingly, RBD-specific IgG avidity dropped at 6 months to levels similar to those observed at one-month post-vaccination (Figure S3C), suggesting that neutralization of the ACE2-Spike binding domain declines with time in both groups. In addition, a cPass surrogate virus neutralization assay confirmed that BCG did not influence the abundance of neutralizing antibodies within immunized mice (Figure 4F) compared to mice vaccinated with Ad(Spike) alone. Intranasal vaccination with Ad(Spike) also resulted in detectable levels of antigen-specific serum IgA in both groups (Figure 4G), prompting us to investigate levels of mucosal antibodies. Bronchoalveolar lavage fluid (BALF)-derived Spike-specific IgG antibodies were present and statistically greater than negative controls at 2 and 6 months post-vaccination (Figure 4H). This was observed also for RBD-specific IgG responses in BALF prior to infection (Figure S3H). Specifically, Spike-specific IgG1 (Figure 4I) and IgG2c (Figure 4J), as well as their ratio (Figure 4K), did not differ between the two vaccinated groups. The same pattern was observed for RBD-specific IgG1 (Figure S3I), IgG2c (Figure S3J), and the ratio of the two (Figure S3K). The mean neutralizing activity of BCG-pre-immunized and vaccinated animals was 2-fold greater than those animals that were solely Ad(Spike) vaccinated, though this difference was not significant (Figure 4L). This trend of higher antibody levels in the BALF from pre-immunized and vaccinated animals was again seen with the Spike-specific IgA (Figure 4M) and RBD-specific IgA (Figure S3L) titers; though, again the difference was not statistically significant.
Intranasal vaccination should also induce mucosal immunity in upper branches of the respiratory tract, so we evaluated both IgG and IgA responses directed against Spike and RBD proteins in mouse turbinate (Figure S4). At 6 months post-vaccination, both experimental groups showed Spike and RBD specific immunoglobulins, whereas the PBS and BCG(e)+Ad(e) groups remained at baseline. Interestingly, Ad(Spike) promoted IgA production in mouse nares, whereas IgG was found in animals which were pre-immunized with BCG(e).
Taken together, these data show that pre-immunization with BCG-Danish does not promote Ad(Spike)-induced protection by modulating the generation, quantity, or quality of circulating or mucosal humoral responses.
BCG pre-immunization promotes long-term cellular immunity in the lungs
An important aspect of vaccination is to generate robust and lasting tissue-resident memory T cells (TRM) in order to confer protection.35 Since we observed that BCG-Danish did not impact the long-term protective antibody response that Ad(Spike) generated against SARS-CoV-2, we next investigated if BCG-Danish pre-immunization potentiated the generation of memory CD4+ and CD8+ TRM cells in vaccinated mice. Six months post-vaccination, isolated lung T cells were activated with SARS-CoV-2 Spike protein peptides (Figures 5A and S5). As shown in Figure 5B, at 6 months post-vaccination, the frequencies of lung CD8+ T cells from immunized groups were significantly greater than controls regardless of BCG administration. However, when we assessed the production of Granzyme B (GrB) and IFNγ in activated CD69+ CD8+ TRM cells, we observed that mice that received BCG(e) prior to immunization with Ad(Spike) produced a higher frequency of GrB- (Figure 5C) and IFNγ-secreting CD8+ T cells (Figure 5D) compared to the Ad(Spike)-alone group 6 months post-vaccination, suggesting that their responses are more cytotoxic in nature. In parallel, we observed an increase in activation (CD69+) of CD4+ T cells upon peptide restimulation in the immunized group that was previously exposed to BCG (Figure 5E). In fact, mice pre-immunized with BCG, regardless of Ad(Spike) vaccination, had increased frequencies of IFNγ+ CD4+ (Figure 5F) and IL17A+ CD4+ T cells in the lung (Figure 5G), although these trends did not reach statistical significance. Coincidently, we also observed significantly less FoxP3+ regulatory T (TREG) cells among CD69+ CD4+ T cells (Figure 5H), suggesting that BCG promotes the generation of cytotoxic over suppressive T cell responses in BCG(e)+Ad(Spike)-immunized mice relative to the other groups. Collectively, these results demonstrate that BCG pre-immunization acts on the long-term potency of the Ad(Spike) vaccine by promoting the generation of cytotoxic and Th1 responses over suppressive FoxP3+ TREG cells in the lungs of infected mice.
Figure 5.
BCG potentiates the generation of long-lasting cellular immunity
Six months post-vaccination, cell mediated responses from lung cells were analyzed following 18 h restimulation with an S-protein peptide pool.
(A) Expression of CD69 and GrB in vaccinated animals compared to controls (Ad-S = Ad(Spike)).
(B–D) Total frequency of CD69+CD8+ T cells. The frequency of CD69+CD8+ T cells which are (C) GrB+ and (D) IFNɣ+.
(E) Frequency of CD69+ cells among the CD4+ population.
(F–H) IFNɣ, (G) IL17a, and (H) FoxP3 expression was also determined from CD69+CD4+ T cells. N = 7. Data points represent individual mice, means ± SD are shown. For (B)–(H), one-way ANOVA with Tukey’s multiple comparisons: ∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001; ∗∗∗∗p < 0.0001; ns = not significant.
Ad(Spike) cross-reactive antibodies persist and are not affected by BCG pre-immunization
Next, we assessed if the administration of BCG affected the ability of Spike antibodies generated by an i.n. Ad(Spike) vaccine based on the ancestral (Wuhan) sequence to subsequently recognize epitopes from SARS-CoV-2 alpha (B.1.1.7), beta (B.1.351), gamma (P.1), delta (B.1.617.2), and omicron variants (B.1.1.529, and BA.2). Six months post-vaccination, serum IgG titers against the ancestral S-protein within Ad(Spike)- and BCG(e)+Ad(Spike)-vaccinated animals were not significantly different between the two vaccinated groups (Figure 6A). To ensure BCG pre-immunization does not affect the cross-reactivity of these antibodies, we conducted ELISAs against S-proteins from variant strains of SARS-CoV-2. We found no significant reduction in the antibody binding capacity of serum from vaccinated animals that were vaccinated with Ad(Spike) (Figure 6B) or pre-immunized with BCG and then vaccinated with Ad(Spike) (Figure 6C) against any of the SARS-CoV-2 strains. These results confirm that BCG administration does not affect the generation of cross-reactive anti-Spike antibodies.
Figure 6.
Production of cross-reactive antibodies to variant spike proteins is not altered by BCG
Spike-specific IgG was calculated from serum from vaccinated animals at 6 months post-vaccination by ELISA.
(A–C) Antibodies are shown against Spike Wuhan (Ancestral) strain. Cross-reactive antibodies from (B) Ad(Spike) vaccinated and (C) BCG pre-immunized and Ad(Spike) vaccinated animals were then assessed and compared to the ancestral strain. Antibodies binding strains B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma), B.1.617.2 (delta), B.1.1.529 (omicron), and BA.2 (omicron) were assessed. N = 7. Data points represent individual mice, means ± SD are shown. For (A), (B), Kruskal-Wallis test with Dunn’s multiple comparisons: ∗p < 0.05; ns = not significant. For (C), one-way ANOVA with Tukey’s multiple comparisons: ∗p < 0.05; ns = not significant.
Discussion
A single intranasal dose of a human adenovirus vectored vaccine has previously been shown to be sufficient to protect mice from SARS-CoV-2 infection and severe disease.36 However, the durability of protection from these vaccines remains to be established as most studies utilize short-term challenge models. Using an AdV vectored vaccine that expresses the Spike protein of the Wuhan isolate, and then challenging with the mouse-permissive Beta variant of SARS-CoV-2, we demonstrated that a single i.n. dose of a human AdV-based vaccine could cross-protect mice from severe disease months after immunization. Specifically, our Ad(Spike) vaccine successfully prevented mice from developing severe pulmonary histological changes when challenged with SARS-CoV-2 at 2 and 6 months post-vaccination. However, the ability of vaccinated mice to limit initial infection was reduced by 6 months post-immunization, confirming what was observed in a recent meta-analysis.6 Thus, we aimed to develop a strategy to prolong the immunity provided by i.n. administration of Ad(Spike) by harnessing the non-specific immune-enhancing effects of the widely available BCG vaccine.37,38 As hypothesized, herein we demonstrate that BCG improves the long-term protection and reduced viral shedding conferred by a single i.n. Ad(Spike) dose by potentiating cellular, rather than humoral, immunity against the Spike antigen in the lungs. Although our initial data to date demonstrates BCG-induced immune-enhancement for up to 6 months, future work will extend the interval of time between vaccination and challenge. In addition, we plan to test additional BCG administration routes (e.g.,: s.c.), as well as varied i.n. Ad(Spike) vaccination doses. The latter is important due to potential safety concerns over the use of AdV-vectored vaccines, and we anticipate that our combination vaccine strategy may enable us to reduce the effective vaccine dose (or dosing frequency) required to maintain a protective effect. Overall, our current research findings may also lay the groundwork for the use of BCG as a more general vaccine enhancement strategy that is applicable to a broad range of viral infections and vaccination modalities, including those based on lipid nanoparticle encapsulated messenger RNA.
