Open access peer-reviewed chapter

Omicron: A SARS-CoV-2 Variant

Written By

Khushi Gandhi

Submitted: 10 September 2023 Reviewed: 10 September 2023 Published: 08 November 2023

DOI: 10.5772/intechopen.1003043

From the Edited Volume

New COVID-19 Variants - Diagnosis and Management in the Post-Pandemic Era

Ozgur Karcioglu

Chapter metrics overview

41 Chapter Downloads

View Full Metrics

Abstract

COVID-19 is a viral disease caused by SARS-CoV-2. Various variants of SARS-CoV-2 were responsible for causing havoc worldwide resulting in approximately 6.9 million deaths across the globe to date. Since the end of 2021, Omicron (B.1.1.529) has been the recent most variant to be studied and understood to a greater extent. Omicron was found to be the most mutated variant, which enhanced its pathogenic characteristics. Its highly contagious nature and the ability to evade immunity have made it a cause of global concern. The variant also poses a serious risk of reinfection. Furthermore, vaccines developed in response to COVID-19 were found to be less successful with Omicron infections. For the development of targeted vaccines and efficient therapeutic methods, it is necessary to understand the pathogenesis of Omicron. Numerous studies have been conducted to analyze the molecular properties of this variant. This chapter summarizes the biological and molecular properties of this variant and its successive mutations. Further, the clinical traits of the variant, including its pathogenicity, transmissibility and response to body’s immune actions and vaccines are discussed. Precisely examining the mechanisms through which the variant infects and propagates inside the host can aid in preventing the illness and lead to successful management of its global spread.

Keywords

  • Omicron
  • SARS-CoV-2
  • COVID-19
  • mutation
  • transmission
  • pathogenicity
  • immune evasion

1. Introduction

Coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first emerged in Wuhan, China in December 2019. The patients infected with virus showed symptoms similar to severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS), including cough, fever and chest discomfort. In January 2020, the whole genome sequence was performed on the novel coronavirus. The outbreak then reached its peak in China in February and later spread worldwide [1, 2]. Since then, there have been 770+ million confirmed cases and 6.9 million deaths globally [3]. Coronaviruses are RNA viruses belonging to the family Coronaviridae and order Nidovirales. Nidovirales are known to have large genomes, high replicative rate and exhibit various enzymatic activities [2].

Mutations are a natural phenomenon commonly seen during the course of replication. SARS-CoV-2 has a great capacity of undergoing mutations and resulting in antigenic changes over time. It is known to evolve at the rate of one substitution approximately every 11 days [4]. New variants of SARS-CoV-2 have hence emerged through accumulation of these mutations. These variations, however, contribute to increased transmissibility, disease progression and reduced treatment efficacy among other concerns [5]. Specific strains of SARS-CoV-2 have been declared as variants of concern (VOC) due to their highly infectious nature and ability to cause re-infections. These VOCs tend to have mutations that lead to alterations in receptor binding, higher transmissibility, reduced antibody neutralization, decreased therapeutic effects and potentially increased disease severity [6].

The first emerged VOC of SARS-CoV-2 is the Alpha (B.1.1.7) variant. It was first detected in the UK in September 2020. It showcased 14 non-synonymous mutations and three deletions. Eight of these were on the spike protein, resulting in increased affinity towards the ACE2 receptors through which the virus entered the host’s cells. The variant also showed higher viral load and mortality [4, 5, 7]. The Beta (B.1.351) variant was the next VOC, emerging in South Africa in October 2020. It too possessed nine mutations on the spike protein, with three being on the receptor binding domain (RBD). It exhibited increased transmissibility; however, similar effects were not seen in terms of disease severity and fatality [7, 8, 9]. The Gamma (P.1 or B.1.1.28), first detected in Brazil in January 2021, showed 10 mutations on the spike protein, with three on the RBD. The gamma variant significantly reduced treatment efficiency with previously used monoclonal antibody (mAb) cocktail showing reduced neutralization [5, 8, 10]. Fourth VOC of SARS-CoV-2—the Delta (B.1.617.2) emerged in India in May 2021. It had several mutations at the spike protein. Interestingly, along with the RBD, N-terminal domain (NTD) region of the spike protein also showed numerous amino acid alterations. This was not seen in the earlier variants. The Delta variant too showed reduced susceptibility to priorly used antibodies and exhibited a high transmissibility rate [8, 11, 12].

Omicron (B.1.1.529) is the recent most VOC of SARS-CoV-2. It was first reported in South Africa in November 2021. Omicron exhibited far more mutations than any other variant that emerged before it. It also demonstrated a very high transmission rate and by the beginning of December, it had spread worldwide, becoming the dominant variant of SARS-CoV-2. It contributed to 99.7% of the confirmed cases in the months of February and March 2022. Omicron could not only successfully escape immunological responses but also alter them. Antibodies formed from previous infections or vaccines offered little to no protection against the variant. Moreover, Omicron posed a high risk of reinfection. The only relief was the reduced severity of illness and hence reduced mortality. Over time, sub-variants of Omicron too emerged [4, 13, 14, 15, 16]. Specific mutations on structural proteins enabled Omicron to exhibit such attributes. Omicron’s molecular structure, mutations and pathogenic traits are discussed in detail later (Figure 1).

Figure 1.

Evolution of the VoCs of SARS-CoV-2. Mutations in the structural proteins, especially S protein of the SARS-CoV-2 led to the emergence of different variants, which because of their threatening nature, were titled variants of concern. SARS-C0V-2, severe acute respiratory syndrome [17].

Advertisement

2. Omicron at the molecular level

The novel Beta coronavirus, SARS-CoV-2, is an enveloped virus with a single-stranded RNA as the genetic material [18, 19]. It primarily consists of four structural proteins—the membrane (M) protein, the envelope (E) protein, the spike (S) protein and the nucleocapsid (N) protein [20, 21]. The E protein aids in the formation of pentameric ion channels in the virus’s body while the M protein acts as the fundamental structural protein [20, 22]. The N protein plays an essential role in the packaging of the viral RNA in the helical nucleocapsid and formation of the liquid enveloped. It further interacts with the other structural proteins during virion assembly and aids in the virus’s replication [21]. S protein is made up of two subunits—S1 (head) and S2 (stem) [20, 22].

It is the S protein that plays the central role in the binding of the virus with the host’s ACE2 receptors, causing the integration of the membranes of the virus and the host cell [20]. The M protein, through the association with the host’s endoplasmic reticulum, aids in the formation of viral particles, which is performed by the virus’s RNA by using the host’s resources [23]. Thus, the structural proteins (found in the spike, membrane, envelope and the nucleocapsid) contribute to the virulence of SARS-CoV-2 and aid in higher pathogenicity and infectivity [5]. Other than this, the viral genome codes for 16 non-structural and 9 accessory proteins. The non-structural proteins (NSPs) are involved in the replication and translation of the viral RNA while the accessory proteins (ORFs 3a, 3b, 6, 7a, 7b, 8, 9b, 9c and 10) regulate the virus’s interaction with the host and aid in immunity evasion and virulence enhancement [20, 24].

