A rational strategy for the maintenance of antiviral immunity to new SARS-CoV-2 strains

Cover Page


Cite item

Abstract

New variants of SARS-CoV-2 such as Omicron BA.2, BA.4/5, BA.2.12.1 and BA 2.75 are characterized by higher infectivity and the ability to escape virus-neutralizing antibodies against previous coronavirus variants. The S-trimer of BA.2 and its phylogenetic derivatives are characterized by a predominant «Up»-conformation, which facilitates the interaction with ACE2 on target cells and promotes the resistance to neutralizing antibodies. The immunity acquired from the infection with earlier strains is non-sterile for both early and later strains; the booster systemic immunization does not significantly affect the effectiveness of antiviral immunity, and its feasibility is currently being questioned. Studies of the mucosal immune response have shown that intranasal immunization with adenovirus vaccines provides more pronounced protective immunity than systemic reimmunization does. A promising approach is the creation of multivalent inhaled next generation vaccines containing immunoadjuvants that activate B- and T-cell mucosal immunity. Currently, a large number of intranasal vaccines are undergoing phase I/II trials, while the preclinical and preliminary clinical results indicate that this method of vaccination provides a better mucosal immune response at the entry site of the virus than systemic immunization does. This strategy may provide a long-term immune protection against the currently existing and yet unknown new strains of SARS-CoV-2.

Full Text

BACKGROUND

The coronavirus disease 2019 (COVID-19) pandemic, which began in late 2019, globally manifested itself as six distinct waves by the end of summer 2022, with each successive wave driven by the emergence of a new variant of SARS-CoV-2 with unique features. The most significant one was wave 5 at the beginning of 2022, which at its peak gave a daily increase of more than 4 million cases worldwide and more than 200 thousand cases in Russia; it arose as a result of the emergence of the omicron variant that has more than 30 mutations in the S-protein, first in the form of strain BA.1.1.529, which was then replaced by its phylogenetic descendant BA.2 [1–3]. At present, after the BA.1/BA.2 wave, wave 6 is growing in Russia due to omicron variants BA2.12.1, BA.4, BA.5, and BA.2.75 (Centaur) that arose on the basis of BA.2 and are widely spread, characterized by an even more pronounced ability to avoid the immune response of neutralizing antibodies developed against previous strains [4–6]. Although the number of hospitalizations and mortality in the case of infection with omicron strains is significantly lower than with the delta variant (B.1.617.2), the total number of cases increased many times over, including among those who had previously been ill with other strains of SARS-CoV-2 and those vaccinated by all existing types of vaccines cannot but cause concern.

The strategy of vaccination and revaccination, which was quite obvious before the emergence of the omicron variant, enabled the reduction to null the pandemic wave caused by the most pathogenic and lethal delta variant [7]. With the advent of omicron strains that evade effectively vaccine immunity, the strategy of regular revaccination, at first glance, ceased to be so obvious [8], even if a meta-analysis showed the absence of allergic reactions in response to repeated vaccinations [9].

The vaccination with mRNA and adenoviral vaccines protects against severe COVID-19; however, no obvious data on protection against asymptomatic or paucisymptomatic infection with SARS-CoV-2 have been obtained either in the case of infection with the delta variant, or especially omicron [10]. The reason is that the existing vaccine immunity is not sterilizing (i.e., does not prevent infection and virus spread). An outbreak of a pandemic and infection spread among the vaccinated population was first recorded when the delta variant appeared in the summer of 2021 [11]. Nearly 100% vaccination coverage in Europe and the USA did not prevent another wave caused by omicron, which suggests primarily the ability it acquired to avoid virus neutralization by antibodies to the S-protein. Moreover, in the vast majority of cases, the disease was asymptomatic or paucisymptomatic, which is usually associated with the presence of immunity in most of the patients infected, which prevents a severe course with systemic damage to the lungs and other organs [1]. In addition, such an enormous outbreak confirmed once again that the systemic immune response to SARS-CoV-2 (both post-vaccination and convalescent) is not sterilizing, and people with humoral immunity spread the virus in the same way as those naive in relation to new variants of the virus [12].

The high infectivity and extremely low efficiency of immune protection against new omicron strains cannot but inspire concern despite the significant decrease in the incidence of severe disease. Concerns are primarily associated with the possible emergence of new strains characterized by increased pathogenicity, based on existing highly virulent variants. In addition, a relatively low proportion of various severe post-COVID complications associated with damage to the nervous and cardiovascular systems can become a significant absolute number with a large number of patients infected worldwide. Post-COVID syndrome, or the so-called long COVID, is characterized by psychoneurological, autonomic, pulmonary, vascular, endocrine, immune, and other disorders lasting for several months [13–17]; along with an increase in the number of people who have been ill, it is becoming an urgent medical and social problem. All of these concerns require a more modern strategy for creating and maintaining an immune defense against new strains of SARS-CoV-2.

This study aimed to analyze current studies of antiviral immunity to new strains of SARS-CoV-2 and develop a rational strategy for creating and maintaining immunity to the omicron variant and new variants that have not yet emerged.

PHYLOGENESIS OF NEW SARS-CoV-2 STRAINS

By the end of 2021, the World Health Organization (WHO) identified five variants, or clades (phylogenetic groups), of SARS-CoV-2, which were variants-of-concern (VOC) (WHO, 2022), namely, alpha (B. 1.1.7 according to PANGO1 [18], or clade 20I according to Nextstrain2 [19]), beta (B.1.351; clade 20H), gamma (P.1; clade 20J), delta (B.1.617.2, AY; clades 21I and 21J), and omicron (B.1.1.529, BA.1-5; clades 21K, 22C, and 22B) (Table 1) [20].