As SARS-CoV-2 achieves endemicity and continues to evolve, novel vaccines and immunization routes that increase mucosal immunity8,39 are needed. Indeed, several groups have demonstrated that when adenovirus vectored SARS-CoV-2 vaccines are administered intranasally, a single immunization is sufficient to confer protection from infection in naive animals.40,41 One promising option, investigated herein, is based on the ability of the BCG vaccine to act as an indirect promoter of cellular immunity. For many years, BCG has been approved for use in infants across the globe to protect against severe, disseminated forms of tuberculosis. As such, BCG is commonly reported as the world’s most widely used vaccine and has an excellent safety record. Moreover, BCG has also recently been shown to reduce all-cause mortality from unrelated infectious agents, including several viruses, as a result of its non-specific or “off-target”, immune-enhancing effects.37,42 Consistent with these reports, we demonstrate that pre-immunization with BCG can non-specifically prevent waning immunity from a single-dose, i.n. SARS-CoV-2 vaccine by potentiating local vaccine-specific cell mediated responses without hindering humoral immunity.
The possible contribution of widespread BCG vaccination to SARS-CoV-2 protection in human populations is still unclear; yet, there is growing evidence that BCG vaccination provides ‘trained immunity’ to innate mechanisms,37,42,43 which can have antagonistic effects on other unrelated pathogens.19,38,44,45 This is seen in the curious protection from neonatal sepsis and respiratory infections conferred to BCG immunized babies.15 It has since been demonstrated that due to intrinsic pathogen associated molecular pattern activation of toll like receptors, BCG induces the activation and reprogramming of monocytes,33,37,42 resulting in increased expression of cell surface markers and the production of pro-inflammatory cytokines and IFNɣ in response to antigenic stimulation.46 These data contributed to the hypothesis that BCG may provide heterologous potentiation of antigen-presenting cell (APC) function to improve the immunogenicity and efficacy of vaccines, as made evident in human studies of neonatal vaccinations47,48 and adult influenza vaccination.49 In fact, BCG is a natural adjuvant, at least partly a result of its modified peptidoglycan structure and unusual cell-wall lipid composition,50 and has been exploited in several novel vaccine efforts including against SARS-CoV-2.32,51,52 It is particularly impressive that BCG has the potential to deliver benefit to vaccination strategies even when not directly co-administered, as shown herein. Nevertheless, BCG alone does not seem to provide direct protection from SARS-CoV-2. While an early study in humans suggested protective efficacy against SARS-CoV-2 from BCG vaccination,53 a recent case study demonstrated that this protection was not attributable to BCG over the course of the pandemic due to an underreporting of COVID-19 cases in regions with high BCG-vaccination rates.31,54 More recently, a randomized trial involving more than 3,000 healthcare workers did not show a reduced risk of severe COVID-19 amongst those who received BCG compared to the placebo group.21 Another group found that BCG alone failed to provide protection from SARS-CoV-2 in mice and hamsters,19 although this may depend upon the timing and route of administration.55 Nonetheless, our data support the idea that neither BCG alone (nor when combined with an empty AdV vaccine vector) confers significant protection against SARS-CoV-2 challenge in a mouse model of infection.
As noted above, data show that despite short-term efficacy, protection from SARS-CoV-2 infection wanes over time in humans.56,57 Consistent with this, we also observed an attenuation of protection by 6 months post-immunization. Although prior exposure to BCG did not change Ad(Spike) protection 2 months post-vaccination (Figure S2), when mice were primed with BCG and then vaccinated with Ad(Spike), significant protection was maintained for at least 6 months post-vaccination. Due to the numerous possible effects of BCG on the innate and adaptive immune responses, we chose to focus on its impact on the type of humoral immunity generated by a single-dose Ad(Spike) vaccine by first assessing the quality and quantity of serum and BALF antibodies. We observed that regardless of their exposure to BCG, mice that were vaccinated with Ad(Spike) displayed robust serum and BALF Spike-specific antibodies, both IgG and IgA which, while slowly declining, were maintained at high titers to at least 6 months post-vaccination. These antibodies maintained high affinity to the receptor binding domain of the Spike protein, confirming that they were capable of neutralizing viral entry. Reassuringly, these antibodies also displayed cross-reactivity against all Spike variants tested, including the beta variant, which was used in our challenge study. However, despite these seemingly high titers of neutralizing IgG1/2c and IgA antibodies, viral RNA and, to a lesser degree, live viral particles remained high in the absence of BCG. Thus, we hypothesized that the BCG bacilli were promoting cellular rather than humoral immunity in the BCG(e)+Ad(Spike)-immunized mice.
Our antigen-specific assay demonstrated that although Ad(Spike) alone showed a trend toward increased expression of cytotoxic activity in antigen-primed T cells, these differences became significant only in mice that were pre-immunized with BCG. Indeed, BCG has been shown to enhance intramuscular vaccine-induced circulating Spike-specific CD4+ T cells in humans,22 and our results reveal that it can also potentiate the generation of antigen-specific CD4+ T cells in tissues. Many reports suggest BCG acts by training monocytes to have higher expression of MHC-II, CD80, and CD86, thereby enhancing antigen presentation to T cells.58,59 Concomitantly, adaptive immune responses after BCG vaccination also involves the activation of CD8+ T cells.38 Here, we observed a clear potentiation of Spike-specific cytotoxic CD8+ T cells up to at least 6 months after vaccination. Furthermore, cytokines secreted by BCG-exposed monocytes, such as IL1β and IL6, are key contributors to CD4+ T cell differentiation into Th1 and Th17 subsets60,61 which is consistent with the increased CD4+ T cell expression of IFNɣ or IL17a we observed in BCG-immunized mice even in the absence of Ad(Spike). Thus, it is likely that our assay could not distinguish Spike-specific from bystander Th1 and Th17 cells generated prior to Ad(Spike) administration.
Our study used wild-type C57BL/6 mice, which necessitated using a SARS-CoV-2 strain that binds the mouse ACE-2 (mACE-2) receptor. For this reason, we challenged the C57BL/6 mice with the B.1.351 strain, capable of binding mACE-2 and establishing infection.62 Thus, we demonstrated cross-protection delivered by our ancestral strain-based AdV vaccine in a context of SARS-CoV-2 infection due to a heterologous strain, which resembles the situation with human disease.63 Within this study, we offer insight into the effects of prior BCG immunization on reinforcing vaccine efficacy in a mouse model of SARS-CoV-2. Interestingly, due to the long-term persistence of viable BCG in our experimental design (Figure S6), this study also raises the issue of vaccine efficacy in the context of other persistent bacterial co-infections, as well as components of the normal mucosal microbiota, and represents an important future line of inquiry. As preclinical vaccine testing is conducted in naive animals, the role of immunological memory from previous vaccination and persisting infections should be addressed in relation to vaccine efficacy.
Collectively, our results present a vaccination approach that can potentially curb viral pandemics by potentiating the long-term efficacy of a next-generation adenovirus-vectored mucosal vaccine when provided in the context of the safe and widely distributed BCG vaccine. This approach can also have an impact in other mucosal and non-mucosal vaccination strategies as well. Going forward, these strategies may also provide a viable solution to ensure a more rapid and equitable distribution of vaccines among developing nations where BCG is already firmly entrenched within many vaccination programs.
Limitations of the study
Although the C57BL/6 mouse model reflects age- and sex-based differences in human COVID-19 disease,63 it is not the ideal model for SARS-CoV-2 vaccine studies since these mice do not display all the hallmark features of severe pathology and typically recover from infection.64 As follows, the lung changes observed in our model were modest, posing a limitation in our ability to discriminate severe disease between vaccinated groups. Furthermore, although lung-cell memory responses were increased in BCG pre-immunized animals, our long-term infection model of 6 months post-vaccination may have been insufficient to clearly assess the synergistic effects of BCG on Ad(Spike). We propose that the roles these memory responses play may become more obvious in longer-term studies when protection from Ad(Spike) alone is further reduced.
In humans, BCG is most typically administered intradermally; however, as noted above, we chose to immunize animals intraperitoneally to maximize systemic responses. An immunization route closer to that used in humans is the s.c. route and this will be explored in future experiments. In addition, our group utilized a human adenovirus serotype 5 (HAdV5), a commonly used vaccine vector due to its capability of inducing strong immune responses, as well as being able to be produced in large quantities. However, some drawbacks include that pre-existing immunity to HAdV5 is common in many developing countries65 and that there are potential risks associated with the use of adenovirus vectors (e.g., Guillain-Barre syndrome (GBS)) in some individuals. Although antigen-specific T cell responses have been demonstrated in individuals with pre-existing HAdV5 immunity,66 to circumvent this potential challenge, a rarer adenoviral vector could be used, for instance, serotype 26 which showed efficacy in Janssen’s COVID-19 vaccine when deployed in South Africa.67 Interestingly, an increase in GBS was not found when using COVID-19 vaccines based on HAdV5, unlike other vaccination platforms.68 Despite this, considering the extreme infrequency of vaccine-related serious adverse events, most consider the increased risk of GBS miniscule in comparison to the protection against severe and lethal COVID-19 offered by vaccination.69 Using a lower dose of viral-vectored vaccines may help to reduce the risk of adverse events even further and may point to an additional benefit of employing BCG as a means of immune enhancement whereby the combination with BCG may allow for a reduced dosing schedule or a reduction in the viral titer required to achieve effective vaccination.
