scispace - formally typeset
Search or ask a question
Posted ContentDOI

Alum:CpG adjuvant enables SARS-CoV-2 RBD-induced protection in aged mice and synergistic activation of human elder type 1 immunity

TL;DR: In this paper, the aluminum hydroxide (AH) and CpG adjuvant formulation (AH:CpG) was used to enhance the anti-RBD immunogenicity of the SARS-CoV-2 vaccine.
Abstract: Global deployment of vaccines that can provide protection across several age groups is still urgently needed to end the COVID-19 pandemic especially for low- and middle-income countries. While vaccines against SARS-CoV-2 based on mRNA and adenoviral-vector technologies have been rapidly developed, additional practical and scalable SARS-CoV-2 vaccines are needed to meet global demand. In this context, protein subunit vaccines formulated with appropriate adjuvants represent a promising approach to address this urgent need. Receptor-binding domain (RBD) is a key target of neutralizing antibodies (Abs) but is poorly immunogenic. We therefore compared pattern recognition receptor (PRR) agonists, including those activating STING, TLR3, TLR4 and TLR9, alone or formulated with aluminum hydroxide (AH), and benchmarked them to AS01B and AS03-like emulsion-based adjuvants for their potential to enhance RBD immunogenicity in young and aged mice. We found that the AH and CpG adjuvant formulation (AH:CpG) demonstrated the highest enhancement of anti-RBD neutralizing Ab titers in both age groups (∼80-fold over AH), and protected aged mice from the SARS-CoV-2 challenge. Notably, AH:CpG-adjuvanted RBD vaccine elicited neutralizing Abs against both wild-type SARS-CoV-2 and B.1.351 variant at serum concentrations comparable to those induced by the authorized mRNA BNT162b2 vaccine. AH:CpG induced similar cytokine and chemokine gene enrichment patterns in the draining lymph nodes of both young adult and aged mice and synergistically enhanced cytokine and chemokine production in human young adult and elderly mononuclear cells. These data support further development of AH:CpG-adjuvanted RBD as an affordable vaccine that may be effective across multiple age groups. One Sentence Summary Alum and CpG enhance SARS-CoV-2 RBD protective immunity, variant neutralization in aged mice and Th1-polarizing cytokine production by human elder leukocytes.

Summary (3 min read)

INTRODUCTION

  • The coronavirus disease 2019 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) resulted in a serious threat to humanity.
  • To this end, alternative platforms such as inactivated and protein subunit SARS-CoV-2 vaccines have entered different stages of clinical development and in some cases have already been deployed at the population level (11) (12) (13) (14) (15) (16) (17) .
  • Adjuvant formulations of aluminum salts and PRR agonists enhance vaccine immune responses compared to aluminum salts or PRR agonists alone (32) .

AH:CpG-formulated RBD vaccine is immunogenic in aged mice

  • To assess the vaccine response in the context of aging, the immunogenicity of RBD vaccines adjuvanted with AH:PRR agonists was further studied in aged mice (14-month-old) .
  • Similar to young mice, the AH:CpG formulation also elicited the highest humoral immune response after prime-boost immunization in aged mice (Fig 2A-F ).
  • Of note, the vaccine adjuvanted with AH:CpG produced significantly higher hACE2/RBD inhibition and neutralizing titers compared to the vaccine adjuvanted with AS01B, which is known as a potent adjuvant in the human elderly population (33, 34) (.

AH:CpG-formulated RBD vaccine protects aged mice from lethal viral challenge

  • Abs are key to protecting from SARS-CoV-2 infection.
  • Since RBD formulated with AH:CpG elicited high titers of neutralizing Abs, the authors assessed the protection of immunized mice in a challenge model.
  • To this end, the authors employed the mouse-adapted SARS-CoV-2 MA10 virus strain (35) .