The Omicron variant (B.1.1.529) of SARS-CoV-2, upon genome sequence analysis, was found to have numerous synonymous and non-synonymous mutations [25]. It has been the most mutated variant reported to date, with more than 60 substitutions, deletions and insertions [5, 26]. Most of the mutations were observed in the S protein—with 28 amino acid substitutions, 3 deletions and 1 insertion in the Omicron 21 K lineage [16, 26, 27]. The key mutations in the spike protein are: A67V, del69–70, T95I, del142–144, Y145D, del211, 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 [16, 25]. Figure 2 illustrates the mutations observed in the proteins of the Omicron variant.

Figure 2.

Omicron (B.1.1.529) mutation profile. S protein was found to be heavily mutated. Mutations depicted in red are known to enhance the binding affinity with ACE2 receptors, resulting in an increase in the infectivity of the variant. Mutations in green aid in immune evasion while mutations in blue promote viral replication leading to a high viral load and virulence. ORF, open reading frame; NTD, N-terminal domain; RBD, receptor binding domain; RBM, receptor binding motif; SD 1 and 2, sub-domains 1 and 2; S1/S2, Furin cleavage site; E, envelope; M, membrane; N, Nucleocapsid proteins [25].

These mutations in the S protein have been correlated with disease severity, infectivity and immune evasion [5]. Two mutations have been associated with S protein’s enhanced affinity to ACE2 receptors, thus improving the virus’s attachment to the host cell. Three mutations near the cleavage site may aid in boosting Omicron’s transmissibility [1628]. Moreover, 15 of these mutations have been discovered to be in the RBD (residues 319–541), which is the key target of the neutralizing antibodies (nAb) [16, 25, 29]. However, some studies showed that the current nAb will still bind with the mutated spike protein and the mutated RBD of Omicron had a low predisposition towards the antibodies. Hence, antibodies produced by previous SARS-CoV-2 infections or vaccines may still offer some protection against the Omicron variant [16, 30]. Interestingly Omicron exhibited no mutations in the accessory proteins, which are immunological regulators and are often heavily altered. This indicated the selection pressures on Omicron [31]. Further, while comparing the nucleotide changes in Omicron with other variants, Alpha variant was found to be the closest with an identity percentage of 99.63% [16, 32].

2.1 Subvariants of Omicron

Omicron is further classified into four sub-strains by WHO—BA.1, BA.1.1, BA.2 and BA.3. Each sub-strain has different properties with respect to their transmissibility, pathogenicity, growth and infectivity. BA.1 and BA.2 have significant differences in their mutations contributing to virulent attributes. Out of the two, BA.2 is the more transmittable (33% higher rate) and has a significant growth advantage. Five different mutations in the RBD of the spike protein are responsible for this enhancement [5, 33, 34]. Table 1 summarizes and compares the key characteristics of BA.1 and BA.2 sub-variants.

AttributesBA.1 Sub-variantBA.2 Sub-variant
Unique mutations in the S proteinRBD regionG496ST376A, D405N, R408S,
Non-RBD regionA67V, HV69-, T95I, VYY143-, NL211I, 215EPE, T547K, N856K, L981FT19I, LPP24-26-/A27S, V213G,
Transmissibility and infectivityLowerHigher (33%)
Replication efficacyLowerHigher (1.4-fold)
PathogenicityLowerHigher
Case severitySimilar
Therapeutic and vaccine efficacySimilar

Table 1.

Comparative analysis between the Omicron sub-variants BA.1 and BA.2.

Certain differences in the mutations on the S protein of the sub-variants were observed, which resulted in differences in specific characteristics. This included a higher infectivity rate and pathogenicity in BA.2 sub-variant. However, both the sub-variants resulted in similar severity in cases and had a similar response against various vaccines and treatments [35, 36, 37, 38].

Even though all the sub-variants emerged around the same time, BA.1 dominated the cases worldwide. The reason still remains unclear, although some researchers attribute this to the spike protein variations, which regulate the virus’s entry into the host cell and its further propagation [39]. BA.3 has been predominantly observed in hospitalized patients, indicating higher disease severity. However, it caused the lowest number of cases out of the three [40]. BA.4 and BA.5 have also been identified as Omicron sub-variants [41]. All these variants are under study to understand the immunological resilience of Omicron [5].

Advertisement

3. Transmission of Omicron

SARS-CoV-2 transmits primarily through two methods—direct viral spread or droplet infection [5]. Omicron, similar to the other variants, infects the host cells in the similar fashion. However, it has a greater infectivity rate [27]. Numerous studies reported its high transmissibility, for instance, a study from South Africa reported doubling times of 3.38 d, while a study from the UK reported doubling times of 2–2.5 d, with the basic reproduction number more than 3 [42, 43, 44]. With respect to the Delta variant, Omicron is reported to have a 3.2 times higher transmission rate, with an average of 3 days as the doubling time [35, 45]. The mutations in the spike protein have been thought to contribute in making Omicron extremely contagious [16].

Various pieces of evidence have been provided by different studies to substantiate the above-made claim. For instance, an in vitro Chinese study used an AI model to show that the mutations at locations N440K, N501Y and T478K directly contributed to increased infectivity, with tenfold increase in Omicron with respect to the original SARS-CoV-2 variation and a double increase with respect to the Delta variant [46]. Another study found the combination of mutations N501Y and Q498R significantly enhance the S protein’s binding affinity to the ACE2 receptors [47]. Other mutations such as the N679K, D614G and P681H have also been associated with higher infectivity [16, 48].

Advertisement

4. Pathogenicity of Omicron

SARS-CoV-2 infects humans through different cell types of the respiratory system such as the airway and alveolar epithelium cells, the vascular endothelial cells, etc. by binding to the ACE2 receptor [5, 49]. This binding, as discussed earlier, is facilitated by the spike protein. Studies even found high Omicron infection in cells possessing high number of ACE2 receptors [50]. However, Omicron has been found to follow a different route. Omicron enters through the endosomal pathway, instead of the plasma membrane followed by the other variants. This pathway is enhanced by cathepsins instead of TMPRSS2 [51, 52].

A study found that the infection by Omicron was more significant in the cells, which exhibited a lower TMPRSS2 expression. This further indicated that Omicron followed the endosomal entry pathway [53]. This altered entry pathway has been associated with better success in the virus’s replication in the upper respiratory tract [54]. Omicron has also been observed to have a lower cleavage efficiency in comparison to the other emerged variants such as the Gamma and the Alpha variant. Mutations like the N679K and H655Y are the primary cause of the same [35, 55].