STAR★Methods
Key resources table
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
Rabbit anti-SARS-COV2 SARS-COV/SARS-COV-2 Spike RBD Polyclonal Antibody(2019-nCoV) Polyclonal Antibody | MyBioSource, San Diego, CA, USA | Cat: MBS2563840 |
Anti-mouse IgG HRP | Sigma Aldrich | Cat:A0168; RRID: AB_257867 |
HRP-conjugated anti-mouse IgA | Sigma Aldrich | Cat:A4789; RRID: AB_258201 |
Goat anti-mouse IgG1-HRP | SouthernBiotech | RRID:AB_2794426 Cat:1071-05 |
Goat anti-mouse IgG2c-HRP | SouthernBiotech | RRID:AB_2794462 Cat:1078-05 |
Fixable viability dye eFluor 780 | Thermofisher | Cat:65-0865-14 |
FOXP3 Monoclonal Antibody (FJK-16s), PE-Cyanine7, eBioscience™ | Thermofisher | RRID: AB_891552 |
BUV395 Hamster Anti-Mouse CD3e | BD Biosciences | RRID: AB_2738278 Cat: 563565 |
BV510 Rat Anti-Mouse CD8b | BD Biosciences | RRID: AB_2739908 Cat: 740155 |
BUV737 Rat Anti-Mouse IFN-γ | BD Biosciences | RRID: AB_2870098 Cat: 612769 |
Alexa Fluor® 700 anti-mouse CD4 Antibody | BioLegend | RRID: AB_493701 Cat: 100536 |
APC anti-mouse IL-17A Antibody | BioLegend | RRID: AB_536018 Cat: 506916 |
Brilliant Violet 421™ anti-mouse IL-4 Antibody | BioLegend | RRID: AB_2562594 Cat: 504127 |
FITC anti-mouse CD69 Antibody | BioLegend | RRID: AB_313108 Cat: 104505 |
PerCP/Cyanine5.5 anti-mouse TNF-α Antibody | BioLegend | RRID: AB_961434 Cat: 506322 |
PE anti-human/mouse Granzyme B Recombinant Antibody | BioLegend | RRID: AB_2687032 Cat: 372208 |
Rockland Immunochemicals Rabbit TrueBlot: Anti-Rabbit IgG HRP - 18-8816-31 | Rockland Immunochemicals, Pottstown, PA, USA | Cat: 18-8816-31; RRID: AB_2610847 |
Bacterial and virus strains | ||
non-replicating Adenovirus (ΔE1, ΔE3; 1st generation) containing no gene cassette | National Research Council of Canada, Montreal, QC, Canada | N/A |
BCG Danish | ATCC | Cat: 35733 |
Chemicals, peptides, and recombinant proteins | ||
SuperSignal West Pico Plus Chemiluminescent Substrate | Thermofisher | Cat:34580 |
TMB Substrate | Sigma Aldrich | Cat:T0440 |
GolgiPlug | BD Science, San Jose, CA, USA | RRID:AB_2869014 Cat: 555029 |
phorbol 12-myristate 13-acetate | Thermofisher | Cat:356150010 |
Ionomycin | Thermofisher | Cat: J62448.MCR |
Fc block | BD Science | RRID:AB_394656 |
Fixation Buffer | BD Science | RRID: AB_2869005 Cat: 554655 |
Permeabilization buffer | BD Science | RRID: AB_2869011 Cat: 554723 |
PepTivator SARS-CoV-2 Prot_S | Miltenyi Biotec, Bergisch Gladbach, North Rhine-Westphalia, Germany | Cat: 130-126-700 |
PepTivator SARS-CoV-2 Prot_S Complete | Miltenyi Biotec | Cat: 130-127-951 |
SARS-CoV-2 Neutralization Antibody Standard | GenScript | Cat: A02087 |
SARS-CoV-2 Spike (Wuhan) | National Research Council of Canada, Montreal, QC, Canada | N/A |
SARS-CoV-2 RBD (Wuhan) | National Research Council of Canada, Montreal, QC, Canada | N/A |
SARS-CoV-2 Spike (B.1.1.7) | National Research Council of Canada, Montreal, QC, Canada | N/A |
SARS-CoV-2 Spike (B.1.351) | National Research Council of Canada, Montreal, QC, Canada | N/A |
SARS-CoV-2 Spike (P.1) | National Research Council of Canada, Montreal, QC, Canada | N/A |
SARS-CoV-2 Spike (B.1.617.2) | National Research Council of Canada, Montreal, QC, Canada | N/A |
SARS-CoV-2 Spike (B.1.1.529) | National Research Council of Canada, Montreal, QC, Canada | N/A |
SARS-CoV-2 Spike (BA.2) | BEI Resources, Manassas, VA, USA | Cat: NR-56517 |
Critical commercial assays | ||
Q-plex Mouse Cytokine – Screen (16-plex) | Quansys Biosciences, West Logan, UT, USA | Cat:110949MS |
SARS-CoV-2 Surrogate Virus Neutralization Test (sVNT) |
GenScript | Cat:L00847-A |
Deposited data | ||
Available upon request | N/A | N/A |
Experimental models: Cell lines | ||
AdEasier-1 cells | Addgene, Watertown, MA, USA | GenBank ID:AY370909, Addgene plasmid #16399 |
One Shot Top10 Chemically Competent E. coli | ThermoFisher Scientific, Waltham, MA, USA | Cat:C404010 |
HEK293A cells | American Type Culture Collection (ATCC), Manassas, VA, USA | RRID:CVCL_6910 |
SF-BMAd-R cells | National Research Council of Canada, Montreal, QC, Canada | N/A |
Experimental models: Organisms/strains | ||
C57BL/6NCrl mice | Charles River Laboratories, Senneville, QC, Canada | RRID:IMSR_CRL:027 |
Recombinant DNA | ||
AdSpike construct | Integrated DNA Technologies (Coralville, IA, USA) | N/A |
Software and algorithms | ||
FlowJo software (version 10.0.8r1) | Treestar, Ashland, OR, USA | N/A |
GraphPad Prism 9 software | (La Jolla, CA, USA) | N/A |
Resource availability
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Momar Ndao (momar.ndao@mcgill.ca).
Materials availability
This study did not generate new unique reagents.
Experimental model and study participant details
Mice
Six- to eight-week-old female C57BL/6 mice were ordered from Charles River Laboratories (RRID:IMSR_CRL:027) (Senneville, QC, Canada). Mouse housing, husbandry, and environmental enrichment can be found within the McGill standard operating procedures (SOP) #502, #508, and #509.
Animal ethics
All animal procedures were performed in accordance with the Institutional Animal Care and Use Guidelines approved by the Animal Care and Use Committee at McGill University (Animal Use Protocol 8190). Animals were monitored for adverse events for 3 days post-vaccination and weekly until the end of each experiment. Humane intervention points were monitored according to McGill SOP #410. Challenge experiments were performed in compliance with the Canadian Council on Animal Care guidelines and approved by the Animal Care Ethics Committee at the University of Toronto (APR-00005433-v0002-0). All animals were humanely sacrificed at endpoint by anaesthesia with isoflurane before euthanasia by carbon dioxide asphyxiation, followed by pneumothorax and blood collection by cardiac puncture.
Cell lines and reagents
Cell lines were obtained from commercial sources, passed quality control procedures, and were certified and validated by the manufacturer. SF-BMAd-R cells were validated for identity, as human derived.70 All reagents were validated by the manufacturer or has been cited previously in the literature. When available, RRID tags have been listed in the text and in the reagent repository (Table S2).
Mycobacterium bovis BCG Danish culture
Mycobacterium tuberculosis variant bovis BCG (ATCC 35733), provided to us by Dr. Marcel Behr (Research Institute of the McGill University Health Centre), was grown in Middlebrook 7H9 broth (BD, Mississauga, ON, Canada) supplemented with 10% ADC (8.1 g/l NaCl, 50 g/l BSA Fraction V (Millipore Sigma, Billerica, MA, USA), 20 g/l glucose), 0.2% glycerol and 0.05% Tween 80, or on Middlebrook 7H11 agar (BD) supplemented with 10% OADC enrichment (as per ADC plus 0.6 ml/l oleic acid, 3.6mM NaOH). The BCG-Danish strain used in these experiments was transformed with an empty pMV361(hygromycinR) plasmid and was initially selected in the presence of 50 μg/ml hygromycin (Wisent, Saint-Jean-Baptiste, QC, Canada). This plasmid was integrated into the non-essential L5 phage attachment site (attB) located within the BCG chromosome. We termed this strain BCG(e).
Method details
Author checklists
This study has been submitted with both the ARRIVE and MDAR author checklists.
Preparation of M. bovis BCG-Danish cultures for mouse immunization
100 ml BCG cultures were grown in 7H9/ADC medium to an OD600nm = 0.6. After spinning at 3000 rpm, cells were washed twice with PBS containing 0.05% Tween-80 (PBS-Tw) and resuspended in 6 ml of this buffer. After passing 10 times through a 22G x1″ and 10 times through a 27G x1/2″ needle, the suspension was mixed with 4 ml of sterile 50% glycerol in PBS. Aliquots were made and frozen at −80°C. Prior to immunization, an aliquot was thawed, and 10-fold serial dilutions were plated on 7H11/OADC+ HYG for quantification, yielding a value of ∼1.5 x108 cfu/ml. On the day of the immunization, these glycerol stocks were diluted (1/20) in PBS-Tw and 200 ul (containing ∼1.5 × 106 cfu) were injected i.p. into the lower right quadrant of the abdomen of the mice using a 28Gx1/2″ needle and an insulin syringe. Inocula were quantified by plating 10-fold serial dilutions on 7H11/OADC+ HYG.