AH:CpG-formulated RBD and Spike mRNA vaccines elicit comparable levels of neutralizing antibodies against wild type SARS-CoV-2 and variants

  • Recently, it has been reported that SARS-CoV-2 mRNA vaccines are more immunogenic than RBD adjuvanted with oil-in-water emulsions (36) .
  • Along with CpG-2395, the authors also tested CpG-1018, which is included in the Heplisav-B vaccine and has also been tested in combination with Spike/RBD and AH in SARS-CoV-2 studies including human vaccine trials (12, 16, 37) .
  • In accordance with previously published data, the mRNA vaccine was highly immunogenic, while RBD formulated with AddaS03 failed to induce (38) (39) (40) (41) .
  • A recent report showed that the mRNA BNT162b2 vaccine maintained its effectiveness against severe COVID-19 with the B.1.351 variant at greater than 90% (42) .
  • As expected, the authors observed reduced titers against the variants, especially against the B.1.351 (Fig.

Innate signaling potentiated by AH:CpG formulation is well preserved in aged mice

  • Lymph nodes (LNs) are critical sites for the interaction between innate and adaptive immune systems and orchestrate the development of vaccine immune responses (43, 44) .
  • To gain further insights into the mechanism of action of the AH:CpG formulation, the authors collected draining LNs (dLNs) 24 hours post injection of AH:CpG or either adjuvant alone.
  • CpG induced comparable dLN expansion in both age groups (Fig, also known as CpG and AH.

AH:CpG synergistically enhances proinflammatory cytokines from human elderly PBMCs

  • To this end, the authors stimulated human peripheral blood mononuclear cells isolated from young adults was not certified by peer review) is the author/funder.
  • The copyright holder for this preprint (which this version posted May 21, 2021.

DISCUSSION

  • The risk of COVID-19-related morbidity and mortality increases with age (47, 48) .
  • Of note, RBD adjuvanted with AH:CpG elicited levels of neutralizing Abs comparable to the clinical-grade BNT162b2 Spike mRNA vaccine.
  • Was not certified by peer review) is the author/funder.

MATERIALS AND METHODS

  • The aim of this study was to assess optimal combinations of RBD antigen and adjuvants in pre-clinical models that take age-dependent vaccine immune responses and COVID-19 susceptibility into account.
  • To this end, the authors used age-specific mouse in vivo and human in vitro models.
  • Affinity tags were cleaved off from eluted protein samples by HRV 3C protease, and tag removed proteins were further purified by size-exclusion chromatography using a Superose 6 10/300 column for full length Spike and a Superdex 75 10/300 Increase column for RBD domain in a PBS buffer (pH 7.4).
  • Each PRR agonist was formulated with and without aluminum hydroxide.
  • Blood samples were collected 2 weeks post-immunization.

Adjuvants and immunization. The

  • Briefly, high-binding flat-bottom 96-well plates (Corning, NY) were coated with 50 ng/well RBD or 25 ng/well Spike and incubated overnight at 4 °C.
  • Plates were washed three times and incubated for 1 hour at RT with HRP-conjugated anti-mouse IgG, IgG1, IgG2a, or IgG2c (Southern Biotech).
  • The copyright holder for this preprint (which this version posted May 21, 2021.
  • Each serum sample was diluted 1:160, pre-incubated with 3 ng of RBD-Fc in 1% BSA PBS for 1 hour at RT, and then transferred to the hACE2-coated plate.
  • The optical density was read at 450 nm with SpectraMax iD3 microplate reader (Molecular Devices).

SARS-CoV-2 neutralization titer determination.

  • All serum samples were heat-inactivated at 56°C for 30 min to remove complement and allowed to equilibrate to RT prior to processing for neutralization titer.
  • Samples were diluted in duplicate to an initial dilution of 1:5 or 1:10 followed by 1:2 serial dilutions (vaccinated sample), resulting in a 12-dilution series with each was not certified by peer review) is the author/funder.
  • The copyright holder for this preprint (which this version posted May 21, 2021.
  • BioRxiv preprint well containing 100 µL. Dilution plates were then transported into the BSL-3 laboratory and 100 µL of diluted SARS-CoV-2 (WA-1, courtesy of Dr. Natalie Thornburg/CDC) inoculum was added to each well to result in a multiplicity of infection (MOI) of 0.01 upon transfer to titering plates, also known as /10.1101/2021.05.20.444848 doi.