 

Table 1

Characteristics of the main variants of SARS-CoV-2 that caused significant morbidity during the pandemic

Name according to WHO

Nomenclature

Country

of occurrence

Date

of occurrence

Mutations in the S-protein

PANGO

NextStrain

Hu-1 (Wuhan isolate)

-

19А

China

November 2019

-

-

В

20

-

-

D614G

-

B.1

20А

-

-

D614G

-

B.1

20B

-

-

D614G

Alpha

B.1.1.7

20I

UK

September 2020

D614G, 69/70del, 144/5del, P618H, T716I, N601Y, S982A, A570D, D1118H

Beta

B.1.351

20H

South Africa

May 2020

D614G, L18F, D80A, D215G, 242-4del, R246I, K417N, E485K, N501Y, A701V

Gamma

P.1

20J

Brazil

November 2020

D614G, L18F, T20N, P26S, D138Y, R190S, K417T, E485K, N501Y, H655Y, T1027I, V1116F

Delta

B.1.617.2

21I, 21J (Delta)

India

Oktober 2020

D614G, T19R, E156G, F157-, R158-, L452R, T478K, P681R, D950N

Omicron

BA.1

21K (Omicron)

South Africa

November 2021

A67V, H69-, V70-, T95I, G142D, V143-, Y144-, Y145-, N211-, L212I, ins214EPE, G339D, S371L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681H, N764K, D796Y, N856K, Q954H, N969K, L981F

Omicron

BA.2

21L (Omicron)

South Africa

November 2021

T19I, L24del, P25del, P26del, A27S, G142D, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, S477N, T478K, E484A, Q493R, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Omicron

BA.2.12.1

22C (Omicron)

USA/Canada

December 2021

T19I, L24del, P25del, P26del, A27S, G142D, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, L452Q, S477N, T478K, E484A, Q493R, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, S704L, N764K, D796Y, Q954H, N969K

Omicron

BA.3

22K (Omicron)

South Africa

November 2021

A67V, H69del, V70del, T95I, G142D, V143del, Y144del, Y145del, N211del, L212I, G339D, S371F, S373P, S375F, D405N, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Omicron

BA.4

22A (Omicron)

South Africa

Yanuary 2022

T19I, L24del, L24del, P25del, P25del, P25del, P26del, P26del, P26del, A27S, H69del, V70del, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, R452K, S477N, T478K, E484A, F486V, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Omicron

BA.5

22B (Omicron)

South Africa

Yanuary 2022

T19I, L24del, L24del, P25del, P25del, P25del, P26del, P26del, P26del, A27S, H69del, V70del, V213G, G339D, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, R452K, S477N, T478K, E484A, F486V, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Omicron

BA.2.75

22D (Omicron)

India

June 2022

T19I, L24del, P25del, P26del, A27S, G142D, K147E, W152R, F157L, I210V, V213G, G257S, G339H, S371F, S373P, S375F, T376A, D405N, R408S, K417N, N440K, G446S, N460K, S477N, T478K, E484A, R493Q, Q498R, N501Y, Y505H, D614G, H655Y, N679K, P681H, N764K, D796Y, Q954H, N969K

Note: Revised from [20]. WHO — World Health Organization.

 

By the beginning of 2022, B.1.617.2 delta, the most virulent and lethal variant of SARS-CoV-2, prevailing worldwide throughout 2021, was almost completely replaced by the new omicron variant BA.1.1.529 (clade 21K) (Fig. 1). Then, by mid-2022, BA.1 was replaced by BA.2 (clade 21L) and then by BA.4/5 (clades 22A and 22B), which subsequently emerged in the Republic of South Africa (South Africa) and spread to other countries concurrently with variant BA.2.12.1 (clade 22C) originating in the USA. As of late August and early September 2022, omicron BA.5 (clade 22B) is the most common strain worldwide [1, 8] (Fig. 1). BA.5, along with strains BA.2.12.1 and BA.4 and some other derivatives of variant BA.2, caused wave 6 of COVID-19 in Russia, which, as of September 21, 2022, causes an official increase of more than 50,000 cases per day.

 

Fig. 1. Evolution of new variants of SARS-CoV-2 superimposed on the “waves” of the pandemic: а — phylogeny of “variants of concern”; b — histogram of the incidence rates of different variants of SARS-CoV-2 in Europe (according to Gissad, https://gisaid.org/) against the background of the morbidity curve in Russia (according to www.yandex.ru). The figures at peaks indicate the daily increase in cases according to the official data.

 

In July 2022, a new omicron strain, BA.2.75, began to spread globally, which was named “Centaur” because of the combination of the beneficial mutations of the BA1/2 and BA4/5 variants. This strain, which was first discovered in India, showed higher distribution dynamics than BA.5 and other phylogenetic descendants of strain BA.2. Currently, BA.2.75 centaur is rapidly spreading worldwide and may be the cause of another wave of increased incidence [4–6].

New SARS-CoV-2 strains are evaluated by their efficiency to spread in humans (disease contagiousness characterizes the so-called reproduction index [R0] defined as the number of individuals to be infected by one typical affected patient) and ability to avoid humoral antiviral immunity and pathogenicity. The latter is directly due to amino acid substitutions in the S-protein, which modify the affinity of the receptor-binding domain for angiotensin-converting enzyme 2 (ACE2) on target cells and determine the dynamics of interaction with virus-neutralizing antibodies.

S-PROTEIN STRUCTURAL ASPECTS IN NEW SARS-CoV-2 VARIANTS

The phenotype and infectivity of new SARS-CoV-2 variants are determined by the structure and functional properties of the S-protein. Just like in other beta-coronaviruses, SARS-CoV-2 S-protein consists of two main domains, namely, receptor-binding domain (RBD) and N-terminal domain (NTD) [21, 22]. RBD interacts directly with ACE2 and ensures that the virus penetrates the target cells; as a result, it is still considered the main target for virus-neutralizing antibodies that block virus entry into cells [23]. Moreover, NTD is the most immunogenic domain of the S-protein, and evidence shows that antibodies recognizing NTD can also be virus-neutralizing [24].

In contrast to the delta variant, which has 7–10 mutations in the S-protein, omicron BA.1 and BA.2 have 37 and 31 mutations in the S-protein, respectively [3] (Table 1). Both omicron stains (BA.1 and BA.2) bind the murine ACE2 receptor with high affinity. By contrast, the wild (Wuhan) variant of SARS-CoV-2 binds to human and cat ACE2, but not to murine ACE2. In this regard, a hypothesis arose of the origin of the omicron variant through evolution with a change of host, i.e., human–cat–mouse–human [2].