Generation of Ad(Spike) vector
The AdSpike construct was developed following a similar protocol as described.71 Briefly, the Spike gene cassette combined a Kozak sequence with the full length of the Spike protein (Genbank accession number QHU36824.1), codon optimized to mouse and human expression avoiding restriction sites Bgl2, Pac1, and Pme1, followed by a Kpn1 restriction site and the poly-A signal “TCTAGACTCGACCTCTGGCTAATAAAGGAAATTTATTTTCATTGCAATAGTGTGTTGGAATTTTTTGTGTCTCTCACTCGGAAGGACATATGGGAGGGCAAATCATTTGCGGCCGCGATATC” (GenScript, Piscataway, NJ, USA). The gene cassette was flanked by Bgl2 sites and synthesized by Integrated DNA Technologies (Coralville, IA, USA) then cloned into the vector, pShuttle-CMV-Cuo.72 Primers to confirm gene sequence can be found in Table S3. The plasmid containing our recombinant non-replicating human adenovirus serotype 5 (E1 and E3 genes removed (ΔE1-, ΔE3-); 1st generation) encoding the S-protein gene was made through homologous recombination in AdEasier-1 cells (strain), a gift from Dr. Bert Vogelstein (Addgene plasmid #16399) (Addgene, Watertown, MA, USA).73 It was then linearized with PacI and transfected into HEK293A cells (RRID:CVCL_6910). Our recombinant adenovirus was then amplified using SF-BMAd-R cells70 in 3 batches (Ad(Spike) 1–3), combined, and purified by ultracentrifugation on CsCl gradients as described previously,74 before titration using a TCID50 assay. A second human adenovirus serotype 5 (ΔE1-, ΔE3-; 1st generation), lacking a gene cassette, was used as a negative control.
Western blot assays
To determine protein expression by Ad(Spike), cell lysates of Ad(Spike) infected HEK293A cells were assessed. Briefly, cells were infected at a multiplicity of infection of 5 particles per cell and incubated for 48–72 hours, pelleted, and then lysed (0.1M Tris, 10 μL EGTA, 50 μL Triton-100, 0.1M NaCl, 1mM EDTA, 25 μL 10% NaDeoxycholate, 1X protease inhibitor, in ddH2O). Cell lysates were then resolved on an SDS-PAGE gel under reducing conditions followed by transfer onto a nitrocellulose membrane. The membrane was subsequently blocked in phosphate buffered saline (PBS) with 0.05% Tween 20 (PBS-T) and 5% milk (Smucker Foods of Canada Corp, Markham, ON, Canada) (PBS-TM). The membrane was then incubated with rabbit anti-SARS-CoV-2 Covid-19 Spike RBD coronavirus polyclonal antibody (RRID:AB_258251) diluted 1:5,000 in PBS-TM overnight at 4°C. The membrane was then washed in PBS-T before incubation with horseradish peroxidase (HRP)-conjugated anti-rabbit IgG (IgG-HRP) (Rockland Immunochemicals, Pottstown, PA, USA) diluted 1:10,000 in PBS-T for one hour at room temperature. After incubation the membrane was washed again and developed using SuperSignal West Pico Plus Chemiluminescent Substrate (ThermoFisher Scientific, Waltham, MA, USA) (Figure S7).
Protein expression and purification
SARS-CoV-2 Spike variants Wuhan, B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma), B.1.617.2 (delta), B.1.1.529 (omicron), and the RBD portion of the Wuhan Spike variant were obtained from the National Research Council of Canada. Recombinant, “tagless” Spike proteins were produced as previously described.75 Recombinant RBD protein was produced as previously described.76,77 Spike variant BA.2 (omicron) was obtained through BEI Resources, NIAID, NIH: Spike Glycoprotein (Stabilized) from SARS-Related Coronavirus 2, BA.2 Lineage (Omicron Variant) with C-Terminal Histidine and Avi Tags, Recombinant from HEK293 Cells, NR-56517.
Immunization and challenge protocol in mice
Each mouse was immunized at weeks 0 and 4 by intraperitoneal (i.p.) injection of 200μL of BCG(e) and i.n. administration of 30μL of adenovirus formulations, respectively. See Table S4 for more precise group descriptions. Group 2 was removed from the long-term challenge experiment since negative control animals displayed similar viremia to the PBS control in the short-term challenge model. Mice were bled from the saphenous vein at weeks 0, 4, 8, 12, and 18. Mice immunized for humoral and cell-mediated immunity assessment (n = 6) were euthanized 6 months after the final immunization and blood, spleens, and lungs were collected. Mice immunized for challenge studies (n = 12) were transferred to the University of Toronto and challenged with 106 TCID50 of SARS-CoV-2 South African strain (B.1.351) 2 (n = 6) or 6 months (n = 6) post-Ad(Spike)-vaccination. TCID50 was determined using the Spearman–Kärber method.78 Oral swabs were taken from mice on days 1, 3, and 5 post-challenge in DMEM. Mice were euthanized 5 days after challenge and lungs were collected.
Quantification of viral load
Quantities of infectious virus was determined by determining the median tissue-culture infectivity dose (TCID50) using methods that have been described previously.79 Briefly, Vero E6 cells were seeded into plates and incubated overnight at 37°C. On the following day, media was removed, and samples were added and serially diluted using ten-fold dilutions. Plates were incubated at 37°C for 1 h. After incubation, the media was removed and replaced with complete DMEM, and plates were incubated at 37°C for 5 days. Cells were examined for cytopathic effect (CPE) at 5 dpi. TCID50 was defined using the Spearman–Kärber method.78
qRT-PCR
Viral RNA loads were calculated as previously described.79 Briefly, viral RNA was extracted using the QIAamp viral RNA kit (Qiagen, Hilden, Germany) according to the manufacturer’s guidelines. SARS-CoV-2 viral RNA detection and quantification was performed using the Luna Universal Probe One-Step RT-qPCR kit (New England Biolabs, Whitby, ON, Canada) on the Rotor-gene Q platform (Qiagen). For quantification, standard curves were generated using a synthetic plasmid containing a segment of the E-gene (GenScript) and interpolation was performed as described by Feld et al.80 The limit of quantification was determined to be 20 copies/mL.
Spike and RBD-specific IgG, IgG1, IgG2c, IgA quantification and IgG avidity assays
Briefly, high binding 96-well plates (Greiner Bio-One, Frickenhausen, Germany) were coated with recombinant Spike or RBD (0.5 μg/mL) in 100 mM bicarbonate/carbonate buffer (pH 9.6) along with various standard curves (IgG, IgG1, IgG2c, IgA: serially diluted from 2000 ng/mL to 1.953 ng/mL) overnight at 4°C. Then, plates were blocked with 2% bovine serum albumin (BSA; Sigma Aldrich, St. Louis, MO, USA) in PBS-T (blocking buffer) for 1 hour at 37°C before samples diluted in blocking buffer were added in duplicate. Nasal wash samples were run in singlet. When running serum for total Spike/RBD-IgG, an additional set of serum samples was run to determine IgG avidity. Plates were incubated for 1 hour at 37°C then washed with PBS (pH 7.4). For IgG avidity assessment, the additional set of samples received 8M urea, while blocking buffer was added to the first set and the standard curve. Plates were covered and incubated for 15 minutes at room temperature protected from light, washed 4 times, and then blocked again with blocking buffer for 1 hour at 37°C. Next, plates were washed with PBS and anti-mouse IgG-HRP (Sigma Aldrich) was diluted 1:20,000 in blocking buffer and applied for 30 minutes at 37°C. For other immunoglobulins, the same protocol was followed without the additional avidity steps and the appropriate HRP-conjugated antibody was applied. Both IgG1- and IgG2c-HRP were diluted 1:20,000 in blocking buffer and applied for 30 minutes at 37°C. For IgA, HRP-conjugated anti-mouse IgA (Sigma Aldrich) was diluted 1:2,000 in blocking buffer and applied for 1 hour at 37°C. Plates were washed a final time with PBS and 3,3′,5,5′-Tetramethyl benzidine (TMB) substrate (Sigma Aldrich) was added to each well. The reaction was stopped after 15 minutes using H2SO4 (0.5M; Fisher Scientific, Waltham, MA, USA) and the optical density (OD) was measured at 450 nm with an EL800 microplate reader (BioTek Instruments Inc., Winooski, VT, USA). Concentrations of Spike/RBD specific antibodies were calculated by extrapolation from respective standard curves and multiplied by the dilution factor. IgG avidity indices were calculated by dividing the IgG titer in the urea conditions by the IgG titer in the non-treated condition.
Surrogate virus neutralization test
Neutralizing antibodies were assessed using the cPass SARS-CoV-2 Neutralization Antibody Detection Kit (GenScript) according to manufacturer’s instructions with the following changes: To collect semi-quantitative results, the kit was run with the SARS-CoV-2 Neutralizing Antibody Calibrator to create a standard curve used to determine the concentration of neutralizing antibodies. BALF samples were run neat or diluted (1:3) and serum samples were diluted (1:150). Samples that gave values above the 30% signal inhibition cut-off value were multiplied by the dilution factor and reported as Units/mL. Data reported according to the manufacturer’s guidelines can be found in Tables S5 (serum) and S6 (BALF).
Turbinate wash, BALF, and lung collection
Six months after the last immunization, unchallenged mice were euthanized, and the lungs were collected. Turbinate washes were collected by combining three washes of 150μL of cold PBS+protease inhibitor (PBS+PI) pushed through the nasal pharynx. Bronchoalveolar lavage fluid (BALF) was collected by combining four lung washes of 0.5mL of cold PBS+PI. Lungs were collected in 1mL cold RPMI. Lungs were digested enzymatically for 30 minutes at 37°C and 5% CO2 with a cocktail of DNase I (200 μg/mL, Sigma Aldrich), LiberaseTM (100 μg/mL, Roche, Indianapolis, IN, USA), hyaluronidase 1a (1 mg/mL, Life Technologies, Carlsbad, CA, USA), and collagenase XI (250μg/ml; Life Technologies) in RPMI-1640 as described previously.81 Cells were then washed with RPMI-1640 media containing 1% Penicillin/Streptomycin and 5% FBS. Sterile, filtered ammonium-chloride-potassium (ACK) buffer was used to lyse red blood cells. Filtration through a 0.7μM strainer was performed and the remaining viable cells were recovered.