Other mutations in the N and S proteins have been linked to the variant’s enhanced cellular permeability. These mutations also aid in more efficient capsid assembly, found to be three-fold better than the Delta variant [56]. The sub-variants of Omicron (BA.1 and BA.2) do not form syncytia (normally produced at the boundary of S1 and S2 during the processing of the S protein), owing to the numerous changes in the S protein. These alterations, combined with the altered entry pathway result in different cellular tropism [35, 54].

Viral load may also be associated with Omicron’s high transmissibility and growth rate. A higher viral load was reported in the upper airway (specifically the nose, trachea and throat). This aggregation of the viral particles in the upper respiratory tract may result from higher growth and increased viral load [35]. Figure 3 illustrates the difference in observed viral load in Omicron in comparison to the other variants.

Figure 3.

Comparison of observed viral load in omicron with the SARS-CoV-2 wild type. The Omicron variant tends to accumulate in the upper respiratory tract, exhibiting higher viral load in the nasal region and larynx. While, the SARS-Cov-2 wild type as well as the other variants settle in the lower respiratory tract, resulting in a higher viral load in the lungs. SARS-CoV-2, severe acute respiratory syndrome [35].

Omicron also has the ability to cause reinfection [42]. Data from South Africa showed that the reinfection rates increased significantly during the Omicron wave in comparison to the Beta and Delta waves [57]. In the UK, it was reported that there was 6 times higher risk of reinfection with Omicron with respect to the Delta variant. A prior COVID-19 infection provided 80% protection against the Delta variant, however, only 19% protection was observed against the Omicron variant [44]. The good news is that fewer cases of Omicron infections required hospitalization, while the frequency of severe cases was more in Alpha and Delta infections [25].

Advertisement

5. Body’s response to infection

Upon infection with SARS-CoV-2, the innate immune cells, primarily consisting of the neutrophils and the macrophages, identify the virus and activate the immune response. Cytokines and chemokines such as Interleukin-6 (IL-6), monocyte chemoattractant protein-1 and Interferon γ (IFN-γ) are then secreted by these immune cells. Monocytes, neutrophils and macrophages, attracted by the released chemicals, reach the site of infection and release cytotoxic chemicals to kill the virus. The virus is fought by cytotoxic CDB+ T cells, CD4+ T helper cells and B cells. The CDB+ T cells eradicate the virus through cytotoxins while the B cells produce nAb. The cellular reaction is efficient and long-lasting and also triggers immuno-protective memory [5, 58, 59, 60, 61]. A study identified the 14 neutralizing IgG antibodies from SARS-CoV-2 infected patients, with BD-368-2 being the most potent [62]. Other antibodies recovered were IgI and IgM. These antibodies were specific to RBD suggesting that anti-RBD antibodies can be used as clinical interventions against SARS-CoV-2 infections [63].

The Omicron variant triggers exactly the same immune reactions as that seen in the wild type of SARS-CoV-2 variant. The infection results in the production of cytotoxic and follicular T helper cells, along with RBD and spike-related IgG+ B cells and memory B cells. The cytotoxic T cells - CDB+ T cells produce proteases and induce apoptosis in infected cells. On the other hand, the B cells derived from the pool of live memory cells interact with the spike protein [5, 35, 53, 64].

5.1 Clinical manifestations of Omicron

Clinical symptoms of the Omicron infection were different from the earlier variants. Most prevalent symptoms included a cough, nasal congestion, sore throat and fatigue. Loss of smell and taste, fever, dizziness, headaches, etc. were less prevalent with respect to the Delta variant [65, 66]. The virus also affects the organs, which extensively express ACE2 such as glandular cells of the gastric, duodenal and rectal epithelium [67]. A study found that the acute symptoms lasted for fewer days in the Omicron outbreak with an average of 6.87 days, while during the Delta wave, they lasted for an average of 8.89 days [68]. Omicron infection was also suggested to exhibit milder symptoms with a lot of the cases being asymptomatic and majority requiring no hospitalization. In total, 2.6% of the Delta variant infected patients were admitted to hospitals while only 1.9% of Omicron-infected patients required hospitalization [35, 68, 69]. A study found that out of the Omicron-infected individuals, 36.1% did not exhibit any antibody response, while 62.7% produced IgG and only 1.2% produced IgM along with IgG [70]. Omicron-infected patients exhibited various hematological disorders such as lymphocytopenia, neutrophilia, anemia, erythrocytopenia, thrombocytopenia etc. These can be used as biomarkers in Omicron infections for a better prognosis [71].

5.2 Severe cases of Omicron infection

In vivo investigations revealed that lower viral load, smaller lung damage and lower mortality rates were observed in Omicron infections in comparison to the other variants [72, 73]. As discussed earlier, the viral load tends to settle in the upper respiratory tract and not so much in the lungs [74]. However, severe cases of Omicron can result in pneumonia and respiratory failure and can even be fatal [75, 76]. Significant comorbidities, including hypertension, bronchitis and diabetes, have been observed in Omicron infections [35].

The major damage to the tissues in COVID-19, however, is caused majorly by the host’s immune reactions and not directly by the virus itself. The host’s immune reactions are crucial in the pathophysiology of COVID-19 and are directly connected with the disease’s progression. Severe SARS-CoV-2 infections have been shown to dysregulate the host’s normal immune responses. As discussed earlier, the infection leads to numerous hematological disorders such as leukocytosis, lymphocytopenia and a high neutrophil-lymphocyte ratio (NLR). This is the first sign of immunological dysregulation, resulting in uncontrolled inflammatory reactions. These processes lead to immune cells’ damage and exaggerated responses by macrophages, eventually resulting in cell death and tissue damage due to the release of cytotoxic substances. Its manifestations are also seen as cytokine storms and organ failures [5, 71, 77, 78, 79].

Cytokine release syndrome (CRS) is often observed in severe cases of COVID-19. CRS is a hyperinflammatory condition resulting from augmented release of inflammatory cytokines such as tumor necrosis factor (TNF), IL -1, −6, −12, −18, − 33 and IFN -I and -II, along with certain chemokines, including CCL -2, −3, −5 and CXCL -8, −9, 10 [77, 80, 81]. CRS results in acute respiratory distress syndrome, characterized by alveolar epithelial and endothelial destruction in lungs followed by breathing difficulty, hypoxemia and pulmonary oedema. This causes organ failure and also becomes fatal in severe cases [5, 82, 83].

5.3 Immune evasion by Omicron

Omicron has exhibited the ability to alter the host’s immune responses. Some mutations enable the variant to weaken the T cells’ capacity to identify and destroy the infected cells. In some instances, Omicron has also been observed to escape the nAb produced from previous infections or vaccines. However, the existing nAb still provides protection against severe illnesses [16, 84, 85]. Certain viral proteins such as the Orf3 and Orf6 were also observed to alter the T cell responses, which affected the IFN activations. Omicron also brought on T cell immunity decline through immunological fatigue and memory deficits. All this resulted in easier reproduction and propagation of the variant [16, 86, 87].

The understanding of the immunopathogenesis of SARS-CoV-2 aided in developing efficient treatment therapies. However, the precise mechanisms through which the Omicron infection progresses are unclear. For the development of Omicron-specific immunological therapies, it is necessary to analyze the distinct pathophysiology during Omicron infections [5, 88].

Advertisement

6. Omicron diagnosis

Provisionally, the infection can be diagnosed according to the symptoms. It is confirmed using reverse transcription polymerase chain reaction (RT-PCR) or other nucleic acid tests. Serological assays can be used to detect a past infection. They detect the antibodies that were produced by the body in response to the infection. Other laboratory tests can further help in detecting the severity of the infection such as lactate dehydrogenase (LDH), C-reactive protein (CRP), ferritin and IL-6 levels. Along with laboratory testing, chest CT scans may be helpful to diagnose COVID-19 in individuals with a high clinical suspicion of infection [89, 90].

For the Omicron variant, RT-PCR still remains the gold standard. The RT-PCR diagnostics kits primarily focus on the E, Rp, Rd. and N genes. The S gene mutation observed in the case of Omicron variant may or may not cause a positive test [5, 91, 92]. To identify and classify the variant, next generation sequencing (NGS) is required. NGS also aids in determining the origin of SARS-CoV-2. It also has the opportunity to rebuild whole viral genomes. However, unlike PCR, NGS is expensive, time-consuming and requires expertise for interpretation [5, 93, 94]. Serological assays use S or N protein-specific antibodies. These assays also provide insights into the disease severity—with higher amounts of IgG, IgM and IgI antibodies being reported in severe cases [95].

Advertisement

7. Omicron treatment and prevention

Important therapeutics for SARS-CoV-2 infection were mAb cocktails such as casirivimab-imdevimab, bamlanivimab-etesevimab that aided in preventing severe disease or hospitalization. These antibodies target the spike protein for neutralization because, as stated before, it is the spike protein that is responsible for the virus’s entry into the host cells by binding to the ACE2 receptors. However, 15 key mutations in the RBD area on the S protein in the Omicron variant may prevent the antibodies from binding with the virus [16, 42]. RBS-A, RBS-B, RBS-C, CR302 and S309 are some of the key targets of the nAb which were replaced in the Omicron variant [96]. This caused scientists worldwide to question the efficacy of such antibodies. In vitro studies too suggested that the Omicron variant is successful in completely escaping neutralization by these antibodies. Nevertheless, mAbs that target sites beyond the RBD, such as sotrovimab and cilgavimab/tixagevimab, have been reported to still remain effective against Omicron in similar studies. Another authorized mAb—bebtelovimab too retained its in vitro activity against the Omicron sub-strains BA.1 and BA.2 [16, 42, 97, 98, 99].

Other antiviral options have also been inspected for emergency use in severe Omicron cases to reduce mortality. Some antivirals proposed are Molnupiravir, Remdesivir, Ensovibep and Camostat. Upon assessment of the above-mentioned and other antiviral molecules against Omicron and its sub-variants, three compounds were found to be particularly efficient. They were Remdesivir, Lufotrelvir and Molnupiravir. Currently, the best antiviral option against omicron is a combination of Nirmatrelvir-Ritonavir. Numerous studies have corroborated the same by analyzing its activity against the variant [35, 100].

7.1 Booster vaccines

The proposal of using booster vaccines was first suggested when in vitro studies found an increase of neutralizing antibody titers against omicron in the serum of individuals provided with the booster dose. Vaccines like BNT162B2/mRNA-1273 (Moderna) and Ad26.COV2.S vaccines were given as boosters which resulted in increase of the nAb titers from 122-fold/44-fold lower against omicron to 4−/sixfold lower. Simultaneously, half of the population in the UK were also given booster doses. These booster doses aided in increasing the vaccine’s effectiveness against symptomatic infections. They also helped in bringing down severe cases, which required hospitalization. Usually, the nAb response used to wane after a couple of months after the second does. This was restored by the booster vaccine [35, 101, 102].

The Technical Advisory Group on COVID-19 Vaccine Composition (TAG-CO-VAC) advised on globalized access of existing COVID-19 vaccines for booster doses, in light of the transmission of Omicron Variant of Concern (VOC). They suggested the vaccine makers to generate and share data on the vaccine’s efficacy against the Omicron variant specifically, along with the other variants. To make the booster doses effective and timely available, the TAG-CO-VAC authorities collaborated with WHO and its expert groups [16].

7.2 Need for new vaccines

As discussed earlier, the Omicron variant is known to escape the mAb, which was effective against the previous variants. Individuals without the booster dose were not able to protect themselves against the variant. Moreover, the booster vaccine also only provided partial protection against it. This indicated the decreasing efficacy of the existing vaccines. Prevalent mutations in Omicron such as Q498R, S477N, Y505H, G496H, T478K, N501Y and E484K are primarily responsible for altering the antigenicity and providing the variant with the capability to evade immune responses. The extensive alterations on the S protein induce stearic hindrance, which in turn affect the antigen–antibody binding. Further, these alterations are unprecedented, making antigen recognition difficult for the antibodies [35, 103, 104, 105].

Hence, several researchers advocated the need for development of novel vaccines, particularly targeting the Omicron variant. Numerous vaccines, predominantly bivalent and RNA vaccines, have since been developed by different researchers or pharmacological companies [17, 35]. A few of the clinical trials working on Omicron vaccines are summarized in Table 2.

S.No.Biological interventionsCountry of originPhaseClinical trial numberStudy title
1Comirnaty® BNT162b2/Omicron BA.4-5 vaccine (Pfizer-BioNTech)
VidPrevtyn® Beta vaccine (Sanofi/GSK)
FranceIIINCT05749926Immunogenicity and Reactogenicity of the Beta-variant Recombinant Protein Booster Vaccine (VidPrevtyn Beta, Sanofi) Compared to a Bivalent mRNA Vaccine (Comirnaty Original/Omicron BA.4-5, BioNTech-Pfizer) in Adults Previously Vaccinated With at Least 3 Doses of COVID-19 mRNA Vaccine
2Omicron COVID-19 Vaccine (Vero Cell), Inactivated
COVID-19 Vaccine (Vero Cell), Inactivated
ChinaIIINCT05374954Study on Sequential Immunization of Omicron Inactivated COVID-19 Vaccine and Prototype Inactivated COVID-19 Vaccine in Population Aged 18 Years Old and Above
3Bivalent BNT162b2 mRNA original/omicron BA.4-5 vaccine
Quadrivalent influenza vaccine
Republic of KoreaIIIINCT05970887Immunogenicity and Safety of Concomitant Administration of Bivalent COVID-19 Vaccines With Influenza Vaccines
4Drug: NVX-CoV2515
Drug: NVX-Cov2373
Drug: NVX-CoV2373+NVX-CoV2515
Drug: NVX-CoV2540
Drug: NVX-CoV2373+NVX-CoV2540
AustraliaIIINCT05372588Phase 3 Boosting Study for the SARS-CoV-2 rS Variant Vaccines
5BIBP Omicron Inactivated COVID-19 vaccine
WIBP Omicron Inactivated COVID-19 vaccine (Vero Cell)
COVID-19 Vaccine (Vero Cell), Inactivated
Hong KongIIINCT05382871Sequential Immunization of Two Doses of Inactivated COVID-19 Vaccine (Omicron) in Vaccinated Population Aged 18 Years and Above
6SCTV01C
SCTV01E
Sinopharm inactivated COVID-19 vaccine
Comirnaty
UAEIIINCT05323461A Study to Evaluate the Immunogenicity and Safety of Two Recombinant Protein COVID-19 Vaccines in Population Aged ≥18 Years as Booster Vaccines
7ABO1020
Placebo
IndonesiaII / IIINCT05636319A Study to Evaluate the Efficacy, Safety, and Immunogenicity of SARS-CoV-2 Variant (BA.4 /5) mRNA Vaccine
8BNT162b2 vaccineIsraelIVNCT05231005Fourth BNT162b2 COVID-19 Vaccine Dose
9mRNA1273 vaccineIsraelIIINCT05230953Fourth COVID-19 Vaccine Dose- mRNA1273
10mRNA-1273
Placebo
mRNA-1273.222
USAII /IIINCT04649151A Study to Evaluate the Safety, Reactogenicity, and Effectiveness of mRNA-1273 Vaccine in Adolescents 12 to <18 Years Old to Prevent COVID-19

Table 2.

Phase III and IV clinical trials studying novel vaccines against the omicron variant. All of these studies are currently active and not recruiting [106].

These vaccines show potential in providing protection against Omicron and its sub-variants. Currently, they are under clinical trials for safety and efficacy testing. Numerous studies are under different phases of trials to create a potent vaccine which not only offers protection against the omicron variant but also prevents the need for booster doses [16, 35].

Advertisement

8. Conclusion and future prospects

The COVID-19 pandemic resulted in a global public health emergency. The virus’s capability to mutate and evolve rapidly became a threatening concern. To keep up with the pace of the virus, the scientific community had to work round the clock for better diagnosis, precise vaccines and treatment methods. While the new Omicron variant exhibited a reduced disease severity, it posed its own set of challenges. Higher transmissibility, increased infectious rate, immune evasion and partial resistance to monoclonal as well as vaccine-induced antibodies are the key challenges that were to be faced. These complications necessitated the development of new technology and alternate approaches to eradicate the virus.

New variants of SARS-CoV-2 were expected to emerge and the virus is still mutating, each time presenting new risks and challenges. Only traditional approaches are not enough to face this challenge, alternate methods have to be incorporated such as the use of bioinformatics and nanotechnology for development of better drugs and/or vaccines. New data are being generated every day by numerous ongoing studies; however, data interpretation and application require time. Persistent and continued research is necessary for a better understanding of the pathogenesis of the new variants, which will eventually lead to the development of efficient preventive and treatment methods.

Advertisement

Acknowledgments

I would like to acknowledge my professors and mentors, especially Dr. Gayatri Sharma at Amity Institute of Biotechnology, Amity University, Noida, India for their guidance and counsel that helped in completing this chapter.

Advertisement

Conflict of interest

The author declares no conflict of interest.

References

  1. 1. Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS-CoV-2 and COVID-19. Nature Reviews. Microbiology. 2021;19(3):141-154
  2. 2. Umakanthan S, Sahu P, Ranade AV, Bukelo MM, Rao JS, Abrahao-Machado LF, et al. Origin, transmission, diagnosis and management of coronavirus disease 2019 (COVID-19). Postgraduate Medical Journal. 2020;96(1142):753-758
  3. 3. World Health Organization. WHO Coronavirus (COVID-19) Dashboard [Internet] [cited 2019 Sep 9]. 2019. Available from: https://covid19.who.int/?adgroupsurvey={adgroupsurvey}&gclid=CjwKCAjwjOunBhB4EiwA94JWsLp1buk5veij5ws1d2h-DS0_O8N_ZEbVaAVVTBBxYnefjJmUseHeOBoCEiwQAvD_BwE
  4. 4. Fernandes Q , Inchakalody VP, Merhi M, Mestiri S, Taib N, Moustafa Abo El-Ella D, et al. Emerging COVID-19 variants and their impact on SARS-CoV-2 diagnosis, therapeutics and vaccines. Annals of Medicine. 2022;54(1):524-540
  5. 5. Bazargan M, Elahi R, Esmaeilzadeh A. OMICRON: Virology, immunopathogenesis, and laboratory diagnosis. The Journal of Gene Medicine. 2022;24(7):e3435
  6. 6. Centers for Disease Control and Prevention. SARS-CoV-2 Variant Classifications and Definitions [Internet] [cited 2023 Sep 9]. 2019. Available from: https://www.cdc.gov/coronavirus/2019-ncov/variants/variant-classifications.html
  7. 7. Zhang Y, Zhang H, Zhang W. SARS-CoV-2 variants, immune escape, and countermeasures. Frontiers in Medicine. 2022;16(2):196-207
  8. 8. Choi JY, Smith DM. SARS-CoV-2 variants of concern. Yonsei Medical Journal. 2021;62(11):961
  9. 9. Rajah MM, Hubert M, Bishop E, Saunders N, Robinot R, Grzelak L, et al. SARS-CoV-2 alpha, beta, and delta variants display enhanced spike-mediated syncytia formation. The EMBO Journal. 2021;40(24):e108944
  10. 10. Magazine N, Zhang T, Wu Y, McGee MC, Veggiani G, Huang W. Mutations and evolution of the SARS-CoV-2 spike protein. Viruses. 2022;14(3):640
  11. 11. McCrone JT, Hill V, Bajaj S, Pena RE, Lambert BC, Inward R, et al. Context-specific emergence and growth of the SARS-CoV-2 Delta variant. Nature. 2022;610(7930):154-160
  12. 12. Zhan Y, Yin H, Yin JY. B.1.617.2 (Delta) variant of SARS-CoV-2: Features, transmission and potential strategies. International Journal of Biological Sciences. 2022;18(5):1844-1851
  13. 13. Wang ML, Lin Y, Hou JF, Yang YP, Chien Y, Sun YC, et al. The Omicron variant wave: Where are we now and what are the prospects? Journal of the Chinese Medical Association. 2023;86(2):135-137
  14. 14. Viana R, Moyo S, Amoako DG, Tegally H, Scheepers C, Althaus CL, et al. Rapid epidemic expansion of the SARS-CoV-2 Omicron variant in southern Africa. Nature. 2022;603(7902):679-686
  15. 15. Guo Y, Han J, Zhang Y, He J, Yu W, Zhang X, et al. SARS-CoV-2 Omicron variant: Epidemiological features, biological characteristics, and clinical significance. Frontiers in Immunology. 2022;13:877101
  16. 16. Rana R, Kant R, Huirem RS, Bohra D, Ganguly NK. Omicron variant: Current insights and future directions. Microbiological Research. 2022;265:127204
  17. 17. Chia TRT, Young BE, Chia PY. The Omicron-transformer: Rise of the subvariants in the age of vaccines. Annals of the Academy of Medicine, Singapore. 2022;51(11):712-729
  18. 18. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, et al. A novel coronavirus from patients with pneumonia in China, 2019. New England Journal of Medicine. 2020;382(8):727-733
  19. 19. Wu C, Chao YW, Jiang Y, Xu HE. Structure genomics of SARS-CoV-2 and its Omicron variant: Drug design templates for COVID-19. Acta Pharmacologica Sinica. 2022;43(12):3021-3033
  20. 20. Bai C, Zhong Q , Gao GF. Overview of SARS-CoV-2 genome-encoded proteins. Science China. Life Sciences. 2022;65(2):280-294
  21. 21. Cao Y, Yang R, Lee I, Zhang W, Sun J, Wang W, et al. Characterization of the SARS-CoV-2 E protein: Sequence, structure, viroporin, and inhibitors. Protein Science. 2021;30(6):1114-1130
  22. 22. Mohamadian M, Chiti H, Shoghli A, Biglari S, Parsamanesh N, Esmaeilzadeh A. COVID-19: Virology, biology and novel laboratory diagnosis. The Journal of Gene Medicine. 2021;23(2):e3303
  23. 23. Kannan S, Shaik Syed Ali P, Sheeza A. Evolving biothreat of variant SARS-CoV-2 – molecular properties, virulence and epidemiology. European Review for Medical and Pharmacological Sciences. 2021;25(12):4405-4412
  24. 24. Arya R, Kumari S, Pandey B, Mistry H, Bihani SC, Das A, et al. Structural insights into SARS-CoV-2 proteins. Journal of Molecular Biology. 2021 Jan;433(2):166725
  25. 25. Tian D, Sun Y, Xu H, Ye Q. The emergence and epidemic characteristics of the highly mutated SARS-CoV-2 Omicron variant. Journal of Medical Virology. 2022;94(6):2376-2383
  26. 26. Shang J, Ye G, Shi K, Wan Y, Luo C, Aihara H, et al. Structural basis of receptor recognition by SARS-CoV-2. Nature. 2020;581(7807):221-224
  27. 27. Callaway E. Heavily mutated Omicron variant puts scientists on alert. Nature. 2021;600(7887):21-21
  28. 28. Bayarri-Olmos R, Jarlhelt I, Johnsen LB, Hansen CB, Helgstrand C, Rose Bjelke J, et al. Functional effects of receptor-binding domain mutations of SARS-CoV-2 B.1.351 and P.1 variants. Frontiers in Immunology. 2021;12
  29. 29. Wang L, Cheng G. Sequence analysis of the emerging SARS-CoV-2 variant Omicron in South Africa. Journal of Medical Virology. 2022;94(4):1728-1733
  30. 30. Ford CT, Jacob Machado D, Janies DA. Predictions of the SARS-CoV-2 Omicron variant (B.1.1.529) spike protein receptor-binding domain structure and neutralizing antibody interactions. Frontiers in Virology. 2022;2
  31. 31. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181(2):271-280.e8
  32. 32. Kandeel M, Mohamed MEM, Abd El-Lateef HM, Venugopala KN, El-Beltagi HS. Omicron variant genome evolution and phylogenetics. Journal of Medical Virology. 2022;94(4):1627-1632
  33. 33. Lyngse FP, Kirkeby CT, Denwood M, Christiansen LE, Mølbak K, Møller CH, et al. Household transmission of SARS-CoV-2 Omicron variant of concern subvariants BA.1 and BA.2 in Denmark. Nature Communications. 2022;13(1):5760
  34. 34. Chen J, Wei GW. Omicron BA.2 (B.1.1.529.2): High potential to becoming the next dominating variant. 2022
  35. 35. Chatterjee S, Bhattacharya M, Nag S, Dhama K, Chakraborty C. A detailed overview of SARS-CoV-2 Omicron: Its sub-variants, mutations and pathophysiology, clinical characteristics, immunological landscape, immune escape, and therapies. Viruses. 2023;15(1):167
  36. 36. Lewnard JA, Hong VX, Patel MM, Kahn R, Lipsitch M, Tartof SY. Clinical outcomes associated with SARS-CoV-2 Omicron (B.1.1.529) variant and BA.1/BA.1.1 or BA.2 subvariant infection in Southern California. Nature Medicine. 2022;28(9):1933-1943