Although the S-trimer of the omicron variant, due to numerous mutations, acquired an increased affinity for ACE2 compared with delta, and protection from it requires a significantly higher concentration of virus-neutralizing antibodies (achieved by triple immunization with an mRNA vaccine or a combination of immunity convalescents with booster immunization [16]), the number of severe cases requiring hospitalization decreased by more than two times at the beginning of a new wave, and the risk of death decreased even more [25]. In the cell culture of the nasal epithelium, the replication of the omicron and delta variants was comparable; however, in alveolocytes and intestinal epithelium, omicron demonstrated a lower level of replication than delta, and it did not correlate with the expression level of transmembrane protease, serine 2 (TMPRSS2 protease) [16]. Thus, the alternative method of penetration into the cell acquired by omicron, which is not associated with the activity of TMPRSS2, is less effective and, possibly, underlies the lower pathogenicity and lethality of the new SARS-CoV-2 variant.

BA.2, which completely replaced BA.1 by the end of March 2022 (Fig. 1), exceeded the Wuhan variant by 11 times in the degree of affinity of the S-protein for ACE2 and exceeds the maternal BA.1 by almost two times [1]. Structural studies have shown that when the BA.2 S-trimer interacts with human ACE2, all three RBDs are predominantly in the open up-conformation, which greatly enhances the efficiency of equimolar (3:3) binding to ACE2 and, thus, increases significantly the transmissibility of this strain.

Some published sequences of the new omicron variant BA.2.75 (Centaur) carry the L452R mutation identified in BA.5, which is associated with the possibility of re-infection of patients, and this gives cause for concern. A study by Cao et al. [4] and several other studies published in August 2022 as preprints on the bioRxiv service investigated the possible mechanisms of increased virulence and avoidance of the immune response by BA.2.75 [4, 5].

Compared with BA.2, BA.2.75 S-trimer carries nine additional mutations, of which five (K147E, W152R, F157L, I210V, and G257S) are in NTD, and the remaining four (D339H, G446S, N460K, and R493Q) are in RBD [4–6] (Table 1).

Among the latest mutations in BA.1, G446S appeared, and the R493Q reversion is noted in the sequences of BA.4/BA.5. Mutations N460K and D339H have not previously been found in prevailing variants and their function are still unknown. An alarming factor is that India’s BA.2.75 is characterized by a more efficient distribution than that of new omicron strains BA.2.38 (BA.2+N417T), BA.2.76 (BA.2+R346T+Y248N), and BA.5 [4]. The increased transmissibility of BA.2.75 suggests that this variant may become prevalent after the global wave driven by BA.4/BA.5.

Compared with BA.5, BA.2.75 has been shown to have a significantly higher affinity for ACE2. In addition, the BA.2.75 spike shows reduced thermal stability and a preferential RBD up-conformation under acidic conditions, which probably contributes to the increased endosomal entry of the virus into cells under acidotic conditions. Bioinformatic analysis of the S-protein of BA.2.75 showed that its RBD domain has a higher (more than 3000 times) affinity for ACE2 than B.1.1.7 (alpha) [26]. Such a high affinity of RBD BA.2.75 for ACE2 suggests the possibility of developing angiotensin intoxication when ACE2 is blocked by SARS-СoV-2 S-protein [27].

Omicron BA.2.75, to a lesser extent than BA.4/BA.5, avoids not only plasma neutralization of convalescents after omicron BA.1/BA.2, but also significantly higher plasma neutralization of convalescents after delta. The plasma of convalescents after infection with BA.5 also neutralizes BA.2.75 much weaker than BA.5 [4]. These data collectively indicate that BA.2.75 centaur may cause a significant increase in the incidence of COVID-19 in the near future. Conversely, on the collection of blood sera from Europeans, BA.2.75 did not demonstrate a more significant avoidance of the immune response than BA.5, which is currently predominant in Europe [5] and may indicate that the next wave will affect third-world countries to a greater extent.

IMMUNE RESPONSE TO NEW SARS-CoV-2 VARIANTS

All epidemically significant variants of omicron, such as BA.1 [28, 29], BA.2 [30, 31], BA.4/BA.5 [32–34], and BA.2.75 [4, 5], are characterized by a pronounced resistance to neutralizing antibodies obtained as a result of vaccination or infection with the previous version of SARS-CoV-2 and therapeutic monoclonal antibodies obtained during the delta wave. Just as omicron BA.1 acquired the ability to avoid the immune response resulting from infection with the delta variant, as a result of numerous amino acid substitutions in the S-protein, subsequent variants acquired the ability to evade virus-neutralizing antibodies developed against previous strains. Specifically, BA.2 is resistant to neutralizing antibodies induced against BA.1 [35–37], BA.5 avoids neutralization by antibodies from sera obtained from outbreaks of BA.1 [34] and BA.2 [38], and the new Indian variant BA.2.75 centaur, which emerged among the latest VOCs, appear to avoid successfully neutralization by antibodies against the BA.5 S-protein [4, 5].

In addition to the ability of new strains to avoid humoral and cellular immune responses, repeated COVID-19 infections are caused by the natural decline of immunity after an illness or after vaccination and a decrease in the titer of virus-neutralizing antibodies. The ability of new strains to avoid an antibody response in this aspect has a quantitative equivalent, and this is the minimum titer of virus-neutralizing antibodies that protects against infection. The more pronounced the ability to avoid the immune response, the higher this titer should be. Specifically, neutralization of the beta variant requires a 6-fold higher titer of virus-neutralizing antibodies than for the wild Wuhan variant, Alpha variant [39], etc., with subsequent variants.

Studies of humoral immunity in SARS-CoV-2 infection have shown that serum antibody levels usually decline significantly within 4–6 months, but remain detectable up to at least 11 months after illness [40]. The titer of antibodies to the S-protein correlates with the frequency of S-specific plasma cells in the bone marrow aspirate, which are at rest. Thus, the presence of long-lived plasma cells (LLPC) and S-specific memory B-cells after COVID-19 was detected. The duration of the humoral immune response is determined by the count and lifespan of memory B-cells and LLPCs in the bone marrow [41].

RATIONAL STRATEGY TO MAINTAIN PROTECTIVE IMMUNITY AGAINST SARS-CoV-2: ENDONASAL IMMUNIZATION

All currently registered SARS-CoV-2 vaccines are administered intramuscularly. Moreover, the mucous membranes of the upper and lower respiratory tracts are the site of entry of SARS-CoV-2; therefore, the local mucosal immune response is very important for protective immunity [42–45]. An alternative non- invasive method of immunization is intranasal vaccination, which is currently actively investigated for the possibility of generating a sterilizing mucosal immune response in COVID-19 [44, 46]. At the moment, several new intranasal vaccine agents are undergoing preclinical and clinical phase I–III trials (Table 2).