Pseudovirus neutralization assay.

  • The SARS-CoV-2 pseudoviruses expressing a luciferase reporter gene were generated in an approach similar to as described previously (75, 76) .
  • To determine the neutralization was not certified by peer review) is the author/funder.
  • The copyright holder for this preprint (which this version posted May 21, 2021.
  • Three-fold serial dilutions of heat inactivated serum or plasma samples were prepared and mixed with 50 µL of pseudovirus.

Splenocyte restimulation assay.

  • Immunized mice were sacrificed 2 weeks after the final immunization, and spleens were collected.
  • To isolate splenocytes, spleens were mashed through a 70 µm cell strainer, and the resulting cell suspensions were washed with PBS and incubated with 2 mL of ACK lysis buffer for 2 minutes at RT to lyse erythrocytes.
  • Splenocytes were washed again with PBS and plated in flat-bottom 96-well plates (2 x 10 6 cells per well).
  • Then, SARS-CoV-2 Spike peptides (PepTivator SARS-CoV-2 Prot_S, Miltenyi Biotec) were added at a final concentration of 0.6 nmol/ml in the presence of 1 μg/mL anti-CD28 antibody (total cell culture volume, 200 µL per well).
  • After 24 (for IL-2 and IL-4) and 96 (for IFNγ) hours, supernatants were harvested, and cytokine levels were measured by ELISA according to the manufacturer's protocol.

SARS-CoV-2 mouse challenge study. Mice were anesthetized by intraperitoneal injection 50

  • Mice were then intranasally inoculated with 1 x 10 3 PFU of mouse-adapted SARS-CoV-2 (MA10, was not certified by peer review) is the author/funder.
  • The copyright holder for this preprint (which this version posted May 21, 2021.
  • SARS-CoV-2 lung titers were quantified by homogenizing harvested lungs in PBS (Quality Biological Inc.) using 1.0 mm glass beads (Sigma Aldrich) and a Beadruptor (Omni International Inc.).
  • A pathologist was blinded to information identifying the treatment groups and fields were examined by light microscopy and analyzed.
  • The severity of interstitial inflammation was evaluated and converted to a score of 0-4 with 0 being no inflammation and 4 being most severe.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

1
Title: Alum:CpG adjuvant enables SARS-CoV-2 RBD-induced protection in aged mice and 1
synergistic activation of human elder type 1 immunity 2
3
One Sentence Summary: Alum and CpG enhance SARS-CoV-2 RBD protective immunity, 4
variant neutralization in aged mice and Th1-polarizing cytokine production by human elder 5
leukocytes. 6
7
Authors: Etsuro Nanishi
1, 2†
, Francesco Borriello
1, 2, 3
, Timothy R. O’Meara
1‡
, Marisa E. 8
McGrath
4‡
, Yoshine Saito
1
, Robert E. Haupt
4
, Hyuk-Soo Seo
5, 6
, Simon D. van Haren
1, 2
, Byron 9
Brook
1, 2
, Jing Chen
7
, Joann Diray-Arce
1, 2
, Simon Doss-Gollin
1
, Maria De Leon
1
, Katherine 10
Chew
1
, Manisha Menon
1
, Kijun Song
5
, Andrew Z. Xu
5
, Timothy M. Caradonna
8
, Jared 11
Feldman
8
, Blake M. Hauser
8
, Aaron G. Schmidt
8, 9
, Amy C. Sherman
1, 10
, Lindsey R. Baden
10
, 12
Robert K. Ernst
11
, Carly Dillen
4
, Stuart M. Weston
4
, Robert M. Johnson
4
, Holly L. Hammond
4
, 13
Romana Mayer
12
, Allen Burke
12
,
Maria E. Bottazzi
13, 14
, Peter J. Hotez
13, 14
, Ulrich Strych
13, 15
, 14
Aiquan Chang
16
, Jingyou Yu
16
, Dan H. Barouch
16
, Sirano Dhe-Paganon
5, 6
, Ivan Zanoni
2, 3
, Al 15
Ozonoff
1, 2
,
Matthew B. Frieman
, Ofer Levy
1, 2, 17§
, David J. Dowling
1, 2§
* 16
17
Affiliations: 18
1
Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, 19
Boston, MA, USA. 20
2
Department of Pediatrics, Harvard Medical School, Boston, MA, USA. 21
3
Division of Immunology, Boston Children’s Hospital, Boston, MA, USA. 22
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted May 21, 2021. ; https://doi.org/10.1101/2021.05.20.444848doi: bioRxiv preprint