  37. 37. Kawaoka Y, Uraki R, Kiso M, Iida S, Imai M, Takashita E, et al. Characterization and antiviral susceptibility of SARS-CoV-2 Omicron/BA.2. Research Sequence. 2022
  38. 38. Yamasoba D, Kimura I, Nasser H, Morioka Y, Nao N, Ito J, et al. Virological characteristics of the SARS-CoV-2 Omicron BA.2 spike. Cell. 2022;185(12):2103-2115.e19
  39. 39. Harvey WT, Carabelli AM, Jackson B, Gupta RK, Thomson EC, Harrison EM, et al. SARS-CoV-2 variants, spike mutations and immune escape. Nature Reviews. Microbiology. 2021;19(7):409-424
  40. 40. Desingu PA, Nagarajan K, Dhama K. Emergence of Omicron third lineage BA.3 and its importance. Journal of Medical Virology. 2022;94(5):1808-1810
  41. 41. Cao Y, Yisimayi A, Jian F, Song W, Xiao T, Wang L, et al. BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. Nature. 2022;608(7923):593-602
  42. 42. Singhal T. The emergence of Omicron: Challenging times are Here again! Indian Journal of Pediatrics. 2022;89(5):490-496
  43. 43. Grabowski F, Kochańczyk M, Lipniacki T. The spread of SARS-CoV-2 variant Omicron with a doubling time of 2.0-3.3 days can Be explained by immune evasion. Viruses. 2022;14(2):294
  44. 44. Ferguson N, Ghani A, Cori A, Hogan A, Hinsley W, Volz E. Report 49: Growth, population distribution and immune escape of Omicron in England. Imperial College London. WHO Collaborating Centre for Infectious Disease Modelling, MRC Centre for Global Infectious Disease Analysis, Jameel Institute; 2021
  45. 45. Long B, Carius BM, Chavez S, Liang SY, Brady WJ, Koyfman A, et al. Clinical update on COVID-19 for the emergency clinician: Presentation and evaluation. The American Journal of Emergency Medicine. 2022;54:46-57
  46. 46. Chen J, Wang R, Gilby NB, Wei GW. Omicron variant (B.1.1.529): Infectivity, vaccine breakthrough, and antibody resistance. Journal of Chemical Information and Modeling. 2022;62(2):412-422
  47. 47. Zahradník J, Marciano S, Shemesh M, Zoler E, Harari D, Chiaravalli J, et al. SARS-CoV-2 variant prediction and antiviral drug design are enabled by RBD in vitro evolution. Nature Microbiology. 2021;6(9):1188-1198
  48. 48. Hodcroft EB. CoVariants: SARS–CoV–2 Mutations and Variants of Interest [Internet] [cited 2023 Sep 9]. 2021. Available from: https://covariants.org/
  49. 49. Shi Y, Wang Y, Shao C, Huang J, Gan J, Huang X, et al. COVID-19 infection: The perspectives on immune responses. Cell Death and Differentiation. 2020;27(5):1451-1454
  50. 50. Callaway E. Are COVID surges becoming more predictable? New Omicron variants offer a hint. Nature. 2022;605(7909):204-206
  51. 51. Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science (1979). 2020;367(6483):1260-1263
  52. 52. Pia L, Rowland-Jones S. Omicron entry route. Nature Reviews. Immunology. 2022;22(3):144-144
  53. 53. Willett BJ, Grove J, MacLean OA, Wilkie C, De Lorenzo G, Furnon W, et al. SARS-CoV-2 Omicron is an immune escape variant with an altered cell entry pathway. Nature Microbiology. 2022;7(8):1161-1179
  54. 54. Syed AM, Ciling A, Taha TY, Chen IP, Khalid MM, Sreekumar B, et al. Omicron mutations enhance infectivity and reduce antibody neutralization of SARS-CoV-2 virus-like particles. Proceedings of the National Academy of Sciences. 2022;119(31):e2200592119
  55. 55. Peacock TP, Goldhill DH, Zhou J, Baillon L, Frise R, Swann OC, et al. The furin cleavage site in the SARS-CoV-2 spike protein is required for transmission in ferrets. Nature Microbiology. 2021;6(7):899-909
  56. 56. Du X, Tang H, Gao L, Wu Z, Meng F, Yan R, et al. Omicron adopts a different strategy from Delta and other variants to adapt to host. Signal Transduction and Targeted Therapy. 2022;7(1):45
  57. 57. Wolter N, Jassat W, Walaza S, Welch R, Moultrie H, Groome M, et al. Early assessment of the clinical severity of the SARS-CoV-2 Omicron variant in South Africa: A data linkage study. The Lancet. 2022;399(10323):437-446
  58. 58. Mortaz E, Tabarsi P, Varahram M, Folkerts G, Adcock IM. The immune response and immunopathology of COVID-19. Frontiers in Immunology. 2020;11
  59. 59. Schultze JL, Aschenbrenner AC. COVID-19 and the human innate immune system. Cell. 2021;184(7):1671-1692
  60. 60. Valizadeh H, Abdolmohammadi-vahid S, Danshina S, Ziya Gencer M, Ammari A, Sadeghi A, et al. Nano-curcumin therapy, a promising method in modulating inflammatory cytokines in COVID-19 patients. International Immunopharmacology. 2020;89:107088
  61. 61. Azkur AK, Akdis M, Azkur D, Sokolowska M, van de Veen W, Brüggen M, et al. Immune response to SARS-CoV-2 and mechanisms of immunopathological changes in COVID-19. Allergy. 2020;75(7):1564-1581
  62. 62. Cao Y, Su B, Guo X, Sun W, Deng Y, Bao L, et al. Potent neutralizing antibodies against SARS-CoV-2 identified by high-throughput single-cell sequencing of convalescent patients’ B cells. Cell. 2020;182(1):73-84.e16
  63. 63. Ju B, Zhang Q , Ge J, Wang R, Sun J, Ge X, et al. Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature. 2020;584(7819):115-119
  64. 64. Kared H, Wolf AS, Alirezaylavasani A, Ravussin A, Solum G, Tran TT, et al. Immune responses in Omicron SARS-CoV-2 breakthrough infection in vaccinated adults. Nature Communications. 2022;13(1):4165
  65. 65. Kupferschmidt K, Vogel G. How bad is Omicron? Some clues are emerging. Science (1979). 2021;374(6573):1304-1305
  66. 66. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. The Lancet. 2020;395(10223):507-513
  67. 67. Zheng YY, Ma YT, Zhang JY, Xie X. COVID-19 and the cardiovascular system. Nature Reviews. Cardiology. 2020;17(5):259-260
  68. 68. Menni C, Valdes AM, Polidori L, Antonelli M, Penamakuri S, Nogal A, et al. Symptom prevalence, duration, and risk of hospital admission in individuals infected with SARS-CoV-2 during periods of Omicron and delta variant dominance: A prospective observational study from the ZOE COVID study. The Lancet. 2022;399(10335):1618-1624
  69. 69. Garrett N, Tapley A, Andriesen J, Seocharan I, Fisher LH, Bunts L, et al. High rate of asymptomatic carriage associated with variant strain Omicron. medRxiv. 2022;399:1618-1624
  70. 70. Halfmann PJ, Iida S, Iwatsuki-Horimoto K, Maemura T, Kiso M, Scheaffer SM, et al. SARS-CoV-2 Omicron virus causes attenuated disease in mice and hamsters. Nature. 2022;603(7902):687-692
  71. 71. Gandhi K, Khare S, Sachdeva R. Significance of biochemical and hematological indicators in predicting COVID-19 prognosis during the Omicron variant phase – A retrospective study. Medicine India. 2022;1:11
  72. 72. Suzuki R, Yamasoba D, Kimura I, Wang L, Kishimoto M, Ito J, et al. Attenuated fusogenicity and pathogenicity of SARS-CoV-2 Omicron variant. Nature. 2022;603(7902):700-705