Quantification of cytokine-secretion in T cells by multi-parametric flow cytometry
Lung lymphocytes were seeded into 96-well flat bottom plates (BD) at 106 cells in 200 uL/well. Duplicate cultures were stimulated with or without a combined preparation of Peptivator Peptide Pools of the complete Spike protein and predicted immunodominant sequences (Miltenyi Biotec, Bergisch Gladbach, North Rhine-Westphalia, Germany) in RPMI (0.3 μg/mL final concentration) for 18 and 96 hours at 37°C + 5% CO2. For the last 6 hours of incubation, protein transport inhibitor was prepared according to the manufacturer’s guidelines (RRID:AB_2869014, BD Science, San Jose, CA, USA) and added to all samples. Cells stimulated with phorbol 12-myristate 13-acetate (Thermofisher Scientific) and ionomycin (Thermofisher Scientific) were processed as positive controls. All staining and fixation steps took place at 4°C protected from light. Briefly, the cells were washed twice with cold PBS and stained with 50μL/well fixable viability dye eFluor 780 (Thermofisher Scientific) diluted at 1:1000 for 20 minutes. Cells were washed once with PBS. All surface stains were diluted 1:50 in PBS and 50μL/well of extracellular cocktail was applied for 30 minutes. The following antibodies made up the extracellular cocktail: CD3-BUV395 (145-2C11, RRID: AB_27382, BD Biosciences, Franklin Lakes, NJ, USA), CD4-AF700 (RM4-5, RRID: AB_49370, BioLegend, San Diego, CA, USA), CD8b-BV510 (H35-17.2, RRID: AB_2739908, BD Biosciences) and CD69-FITC (H1.2F3, RRID: AB_313108, BioLegend). Cells were then washed as before and fixed with the eBioscience FoxP3 transcription factor staining buffer (Thermofisher Scientific) overnight. The next day, plates were washed with 1X permeabilization buffer (perm buffer) (Thermofisher Scientific) and stained with an intracellular cocktail of antibodies diluted 1:50 in perm buffer applied as 50μL/well for 30 minutes. The intracellular cocktail was made up of: FOXP3-Pe-Cy7 (FJK-16s, RRID: AB_891552, Thermofisher Scientific), IFNγ-BUV737 (XMG1.2, RRID: AB_2870098, BD Biosciences), IL-17A-APC (TC11-18H10.1, RRID: AB_536018, BioLegend), IL-4-BV421 (11B11, RRID: AB_2562594, BioLegend), GrB-PE (QA1602, RRID: AB_2687032, BioLegend), and TNFα-PerCP-Cy5.5 (MP6-XT22, RRID: AB_961434, BioLegend). After staining, cells were washed twice with perm buffer and resuspended in PBS 1X and acquired on a BD LSRFortessa X-20 (BD Science). Flow data were analysed using FlowJo software (version 10.0.8r1) (Treestar, Ashland, OR, USA). Our gating strategy is shown in Figure S5.
Histological analysis
Organs collected from the challenged animals at the time of necropsy were placed in 10% phosphate-buffered formalin. Collected tissues were subsequently processed for histopathology, and slides were stained with haematoxylin and eosin (H & E), to assess tissue architecture and inflammation, and Masson’s trichrome, to determine the progression of fibrosis. Sections of lungs were examined and scored by a pathologist who was blinded to the experimental groups. Lungs were evaluated for fibrosis, the presence or absence of features of cell or tissue damage (CTD: necrosis of bronchiolar epithelial cells (BEC), inflammatory cells and/or cellular debris in bronchi, intraepithelial neutrophils, alveolar emphysema), circulatory changes and vascular lesions (CVL: alveolar hemorrhage, significant alveolar edema, vasculitis/vascular endothelialitis), reactive inflammatory patterns (RIP: necrosuppurative bronchitis, intraalveolar neutrophils, and macrophages, mononuclear infiltrates around airways, presence of polymorphonuclear granulocytes, perivascular mononuclear cuffs, and mesothelial reactivity), as well as regeneration and repair (RR: alveolar epithelial hyperplasia/regeneration, BEC hyperplasia/regeneration) as described elsewhere.79,82 After the scoring was completed, lung pathology scores were tabulated. Processed and stained lung slides were digitized using Aperio AT2 (Leica Biosystems, Wetzlar, Germany).
Quantification and statistical analysis
Experimental units are defined as individual animals. Sample sizes were empirically estimated based on previous data considering the anticipated variation of the results and statistical power needed, while also minimizing the number of animals used. C57BL/6 mice were randomly attributed to treatment groups. To minimise potential confounders, mice were matched for age and sex. Blinding: For all challenge experiments, staff performing infections and sample harvesting were blinded to the different groups and were only unblinded after data analysis. Inclusion/Exclusion: Aside from a small number of deaths in one of the control groups (Ad(e)+BCG(e)), no other animals were excluded from the analysis. Statistical analysis was performed using GraphPad Prism 9 software (La Jolla, CA, USA) and statistical details of experiments can be found in figure legends. Data were assessed for normality using Shapiro-Wilk tests. Non-parametric data were analysed by Kruskal-Wallis tests with Dunn’s multiple comparisons. When appropriate, one-way and two-way ANOVAs were employed with Tukey’s multiple comparisons. P values <0.05 were considered significant.
Additional resources
Included in the supplementary data are:
-
(1)
Supplemental figures.
-
(2)
Reagent repository.
-
(3)
ARRIVE Checklist.
-
(4)
MDAR Author Checklist.
Acknowledgments
We would like to thank Annie Beauchamp, Rami Karkout, Sarah Santoso, Louis Cyr, Angela Brewer, and Raidan Alyazidi for their contributions during animal sacrifice and advice for experimental procedures as well as the other members of the Ward/Ndao laboratory for their support. We would also like to thank Dr. Marcel Behr for kindly providing us with the Danish strain of BCG. We would like to thank Heather Tyra and Francois Francoeur at IDT for their assistance in making our gene construct. We would like to thank Nazila Nazemi-Moghaddam and Claire Guilbault of the National Research Council of Canada for their support manufacturing, amplifying, and titrating our recombinant adenovirus. We thank the staff from the high-containment lab (Sunnybrook Hospital) for their technical assistance. We would like to thank the members of the Mammalian Cell Expression Section of the NRC-HHT for their contribution to producing and purifying the recombinant proteins and those members at BEI resources for their contributions of recombinant proteins. In addition, we would like to thank the Immunophenotyping Platform at the Research Institute of the McGill University Health Center (RI-MUHC). We would like to thank Kim Babin and Bruce Lu, from Euroimmun and GenScript, respectively, with their help obtaining cPass neutralizing antibody kits. We would also like to thank Ara Xiii for their help in editing our manuscript. Finally, we would like to thank all the entities which contributed to this work financially including the McGill Interdisciplinary Initiative in Infection and Immunity (MI4) program, the Foundation of the MUHC, the R. Howard Webster Foundation, the Foundation of the Montreal General Hospital, and the RI-MUHC. Graphical abstract and other schematics were made using BioRender.com. This study was supported by the McGill Interdisciplinary Initiative in Infection and Immunity (MI4) Emergency COVID-19 Research Funding Grant ECRF-R2-70 awarded to M.O., M.B.R., and M.Nd.
Author contributions
Study design: D.J.P., P.D., M.B.R., and M.Nd. in collaboration with R.K., G.P.K., M.O., and R.G.
Funding acquisition: M.Nd., M.B.R., and M.O.
Vaccine design: D.J.P. and P.D. with assistance from S.M.E.
Animal vaccinations and immunogenicity experiments: D.J.P., C.K.P., and P.D.
Animal sacrifice, sample collection, and sample processing: D.J.P., P.D., F.A., and L.L.
Histology imaging and scoring: A.L. and P.O.F.
Recombinant protein production: M.S. and Y.D.
Manuscript preparation: D.J.P. and F.A.
Manuscript editing and contribution: P.D., G.G.B., M.Na., C.A.P., R.K., M.B.R., and M.Nd.
Animal challenge experiments: M.Na., G.G.B., and R.K.
All authors have read and approved the final version of this manuscript.
Declaration of interests
The authors declare that there are no competing interests involved in this work.
Inclusion and diversity
We support inclusive, diverse, and equitable conduct of research.
Published: August 11, 2023
Footnotes
Supplemental information can be found online at https://doi.org/10.1016/j.isci.2023.107612.
Contributor Information
Robert Kozak, Email: rkozak@shn.ca.
Michael B. Reed, Email: Michael.reed@mcgill.ca.
Momar Ndao, Email: Momar.ndao@mcgill.ca.
Supplemental information
Data and code availability
-
•
All data associated with this study are present in the paper or the Supplementary Materials. Requests may be made by contacting the corresponding authors.
-
•
This paper does not report original code
-
•
Any additional information required to reanalyze the data reported in this work paper is available from the lead contact upon request.