2
4
Department of Microbiology and Immunology, University of Maryland School of Medicine, 23
Baltimore, MD, USA. 24
5
Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA. 25
6
Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 26
Boston, MA, USA. 27
7
Research Computing Group, Boston Children’s Hospital, Boston, MA, USA. 28
8
Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA. 29
9
Department of Microbiology, Harvard Medical School, Boston, MA, USA. 30
10
Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA. 31
11
Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, 32
MD, USA. 33
12
Department of Pathology, University of Maryland Medical Center, Baltimore, MD, USA. 34
13
Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, 35
Houston, TX, USA.
36
14
National School of Tropical Medicine and Departments of Pediatrics and Molecular Virology 37
& Microbiology, Baylor College of Medicine, Houston, TX, USA. 38
15
National School of Tropical Medicine and Department of Pediatrics, Baylor College of 39
Medicine, Houston, TX, USA. 40
16
Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard 41
Medical School, Boston, MA, USA. 42
17
Broad Institute of MIT & Harvard, Cambridge, MA, USA.
43
44
These authors contributed equally to this manuscript. 45
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted May 21, 2021. ; https://doi.org/10.1101/2021.05.20.444848doi: bioRxiv preprint

3
These authors contributed equally to this manuscript. 46
§
Co-senior authors. 47
48
*Corresponding author: David J. Dowling, Precision Vaccines Program, Division of 49
Infectious Diseases, Boston Children’s Hospital; Harvard Medical School, Rm 842, Boston, MA 50
02115, USA. Tel: +1 617-919-6890. e-mail: david.dowling@childrens.harvard.edu 51
52
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted May 21, 2021. ; https://doi.org/10.1101/2021.05.20.444848doi: bioRxiv preprint

4
ABSTRACT 53
Global deployment of vaccines that can provide protection across several age groups is still 54
urgently needed to end the COVID-19 pandemic especially for low- and middle-income 55
countries. While vaccines against SARS-CoV-2 based on mRNA and adenoviral-vector 56
technologies have been rapidly developed, additional practical and scalable SARS-CoV-2 57
vaccines are needed to meet global demand. In this context, protein subunit vaccines formulated 58
with appropriate adjuvants represent a promising approach to address this urgent need. Receptor-59
binding domain (RBD) is a key target of neutralizing antibodies (Abs) but is poorly 60
immunogenic. We therefore compared pattern recognition receptor (PRR) agonists, including 61
those activating STING, TLR3, TLR4 and TLR9, alone or formulated with aluminum hydroxide 62
(AH), and benchmarked them to AS01B and AS03-like emulsion-based adjuvants for their 63
potential to enhance RBD immunogenicity in young and aged mice. We found that the AH and 64
CpG adjuvant formulation (AH:CpG) demonstrated the highest enhancement of anti-RBD 65
neutralizing Ab titers in both age groups (~80-fold over AH), and protected aged mice from the 66
SARS-CoV-2 challenge. Notably, AH:CpG-adjuvanted RBD vaccine elicited neutralizing Abs 67
against both wild-type SARS-CoV-2 and B.1.351 variant at serum concentrations comparable to 68
those induced by the authorized mRNA BNT162b2 vaccine. AH:CpG induced similar cytokine 69
and chemokine gene enrichment patterns in the draining lymph nodes of both young adult and 70
aged mice and synergistically enhanced cytokine and chemokine production in human young 71
adult and elderly mononuclear cells. These data support further development of AH:CpG-72
adjuvanted RBD as an affordable vaccine that may be effective across multiple age groups. 73
74
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted May 21, 2021. ; https://doi.org/10.1101/2021.05.20.444848doi: bioRxiv preprint

