ArticlePDF Available

SARS-CoV-2 mutations, vaccines, and immunity: implication of variants of concern

Authors:
RESEARCH HIGHLIGHT OPEN
SARS-CoV-2 mutations, vaccines, and immunity: implication
of variants of concern
Ji Yun Noh
1,2
, Hye Won Jeong
2,3
and Eui-Cheol Shin
2,4
Signal Transduction and Targeted Therapy (2021) 6:203 ; https://doi.org/10.1038/s41392-021-00623-2
In a recent study published in Nature, Wang et al.
1
investigated the
neutralizing activities of antibodies elicited by COVID-19 mRNA
vaccines and natural infection with SARS-CoV-2 against SARS-CoV-
2 variants.
The devastating impact of the ongoing COVID-19 pandemic on
public health, the economy, and society has made vaccine
development a top priority for global health. Thus, vaccine
development is progressing at an unprecedented pace as an
urgent response to COVID-19.
Currently, there are four main types of COVID-19 vaccine:
nucleic acid (mRNA and DNA), viral vector, protein subunit, and
inactivated virus. Two COVID-19 mRNA vaccines (BNT162b2
developed by Pzer-BioNTech and mRNA-1273 by Moderna) have
been authorized by the U.S. Food and Drug Administration (FDA)
and European Medicines Agency (EMA). In addition, Ad26.COV2.S
(Johnson & Johnson/Janssen) was approved by the FDA and EMA
and ChAdOx1 nCoV-19 (AstraZeneca) was authorized by the EMA,
both of which are viral vector vaccines.
BNT162b2 and mRNA-1273 are lipid nanoparticle-formulated,
nucleoside-modied RNA vaccines encoding the prefusion spike
glycoprotein of SARS-CoV-2. Both of them have shown favorable
vaccine efcacy (9495%) in preventing COVID-19 in phase 3
clinical trials. However, emerging variants of SARS-CoV-2 and its
global expansion have raised concerns about potentially reduced
protection against variants of concern (VOCs) by current COVID-19
vaccines. Notable variants harboring multiple mutations in the
spike protein have emerged in the United Kingdom (B.1.1.7),
South Africa (B.1.351), and Brazil (P.1). B.1.1.7 variant (20I/501Y.V1),
the most globally widespread VOC, has a N501Y substitution in
the receptor-binding domain (RBD), H69/V70 deletion in the N-
terminal domain, and P681H mutation adjacent to the furin
cleavage site in the spike protein. This variant has been associated
with increased transmissibility. B.1.351 variant (20H/501Y.V2)
contains several mutations, including K417N, E484K, and N501Y.
P.1 variant (B.1.1.28.1) possesses K417T, E484K, and N501Y
substitution in the RBD of the spike protein. These VOCs also
share the D614G mutation, which confers an increased ability for
rapid viral spread.
Wang et al.
1
tested the neutralizing activity of plasma from
vaccinees (BNT162b2, n=6; mRNA-1273, n=14) against pseudo-
type viruses harboring K417N, E484K, N501Y, and a combination
of these three RBD mutations (B.1.351 variant). The study revealed
1- to 3-fold decreased neutralizing activity against E484K, N501Y,
and the K417N:E484K:N501Y combination (p=0.0033, p=0.0002,
and p< 0.0001, respectively), but there was no signicant
difference in neutralizing activity against wild-type and K417N
mutation. This result suggests that COVID-19 mRNA vaccine-
elicited neutralizing antibodies are less effective against emerging
SARS-CoV-2 VOCs with RBD mutations. In addition, convalescent
plasma obtained 6 months after SARS-CoV-2 infection was 0.5- to
20.2-fold less effective in neutralizing the K417N:E484K:N501Y
combination (p< 0.0001).
In a subsequent analysis, the study was extended to SARS-CoV-2
RBD-specic memory B cells. The mRNA vaccines elicited a robust
SARS-CoV-2 RBD-specic memory B-cell response similar to
natural infection. Monoclonal antibodies (mAbs) were expressed
by SARS-CoV-2 RBD-specic memory B cells and had potent
neutralizing activity towards SARS-CoV-2 pseudovirus. However,
among the 17 most potent mAbs, 14 demonstrated reduced or
abolished activities in neutralizing the K417N, E484K, or N501Y
mutations. Selection pressure by mAbs was also detected; these
mutations emerged when recombinant vesicular stomatitis virus/
SARS-CoV-2 S was cultured in the presence of the vaccine-
elicited mAbs.