  73. 73. Shuai H, Chan JFW, Hu B, Chai Y, Yuen TTT, Yin F, et al. Attenuated replication and pathogenicity of SARS-CoV-2 B.1.1.529 Omicron. Nature. 2022;603(7902):693-699
  74. 74. Bouzid D, Visseaux B, Kassasseya C, Daoud A, Fémy F, Hermand C, et al. Comparison of patients infected with delta versus Omicron COVID-19 variants presenting to Paris emergency departments. Annals of Internal Medicine. 2022;175(6):831-837
  75. 75. Feikin DR, Abu-Raddad LJ, Andrews N, Davies MA, Higdon MM, Orenstein WA, et al. Assessing vaccine effectiveness against severe COVID-19 disease caused by Omicron variant. Report from a meeting of the World Health Organization. Vaccine. 2022;40(26):3516-3527
  76. 76. Chen X, Yan X, Sun K, Zheng N, Sun R, Zhou J, et al. Estimation of disease burden and clinical severity of COVID-19 caused by Omicron BA.2 in Shanghai, February-June 2022. Emerging Microbes and Infection. 2022;11(1):2800-2807
  77. 77. Esmaeilzadeh A, Elahi R. Immunobiology and immunotherapy of COVID-19: A clinically updated overview. Journal of Cellular Physiology. 2021;236(4):2519-2543
  78. 78. Liu J, Li S, Liu J, Liang B, Wang X, Wang H, et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. eBioMedicine. 2020;55:102763
  79. 79. Esmaeilzadeh A, Jafari D, Tahmasebi S, Elahi R, Khosh E. Immune-based therapy for COVID-19. 2021; 449-468.
  80. 80. Elahi R, Karami P, Heidary AH, Esmaeilzadeh A. An updated overview of recent advances, challenges, and clinical considerations of IL-6 signaling blockade in severe coronavirus disease 2019 (COVID-19). International Immunopharmacology. 2022;105:108536
  81. 81. Hojyo S, Uchida M, Tanaka K, Hasebe R, Tanaka Y, Murakami M, et al. How COVID-19 induces cytokine storm with high mortality. Inflammation Regeneration. 2020;40(1):37
  82. 82. Esmaeilzadeh A, Rostami S, Yeganeh PM, Tahmasebi S, Ahmadi M. Recent advances in antibody-based immunotherapy strategies for COVID-19. Journal of Cellular Biochemistry. 2021;122(10):1389-1412
  83. 83. Batah SS, Fabro AT. Pulmonary pathology of ARDS in COVID-19: A pathological review for clinicians. Respiratory Medicine. 2021;176:106239
  84. 84. Wilhelm A, Widera M, Grikscheit K, Toptan T, Schenk B, Pallas C, et al. Limited neutralisation of the SARS-CoV-2 Omicron subvariants BA.1 and BA.2 by convalescent and vaccine serum and monoclonal antibodies. eBioMedicine. 2022;82:104158
  85. 85. Cele S, Jackson L, Khoury DS, Khan K, Moyo-Gwete T, Tegally H, et al. SARS-CoV-2 Omicron has extensive but incomplete escape of Pfizer BNT162b2 elicited neutralization and requires ACE2 for infection. medRxiv. 2021;176:106239
  86. 86. Guihot A, Litvinova E, Autran B, Debré P, Vieillard V. Cell-mediated immune responses to COVID-19 infection. Frontiers in Immunology. 2020;82:104158
  87. 87. Iqbal H. The importance of cell-mediated immunity in COVID-19 – An opinion. Medical Hypotheses. 2020;143:110152
  88. 88. Boechat JL, Chora I, Morais A, Delgado L. The immune response to SARS-CoV-2 and COVID-19 immunopathology – Current perspectives. Pulmonology. 2021;27(5):423-437
  89. 89. Gowrisankar A, Priyanka TMC, Banerjee S. Omicron: A mysterious variant of concern. The European Physical Journal Plus. 2022;137(1):100
  90. 90. Li C, Zhao C, Bao J, Tang B, Wang Y, Gu B. Laboratory diagnosis of coronavirus disease-2019 (COVID-19). Clinica Chimica Acta. 2020;510:35-46
  91. 91. Tahmasebi S, Khosh E, Esmaeilzadeh A. The outlook for diagnostic purposes of the 2019-novel coronavirus disease. Journal of Cellular Physiology. 2020;235(12):9211-9229
  92. 92. Ettaboina SK, Nakkala K, Laddha KS. A mini review on SARS-COVID-19-2 Omicron variant (B.1.1.529). SciMed Journal. 2021;3(4):399-406
  93. 93. Chen X, Kang Y, Luo J, Pang K, Xu X, Wu J, et al. Next-generation sequencing reveals the progression of COVID-19. Frontiers in Cellular and Infection Microbiology. 2021;11:11
  94. 94. Allam M, Ismail A, Khumalo ZTH, Kwenda S, van Heusden P, Cloete R, et al. Genome sequencing of a severe acute respiratory syndrome coronavirus 2 isolate obtained from a south African patient with coronavirus disease 2019. Microbiology Resources Announcement. 2020;9(27):632490
  95. 95. Li D, Li J. Immunologic testing for SARS-CoV-2 infection from the antigen perspective. Journal of Clinical Microbiology. 2021;59(5):e00572-e00620
  96. 96. Yuan M, Liu H, Wu NC, Wilson IA. Recognition of the SARS-CoV-2 receptor binding domain by neutralizing antibodies. Biochemical and Biophysical Research Communications. 2021;538:192-203
  97. 97. Hurt AC, Wheatley AK. Neutralizing antibody therapeutics for COVID-19. Viruses. 2021;13(4):628
  98. 98. Cameroni E, Saliba C, Bowen JE, Rosen LE, Culap K, Pinto D, et al. Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift. bioRxiv. 2021;538:192-203
  99. 99. Tao K, Tzou PL, Kosakovsky Pond SL, Ioannidis JPA, Shafer RW. Susceptibility of SARS-CoV-2 Omicron variants to therapeutic monoclonal antibodies: Systematic review and meta-analysis. Microbiology Spectroscopy. 2022;10(4):628
  100. 100. Zhou H, Møhlenberg M, Thakor JC, Tuli HS, Wang P, Assaraf YG, et al. Sensitivity to vaccines, therapeutic antibodies, and viral entry inhibitors and advances to counter the SARS-CoV-2 Omicron variant. Nature. Dec 2022;602:664-670
  101. 101. Garcia-Beltran WF, Denis KJ, Hoelzemer A, Lam EC, Nitido AD, Sheehan ML, et al. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant. Cell. 2022;185(3):457-466.e4
  102. 102. Shrestha LB, Foster C, Rawlinson W, Tedla N, Bull RA. Evolution of the SARS-CoV-2 Omicron variants BA.1 to BA.5: Implications for immune escape and transmission. Reviews in Medical Virology. 2022;32(5):e0001422
  103. 103. Zhang L, Cao L, Gao XS, Zheng BY, Deng YQ , Li JX, et al. A proof of concept for neutralizing antibody-guided vaccine design against SARS-CoV-2. National Science Review. 2021;185:457-466.e4
  104. 104. Sun C, Kang YF, Liu YT, Kong XW, Xu HQ , Xiong D, et al. Parallel profiling of antigenicity alteration and immune escape of SARS-CoV-2 Omicron and other variants. Signal Transduction and Targeted Therapy. 2022;7(1):42
  105. 105. Cerutti G, Guo Y, Liu L, Liu L, Zhang Z, Luo Y, et al. Cryo-EM structure of the SARS-CoV-2 Omicron spike. Cell Reports. 2022;38(9):110428
  106. 106. U.S. National Library of Medicine. ClincalTrial.gov [Internet]. [cited 2023 Sep 10]. 2023. Available from: https://classic.clinicaltrials.gov/ct2/results?term=vaccine&cond=Omicron&recrs=d&age_v=&gndr=&type=&rslt=&phase=2&phase=3&Search=Apply

Written By

Khushi Gandhi

Submitted: 10 September 2023 Reviewed: 10 September 2023 Published: 08 November 2023