Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Jul:347:476-488.
doi: 10.1016/j.jconrel.2022.05.023. Epub 2022 May 20.

Nanoparticle-delivered TLR4 and RIG-I agonists enhance immune response to SARS-CoV-2 subunit vaccine

Affiliations

Nanoparticle-delivered TLR4 and RIG-I agonists enhance immune response to SARS-CoV-2 subunit vaccine

Alexandra Atalis et al. J Control Release. 2022 Jul.

Abstract

Despite success in vaccinating populations against SARS-CoV-2, concerns about immunity duration, continued efficacy against emerging variants, protection from infection and transmission, and worldwide vaccine availability remain. Molecular adjuvants targeting pattern recognition receptors (PRRs) on antigen-presenting cells (APCs) could improve and broaden the efficacy and durability of vaccine responses. Native SARS-CoV-2 infection stimulates various PRRs, including toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors. We hypothesized that targeting PRRs using molecular adjuvants on nanoparticles (NPs) along with a stabilized spike protein antigen could stimulate broad and efficient immune responses. Adjuvants targeting TLR4 (MPLA), TLR7/8 (R848), TLR9 (CpG), and RIG-I (PUUC) delivered on degradable polymer NPs were combined with the S1 subunit of spike protein and assessed in vitro with isogeneic mixed lymphocyte reactions (isoMLRs). For in vivo studies, the adjuvant-NPs were combined with stabilized spike protein or spike-conjugated NPs and assessed using a two-dose intranasal or intramuscular vaccination model in mice. Combination adjuvant-NPs simultaneously targeting TLR and RIG-I receptors (MPLA+PUUC, CpG+PUUC, and R848+PUUC) differentially induced T cell proliferation and increased proinflammatory cytokine secretion by APCs in vitro. When delivered intranasally, MPLA+PUUC NPs enhanced CD4+CD44+ activated memory T cell responses against spike protein in the lungs while MPLA NPs increased anti-spike IgA in the bronchoalveolar (BAL) fluid and IgG in the blood. Following intramuscular delivery, PUUC NPs induced strong humoral immune responses, characterized by increases in anti-spike IgG in the blood and germinal center B cell populations (GL7+ and BCL6+ B cells) in the draining lymph nodes (dLNs). MPLA+PUUC NPs further boosted spike protein-neutralizing antibody titers and T follicular helper cell populations in the dLNs. These results suggest that protein subunit vaccines with particle-delivered molecular adjuvants targeting TLR4 and RIG-I could lead to robust and unique route-specific adaptive immune responses against SARS-CoV-2.

Keywords: Adaptive immune response; COVID-19 protein subunit vaccine; Combination adjuvant; Intranasal versus intramuscular vaccination; Monophosphoryl lipid A; SARS-CoV-2 spike protein.

PubMed Disclaimer

Conflict of interest statement

There are no conflicts of interest to disclose.