References
- 1.WHO Coronavirus (COVID-19) Dashboard | WHO Coronavirus (COVID-19) Dashboard With Vaccination Data. https://covid19.who.int/
- 2.Zhu N., Zhang D., Wang W., Li X., Yang B., Song J., Zhao X., Huang B., Shi W., Lu R., et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020;382:727–733. doi: 10.1056/NEJMOA2001017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Al-Aly Z., Bowe B., Xie Y. Long COVID after breakthrough SARS-CoV-2 infection. Nat. Med. 2022;28:1461–1467. doi: 10.1038/s41591-022-01840-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zheng C., Shao W., Chen X., Zhang B., Wang G., Zhang W. Real-world effectiveness of COVID-19 vaccines: a literature review and meta-analysis. Int. J. Infect. Dis. 2022;114:252–260. doi: 10.1016/J.IJID.2021.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Eyre D.W., Taylor D., Purver M., Chapman D., Fowler T., Pouwels K.B., Walker A.S., Peto T.E.A. Effect of Covid-19 Vaccination on Transmission of Alpha and Delta Variants. N. Engl. J. Med. 2022;386:744–756. doi: 10.1056/NEJMOA2116597/SUPPL_FILE/NEJMOA2116597_DISCLOSURES.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Feikin D.R., Higdon M.M., Abu-Raddad L.J., Andrews N., Araos R., Goldberg Y., Groome M.J., Huppert A., O’Brien K.L., Smith P.G., et al. Duration of effectiveness of vaccines against SARS-CoV-2 infection and COVID-19 disease: results of a systematic review and meta-regression. Lancet. 2022;399:924–944. doi: 10.1016/S0140-6736(22)00152-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brosh-Nissimov T., Orenbuch-Harroch E., Chowers M., Elbaz M., Nesher L., Stein M., Maor Y., Cohen R., Hussein K., Weinberger M., et al. BNT162b2 vaccine breakthrough: clinical characteristics of 152 fully vaccinated hospitalized COVID-19 patients in Israel. Clin. Microbiol. Infect. 2021;27:1652–1657. doi: 10.1016/J.CMI.2021.06.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Travis C.R. As Plain as the Nose on Your Face: The Case for A Nasal (Mucosal) Route of Vaccine Administration for Covid-19 Disease Prevention. Front. Immunol. 2020;11:591897. doi: 10.3389/FIMMU.2020.591897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Dhama K., Dhawan M., Tiwari R., Emran T.B., Mitra S., Rabaan A.A., Alhumaid S., Alawi Z.A., Al Mutair A. COVID-19 intranasal vaccines: current progress, advantages, prospects, and challenges. Hum. Vaccines Immunother. 2022;18:2045853. doi: 10.1080/21645515.2022.2045853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hassan A.O., Kafai N.M., Dmitriev I.P., Fox J.M., Smith B.K., Harvey I.B., Chen R.E., Winkler E.S., Wessel A.W., Case J.B., et al. A Single-Dose Intranasal ChAd Vaccine Protects Upper and Lower Respiratory Tracts against SARS-CoV-2. Cell. 2020;183:169–184.e13. doi: 10.1016/J.CELL.2020.08.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hassan A.O., Feldmann F., Zhao H., Curiel D.T., Okumura A., Tang-Huau T.L., Case J.B., Meade-White K., Callison J., Chen R.E., et al. A single intranasal dose of chimpanzee adenovirus-vectored vaccine protects against SARS-CoV-2 infection in rhesus macaques. Cell Rep. Med. 2021;2:100230. doi: 10.1016/j.xcrm.2021.100230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Madhavan M., Ritchie A.J., Aboagye J., Jenkin D., Provstgaad-Morys S., Tarbet I., Woods D., Davies S., Baker M., Platt A., et al. Tolerability and immunogenicity of an intranasally-administered adenovirus-vectored COVID-19 vaccine: An open-label partially-randomised ascending dose phase I trial. EBioMedicine. 2022;85:104298. doi: 10.1016/J.EBIOM.2022.104298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Martinez L., Cords O., Liu Q., Acuna-Villaorduna C., Bonnet M., Fox G.J., Carvalho A.C.C., Chan P.C., Croda J., Hill P.C., et al. Infant BCG vaccination and risk of pulmonary and extrapulmonary tuberculosis throughout the life course: a systematic review and individual participant data meta-analysis. Lancet Global Health. 2022;10:e1307–e1316. doi: 10.1016/S2214-109X(22)00283-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Roth A., Gustafson P., Nhaga A., Djana Q., Poulsen A., Garly M.L., Jensen H., Sodemann M., Rodriques A., Aaby P. BCG vaccination scar associated with better childhood survival in Guinea-Bissau. Int. J. Epidemiol. 2005;34:540–547. doi: 10.1093/IJE/DYH392. [DOI] [PubMed] [Google Scholar]
- 15.Stensballe L.G., Nante E., Jensen I.P., Kofoed P.E., Poulsen A., Jensen H., Newport M., Marchant A., Aaby P. Acute lower respiratory tract infections and respiratory syncytial virus in infants in Guinea-Bissau: a beneficial effect of BCG vaccination for girls: Community based case–control study. Vaccine. 2005;23:1251–1257. doi: 10.1016/J.VACCINE.2004.09.006. [DOI] [PubMed] [Google Scholar]
- 16.Netea M.G., Quintin J., Van Der Meer J.W.M. Trained immunity: A memory for innate host defense. Cell Host Microbe. 2011;9:355–361. doi: 10.1016/j.chom.2011.04.006. [DOI] [PubMed] [Google Scholar]
- 17.Moorlag S.J.C.F.M., Arts R.J.W., van Crevel R., Netea M.G. Non-specific effects of BCG vaccine on viral infections. Clin. Microbiol. Infect. 2019;25:1473–1478. doi: 10.1016/J.CMI.2019.04.020. [DOI] [PubMed] [Google Scholar]
- 18.O’Neill L.A.J., Netea M.G. BCG-induced trained immunity: can it offer protection against COVID-19? Nat. Rev. Immunol. 2020;20:335–337. doi: 10.1038/s41577-020-0337-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kaufmann E., Khan N., Tran K.A., Ulndreaj A., Pernet E., Fontes G., Lupien A., Desmeules P., McIntosh F., Abow A., et al. BCG vaccination provides protection against IAV but not SARS-CoV-2. Cell Rep. 2022;38:110502. doi: 10.1016/J.CELREP.2022.110502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Messina N.L., Germano S., McElroy R., Rudraraju R., Bonnici R., Pittet L.F., Neeland M.R., Nicholson S., Subbarao K., Curtis N., BRACE trial Off-target effects of bacillus Calmette–Guérin vaccination on immune responses to SARS-CoV-2: implications for protection against severe COVID-19. Clin. Transl. Immunology. 2022;11:e1387. doi: 10.1002/CTI2.1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pittet L.F., Messina N.L., Orsini F., Moore C.L., Abruzzo V., Barry S., Bonnici R., Bonten M., Campbell J., Croda J., et al. Randomized Trial of BCG Vaccine to Protect against Covid-19 in Health Care Workers. N. Engl. J. Med. 2023;388:1582–1596. doi: 10.1056/NEJMOA2212616/SUPPL_FILE/NEJMOA2212616_DATA-SHARING.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Rakshit S., Adiga V., Ahmed A., Parthiban C., Chetan Kumar N., Dwarkanath P., Shivalingaiah S., Rao S., D’Souza G., Dias M., et al. Evidence for the heterologous benefits of prior BCG vaccination on COVISHIELD™ vaccine-induced immune responses in SARS-CoV-2 seronegative young Indian adults. Front. Immunol. 2022;13:5362. doi: 10.3389/FIMMU.2022.985938/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Van Doremalen N., Purushotham J.N., Schulz J.E., Holbrook M.G., Bushmaker T., Carmody A., Port J.R., Yinda C.K., Okumura A., Saturday G., et al. Intranasal ChAdOx1 nCoV-19/AZD1222 vaccination reduces viral shedding after SARS-CoV-2 D614G challenge in preclinical models. Sci. Transl. Med. 2021;13:eabh0755. doi: 10.1126/SCITRANSLMED.ABH0755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Shin H., Iwasaki A. A vaccine strategy that protects against genital herpes by establishing local memory T cells. Nature. 2012;491:463–467. doi: 10.1038/nature11522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kremer L., Riveau G., Baulard A., Capron A., Locht C. Neutralizing antibody responses elicited in mice immunized with recombinant bacillus Calmette-Guérin producing the Schistosoma mansoni glutathione S-transferase. J. Immunol. 1996;156:4309–4317. doi: 10.4049/JIMMUNOL.156.11.4309. [DOI] [PubMed] [Google Scholar]
- 26.Langermann S., Palaszynski S., Sadziene A., Stover C.K., Koenig S. Systemic and mucosal immunity induced by BCG vector expressing outer-surface protein A of Borrelia burgdorferi. Nature. 1994;372:552–555. doi: 10.1038/372552a0. [DOI] [PubMed] [Google Scholar]