5
INTRODUCTION 75
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory 76
syndrome coronavirus 2 (SARS-CoV-2) resulted in a serious threat to humanity. Rapid 77
deployment of safe and effective vaccines is proving key to reducing morbidity and mortality of 78
COVID-19, especially in high-risk populations such as the older adults (1). Novel vaccine 79
technologies including mRNA and adenoviral vector vaccines have dramatically accelerated the 80
process of vaccine development, shown high efficacy in preclinical and clinical studies, and 81
therefore been granted Emergency Use Authorization by the Food and Drug Administration (2-9). 82
Unfortunately, worldwide access to these vaccines may be limited by the need for ultra-cold 83
storage (mRNA vaccines), cost, and concerns regarding global scalability especially in the third 84
world (1). This situation not only represents a major ethical problem but may also promote the 85
emergence of vaccine-resistant SARS-CoV-2 strains due to high infection rates in unvaccinated 86
regions (10). Thus, ongoing efforts are needed to investigate additional affordable, easily 87
scalable, and effective vaccine approaches against SARS-CoV-2 to improve global access. To 88
this end, alternative platforms such as inactivated and protein subunit SARS-CoV-2 vaccines 89
have entered different stages of clinical development and in some cases have already been 90
deployed at the population level (11-17). These approaches may play an essential role in the 91
global fight against COVID-19 since they utilize well-established technologies, do not require 92
low temperature storage, and have proven safety and effectiveness in various age groups 93
including young children and the elderly. 94
95
With the exception of inactivated viruses, most SARS-CoV-2 vaccine candidates aim to target 96
the SARS-CoV-2 Spike glycoprotein, as it is required for binding to the human receptor 97
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted May 21, 2021. ; https://doi.org/10.1101/2021.05.20.444848doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI
TL;DR: Results show that the vaccine formulation composed of U-Omp19 and alum as adjuvants has a better performance: it significantly increased mucosal and systemic neutralizing antibodies in comparison to antigen plus alum, AddaVax, or AddaS03.
Abstract: In this work, we evaluated recombinant receptor binding domain (RBD)-based vaccine formulation prototypes with potential for further clinical development. We assessed different formulations containing RBD plus alum, AddaS03, AddaVax, or the combination of alum and U-Omp19: a novel Brucella spp. protease inhibitor vaccine adjuvant. Results show that the vaccine formulation composed of U-Omp19 and alum as adjuvants has a better performance: it significantly increased mucosal and systemic neutralizing antibodies in comparison to antigen plus alum, AddaVax, or AddaS03. Antibodies induced with the formulation containing U-Omp19 and alum not only increased their neutralization capacity against the ancestral virus but also cross-neutralized alpha, lambda, and gamma variants with similar potency. Furthermore, the addition of U-Omp19 to alum vaccine formulation increased the frequency of RBD-specific geminal center B cells and plasmablasts. Additionally, U-Omp19+alum formulation induced RBD-specific Th1 and CD8+ T-cell responses in spleens and lungs. Finally, this vaccine formulation conferred protection against an intranasal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge of K18-hACE2 mice.