Figures

Unlabelled Image
Graphical abstract
Fig. 1
Fig. 1
TLR and RIG-I signaling pathways intersect and can be activated by nanoparticles with encapsulated and surface-loaded pathogen-associated molecular patterns. A) Schematic of signaling pathways initiated by TLRs 1–9, and RIG-I receptors including adaptor proteins MyD88, TRIF, TRAF3, and MAVS and transcription factors NF-κB and various IRFs which regulate the transcription of proinflammatory genes. B) Left: Depiction of poly(lactic-co-glycolic acid)-polyethyleneimine nanoparticles (PLGA-PEI NPs) with encapsulated hydrophobic molecules (R848 or MPLA) and surface-loaded nucleic acids (CpG or PUUC) for adjuvant delivery during vaccination. Right: PLGA-PEI NPs with surface-loaded SARS-CoV-2 spike protein for antigen delivery during vaccination.
Fig. 2
Fig. 2
GM-CSF and FLT3L BMDCs secrete different cytokine profiles and activate CD8+ T cells in response to S1 protein with combination adjuvants. A) t-SNE plots of GM-CSF and FLT3L BMDCs with labeled clusters of APC subsets. Macrophages/Mo-DCs are CD64+ F4/80, Conventional DCs are CD11c+ MHCII+ CD64lo F4/80lo, Monocytes are Ly6Chi CD11c, Neutrophils are Ly6Ghi Ly6C+, plasmacytoid DCs are B220+ Ly6C+ CD11c+ MHCIIlo. BG) Cytokine concentrations (pg/mL) of IL-1β, IL-27, IL-12p70, IFN-β, and IFN-λ3 in supernatants of BMDC culture after incubation with adjuvanted NPs for 24 h. HI) Percentage of live CD3+CD4+ T cells or CD3+CD8+ T cells proliferating in presence of GM-CSF BMDCs, gated on diminished CFSE signal (Fig. S1). “No Adjuvant” condition is blank NPs. J, K) Percentage of live CD3+CD4+ T cells or CD3+CD8+ T cells proliferating in presence of FLT3L BMDCs. *p < 0.05. **p < 0.01, ***p < 0.001, ****p < 0.0001 based on a One-Way ANOVA and Tukey Test for multiple comparison.
Fig. 3
Fig. 3
MPLA+PUUC NPs increase T cell responses in the lung when delivered intranasally with spike protein. On days 0 (1st dose) and 28 (2nd dose), female BALB/c mice were immunized I.N. with unformulated (i.e., soluble) or NP-conjugated spike protein (1 μg) and PLGA-PEI NPs (4 mg) loaded with MPLA (24 μg), PUUC (17 μg), and MPLA+PUUC (20 μg, 17 μg). Mice were euthanized and lungs were collected on day 35, one week after the 2nd dose. Lung cells were restimulated with spike peptide pools for 6 h and stained for analysis by flow cytometry. Percentages of cells expressing A) CD4+CD44+ out of CD45+ cells, B) IFNγ+ out of CD4+CD44+ cells, C) TNFα+ out of CD4+CD44+ cells, D) CD69+CD103+ (tissue resident memory T cells) out of CD45+ cells. BAL fluid from vaccinated mice using soluble spike antigen was assayed for anti-spike E) IgG and F) IgA with ELISA. G) Sera were assayed for anti-spike IgG with ELISA (error bars represent the SEM). p values are *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001 calculated using AD) One-way ANOVA with Tukey post-hoc test, E, F) Kruskal-Wallis with Dunn's post-hoc test for nonparametric data, or G) Two-way ANOVA with Tukey post-hoc test.
Fig. 4
Fig. 4
PUUC NPs delivered intramuscularly with spike protein enhance humoral responses. Female BALB/c mice were immunized I.M. into both tibialis anterior muscles at day 0 (1st dose) with soluble spike protein at doses of 80 ng, 200 ng, 1000 ng with or without adjuvant-NPs (4 mg) loaded with PUUC (+P, 20 ng PUUC dose). Peripheral blood was sampled on day 26. On day 28, mice received a 2nd dose of protein subunit vaccines. Mice received the same formulations, except for two groups that received 80 ng spike protein as a 1st dose received 1000 ng spike protein as a 2nd antigen dose (80/1000 and 80/1000 +P). Mice were euthanized on day 36 for to collect blood and popliteal LNs. A) Anti-spike IgG in post-1st dose sera at various dilutions measured by absorbance at 450 nm during ELISA assays and B) comparison of area under the curve (AUC). CD) Anti-spike IgG in post-2nd dose sera measured by absorbance at 450 nm and comparison of AUC. E) ACE-2 signal measured by absorbance at 450 nm in spike protein neutralization assay with post-2nd dose sera. Absorbance was normalized to a blank well in each row of a 384 well plate to correct for plate effects. Lower absorbance values indicate higher spike-neutralizing antibody levels in sera. Percentages of cells expressing F) Bcl6+ out of B220+ cells, G) GL7+ out of B220+ cells and H) CXCR5+ out of B220 cells from combined popliteal lymph nodes. B,D) Normality was assessed with the Kolmogorov-Smirnov test. Statistical significance was determined with the Kruskal-Wallis test and Dunn's post-hoc test for multiple comparisons. EH) Statistical significance calculated with One-Way ANOVA and Tukey post-hoc test. *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001 for all graphs.
Fig. 5
Fig. 5
Key takeaways from intranasal and intramuscular two-dose vaccination models. Top: MPLA-PUUC NPs administered with spike-NPs intranasally induce local T cell responses, characterized by increases in IFNγ+ and TNFα+ CD4 T cells and tissue resident memory cells after lung cell restimulation with spike peptide pools. Middle: MPLA NPs administered with soluble spike protein intranasally induce systemic and localized humoral responses, characterized by increases in serum and BAL fluid antibodies. Bottom: PUUC NPs injected with soluble spike protein intramuscularly induce systemic humoral responses, characterized by increases in serum antibodies and germinal center B cells in the popliteal lymph nodes.

Update of

Similar articles

Cited by

References

    1. R&D Blue Print . 2021. COVID-19 Vaccine Tracker and Landscape.
    1. Awate S., Babiuk L.A.B., Mutwiri G. Mechanisms of action of adjuvants. Front. Immunol. 2013;4:114. - PMC - PubMed
    1. Reed S.G., Orr M.T., Fox C.B. Key roles of adjuvants in modern vaccines. Nat. Med. 2013;19:1597–1608. - PubMed
    1. Saravia J., Chapman N.M., Chi H. Helper T cell differentiation. Cell. Mol. Immunol. 2019;16:634–643. - PMC - PubMed
    1. Maddur M.S., Bayry J. B cells drive Th2 responses by instructing human dendritic cell maturation. Oncoimmunology. 2015;4 e1005508. - PMC - PubMed

Publication types