 

Table 2

Intranasal vaccines against SARS-CoV-2 undergoing preclinical and clinical trials

Vaccine

candidate name

Vaccine product basis

Developer

CT phase

CT identifier

Source

Vaccines based on recombinant adenoviruses

AZD1222

(ChAdOx1 nCoV-19)

Recombinant viral

vector СhAd expressing S-protein

Imperial College London, University of Oxford AstraZeneca (UK)

I

NCT05007275

NCT04816019

[47]

ChAd-SARS-CoV-2-S

Recombinant viral

vector СhAd expressing stabilized S-protein

Washington University School of Medicine (USA)

I

NCT04751682

[48]

Ad5-nCoV

Recombinant viral

vector Ad5 expressing the RBD domain

of the S-protein

CanSino Biologics Inc. jointly with Beijing Institute of Biotechnology (China)

I/II

NCT04840992

[49, 50]

AdCOVID

Recombinant viral

vector Ad5 expressing the RBD domain of the S-protein

Altimmune, Inc. (USA)

I

NCT04679909

[51]

BBV154

Recombinant viral

vector СhAd expressing stabilized S-protein

Bharat Biotech International Limited (India)

III

NCT05522335

[52]

Gam-COVID-Vac (Sputnik)

Recombinant viral

vector Ad5 expressing S-protein

The Gamaleya National Center of Epidemiology and Microbiology (Russia)

I/II

NCT05248373

[53]

Vaccines based on attenuated viruses

COVI-VAC

Live-attenuated

SARS-CoV-2

CODAGENIX Inc (USA)

I

NCT04619628

[54]

DelNS1-NCoV-RBD LAIV

Live-attenuated

SARS-CoV-2

Beijing Wantai Biological Pharmacy Enterprise jointly with Hong Kong University (China)

I

NCT04809389

[55]

MV-014-212

Live-attenuated respiratory syncytial virus vaccine expressing SARS-CoV-2 S-protein

Meissa Vaccines, Inc. (USA)

I

NCT04798001

[56]

ACM-001

Protein subunit vaccine (ACM-CpG) based

on S-protein from strain B.1.351 and adjuvant CpG7909, packaged in an artificial cell membrane

ACM Biolabs (Singapore)

I

NCT05385991

[57]

CROWNase

SARS-CoV-2 S-protein envelope degrading enzyme

Illinois Institute of Technology (USA)

-

Preclinical study

[58]

CovOMV

Neisseria meningitidis outer membrane vesicles mixed with recombinant S-protein

Intravacc (Netherlands)

-

Preclinical study

[59]

STINGa-

S-trimer in PEGylated liposomes

AuraVax Therapeutics (USA)

-

Preclinical study

[60]

Note: Revised and supplemented from [44]. ChAd — chimpanzee adenovirus; Ad5 — adenovirus 5 serotype.

 

The most promising results are demonstrated by intranasal adenovirus-based vaccines, probably because the mucous membranes are the entry sites of adenoviruses, and viral particles can serve as natural adjuvants for intranasal immunization. Two adenoviral vectors are used as carriers in currently ongoing clinical trials of vaccine agents, namely, Ad5 (AdCOVID [51], Ad5-nCoV [49, 50], and Sputnik V [53]) based on adenovirus serotype 5 and ChAd (AZD1222 [47], ChAd-SARS-CoV-2-S [48], and BBV154 [52]) based on chimpanzee adenovirus.

Preclinical studies of adenovirus vaccines based on Ad5 and ChAd when administered intranasally have shown their ability to induce persistent systemic and local mucosal immunity, characterized by high titers of secretory anti-RBD IgA and serum virus-neutralizing antibodies, increased levels of specific CD4+ T-cells and CD8+ cytotoxic cells, and increased level of Th1 cytokines [48, 51, 52, 61]. Intranasal booster vaccination after intramuscular vaccination induces the development of durable immunity against new strains of SARS-CoV-2, an increase in specific T- and B-cells, including in the secretion of mucous membranes [44, 45].

Phase I/II clinical trials for the safety and efficacy of adenovirus vaccines are currently performed in the US, UK, India, and Russia (Table 2). The Russian intranasal vaccine is being developed based on component 2 (Ad5) of the registered Sputnik V vaccine at the Gamaleya National Center of Epidemiology and Microbiology [53].

Various variants of attenuated viral vaccines can serve as an alternative to adenovirus vaccines (Table 2). An attenuated live vaccine was obtained by passaging SARS-CoV-2 in Vero cells at a temperature reduced from 37°C to 22°C. A single administration of such a vaccine to humanized K18-hACE2 mice, in which SARS-CoV-2 causes a lethal infection, made them insensitive to infection due to a pronounced B- and T-cell immune response and a high titer of secreted IgA [62]. The advantage of a live-attenuated endonasal vaccine based on SARS-CoV-2 is a polyclonal immune response to all antigens of the virus, which can more effectively activate T-cell immunity.

The development of a T-cell immune response is another promising strategy for acquiring long-term immunity covering different SARS-CoV-2 variants. Conserved peptide epitopes of the nucleocapsid (N-protein) may be no less important than the peptide epitopes of the S-protein, and probably more important, for the implementation of the T-cell response, because the latter is the most immunogenic antigen of SARS-CoV-2. In BALB/c mice, intranasal immunization with recombinant adenovirus serotype 5 expressing the SARS-CoV-2 N-protein is accompanied by a significant activation of the T-cell response in the bronchoalveolar tree. Moreover, after such intranasal immunization, specific CD4+ T-cells were detected in the spleen, which, along with an increase in the titer of specific antibodies, indicated the triggering of a systemic humoral immune response [63].

Along with adenoviral vectors and attenuated vaccine agents, new nanotechnology-based vaccine platforms are being actively developed. Liposomal nanoconjugates [57, 60] are tested in preclinical studies, including those with shRNA [64], various organic nanoparticles, for example, nanoparticles based on inulin acetate, a plant polymer that can activate the TLR4 receptor [65], or nanoparticles of chitosan conjugated with RBD, which increases significantly its immunogenicity compared with soluble RBD [66], vesicles based on bacterial membranes [59], and other approaches that activate the immune response.