Similarly, Chen et al.
2
reported that sera from BNT162b2-
vaccinated individuals showed reduced neutralizing activities
against emerging SARS-CoV-2 variants. They observed signicantly
decreased neutralizing potency of sera from the vaccinees against
B.1.1.7 isolate (2-fold), E484K/N501Y/D614G recombinant variant
(4-fold), and two chimeric SARS-CoV-2 strains encoding
B.1.351 spike (10-fold) and P.1 spike (2.2-fold) compared to the
D614G variant in Vero-hACE2-TMPRSS2 cells. A notable reduction
in neutralizing activity was not shown with the K417N/D614G
variant, suggesting that sera from recipients of the BNT162b2
vaccine had a lower neutralizing capacity against E484K and
N501Y-containing viruses.
In addition, Wang et al.
3
demonstrated a substantial loss of
neutralizing activity against the B.1.351 strain in convalescent
plasma (9.4-fold) and sera from vaccinees who received mRNA
vaccines (10.312.4-fold). E484K seemed to be the main
contributor to the neutralization resistance. Furthermore, the
neutralizing potency of mAb therapeutics in clinical use or
under clinical investigation decreased against the B.1.351
variant.
Received: 17 March 2021 Revised: 2 April 2021 Accepted: 24 April 2021
1
Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea;
2
Graduate
School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea;
3
Division of Infectious Diseases,
Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea and
4
The Center for Epidemic Preparedness, KAIST, Daejeon,
Republic of Korea
Correspondence: Eui-Cheol Shin (ecshin@kaist.ac.kr)
These authors contributed equally: Ji Yun Noh, Hye Won Jeong.
www.nature.com/sigtrans
Signal Transduction and Targeted Therapy
©The Author(s) 2021
1234567890();,:
Taken together, the results from recent studies suggest that
the emergence of resistant SARS-CoV-2 variants may nullify the
effects of current COVID-19 vaccines. However, COVID-19
vaccines can elicit not only neutralizing antibodies, but also
SARS-CoV-2-specicCD4
+
and CD8
+
T-cell responses. Vaccina-
tion with various vaccine platforms, including mRNA and viral
vectors, has been shown to elicit SARS-CoV-2-specicCD4
+
and
CD8
+
T-cell responses (Fig. 1). In principle, it is more difcult to
evade T-cell responses than a neutralizing antibody response
because multiple T-cell epitopes are scattered across viral
proteins, whereas neutralizing antibody targets a narrow region
in the viral protein. Although SARS-CoV-2 mutations that
abrogate binding to major histocompatibility complex have
been reported,
4
Tarke et al.
5
recently reported an insignicant
impact of SARS-CoV-2 variants on both CD4
+
and CD8
+
T-cell
responses in COVID-19 convalescents and recipients of COVID-
19 mRNA vaccines. T-cell responses to the variants B.1.1.7,
B.1.351, P.1, and CAL.20C (emerged in Southern California) were
not differ from those to the ancestral strain of SARS-CoV-2. Most
SARS-CoV-2 T-cell epitopes were conserved despite the muta-
tions in the variants.
The future of the current COVID-19 pandemic is unpredict-
able. Careful surveillance for the emergence of variants and a
thorough investigation of its impact on public health will be
continuously required. Continuous emergence of SARS-CoV-2
VOCs have implications for updating current COVID-19 vaccines
and the development of vaccines providing broader protection.
Even if SARS-CoV-2 variants escape the neutralizing antibodies
elicited by current COVID-19 vaccines, T-cell immunity may be
helpful in reducing the disease burden of COVID-19 by
attenuating disease severity and decreasing mortality. The
real-world effectiveness of the COVID-19 vaccines, especially in
preventing hospitalization, complications, and death, should be
assessed in the near future. Above all, public health policies
should be implemented to ensure that concerns about SARS-
CoV-2 variants and possible reduced vaccine efcacy towards
VOCs do not lead to blunted COVID-19 vaccination rates.
ACKNOWLEDGEMENTS
This research was supported by the 2020 Joint Research Project of Institutes of
Science and Technology, the Korea Health Technology R&D Project through the
Korea Health Industry Development Institute (KHIDI), funded by the Ministry of
Health & Welfare, Republic of Korea (grant number: HI20C0452), and the National
Research Foundation of Korea (NRF) grant funded by the Korea government (MISP)
(2020R1A5A2017476).