- 27.Grode L., Seiler P., Baumann S., Hess J., Brinkmann V., Nasser Eddine A., Mann P., Goosmann C., Bandermann S., Smith D., et al. Increased vaccine efficacy against tuberculosis of recombinant Mycobacterium bovis bacille Calmette-Guérin mutants that secrete listeriolysin. J. Clin. Invest. 2005;115:2472–2479. doi: 10.1172/JCI24617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Palendira U., Bean A.G.D., Feng C.G., Britton W.J. Lymphocyte recruitment and protective efficacy against pulmonary mycobacterial infection are independent of the route of prior Mycobacterium bovis BCG immunization. Infect. Immun. 2002;70:1410–1416. doi: 10.1128/IAI.70.3.1410-1416.2002/ASSET/754FC338-A11C-46D2-8FB5-7810DA6CD3CA/ASSETS/GRAPHIC/II0321253006.JPEG. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jeyanathan M., Vaseghi-Shanjani M., Afkhami S., Grondin J.A., Kang A., D’Agostino M.R., Yao Y., Jain S., Zganiacz A., Kroezen Z., et al. Parenteral BCG vaccine induces lung-resident memory macrophages and trained immunity via the gut-lung axis. Nat. Immunol. 2022;23:1687–1702. doi: 10.1038/S41590-022-01354-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Moliva J.I., Turner J., Torrelles J.B. Prospects in Mycobacterium bovis Bacille Calmette et Guérin (BCG) Vaccine Diversity and Delivery: Why does BCG fail to protect against Tuberculosis? Vaccine. 2015;33:5035–5041. doi: 10.1016/J.VACCINE.2015.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ledesma J.R., Lurie P., Yorlets R.R., Daly G., Chrysanthopoulou S., Lurie M.N. Spurious early ecological association suggesting BCG vaccination effectiveness for COVID-19. PLoS One. 2022;17:e0274900. doi: 10.1371/JOURNAL.PONE.0274900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Counoupas C., Johansen M.D., Stella A.O., Nguyen D.H., Ferguson A.L., Aggarwal A., Bhattacharyya N.D., Grey A., Hutchings O., Patel K., et al. A single dose, BCG-adjuvanted COVID-19 vaccine provides sterilising immunity against SARS-CoV-2 infection. npj Vaccines. 2021;6:143–210. doi: 10.1038/s41541-021-00406-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bannister S., Kim B., Domínguez-Andrés J., Kilic G., Ansell B.R.E., Neeland M.R., Moorlag S.J.C.F.M., Matzaraki V., Vlahos A., Shepherd R., et al. Neonatal BCG vaccination is associated with a long-term DNA methylation signature in circulating monocytes. Sci. Adv. 2022;8:eabn4002. doi: 10.1126/SCIADV.ABN4002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jackson S.W., Jacobs H.M., Arkatkar T., Dam E.M., Scharping N.E., Kolhatkar N.S., Hou B., Buckner J.H., Rawlings D.J. B cell IFN-γ receptor signaling promotes autoimmune germinal centers via cell-intrinsic induction of BCL-6. J. Exp. Med. 2016;213:733–750. doi: 10.1084/JEM.20151724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wirsching S., Harder L., Heymanns M., Gröndahl B., Hilbert K., Kowalzik F., Meyer C., Gehring S. Long-Term, CD4+ Memory T Cell Response to SARS-CoV-2. Front. Immunol. 2022;13:800070. doi: 10.3389/FIMMU.2022.800070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.He X., Chandrashekar A., Zahn R., Wegmann F., Yu J., Mercado N.B., McMahan K., Martinot A.J., Piedra-Mora C., Beecy S., et al. Low-dose Ad26.COV2.S protection against SARS-CoV-2 challenge in rhesus macaques. Cell. 2021;184:3467–3473.e11. doi: 10.1016/J.CELL.2021.05.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Arts R.J.W., Moorlag S.J.C.F.M., Novakovic B., Li Y., Wang S.Y., Oosting M., Kumar V., Xavier R.J., Wijmenga C., Joosten L.A.B., et al. BCG Vaccination Protects against Experimental Viral Infection in Humans through the Induction of Cytokines Associated with Trained Immunity. Cell Host Microbe. 2018;23:89–100.e5. doi: 10.1016/J.CHOM.2017.12.010. [DOI] [PubMed] [Google Scholar]
- 38.Covián C., Fernández-Fierro A., Retamal-Díaz A., Díaz F.E., Vasquez A.E., Lay M.K., Riedel C.A., González P.A., Bueno S.M., Kalergis A.M. BCG-Induced Cross-Protection and Development of Trained Immunity: Implication for Vaccine Design. Front. Immunol. 2019;10:2806. doi: 10.3389/FIMMU.2019.02806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tiboni M., Casettari L., Illum L. Nasal vaccination against SARS-CoV-2: Synergistic or alternative to intramuscular vaccines? Int. J. Pharm. 2021;603:120686. doi: 10.1016/J.IJPHARM.2021.120686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hassan A.O., Shrihari S., Gorman M.J., Ying B., Yuan D., Raju S., Chen R.E., Dmitriev I.P., Kashentseva E., Adams L.J., et al. An intranasal vaccine durably protects against SARS-CoV-2 variants in mice. Cell Rep. 2021;36:109452. doi: 10.1016/J.CELREP.2021.109452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Afkhami S., D’Agostino M.R., Zhang A., Stacey H.D., Marzok A., Kang A., Singh R., Bavananthasivam J., Ye G., Luo X., et al. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell. 2022;185:896–915.e19. doi: 10.1016/J.CELL.2022.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kong L., Moorlag S.J.C.F.M., Lefkovith A., Li B., Matzaraki V., van Emst L., Kang H.A., Latorre I., Jaeger M., Joosten L.A.B., et al. Single-cell transcriptomic profiles reveal changes associated with BCG-induced trained immunity and protective effects in circulating monocytes. Cell Rep. 2021;37:110028. doi: 10.1016/J.CELREP.2021.110028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kaufmann E., Sanz J., Dunn J.L., Khan N., Mendonça L.E., Pacis A., Tzelepis F., Pernet E., Dumaine A., Grenier J.C., et al. BCG Educates Hematopoietic Stem Cells to Generate Protective Innate Immunity against. Tuberculosis. Cell. 2018;172:176–190.e19. doi: 10.1016/J.CELL.2017.12.031. [DOI] [PubMed] [Google Scholar]
- 44.Walk J., de Bree L.C.J., Graumans W., Stoter R., van Gemert G.J., van de Vegte-Bolmer M., Teelen K., Hermsen C.C., Arts R.J.W., Behet M.C., et al. Outcomes of controlled human malaria infection after BCG vaccination. Nat. Commun. 2019;10:874–878. doi: 10.1038/s41467-019-08659-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Singh A.K., Chaube B., Zhang X., Sun J., Citrin K.M., Canfrán-Duque A., Aryal B., Rotllan N., Varela L., Lee R.G., et al. BCG turns 100: its nontraditional uses against viruses, cancer, and immunologic diseases. J. Clin. Invest. 2021;131:e140989. doi: 10.1172/JCI148291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kleinnijenhuis J., Quintin J., Preijers F., Joosten L.A.B., Ifrim D.C., Saeed S., Jacobs C., Van Loenhout J., De Jong D., Stunnenberg H.G., et al. Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc. Natl. Acad. Sci. USA. 2012;109:17537–17542. doi: 10.1073/PNAS.1202870109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Libraty D.H., Zhang L., Woda M., Acosta L.P., Obcena A., Brion J.D., Capeding R.Z. Neonatal BCG vaccination is associated with enhanced T-helper 1 immune responses to heterologous infant vaccines. Trials Vaccinol. 2014;3:1–5. doi: 10.1016/J.TRIVAC.2013.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ota M.O.C., Vekemans J., Schlegel-Haueter S.E., Fielding K., Sanneh M., Kidd M., Newport M.J., Aaby P., Whittle H., Lambert P.-H., et al. Influence of Mycobacterium bovis bacillus Calmette-Guérin on antibody and cytokine responses to human neonatal vaccination. J. Immunol. 2002;168:919–925. doi: 10.4049/JIMMUNOL.168.2.919. [DOI] [PubMed] [Google Scholar]
- 49.Leentjens J., Kox M., Stokman R., Gerretsen J., Diavatopoulos D.A., Van Crevel R., Rimmelzwaan G.F., Pickkers P., Netea M.G. BCG Vaccination Enhances the Immunogenicity of Subsequent Influenza Vaccination in Healthy Volunteers: A Randomized, Placebo-Controlled Pilot Study. J. Infect. Dis. 2015;212:1930–1938. doi: 10.1093/INFDIS/JIV332. [DOI] [PubMed] [Google Scholar]
- 50.Dubé J.Y., McIntosh F., Zarruk J.G., David S., Nigou J., Behr M.A. Synthetic mycobacterial molecular patterns partially complete Freund’s adjuvant. Sci. Rep. 2020;10:1–14. doi: 10.1038/s41598-020-62543-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nieuwenhuizen N.E., Kaufmann S.H.E. Next-generation vaccines based on Bacille Calmette-Guérin. Front. Immunol. 2018;9:121. doi: 10.3389/FIMMU.2018.00121/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mouhoub E., Domenech P., Ndao M., Reed M.B. The Diverse Applications of Recombinant BCG-Based Vaccines to Target Infectious Diseases Other Than Tuberculosis: An Overview. Front. Microbiol. 2021;12:3199. doi: 10.3389/FMICB.2021.757858/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Escobar L.E., Molina-Cruz A., Barillas-Mury C. BCG vaccine protection from severe coronavirus disease 2019 (COVID-19) Proc. Natl. Acad. Sci. USA. 2020;117:17720–17726. doi: 10.1073/PNAS.2008410117/SUPPL_FILE/PNAS.2008410117.SD01.XLSX. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Arlehamn C.S.L., Sette A., Peters B. Lack of evidence for BCG vaccine protection from severe COVID-19. Proc. Natl. Acad. Sci. USA. 2020;117:25203–25204. doi: 10.1073/PNAS.2016733117/ASSET/BAFD4353-75D8-4716-AEBA-9C91E0FBCD85/ASSETS/PNAS.2016733117.FP.PNG. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hilligan K.L., Namasivayam S., Clancy C.S., O’Mard D., Oland S.D., Robertson S.J., Baker P.J., Castro E., Garza N.L., Lafont B.A.P., et al. Intravenous administration of BCG protects mice against lethal SARS-CoV-2 challenge. J. Exp. Med. 2022;219:e20211862. doi: 10.1084/JEM.20211862/212903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Peng Q., Zhou R., Wang Y., Zhao M., Liu N., Li S., Huang H., Yang D., Au K.K., Wang H., et al. Waning immune responses against SARS-CoV-2 variants of concern among vaccinees in Hong Kong. EBioMedicine. 2022;77:103904. doi: 10.1016/j.ebiom.2022.103904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Altmann D.M., Boyton R.J. Waning immunity to SARS-CoV-2: implications for vaccine booster strategies. Lancet Respir. Med. 2021;9:1356–1358. doi: 10.1016/S2213-2600(21)00458-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yao Y., Jeyanathan M., Haddadi S., Barra N.G., Vaseghi-Shanjani M., Damjanovic D., Lai R., Afkhami S., Chen Y., Dvorkin-Gheva A., et al. Induction of Autonomous Memory Alveolar Macrophages Requires T Cell Help and Is Critical to Trained Immunity. Cell. 2018;175:1634–1650.e17. doi: 10.1016/J.CELL.2018.09.042. [DOI] [PubMed] [Google Scholar]
- 59.Jeljeli M., Riccio L.G.C., Doridot L., Chêne C., Nicco C., Chouzenoux S., Deletang Q., Allanore Y., Kavian N., Batteux F. Trained immunity modulates inflammation-induced fibrosis. Nat. Commun. 2019;10:1–15. doi: 10.1038/s41467-019-13636-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Woodworth J.S., Clemmensen H.S., Battey H., Dijkman K., Lindenstrøm T., Laureano R.S., Taplitz R., Morgan J., Aagaard C., Rosenkrands I., et al. A Mycobacterium tuberculosis-specific subunit vaccine that provides synergistic immunity upon co-administration with Bacillus Calmette-Guérin. Nat. Commun. 2021;12:6658–6713. doi: 10.1038/s41467-021-26934-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Rakshit S., Ahmed A., Adiga V., Sundararaj B.K., Sahoo P.N., Kenneth J., D’Souza G., Bonam W., Johnson C., Franken K.L., et al. BCG revaccination boosts adaptive polyfunctional Th1/Th17 and innate effectors in IGRA+ and IGRA- Indian adults. JCI Insight. 2019;4:e130540. doi: 10.1172/JCI.INSIGHT.130540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Pan T., Chen J., Zhang T., Liu S., He S., Lv H., Yan H., Yan S., Liu J., Zhang Y., et al. Infection of wild-type mice by SARS-CoV-2 B.1.351 variant indicates a possible novel cross-species transmission route. ISA Trans. 2021;128:1–10. doi: 10.1038/s41392-021-00848-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Davis M.A., Voss K., Turnbull J.B., Gustin A.T., Knoll M., Muruato A., Hsiang T.Y., Dinnon Iii K.H., Leist S.R., Nickel K., et al. A C57BL/6 Mouse Model of SARS-CoV-2 Infection Recapitulates Age- and Sex-Based Differences in Human COVID-19 Disease and Recovery. Vaccines. 2022;11:47. doi: 10.3390/VACCINES11010047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Currey J.M., Rabito F., Maness N.J., Blair R.V., Rappaport J., Qin X., Kolls J.K., Srivastava A.K. C57BL/6J Mice Are Not Suitable for Modeling Severe SARS-CoV-2 Beta and Gamma Variant Infection. Viruses. 2022;14 doi: 10.3390/V14050966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Barouch D.H., Tomaka F.L., Wegmann F., Stieh D.J., Alter G., Robb M.L., Michael N.L., Peter L., Nkolola J.P., Borducchi E.N., et al. Evaluation of a Mosaic HIV-1 Vaccine in a Randomized, Double-Blinded, Placebo-Controlled Phase I/IIa Clinical Trial and in Rhesus Monkeys. Lancet. 2018;392:232–243. doi: 10.1016/S0140-6736(18)31364-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Smaill F., Jeyanathan M., Smieja M., Medina M.F., Thanthrige-Don N., Zganiacz A., Yin C., Heriazon A., Damjanovic D., Puri L., et al. A human type 5 adenovirus-based tuberculosis vaccine induces robust T cell responses in humans despite preexisting anti-adenovirus immunity. Sci. Transl. Med. 2013;5:205ra134. doi: 10.1126/SCITRANSLMED.3006843/SUPPL_FILE/5-205RA134_SM.PDF. [DOI] [PubMed] [Google Scholar]
- 67.Bekker L.G., Garrett N., Goga A., Fairall L., Reddy T., Yende-Zuma N., Kassanjee R., Collie S., Sanne I., Boulle A., et al. Effectiveness of the Ad26.COV2.S vaccine in health-care workers in South Africa (the Sisonke study): results from a single-arm, open-label, phase 3B, implementation study. Lancet. 2022;399:1141–1153. doi: 10.1016/S0140-6736(22)00007-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Rzymski P. Guillain-Barré syndrome and COVID-19 vaccines: focus on adenoviral vectors. Front. Immunol. 2023;14:1183258. doi: 10.3389/FIMMU.2023.1183258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.García-Grimshaw M., Galnares-Olalde J.A., Bello-Chavolla O.Y., Michel-Chávez A., Cadena-Fernández A., Briseño-Godínez M.E., Antonio-Villa N.E., Núñez I., Gutiérrez-Romero A., Hernández-Vanegas L., et al. Incidence of Guillain–Barré syndrome following SARS-CoV-2 immunization: Analysis of a nationwide registry of recipients of 81 million doses of seven vaccines. Eur. J. Neurol. 2022;29:3368–3379. doi: 10.1111/ENE.15504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gilbert R., Guilbault C., Gagnon D., Bernier A., Bourget L., Elahi S.M., Kamen A., Massie B. Establishment and validation of new complementing cells for production of E1-deleted adenovirus vectors in serum-free suspension culture. J. Virol. Methods. 2014;208:177–188. doi: 10.1016/J.JVIROMET.2014.08.013. [DOI] [PubMed] [Google Scholar]
- 71.Perera D.J., Hassan A.S., Liu S.S., Elahi S.M., Gadoury C., Weeratna R.D., Gilbert R., Ndao M. A low dose adenovirus vectored vaccine expressing Schistosoma mansoni Cathepsin B protects from intestinal schistosomiasis in mice. EBioMedicine. 2022;80:104036. doi: 10.1016/J.EBIOM.2022.104036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Mullick A., Xu Y., Warren R., Koutroumanis M., Guilbault C., Broussau S., Malenfant F., Bourget L., Lamoureux L., Lo R., et al. The cumate gene-switch: a system for regulated expression in mammalian cells. BMC Biotechnol. 2006;6:1–18. doi: 10.1186/1472-6750-6-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.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. doi: 10.1073/PNAS.95.5.2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Elahi S.M., Nazemi-Moghaddam N., Guilbault C., Simoneau M., Gilbert R. Complementary Cell Lines for Protease Gene-Deleted Single-Cycle Adenovirus Vectors. Cells. 2023;12:619. doi: 10.3390/CELLS12040619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Akache B., Renner T.M., Stuible M., Rohani N., Cepero-Donates Y., Deschatelets L., Dudani R., Harrison B.A., Gervais C., Hill J.J., et al. Immunogenicity of SARS-CoV-2 spike antigens derived from Beta & Delta variants of concern. npj Vaccines. 2022;1:118–127. doi: 10.1038/s41541-022-00540-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Colwill K., Galipeau Y., Stuible M., Gervais C., Arnold C., Rathod B., Abe K.T., Wang J.H., Pasculescu A., Maltseva M., et al. A scalable serology solution for profiling humoral immune responses to SARS-CoV-2 infection and vaccination. Clin. Transl. Immunology. 2022;11:e1380. doi: 10.1002/CTI2.1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Forest-Nault C., Koyuturk I., Gaudreault J., Pelletier A., L'Abbé D., Cass B., Bisson L., Burlacu A., Delafosse L., Stuible M., et al. Impact of the temperature on the interactions between common variants of the SARS-CoV-2 receptor binding domain and the human ACE2. Sci. Rep. 2022;12:11520–11611. doi: 10.1038/s41598-022-15215-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Reed L.J., Muench H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 1938;27:493–497. doi: 10.1093/OXFORDJOURNALS.AJE.A118408. [DOI] [Google Scholar]
- 79.Babuadze G.G., Fausther-Bovendo H., deLaVega M.A., Lillie B., Naghibosadat M., Shahhosseini N., Joyce M.A., Saffran H.A., Lorne Tyrrell D., Falzarano D., et al. Two DNA vaccines protect against severe disease and pathology due to SARS-CoV-2 in Syrian hamsters. npj Vaccines. 2022;7:1–11. doi: 10.1038/s41541-022-00461-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Feld J.J., Kandel C., Biondi M.J., Kozak R.A., Zahoor M.A., Lemieux C., Borgia S.M., Boggild A.K., Powis J., McCready J., et al. Peginterferon lambda for the treatment of outpatients with COVID-19: a phase 2, placebo-controlled randomised trial. Lancet Respir. Med. 2021;9:498–510. doi: 10.1016/S2213-2600(20)30566-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Nakada E.M., Shan J., Kinyanjui M.W., Fixman E.D. Adjuvant-dependent regulation of interleukin-17 expressing γδ T cells and inhibition of Th2 responses in allergic airways disease. Respir. Res. 2014;15:1–14. doi: 10.1186/S12931-014-0090-5/FIGURES/6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Gruber A.D., Osterrieder N., Bertzbach L.D., Vladimirova D., Greuel S., Ihlow J., Horst D., Trimpert J., Dietert K. Standardization of Reporting Criteria for Lung Pathology in SARS-CoV-2-infected Hamsters: What Matters? Am. J. Respir. Cell Mol. Biol. 2020;63:856–859. doi: 10.1165/RCMB.2020-0280LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
-
•
All data associated with this study are present in the paper or the Supplementary Materials. Requests may be made by contacting the corresponding authors.
-
•
This paper does not report original code
-
•
Any additional information required to reanalyze the data reported in this work paper is available from the lead contact upon request.