A key role in the local humoral and cellular immune response on the mucous membranes of the bronchopulmonary tree belongs to cytokines from the pro-inflammatory superfamily tumor necrosis factor, namely, B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL), as well as chemokines CXCL13, CCL19, and CCL21, which induce a local response of T- and B-cells in bronchial lymphoid tissue [43]. The addition of BAFF/APRIL sequences and/or the listed chemokines to the composition of new polyvalent nasal vaccines was assumed to significantly increase the efficiency of the mucosal immune response.

A separate area is the development of polyvalent nasal vaccines of a new-generation. For example, a trivalent vaccine containing the sequences of the S1 domain (RBD+NTD) of the S-protein, full-length nucleocapsid protein, and nsp12 fragment (RNA-dependent RNA polymerase, RdRp) was created based on adenovirus vectors Ad5 and ChAd68 [67]. The S1 domain in the construct was fused to the transmembrane domain of the vesicular stomatitis virus G protein, which provides trimerization and exosomal targeting [68] for a better immune response. The full-length N-protein richest in T-cell peptide epitopes and the selected RdRp fragment, which, according to bioinformatic analysis, exhibits the highest affinity for T-cell receptors, were included in the vaccine to activate cellular immunity. Intranasal (but not intramuscular) immunization with a single dose of such a trivalent vaccine leads to the formation of protective mucosal immunity against both B.1.1.7 and B.1.351 VOCs [67]. Thus, intranasal immunization with new-generation multivalent vaccines may be an effective future vaccination strategy against COVID-19.

IS THE CREATION OF STERILIZING IMMUNITY AGAINST NEW STRAINS REAL?

To analyze mucosal immunity to new SARS-CoV-2 strains in humans, Tang et al. [69] evaluated S-specific total and virus-neutralizing antibodies, as well as B- and T-cell immune responses in bronchoalveolar lavages and in the blood of patients vaccinated with mRNA vaccines and patients who recovered from COVID-19. In vaccinated patients, the levels of neutralizing antibodies against strain B (D614G), strain delta (B.1.617.2), and omicron BA.1.1 in the bronchoalveolar lavage were significantly lower than those in patients who had recovered from COVID-19, despite a comparable virus-neutralizing activity of blood [69]. Notably, vaccination with mRNA vaccines induced circulating S-specific B- and T-cell immunity; however, unlike COVID-19 convalescents, these responses were absent in the bronchoalveolar lavage of vaccinated individuals. By using a mouse immunization model, systemic mRNA vaccination induces weak mucosal immunity, especially against omicron BA.1.1. The combination of systemic mRNA vaccination with endonasal immunization with pseudotyped S-adenovirus induced the production of mucosal virus-neutralizing antibodies not only against delta but also against omicron BA.1.1 and reduced significantly the viral load in experimental animals. Thus, in a rational strategy for developing antiviral immunity to new SARS-CoV-2 strains, endonasal vaccines that create sterilizing immunity in the respiratory tract against SARS-CoV-2, including the latest versions of omicron and new potentially dangerous strains, can come into prominence [69].

CONCLUSION

New SARS-CoV-2 variants, characterized by high contagiousness and the ability to evade virus-neutralizing antibodies, require a new antiviral defense strategy. Such a strategy could be the activation of the mucosal immune response of the bronchoalveolar tree by intranasal and/or inhalation immunization with vector vaccines along with the development of new-generation multivalent vaccines that activate specific B- and T-cells and promote the production of broadly neutralizing secretory antibodies that provide sterile immunity.

ADDITIONAL INFORMATION

Author contribution. Baklaushev V.P. — the concept of the review, literature search, manuscript writing and editing. Yusubalieva G.M., Bychinin M.V. — literature search, manuscript writing and editing. Yusubalieva S.M. — literature search, manuscript editing, preparation of tables. Kalsin V.A. — literature search, manuscript editing. Troitsky A.V. — general guidance, manuscript editing. The authors made a substantial contribution to the conception of the work, acquisition, analysis, interpretation of data for the work, drafting and revising the work, final approval of the version to be published and agree to be accountable for all aspects of the work.

Funding source. The work was carried out with the financial support of the Ministry of Science and Higher Education of the Russian Federation (contract No. 075-15-2021-1086, Contract No. RF----193021X0015, 15.IP.21.0015).

Competing interests. This study was not supported by any external sources of funding.

Acknowledgements. The authors are grateful to A.V. Petrov for preparing the drawing.

 

1 Access mode: https://cov-lineages.org

2 Access mode: https://nextstrain.org

×

About the authors

Vladimir P. Baklaushev

Federal Scientific and Clinical Center for Specialized Medical Assistance and Medical Technologies of the Federal Medical Biological Agency; Pulmonology Scientific Research Institute under Federal Medical and Biological Agency of Russian Federation; Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences

Email: baklaushev.vp@fnkc-fmba.ru
ORCID iD: 0000-0003-1039-4245
SPIN-code: 3968-2971
https://fnkc-fmba.ru/about/komanda-upravleniya/

MD, PhD

Russian Federation, 28, Orekhovy blvd, Moscow, 115682; Moscow; Moscow

Gaukhar M. Yusubalieva

Federal Scientific and Clinical Center for Specialized Medical Assistance and Medical Technologies of the Federal Medical Biological Agency; Pulmonology Scientific Research Institute under Federal Medical and Biological Agency of Russian Federation

Email: gaukhar@gaukhar.org
ORCID iD: 0000-0003-3056-4889
SPIN-code: 1559-5866

MD, PhD

Russian Federation, 28, Orekhovy blvd, Moscow, 115682; Moscow

Mikhail V. Bychinin

Federal Scientific and Clinical Center for Specialized Medical Assistance and Medical Technologies of the Federal Medical Biological Agency

Email: drbychinin@gmail.com
ORCID iD: 0000-0001-8461-4867
SPIN-code: 6524-9947

MD, PhD

Russian Federation, 28, Orekhovy blvd, Moscow, 115682

Saule M. Yusubalieva

Astana Medical University

Email: sm_yusubalieva@mail.ru
ORCID iD: 0000-0002-8258-8148
Kazakhstan, Astana

Vladimir A. Kalsin

Federal Scientific and Clinical Center for Specialized Medical Assistance and Medical Technologies of the Federal Medical Biological Agency; Pulmonology Scientific Research Institute under Federal Medical and Biological Agency of Russian Federation