ADDITIONAL INFORMATION
Competing interests: The authors declare no competing interests.
REFERENCES
1. Wang, Z. et al. mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating
variants. Nature 592, 616622 (2021).
2. Chen, R. E. et al. Resistance of SARS-CoV-2 variants to neutralization by monoclonal
and serum-derived polyclonal antibodies. Nat. Med. 27, 717726 (2021).
3. Wang, P. et al. Antibody Resistance of SARS-CoV-2 Variants B.1.351 and B.1.1.7.
Nature 593, 130135 (2021).
4. Agerer, B. et al. SARS-CoV-2 mutations in MHC-I-restricted epitopes evade CD8(+)
T cell responses. Sci. Immunol. 6, eabg6461 (2021).
5. Tarke, A. et al. Negligible impact of SARS-CoV-2 variants on CD4
+
and CD8
+
T cell
reactivity in COVID-19 exposed donors and vaccinees. Preprint at bioRxiv https://
doi.org/10.1101/2021.02.27.433180 (2021).
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons license, and indicate if changes were made. The images or other third party
material in this article are included in the articles Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the
articles Creative Commons license and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder. To view a copy of this license, visit http://creativecommons.
org/licenses/by/4.0/.
© The Author(s) 2021
Fig. 1 COVID-19 vaccines elicit SARS-CoV-2-specic CD4
+
and CD8
+
T-cell responses as well as neutralizing antibodies. Even though
antibodies elicited by current COVID-19 mRNA vaccines had shown diminished neutralizing activities against SARS-CoV-2 variants, T-cell
responses may have a role for host protection against SARS-CoV-2 variants
SARS-CoV-2 mutations, vaccines, and immunity: implication of variants of. . .
Noh et al.
2
Signal Transduction and Targeted Therapy (2021) 6:203
... 9 In addition, prior to 2020, little was known about the new strain of virus other than it was more highly contagious and caused a more serious respiratory illness than the related strains of coronavirus that had caused MERS and SARS years earlier. [9][10][11]27 The WHO declared it to be a pandemic in early 2020, and as of late February, 2023, the number of COVID-19 cases reported worldwide had reached almost 680 million. In addition, the number of known deaths from COVID-19 was over 6 million. ...
... Scientific research of RNA coronaviruses impacting the current pandemic has been developing for over two decades and of extreme necessity became further intensified with the onset of COVID-19 by early 2020. [8][9][10] These research studies culminated in numerous clinical trials of multiple newly developed COVID-19 vaccines in 2020, some using the recently developed mRNA and other molecular models, and with recommendations for booster shots that encompassed recently evolved epitopes discovered in some of the more prevalent mutant substrains as the pandemic progressed. Episodes of COVID-19 illness later were reported to occur commonly in individuals who had been vaccinated with the newly developed vaccines in addition to many who had contracted the virus and previously experienced live infections early in the pandemic. ...
... 9,23-27 Those deemed at greater risk typically require additional emphasis for early treatment in an attempt to limit post infection sequelae including the development of long COVID, where a variety of symptoms may persist for a year or more following COVID-19 infection. [8][9][10][11]27 The most prevalent clinical symptoms of COVID-19 infections often overlap for patients with chronic autoimmune diseases, obesity, and other common comorbidities, therefore supportive attention and appropriate care must be taken to address and identify the specific symptomology as accurately as possible because the pathophysiologic mechanisms associated with COVID-19 illness and other comorbidities may differ. 25 Thus, it becomes important to discriminate among the possible therapeutic regimens for COVID-19 and other overlapping conditions that may be prescribed to best effect the success of the therapeutic regimen for each concurrent disease entity. ...