10 citations

DOI
05 Nov 2021
TL;DR: In this article, the authors described the production of insect-cell expressed SARS-CoV-2 spike protein ectodomain constructs and examined their immunogenicity in mice, and demonstrated that, when formulated with CoVaccine HTTM adjuvant, an oil-in-water nanoemulsion compatible with lyophilization, their vaccine candidates elicit a broad-spectrum IgG response, high neutralizing antibody (NtAb) titers against SARS CoV2 prototype and variants of concern, specifically B.1.351 (Beta) and
Abstract: The speed at which several COVID-19 vaccines went from conception to receiving FDA and EMA approval for emergency use is an achievement unrivaled in the history of vaccine development. Mass vaccination efforts using the highly effective vaccines are currently underway to generate sufficient herd immunity and reduce transmission of the SARS-CoV-2 virus. Despite the most advanced vaccine technology, global recipient coverage, especially in resource-poor areas remains a challenge as genetic drift in naive population pockets threatens overall vaccine efficacy. In this study, we described the production of insect-cell expressed SARS-CoV-2 spike protein ectodomain constructs and examined their immunogenicity in mice. We demonstrated that, when formulated with CoVaccine HTTM adjuvant, an oil-in-water nanoemulsion compatible with lyophilization, our vaccine candidates elicit a broad-spectrum IgG response, high neutralizing antibody (NtAb) titers against SARS-CoV-2 prototype and variants of concern, specifically B.1.351 (Beta) and P.1. (Gamma), and an antigen-specific IFN-γ secreting response in outbred mice. Of note, different ectodomain constructs yielded variations in NtAb titers against the prototype strain and some VOC. Dose response experiments indicated that NtAb titers increased with antigen dose, but not adjuvant dose, and may be higher with a lower adjuvant dose. Our findings lay the immunological foundation for the development of a dry-thermostabilized vaccine that is deployable without refrigeration.

10 citations

Posted ContentDOI
11 Oct 2021-bioRxiv
TL;DR: In this article, a vaccination strategy using a combination of Alum and polyinosinic:polycytidylic acid (Poly(I:C)) adjuvants plus the SARS-CoV-2 spike protein in a prefusion trimeric conformation by an intradermal (ID) route was assessed.
Abstract: The SARS-CoV-2 pandemic has had a social and economic impact worldwide, and vaccination is an efficient strategy for diminishing those damages. New adjuvant formulations are required for the high vaccine demands, especially adjuvant formulations that induce a Th1 phenotype. Herein we assess a vaccination strategy using a combination of Alum and polyinosinic:polycytidylic acid (Poly(I:C)) adjuvants plus the SARS-CoV-2 spike protein in a prefusion trimeric conformation by an intradermal (ID) route. We found high levels of IgG anti-spike antibodies in the serum by enzyme linked immunosorbent assay (ELISA) and high neutralizing titers against SARS-CoV-2 in vitro by neutralization assay, after one or two boosts. By evaluating the production of IgG subtypes, as expected, we found that formulations containing Poly(I:C) induced IgG2a whereas Alum did not. The combination of these two adjuvants induced high levels of both IgG1 and IgG2a. In addition, cellular immune responses of CD4+ and CD8+ T cells producing interferon-gamma were equivalent, demonstrating that the Alum + Poly(I:C) combination supported a Th1 profile. Based on the high neutralizing titers, we evaluated B cells in the germinal centers, which are specific for receptor-binding domain (RBD) and spike, and observed that more positive B cells were induced upon the Alum + Poly(I:C) combination. Moreover, these B cells produced antibodies against both RBD and non-RBD sites. We also studied the impact of this vaccination preparation (spike protein with Alum + Poly(I:C)) in the lungs of mice challenged with inactivated SARS-CoV-2 virus. We found a production of IgG, but not IgA, and a reduction in neutrophil recruitment in the bronchoalveolar lavage fluid (BALF) of mice, suggesting that our immunization scheme reduced lung inflammation. Altogether, our data suggest that Alum and Poly(I:C) together is a possible adjuvant combination for vaccines against SARS-CoV-2 by the intradermal route.

2 citations

Posted ContentDOI
06 Oct 2021-bioRxiv
TL;DR: In this article, a vaccination strategy using a combination of Alum and polyinosinic:polycytidylic acid (Poly(I:C)) adjuvants plus the SARS-CoV-2 spike protein in a prefusion trimeric conformation by an intradermal (ID) route was assessed.
Abstract: The SARS-CoV-2 pandemic has had a social and economic impact worldwide, and vaccination is an efficient strategy for diminishing those damages. New adjuvant formulations are required for the high vaccine demands, especially adjuvant formulations that induce a Th1 phenotype. Herein we assess a vaccination strategy using a combination of Alum and polyinosinic:polycytidylic acid (Poly(I:C)) adjuvants plus the SARS-CoV-2 spike protein in a prefusion trimeric conformation by an intradermal (ID) route. We found high levels of IgG anti-spike antibodies in the serum by enzyme linked immunosorbent assay (ELISA) and high neutralizing titers against SARS-CoV-2 in vitro by neutralization assay, after one or two boosts. By evaluating the production of IgG subtypes, as expected, we found that formulations containing Poly(I:C) induced IgG2a whereas Alum did not. The combination of these two adjuvants induced high levels of both IgG1 and IgG2a. In addition, cellular immune responses of CD4+ and CD8+ T cells producing interferon-gamma were equivalent, demonstrating that the Alum + Poly(I:C) combination supported a Th1 profile. Based on the high neutralizing titers, we evaluated B cells in the germinal centers, which are specific for receptor-binding domain (RBD) and spike, and observed that more positive B cells were induced upon the Alum + Poly(I:C) combination. Moreover, these B cells produced antibodies against both RBD and non-RBD sites. We also studied the impact of this vaccination preparation (spike protein with Alum + Poly(I:C)) in the lungs of mice challenged with inactivated SARS-CoV-2 virus. We found a production of IgG, but not IgA, and a reduction in neutrophil recruitment in the bronchoalveolar lavage fluid (BALF) of mice, suggesting that our immunization scheme reduced lung inflammation. Altogether, our data suggest that Alum and Poly(I:C) together is a possible adjuvant combination for vaccines against SARS-CoV-2 by the intradermal route.

2 citations

References
More filters
Journal ArticleDOI
TL;DR: A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older and safety over a median of 2 months was similar to that of other viral vaccines.
Abstract: Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a world...

10,274 citations

Journal ArticleDOI
08 Jul 2020-Nature
TL;DR: A range of clinical factors associated with COVID-19-related death is quantified in one of the largest cohort studies on this topic so far and includes people of white ethnicity, Black and South Asian people were at higher risk, even after adjustment for other factors.
Abstract: Coronavirus disease 2019 (COVID-19) has rapidly affected mortality worldwide1. There is unprecedented urgency to understand who is most at risk of severe outcomes, and this requires new approaches for the timely analysis of large datasets. Working on behalf of NHS England, we created OpenSAFELY-a secure health analytics platform that covers 40% of all patients in England and holds patient data within the existing data centre of a major vendor of primary care electronic health records. Here we used OpenSAFELY to examine factors associated with COVID-19-related death. Primary care records of 17,278,392 adults were pseudonymously linked to 10,926 COVID-19-related deaths. COVID-19-related death was associated with: being male (hazard ratio (HR) 1.59 (95% confidence interval 1.53-1.65)); greater age and deprivation (both with a strong gradient); diabetes; severe asthma; and various other medical conditions. Compared with people of white ethnicity, Black and South Asian people were at higher risk, even after adjustment for other factors (HR 1.48 (1.29-1.69) and 1.45 (1.32-1.58), respectively). We have quantified a range of clinical factors associated with COVID-19-related death in one of the largest cohort studies on this topic so far. More patient records are rapidly being added to OpenSAFELY, we will update and extend our results regularly.

4,263 citations

Journal ArticleDOI
TL;DR: The mRNA-1273 vaccine as discussed by the authors is a lipid nanoparticle-encapsulated mRNA-based vaccine that encodes the prefusion stabilized full-length spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes Covid-19.
Abstract: Background Vaccines are needed to prevent coronavirus disease 2019 (Covid-19) and to protect persons who are at high risk for complications. The mRNA-1273 vaccine is a lipid nanoparticle-encapsulated mRNA-based vaccine that encodes the prefusion stabilized full-length spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes Covid-19. Methods This phase 3 randomized, observer-blinded, placebo-controlled trial was conducted at 99 centers across the United States. Persons at high risk for SARS-CoV-2 infection or its complications were randomly assigned in a 1:1 ratio to receive two intramuscular injections of mRNA-1273 (100 μg) or placebo 28 days apart. The primary end point was prevention of Covid-19 illness with onset at least 14 days after the second injection in participants who had not previously been infected with SARS-CoV-2. Results The trial enrolled 30,420 volunteers who were randomly assigned in a 1:1 ratio to receive either vaccine or placebo (15,210 participants in each group). More than 96% of participants received both injections, and 2.2% had evidence (serologic, virologic, or both) of SARS-CoV-2 infection at baseline. Symptomatic Covid-19 illness was confirmed in 185 participants in the placebo group (56.5 per 1000 person-years; 95% confidence interval [CI], 48.7 to 65.3) and in 11 participants in the mRNA-1273 group (3.3 per 1000 person-years; 95% CI, 1.7 to 6.0); vaccine efficacy was 94.1% (95% CI, 89.3 to 96.8%; P Conclusions The mRNA-1273 vaccine showed 94.1% efficacy at preventing Covid-19 illness, including severe disease. Aside from transient local and systemic reactions, no safety concerns were identified. (Funded by the Biomedical Advanced Research and Development Authority and the National Institute of Allergy and Infectious Diseases; COVE ClinicalTrials.gov number, NCT04470427.).

2,721 citations

Journal ArticleDOI
TL;DR: The HPV- 16/18 AS04-adjuvanted vaccine showed high efficacy against CIN2+ associated with HPV-16/18 and non-vaccine oncogenic HPV types and substantial overall effect in cohorts that are relevant to universal mass vaccination and catch-up programmes.

1,569 citations

Journal ArticleDOI
TL;DR: These emerging principles of innate control of adaptive immunity are discussed, which are variations on a common design principle wherein the cells that sense infections produce one set of cytokines to induce lymphocytes to produce another set ofinflammatory cytokines, which in turn activate effector responses.
Abstract: Microbial infections are recognized by the innate immune system both to elicit immediate defense and to generate long-lasting adaptive immunity. To detect and respond to vastly different groups of pathogens, the innate immune system uses several recognition systems that rely on sensing common structural and functional features associated with different classes of microorganisms. These recognition systems determine microbial location, viability, replication and pathogenicity. Detection of these features by recognition pathways of the innate immune system is translated into different classes of effector responses though specialized populations of dendritic cells. Multiple mechanisms for the induction of immune responses are variations on a common design principle wherein the cells that sense infections produce one set of cytokines to induce lymphocytes to produce another set of cytokines, which in turn activate effector responses. Here we discuss these emerging principles of innate control of adaptive immunity.

1,481 citations

Related Papers (5)
Frequently Asked Questions (6)
Q1. What was the RNA used to determine the RNA content of dLNs?

(B–E) RNA isolated from dLNs was subjected to a quantitative real-1116 time PCR array comprised of 157 genes related to cytokines, chemokines, and type 1 IFN 1117 responses. 

7. AH:CpG synergistically enhances proinflammatory cytokine production from 1130 human adult and elderly PBMCs 1131 Human PBMCs collected from young adult (A, C) and elderly individuals (B, D) were cultured 1132 in vitro for 24 h with CpG alone (4, 10, 20, 40, and 100 μg/mL), aluminum hydroxide (AH) 1133 alone (8, 20, 40, 80, and 200 μg/mL), or a combination of both. 

G. Sen, Q. Chen, C. M. Snapper, Immunization of aged mice with a pneumococcal 906 conjugate vaccine combined with an unmethylated CpG-containing oligodeoxynucleotide 907 restores defective immunoglobulin G antipolysaccharide responses and specific CD4+-T-908 cell priming to young adult levels. 

(A–F) Serum samples were 1039 collected on Day 28, and (A) Anti-RBD IgG, (B) IgG1, (C) IgG2a, (D) IgG2a/IgG1 ratio, (E) 1040 hACE2/RBD inhibition rate, and (F) anti-Spike IgG were assessed. 

(E) Enrichment analysis of differentially expressed genes using the blood transcriptional 1126 modules (Li et al., 2013- PMC: 24336226) was performed from the significant gene results after 1127 the generalized linear model by treatment. 

Level of 1139 synergy was calculated using an adapted Loewe definition of additivity (D <1: synergy, D=1: 1140 additivity, D >1: antagonism).