Email: vkalsin@mail.ru
ORCID iD: 0000-0003-2705-3578
SPIN-code: 1046-8801
Russian Federation, 28, Orekhovy blvd, Moscow, 115682; Moscow

Aleksandr V. Troitskiy

Federal Scientific and Clinical Center for Specialized Medical Assistance and Medical Technologies of the Federal Medical Biological Agency

Author for correspondence.
Email: dr.troitskiy@gmail.com
ORCID iD: 0000-0003-2143-8696
SPIN-code: 2670-6662

MD, PhD

Russian Federation, 28, Orekhovy blvd, Moscow, 115682

References

  1. Xu Y, Wu C, Cao X, et al. Structural and biochemical mechanism for increased infectivity and immune evasion of Omicron BA.2 variant compared to BA.1 and their possible mouse origins. Cell Res. 2022;32(7):609–620. doi: 10.1038/s41422-022-00672-4
  2. Sun Y, Lin W, Dong W, Xu J. Origin and evolutionary analysis of the SARS-CoV-2 Omicron variant. J Biosaf Biosecur. 2022; 4(1):33–37. doi: 10.1016/j.jobb.2021.12.001
  3. Li Q, Zhang M, Liang Z, et al. Antigenicity comparison of SARS-CoV-2 Omicron sublineages with other variants contained multiple mutations in RBD. MedComm. 2022;3(2):e130. doi: 10.1002/mco2.130
  4. Cao Y, Song W, Wang L, et al. Characterizations of enhanced infectivity and antibody evasion of Omicron BA.2.75. bioRxiv. 2022. doi: 10.1101/2022.07.18.500332
  5. Sheward DJ, Kim C, Fischbach J, et al. Evasion of neutralizing antibodies by Omicron sublineage BA.2.75. bioRxiv. 2022. doi: 10.1101/2022.07.19.500716
  6. Saito A, Tamura T, Zahradnik J, et al. Virological characteristics of the SARS-CoV-2 Omicron BA.2.75. bioRxiv. 2022. doi: 10.1101/2022.08.07.503115
  7. Carvalho T, Krammer F, Iwasaki A. The first 12 months of COVID-19: a timeline of immunological insights. Nat Rev Immunol. 2021;21(4):245–256. doi: 10.1038/s41577-021-00522-1
  8. Liu SL, Iketani Y, Guo JF, et al. Striking antibody evasion manifested by the Omicron variant of SARS-CoV-2. Nature. 2022; 602(7898):676–681. doi: 10.1038/s41586-021-04388-0
  9. Chu DK, Abrams EM, Golden DB, et al. Risk of second allergic reaction to SARS-CoV-2 vaccines: a systematic review and meta-analysis. JAMA Intern Med. 2022;182(4):376–385. doi: 10.1001/jamainternmed.2021.8515
  10. Gupta RK, Topol EJ. COVID-19 vaccine breakthrough infections. Science. 2021;374(6575):1561–1562. doi: 10.1126/science.abl8487
  11. Siddle KJ, Krasilnikova LA, Moreno GK, et al. Transmission from vaccinated individuals in a large SARS-CoV-2 Delta variant outbreak. Cell. 2022;185(3):485–492.e10. doi: 10.1016/j.cell.2021.12.027
  12. Mostaghimi D, Valdez CN, Larson HT, et al. Prevention of host-to-host transmission by SARS-CoV-2 vaccines. Lancet Infect Dis. 2022;22(2):e52–e58. doi: 10.1016/S1473-3099(21)00472-2
  13. Белопасов В.В., Яшу Я., Самойлова Е.М., Баклаушев В.П. Поражение нервной системы при СOVID-19 // Клиническая практика. 2020. Т. 11, № 2. C. 60–80. [Belopasov VV, Yashu Y, Samoylova EM, Baklaushev VP. Lesion of the nervous system in COVID-19. J Clin Pract. 2020;11(2):60–80. (In Russ).] doi: 10.17816/clinpract34851
  14. Белопасов В.В., Журавлева Е.Н., Нугманова Н.П., Абдрашитова А.Т. Постковидные неврологические синдромы // Клиническая практика. 2021. Т. 12, № 2. C. 69–82. [Belopasov VV, Zhuravleva EN, Nugmanova NP, Abdrashitova AT. Postcovid neurological syndromes. J Clin Pract. 2021;12(2):69–82. (In Russ).] doi: 10.17816/clinpract71137
  15. Mehandru S, Merad M. Pathological sequelae of long-haul COVID. Nat Immunol. 2022;23(2):194–202. doi: 10.1038/s41590-021-01104-y
  16. Meng B, Abdullahi A, Ferreira IA, et al. Altered TMPRSS2 usage by SARS-CoV-2 Omicron impacts infectivity and fusogenicity. Nature. 2022;603(7902):706–714. doi: 10.1038/s41586-022-04474-x
  17. Desai AD, Lavelle M, Boursiquot BC, Wan EY. Long-term complications of COVID-19. Am J Physiol Cell Physiol. 2022; 322(1):C1–C11. doi: 10.1152/ajpcell.00375.2021
  18. Rambaut A, Holmes EC, O’Toole Á, et al. A dynamic nomenclature proposal for SARS-CoV-2 lineages to assist genomic epidemiology. Nat Microbiol. 2020;5(11):1403–1407. doi: 10.1038/s41564-020-0770-5
  19. Hadfield J, Megill C, Bell SM, et al. Nextstrain: real-time tracking of pathogen evolution. Bioinformatics. 2018;34(23):4121–4123. doi: 10.1093/bioinformatics/bty407
  20. BV-BRC [Internet]. SARS-COV-2 variants and lineages of concern. Available from: https://www.bv-brc.org/view/VariantLineage/. Accessed: 15.06.2022.
  21. Harvey WT, Carabelli AM, Jackson B, et al. SARS-CoV-2 variants, spike mutations and immune escape. Nat Rev Microbiol. 2021;19(7):409–424. doi: 10.1038/s41579-021-00573-0
  22. Mittal A, Khattri A, Verma V. Structural and antigenic variations in the spike protein of emerging SARS-CoV-2 variants. PLoS Pathog. 2022;18(2):e1010260. doi: 10.1371/journal.ppat.1010260
  23. Jackson CB, Farzan M, Chen B, Choe H. Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 2022;23(1): 3–20. doi: 10.1038/s41580-021-00418-x
  24. Cerutti G, Guo Y, Zhou T, et al. Potent SARS-CoV-2 neutralizing antibodies directed against spike N-terminal domain target a single supersite. Cell Host Microbe. 2021;29(5):819–833. e817. doi: 10.1016/j.chom.2021.03.005