Preprint
Full-text available
Discovery and development of effective strategies to treat emerging and often drug resistant strains of viral and microbial pathogens presents one of the greatest challenges of the 21st century. Despite nearly a century of progress in developments in antimicrobial and antiviral therapies, infectious diseases still account for a substantial proportion of deaths worldwide, including over 100 million globally in the recent pandemic to date. In recent decades, the plague of antimicrobial resistance also represents an additional and alarming signal for both human and animal healthcare and signals a renewed sense of urgency in addressing this issue. This has made the search for novel classes of antibiotics, antivirals and therapeutics found to be capable of bypassing the microbial and viral resistance mechanisms necessary to in order to replenish our current arsenal of antimicrobial and antiviral drugs and update our therapeutic regimens. In addition, the tremendous impact exerted by viral infections and related pathologies on our lives during the recent decade has forced scientists to acknowledge the opportunities and challenges associated with tackling infectious diseases by developing effective antiviral agents endowed with novel mechanisms of action. The discovery of new antimicrobial/antiviral agents, as well as the repurposing of existing drugs and therapeutic options will be crucial to fight the ever-increasing resistance of “superbugs”, pathogenic fungi, viruses, and parasites. Proponents of modern biophoton therapy proposed in this paper can trace its origins to the Russian scientist Alexander Gurwitsch, who a century ago observed ultra-weak emissions of light emerging from an onion root and soon found additional living tissues to be emanating similar energetic phenomena, thus energizing research which continues to the present day and projected toward the development of useful applications of the new-found biological photonic phenomena. The purpose of the present paper is to introduce the past experiences, potential applications and highly successful outcomes of biophotonic therapy as an option to treat infectious diseases including HIV, Hepatitis C, MRSA, and others, with emphasis on efficacy of using biophotonic therapies to treat and ameliorate the drug resistant variants of the above infectious agents, including potential applications for the current COVID-19 pandemic and in preparation for the emergence of the next epidemic or pandemic.
... Moreover, their effect on these variants is still unclear. 15 Based on the WHO announcement, variants of the virus that are associated with one of the following changes have been considered as variants of concern (VOCs): 1-higher transmissibility or harmful transition in COVID-19 epidemiology, 2-enhanced pathogenicity or alteration in the clinical manifestations of the disease, 3-reduction in the efficiency of available diagnostics, vaccines, and therapeutics. These variants include B.1.351 ...
... (South Africa), which have multiple mutations in the surface glycoprotein as shown in Table 1. [15][16][17][18][19] Using immunoinformatics methods, this study aimed to develop a multi-epitope vaccine against COVID-19. This innovative vaccine is composed of both conserved and mutant epitopes; hence it could be effective against wild-type and mutant variants of SARS-CoV-2. ...
... However, there are serious concerns about their efficiency against the emerging VOCs strains containing different surface glycoprotein mutations. 15 Recent studies have also revealed that neutralized antibody titers are reduced for the emerging VOCs. 44,45 Traditional and modern approaches have been adopted for COVID-19 vaccine development. ...
Article
Full-text available
Background: The newly emerged coronavirus, SARS-CoV-2, the causative agent of the COVID-19 disease, appeared in Wuhan, China in December 2019 and led to the death of millions of people. The pandemic has increased the demand for effective vaccines and treatments worldwide. Due to the changes in the virus genome caused by mutation, variants with higher transmission ability and pathogenicity have emerged and raised excessive concerns about the efficiency of the developed vaccines. This study aimed to design a multi-epitope vaccine that targets the primary SARS-CoV-2 strain and its five variants of concern using immunoinformatics approaches. Methods: B-cell, cytotoxic T lymphocytes (CTLs) and helper T lymphocytes epitopes of the conserved and mutated SARS-CoV-2 surface glycoproteins were predicted using immunoinformatics approaches. These epitopes were attached utilizing appropriate linkers, and to provoke the immune response efficiently, the cholera toxin B subunit was added as an adjuvant. Results: Analyses by different bioinformatics softwares revealed that the designed vaccine’s high safety, stability, and efficacy. Conclusion: In this study, the vaccine designed by immunoinformatics tools showed specific reactivity to Toll-like receptor 4. Given the validation of other epitope-based vaccines, our vaccine might be capable of providing strong immunity in a wide range of populations.
... Vaccination is one of the most effective tools to protect people against severe illness, hospitalization, and mortality [3]. Since the first report of COVID-19, emergence of new circulating variants of the COVID-19 virus harboring mutations in the spike molecule, as the main target of neutralizing antibodies, has raised significant concern about the efficacy of current vaccines [25]. ...
Article
Full-text available
Background Waning immunity and emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlight the need for further research in vaccine development. Methods A recombinant fusion protein containing the receptor-binding domain (RBD) fused to the human IgG1 Fc (RBD-Fc) was produced in CHO-K1 cells. RBD-Fc was emulsified with four adjuvants to evaluate its immunogenicity. The RBD-specific humoral and cellular immune responses were assessed by ELISA. The virus neutralizing potency of the vaccine was investigated using four neutralization methods. Safety was studied in mice and rabbits, and Antibody-Dependent Enhancement (ADE) effects were investigated by flow cytometry. Results RBD-Fc emulsified in Alum induced a high titer of anti-RBD antibodies with remarkable efficacy in neutralizing both pseudotyped and live SARS-CoV-2 Delta variant. The neutralization potency dropped significantly in response to the Omicron variant. RBD-Fc induced both TH2 and particularly TH1 immune responses. Histopathologic examinations demonstrated no substantial pathologic changes in different organs. No changes in serum biochemical and hematologic parameters were observed. ADE effect was not observed following immunization with RBD-Fc. Conclusion RBD-Fc elicits highly robust neutralizing antibodies and cellular immune responses, with no adverse effects. Therefore, it could be considered a promising and safe subunit vaccine against SARS-CoV-2.
... Utilizing virus particles that have been inactivated or have undergone attenuation and are expressing the spike protein can result in a broad immune response against several viral antigens [131]. Another strategy is to introduce the spike protein gene into host cells using viral vectors like adenovirus or vesicular stomatitis virus to start an immunological reaction against the virus [131,132]. A more recent strategy makes use of mRNA technology to transport the spike protein's genetic code to host cells, enabling those cells to manufacture the protein and trigger an immune response [133,134]. ...
Article
Full-text available
The emergence of SARS-CoV-2, the virus responsible for the COVID-19 pandemic, has sparked intense research on its spike protein, which is essential for viral entrance into host cells. Viral reproduction and transmission, host immune response regulation, receptor recognition and host cell entrance mechanisms, as well as structural and functional effects have all been linked to mutations in the spike protein. Spike protein mutations can also result in immune evasion mechanisms that impair vaccine effectiveness and escape, and they are linked to illness severity and clinical consequences. Numerous studies have been conducted to determine the effects of these mutations on the spike protein structure and how it interacts with host factors. These results have important implications for the design and development of medicines and vaccines based on spike proteins as well as for the assessment of those products’ efficiency against newly discovered spike protein mutations. This paper gives a general overview of how spike protein mutations are categorized and named. It further looks at the links between spike protein mutations and clinical outcomes, illness severity, unanswered problems, and future research prospects. Additionally, explored are the effects of these mutations on vaccine effectiveness as well as the possible therapeutic targeting of spike protein mutations.
... First-generation COVID-19 vaccines, derived from the spike or RBD of SARS-CoV-2 ancestral strain, have shown waning effectiveness against VOCs, especially Omicron (2). E484K spike mutation shared by Beta and Gamma is a main contributor to neutraliza tion evasion (19,20). In addition, to stabilize spike in the prefusion conformation, we introduced furin cleavage site mutation (dFurin: 681-PRRAR-685 to 681-HSRAG-685) and two proline substitutions K986P and V987P (2P) (21,22). ...
Article
Full-text available
Developing broad-spectrum vaccines and optimal vaccination strategies is crucial to controlling the COVID-19 pandemic. Here, we generated a chimpanzee adenoviral vector-based COVID-19 vaccine carrying broad-spectrum immunogens, modified full-length spike, and conserved T-cell epitopes of SARS-CoV-2, and assessed its immune response in mice through intramuscular (i.m.), intranasal (i.n.), or combined immunization routes (i.m. + i.n., or i.n. + i.m.). Compared to other vaccination strategies, the two combined regimens elicited higher neutralizing antibody (NAb) responses to all variants. Compared to i.n. + i.m. regimen, the i.m. + i.n. regimen stimulated a stronger secondary GC response, which is more pivotal to high-quality antibody production than the primary GC response. Moreover, the i.m. + i.n. regimen was adept at mediating systemic cellular immunity, while the i.n. + i .m. regimen tended to elicit lung tissue-resident memory T (T RM ) cell responses. Overall, the two combined regimens induced comprehensive but distinct immune responses consisting of lgA, lgG, NAbs, GC B cells, long-lived plasma cells, T RM cells, and systemic memory T cells, which conferred complete protection against BA.2 infection in hACE2 transgenic mice, and warranted further investigation as potential universal vaccination strategies. IMPORTANCE The development of broad-spectrum SARS-CoV-2 vaccines will reduce the global economic and public health stress from the COVID-19 pandemic. The use of conserved T-cell epitopes in combination with spike antigen that induce humoral and cellular immune responses simultaneously may be a promising strategy to further enhance the broad spectrum of COVID-19 vaccine candidates. Moreover, this research suggests that the combined vaccination strategies have the ability to induce both effective systemic and mucosal immunity, which may represent promising strategies for maximizing the protective efficacy of respiratory virus vaccines.
... Utilizing virus particles that have been inactivated or have undergone attenuation and are expressing the spike protein can result in a broad immune response against several viral antigens [148]. Another strategy is to introduce the spike protein gene into host cells using viral vectors like adenovirus or vesicular stomatitis virus to start an immunological reaction against the virus [148,149]. 14 A more recent strategy makes use of mRNA technology to transport the spike protein's genetic code to host cells, enabling those cells to manufacture the protein and trigger an immune response [150,151]. ...
Preprint
Full-text available
The SARS-CoV-2 virus, which is responsible for the COVID-19 pandemic which emerged and spread, has sparked intense research on its spike protein, which is essential for viral entrance into host cells. Viral reproduction and transmission, host immune response regulation, receptor recognition and host cell entrance mechanisms, as well as structural and functional effects have all been linked to mutations in the spike protein. Spike protein mutations can also result in immune evasion mechanisms that impair vaccine effectiveness and escape, and they are linked to illness severity and clinical consequences. Numerous studies have been conducted to determine the effects of these mutations on the spike protein structure and how it interacts with host factors. These results have important implications for the design and development of medicines and vaccines based on spike proteins as well as for the assessment of those products' efficiency against newly discovered spike protein mutations. The paper gives a general overview of how spike protein mutations are categorized and named, as well as the genomic and phylogenetic techniques that have been used to track their genesis and dissemination. Additionally, it looks at the links between spike protein mutations and clinical outcomes, illness severity, unanswered problems, and future research prospects. Additionally, explored are the effects of these mutations on vaccine effectiveness as well as the possible therapeutic targeting of spike protein mutations.
Article
The protective efficacies of current licensed vaccines against COVID-19 have significantly reduced as a result of SARS-CoV-2 variants of concern (VOCs) which carried multiple mutations in the Spike (S) protein. Considering that these vaccines were developed based on the S protein of the original SARS-CoV-2 Wuhan strain, we designed a recombinant plasmid DNA vaccine based on highly conserved and immunogenic B and T cell epitopes against SARS-CoV-2 Wuhan strain and the Omicron VOC. Literature mining and bioinformatics were used to identify 6 immunogenic peptides from conserved regions of the SARS-CoV-2 S and membrane (M) proteins. Nucleotide sequences encoding these peptides representing highly conserved B and T cell epitopes were cloned into a pVAX1 vector to form the pVAX1/S2-6EHGFP recombinant DNA plasmid vaccine. The DNA vaccine was intranasally or intramuscularly administered to BALB/c mice and evaluations of humoral and cellular immune responses were performed. The intramuscular administration of pVAX1/S2-6EHGFP was associated with a significantly higher percentage of CD8+ T cells expressing IFN-γ when compared with the empty vector and PBS controls. Intramuscular or intranasal administrations of pVAX1/S2-6EHGFP resulted in robust IgG antibody responses. Sera from mice intramuscularly immunized with pVAX1/S2-6EHGFP were found to elicit neutralizing antibodies capable of SARS-CoV-2 Omicron variant with the ACE2 cell surface receptor. This study demonstrated that the DNA vaccine construct encoding highly conserved immunogenic B and T cell epitopes was capable of eliciting potent humoral and cellular immune responses in mice.
Article
Full-text available
SARS-CoV-2-reactive T cells are detected in some healthy unexposed individuals. Human studies indicate these T cells could be elicited by the common cold coronavirus OC43. To directly test this assumption and define the role of OC43-elicited T cells that are cross-reactive with SARS-CoV-2, we develop a model of sequential infections with OC43 followed by SARS-CoV-2 in HLA-B*0702 and HLA-DRB1*0101 Ifnar1−/− transgenic mice. We find that OC43 infection can elicit polyfunctional CD8⁺ and CD4⁺ effector T cells that cross-react with SARS-CoV-2 peptides. Furthermore, pre-exposure to OC43 reduces subsequent SARS-CoV-2 infection and disease in the lung for a short-term in HLA-DRB1*0101 Ifnar1−/− transgenic mice, and a longer-term in HLA-B*0702 Ifnar1−/− transgenic mice. Depletion of CD4⁺ T cells in HLA-DRB1*0101 Ifnar1−/− transgenic mice with prior OC43 exposure results in increased viral burden in the lung but no change in virus-induced lung damage following infection with SARS-CoV-2 (versus CD4⁺ T cell-sufficient mice), demonstrating that the OC43-elicited SARS-CoV-2 cross-reactive T cell-mediated cross-protection against SARS-CoV-2 is partially dependent on CD4⁺ T cells. These findings contribute to our understanding of the origin of pre-existing SARS-CoV-2-reactive T cells and their effects on SARS-CoV-2 clinical outcomes, and also carry implications for development of broadly protective betacoronavirus vaccines.
Article
Here, we examine peripheral blood memory T cell responses against the SARS-CoV-2 BA.4/BA.5 variant spike among vaccinated individuals with or without Omicron breakthrough infections. We provide evidence supporting a lack of original antigenic sin in CD8 ⁺ T cell responses targeting the spike. We show that BNT162b2-induced memory T cells respond to the BA.4/BA.5 spike. Among individuals with BA.1/BA.2 breakthrough infections, IFN-γ–producing CD8 ⁺ T cell responses against the BA.4/BA.5 spike increased. In a subgroup with BA.2 breakthrough infections, IFN-γ–producing CD8 ⁺ T cell responses against the BA.2-mutated spike region increased and correlated directly with responses against the BA.4/BA.5 spike, indicating that BA.2 spike–specific CD8 ⁺ T cells elicited by BA.2 breakthrough infection cross-react with the BA.4/BA.5 spike. We identified CD8 ⁺ T cell epitope peptides that are present in the spike of BA.2 and BA.4/BA.5 but not the original spike. These peptides are fully conserved in the spike of now-dominant XBB lineages. Our study shows that breakthrough infection by early Omicron subvariants elicits CD8 ⁺ T cell responses that recognize epitopes within the spike of newly emerging subvariants.
Article
Full-text available
Unlike conventional mRNA vaccines,intranasal vaccines display several advantages, including the ability to generatestrong mucosal immunity. However,the formulation challenges associated with intranasal mRNA vaccines have so farhindered their extensive application and further research in the area of formulationdevelopment and optimisation are required before translation. This study aimsto optimise the manufacturing conditions of inhalable dry powders by examining theimpact of temperature and lipid composition on liposome particle size and theeffect of spray-drying parameters on the particle size of spray-dried powders. The results indicate that liposomesgenerated at 20°C exhibited notably larger sizes than those produced at 55°C,irrespective of the lipid composition. Notably, dry powder formulations featuringlarger particle sizes were achieved at a low gas flow rate of 25mm.
Article
Full-text available
The COVID-19 pandemic has ravaged the globe, and its causative agent, SARS-CoV-2, continues to rage. The prospects of ending this pandemic rest on the development of effective interventions. Single and combination monoclonal antibody (mAb) therapeutics have received emergency use authorization1–3, with more in the pipeline4–7. Furthermore, multiple vaccine constructs have shown promise8, including two with ~95% protective efficacy against COVID-199,10. However, these interventions were directed toward the initial SARS-CoV-2 that emerged in 2019. The recent emergence of new SARS-CoV-2 variants B.1.1.7 in the UK11 and B.1.351 in South Africa12 is of concern because of their purported ease of transmission and extensive mutations in the spike protein. We now report that B.1.1.7 is refractory to neutralization by most mAbs to the N-terminal domain (NTD) of the spike and relatively resistant to a few mAbs to the receptor-binding domain (RBD). It is not more resistant to convalescent plasma or vaccinee sera. Findings on B.1.351 are more worrisome in that this variant is not only refractory to neutralization by most NTD mAbs but also by multiple individual mAbs to the receptor-binding motif on RBD, largely owing to an E484K mutation. Moreover, B.1.351 is markedly more resistant to neutralization by convalescent plasma (9.4 fold) and vaccinee sera (10.3-12.4 fold). B.1.351 and emergent variants13,14 with similar spike mutations present new challenges for mAb therapy and threaten the protective efficacy of current vaccines.
Article
Full-text available
CD8 ⁺ T cell immunity to SARS-CoV-2 has been implicated in COVID-19 severity and virus control. Here, we identified nonsynonymous mutations in MHC-I-restricted CD8 ⁺ T cell epitopes after deep sequencing of 747 SARS-CoV-2 virus isolates. Mutant peptides exhibited diminished or abrogated MHC-I binding in a cell-free in vitro assay. Reduced MHC-I binding of mutant peptides was associated with decreased proliferation, IFN-γ production and cytotoxic activity of CD8 ⁺ T cells isolated from HLA-matched COVID-19 patients. Single cell RNA sequencing of ex vivo expanded, tetramer-sorted CD8 ⁺ T cells from COVID-19 patients further revealed qualitative differences in the transcriptional response to mutant peptides. Our findings highlight the capacity of SARS-CoV-2 to subvert CD8 ⁺ T cell surveillance through point mutations in MHC-I-restricted viral epitopes.
Article
Full-text available
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global COVID-19 pandemic. Rapidly spreading SARS-CoV-2 variants may jeopardize newly introduced antibody and vaccine countermeasures. Here, using monoclonal antibodies (mAbs), animal immune sera, human convalescent sera and human sera from recipients of the BNT162b2 mRNA vaccine, we report the impact on antibody neutralization of a panel of authentic SARS-CoV-2 variants including a B.1.1.7 isolate, chimeric strains with South African or Brazilian spike genes and isogenic recombinant viral variants. Many highly neutralizing mAbs engaging the receptor-binding domain or N-terminal domain and most convalescent sera and mRNA vaccine-induced immune sera showed reduced inhibitory activity against viruses containing an E484K spike mutation. As antibodies binding to spike receptor-binding domain and N-terminal domain demonstrate diminished neutralization potency in vitro against some emerging variants, updated mAb cocktails targeting highly conserved regions, enhancement of mAb potency or adjustments to the spike sequences of vaccines may be needed to prevent loss of protection in vivo.
Preprint
Full-text available
The emergence of SARS-CoV-2 variants highlighted the need to better understand adaptive immune responses to this virus. It is important to address whether also CD4+ and CD8+ T cell responses are affected, because of the role they play in disease resolution and modulation of COVID-19 disease severity. Here we performed a comprehensive analysis of SARS-CoV-2-specific CD4+ and CD8+ T cell responses from COVID-19 convalescent subjects recognizing the ancestral strain, compared to variant lineages B.1.1.7, B.1.351, P.1, and CAL.20C as well as recipients of the Moderna (mRNA-1273) or Pfizer/BioNTech (BNT162b2) COVID-19 vaccines. Similarly, we demonstrate that the sequences of the vast majority of SARS-CoV-2 T cell epitopes are not affected by the mutations found in the variants analyzed. Overall, the results demonstrate that CD4+ and CD8+ T cell responses in convalescent COVID-19 subjects or COVID-19 mRNA vaccinees are not substantially affected by mutations found in the SARS-CoV-2 variants.
Article
Full-text available
Here we report on the antibody and memory B cell responses in a cohort of 20 volunteers who received either the Moderna (mRNA-1273) or Pfizer-BioNTech (BNT162b2) vaccines1-4. Eight weeks after the second vaccine injection volunteers showed high levels of IgM, and IgG anti-SARS-CoV-2 spike protein (S) and receptor binding domain (RBD) binding titers. Moreover, the plasma neutralizing activity, and the relative numbers of RBD-specific memory B cells were equivalent to individuals who recovered from natural infection5,6. However, activity against SARS-CoV-2 variants encoding E484K or N501Y or the K417N:E484K:N501Y combination was reduced by a small but significant margin. Vaccine-elicited monoclonal antibodies (mAbs) potently neutralize SARS-CoV-2, targeting a number of different RBD epitopes in common with mAbs isolated from infected donors5-8. However, neutralization by 14 of the 17 most potent mAbs tested was reduced or abolished by either K417N, or E484K, or N501Y mutations. Notably, the same mutations were selected when recombinant vesicular stomatitis virus (rVSV)/SARS-CoV-2 S was cultured in the presence of the vaccine elicited mAbs. Taken together the results suggest that the monoclonal antibodies in clinical use should be tested against newly arising variants, and that mRNA vaccines may need to be updated periodically to avoid potential loss of clinical efficacy.
Negligible impact of SARS-CoV-2 variants on CD4+and CD8+ T cell reactivity in COVID-19 exposed donors and vaccinees
  • A Tarke