  25. Nyberg T, Ferguson NM, Nash SG, et al. Comparative analysis of the risks of hospitalisation and death associated with SARS-CoV-2 omicron (B.1.1.529) and delta (B.1.617.2) variants in England: a cohort study. Lancet. 2022;399(10332):1303–1312. doi: 10.1016/S0140-6736(22)00462-7
  26. Zappa M, Verdecchia P, Angeli F. Knowing the new Omicron BA.2.75 variant (‘Centaurus’): a simulation study. Eur J Intern Med. 2022:S0953-6205(22)00286-2. doi: 10.1016/j.ejim.2022.08.009
  27. Sfera A, Osorio C, Jafri N, et al. Intoxication with endogenous angiotensin II: a COVID-19 hypothesis. Front Immunol. 2020; 11:1472. doi: 10.3389/fimmu.2020.01472
  28. Cele А, Jackson L, Khoury DS, et al. Omicron extensively but incompletely escapes Pfizer BNT162b2 neutralization. Nature. 2021;602(7898):654–656. doi: 10.1038/s41586-021-04387-1
  29. Dejnirattisai W, Huo J, Zhou D, et al. SARS-CoV-2 Omicron-B.1.1.529 leads to widespread escape from neutralizing antibody responses. Cell. 2022;185(3):467–484.e415. doi: 10.1016/j.cell.2021.12.046
  30. Bruel T, Hadjadj J, Maes P, et al. Serum neutralization of SARS-CoV-2 Omicron sublineages BA.1 and BA.2 in patients receiving monoclonal antibodies. Nat Med. 2022;28(6):1297–1302. doi: 10.1038/s41591-022-01792-5
  31. Takashita E, Kinoshita N, Yamayoshi S, et al. Efficacy of antiviral agents against the SARS-CoV-2 Omicron Subvariant BA.2. N Engl J Med. 2022;386(15)1475–1477. doi: 10.1056/NEJMc2201933
  32. Arora P, Kempf A, Nehlmeier I, et al. Augmented neutralisation resistance of emerging omicron subvariants BA.2.12.1, BA.4, and BA.5. Lancet Infect Dis. 2022;22(8):1117–1118. doi: 10.1016/S1473-3099(22)00422-4
  33. Cao Y, Yisimayi A, Jian F, et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by omicron infection. Nature. 2022; 608(7923):593–602. doi: 10.1038/s41586-022-04980-y
  34. Tuekprakhon A, Nutalai R, Dijokaite-Guraliuc A, et al. Antibody escape of SARS-CoV-2 omicron BA.4 and BA.5 from vaccine and BA.1 serum. Cell. 2022;185(14):2422–2433.e13. doi: 10.1016/j.cell.2022.06.005
  35. Medits I, Springer DN, Graninger M, et al. Different neutralization profiles after primary SARS-CoV-2 Omicron BA.1 and BA.2 Infections. Front Immunol. 2022;13:946318. doi: 10.3389/fimmu.2022.946318
  36. Qu P, Faraone J, Evans JP, et al. Neutralization of the SARS-CoV-2 Omicron BA.4/5 and BA.2.12.1 subvariants. N Engl J Med. 2022;386(26):2526–2528. doi: 10.1056/NEJMc2206725
  37. Yamasoba D, Kimura I, Nasser H, et al. Virological characteristics of the SARS-CoV-2 omicron BA.2 spike. Cell. 2022;185(12): 2103–2115.e19. doi: 10.1016/j.cell.2022.04.035
  38. Hachmann NP, Miller J, Collier AY, et al. Neutralization escape by SARS-CoV-2 omicron subvariants BA.2.12.1, BA.4, and BA.5. N Engl J Med. 2022;387(1):86–88. doi: 10.1056/NEJMc2206576
  39. Lustig Y, Nemet I, Kliker L, et al. Neutralizing response against variants after SARS-CoV-2 infection and one dose of BNT162b2. N Engl J Med. 2021;384(25):2453–2454. doi: 10.1056/NEJMc2104036
  40. Turner JS, Kim W, Kalaidina E, et al. SARS-CoV-2 infection induces long-lived bone marrow plasma cells in humans. Nature. 2021;595(7867):421–425. doi: 10.1038/s41586-021-03647-4
  41. Nguyen DC, Lamothe PA, Woodruff MC, et al. COVID-19 and plasma cells: is there long-lived protection? Immunol Rev. 2022;309(1):40–63. doi: 10.1111/imr.13115
  42. Mettelman RC, Allen EK, Thomas PG. Mucosal immune responses to infection and vaccination in the respiratory tract. Immunity. 2022;55(5):749–780. doi: 10.1016/j.immuni.2022.04.013
  43. Alturaiki W. Considerations for novel COVID-19 mucosal vaccine development. Vaccines (Basel). 2022;10(8):1173. doi: 10.3390/vaccines10081173
  44. Dhama K, Dhawan M, Tiwari R, et al. COVID-19 intranasal vaccines: current progress, advantages, prospects, and challenges. Hum Vaccin Immunother. 2022;18(5):2045853. doi: 10.1080/21645515.2022.2045853
  45. 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
  46. Kumar A, Kumar A. Mucosal and transdermal vaccine delivery strategies against COVID-19. Drug Deliv Transl Res. 2022; 12(5):968–972. doi: 10.1007/s13346-021-01001-9
  47. Van Doremalen N, Purushotham JN, Schulz JE, 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(607):eabh0755. doi: 10.1126/scitranslmed.abh0755
  48. Hassan AO, Shrihari S, Gorman MJ, et al. An intranasal vaccine durably protects against SARS-CoV-2 variants in mice. Cell Rep. 2021;36(4):109452. doi: 10.1016/j.celrep.2021.109452
  49. Clinicaltrials.gov. A randomized, double-blind, placebo-controlled phase I/II clinical trial to evaluate the safety and immunogenicity of Ad5-nCov for inhalation in adults 18 years of age and older. CanSino Biologics Inc., 2021. Available from: https://clinicaltrials.gov/ct2/show/NCT04840992. Accessed: 15.06.2022.
  50. Wu S, Huang J, Zhang Z, et al. Safety, tolerability, and immunogenicity of an aerosolised adenovirus type-5 vector-based COVID-19 vaccine (Ad5-nCov) in adults: preliminary report of an open-label and randomised phase 1 clinical trial. Lancet Infect Dis. 2021;21(12):1654–1664. doi: 10.1016/S1473-3099(21)00396-0
  51. King RG, Silva-Sanchez A, Peel JN, et al. Single-dose intranasal administration of AdCOVID elicits systemic and mucosal immunity against SARS-CoV-2 and fully protects mice from lethal challenge. Vaccines (Basel). 2021;9(8):881. doi: 10.3390/vaccines9080881
  52. Hassan AO, Kafai NM, Dmitriev IP, et al. A single-dose intranasal ChAd vaccine protects upper and lower respiratory tracts against SARS-CoV-2. Cell. 2020;183(1):169–184.e13. doi: 10.1016/j.cell.2020.08.026
  53. Safety, tolerability and immunogenicity of Gam-COVID-Vac vaccine in a nasal spray (SPRAY). Available from: https://clinicaltrials.gov/ct2/show/results/NCT05248373. Accessed: 15.06.2022.
  54. Safety and Immunogenicity of COVI-VAC, a live attenuated vaccine against COVID-19. Available from: https://clinicaltrials.gov/ct2/show/NCT04619628. Accessed: 15.06.2022.
  55. A phase 1, randomized, double-blinded, placebo-controlled, dose-escalation and dose-expansion study to evaluate the safety and immunogenicity of DelNS1-NCoV-RBD LAIV for COVID-19 in Healthy Adults. The University of Hong Kong, Hong Kong; 2022. Available from: https://clinicaltrials.gov/ct2/show/NCT04809389. Accessed: 15.06.2022.
  56. Safety and immunogenicity of an intranasal RSV vaccine expressing SARS-CoV-2 spike protein (COVID-19 Vaccine) in adults. Available from: https://clinicaltrials.gov/ct2/show/NCT04798001. Accessed: 15.06.2022.
  57. Lam JH, Shivhare D, Chia TW, et al. Next-generation intranasal Covid-19 vaccine: a polymersome-based protein subunit formulation that provides robust protection against multiple variants of concern and early reduction in viral load of the upper airway in the golden Syrian hamster model. bioRxiv. 2022. doi: 10.1101/2022.02.12.480188
  58. Illinois Institute of Technology. Promising new COVID-19 treatment in development at Illinois Tech. Available from: www.iit.edu/news/promising-new-covid-19-treatment-development-illinois-tech. Accessed: 15.06.2022.
  59. Gaspar EB, Prudencio CR, De Gaspari E. Experimental studies using OMV in a new platform of SARS-CoV-2 vaccines. Hum Vaccines Immunother. 2021;17(9):2965–2968. doi: 10.1080/21645515.2021.1920272
  60. AuraVax Therapeutics licences intranasal vaccine adjuvant technology from Massachusetts General Hospital. Available from: www.oindpnews.com/2021/01/auravax-therapeutics-licences-intranasal-vaccine-adjuvant-technology-from-massachusetts-general-Hosp. Accessed: 15.06.2022.
  61. Kim E, Weisel FJ, Balmert SC, et al. A single subcutaneous or intranasal immunization with adenovirus-based SARS-CoV-2 vaccine induces robust humoral and cellular immune responses in mice. Eur J Immunol. 2021;51(7):1774–1784. doi: 10.1002/eji.202149167
  62. Seo SH, Jang Y. Cold-Adapted live attenuated sars-cov-2 vaccine completely protects human ace2 transgenic mice from sars-cov-2 infection. Vaccines. 2020;8(4):584. doi: 10.3390/vaccines8040584
  63. He J, Huang JR, Zhang YL, Zhang J. SARS-CoV-2 nucleocapsid protein intranasal inoculation induces local and systemic T cell responses in mice. J Med Virol. 2021;93(4):1923–1925. doi: 10.1002/jmv.26769
  64. Acharya R. Prospective vaccination of COVID-19 using shRNA-plasmid-LDH nanoconjugate. Med Hypotheses. 2020;143: 110084. doi: 10.1016/j.mehy.2020.110084
  65. Bakkari MA, Valiveti CK, Kaushik RS, Tummala H. Toll-like receptor-4 (TLR4) agonist-based intranasal nanovaccine delivery system for inducing systemic and mucosal immunity. Mol Pharm. 2021;18(6):2233–2241. doi: 10.1021/acs.molpharmaceut.0c01256
  66. Jearanaiwitayakul T, Seesen M, Chawengkirttikul R, et al. Intranasal administration of RBD nanoparticles confers induction of mucosal and systemic immunity against SARS-CoV-2. Vaccines. 2021;9(7):768. doi: 10.3390/vaccines9070768
  67. Afkhami S, D’Agostino MR, Zhang A, 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(5):896–915.e19. doi: 10.1016/j.cell.2022.02.005
  68. Bliss CM, Parsons AJ, Nachbagauer R, et al. Targeting antigen to the surface of EVs improves the in vivo immunogenicity of human and non-human adenoviral vaccines in mice. Mol Ther Methods Clin Dev. 2020;16:108–125. doi: 10.1016/j.omtm.2019.12.003
  69. Tang J, Zeng C, Cox TM, et al. Respiratory mucosal immunity against SARS-CoV-2 following mRNA vaccination. Sci Immunol. 2022;eadd4853. doi: 10.1126/sciimmunol.add4853

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Fig. 1. Evolution of new variants of SARS-CoV-2 superimposed on the “waves” of the pandemic: а — phylogeny of “variants of concern”; б — histogram of the incidence rates of different variants of SARS-CoV-2 in Europe (according to Gissad, https://gisaid.org/) against the background of the morbidity curve in Russia (according to www.yandex.ru). The figures at peaks indicate the daily increase in cases according to the official data.

Download (1MB)

Copyright (c) 2022 Baklaushev V.P., Yusubalieva G.M., Bychinin M.V., Yusubalieva S.M., Kalsin V.A., Troitskiy A.V.

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 38032 от 11 ноября 2009 года.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies