Skip to main content

A review of epidemiology, clinical features and disease course, transmission dynamics, and neutralization efficacy of SARS-CoV-2 variants

Abstract

Background

After the first detection in November 2019, SARS-CoV-2 has spread rapidly over the continents and started the pandemic of the millennium. In addition to several novels and repurposed monoclonal antibodies (mAbs) as a therapeutic option against COVID-19, scientists from across the world have developed several candidate vaccines, developed mainly targeting the Wuhan strain, with very promising results to combat this pandemic. Unfortunately like any RNA viruses, SARS CoV-2 has also gone through the accumulation of hundreds and thousands of mutations in their genome lead to the development of several variants of concerns (VOC) and variants of interests (VOI), resulting in increased transmissibility and virulence of the virus, along with their capacity to escape cross-protection. Seemingly, the main hindrance of containing this pandemic right now is the effectiveness of currently available vaccines and mAbs against newly emerging variants. Therefore, it is important to monitor variants epidemiology, transmission dynamics, clinical characteristics, as well as their immune evasion capacity to implement appropriate vaccine strategy and other containment measures.

Body

In this review, we tried to focus on variants characteristics and to what extent they can escape immunity, provided by both available vaccinated sera and convalescent sera. A stringent literature review was performed using various databases, mentioned in the methodology portion. The current geographical distribution of these variants of SARS CoV-2 has been presented using a heat map. Findings from published articles comparing these variants, in terms of genome epidemiology, transmissibility, viral load dynamics, and association with different waves have been described briefly. Due strength was given while describing variants neutralization potency against current vaccines, mAbs, and also against convalescent sera. Data from both clinical trials and in vitro/ex-vivo studies have been discussed here. Comparative findings from several articles were brought into one concise paper. After careful reviewing of all the available data, it was clear that, without hesitation, we should strengthen our vaccination strategy, because the severity of COVID 19 is reasonably lower, irrespective of variants and vaccine used.

Conclusion

We hope that many falsified myths and beliefs regarding vaccine immunity and emerging variants will be clarified in light of this available evidence, which we summarized in our paper.

Background

The first cases of SARS-CoV-2 infection were detected near a seafood market in Wuhan, China [1]. On March 11, 2020, the World Health Organization (WHO) declared the disease as a global pandemic [2]. To date, more than 164 million cases have been detected throughout the world with the death toll amounting to more than 3.4 million [3].

With the time being passed, SARS-CoV-2 has gone through several genetic evolutions with no significant impact on cross-immunity, until the recently emerged strains, namely, Alpha variant (previously known as UK variant) (B.1.1.7), Beta variant (previously known as South African variant) (B.1.351), Gamma variant (previously known as Brazilian variant) (P.1), Delta variant (previously known as Indian variant) (B.1.617), and other variants of interest (VOI) [4, 5]. They are raising concerns as many of their mutations are positioned in the S1 portion of SARS-CoV-2 spike protein, against which the majority of candidate vaccines and monoclonal antibodies (mAb) are directed, and thus they can affect antibody-mediated immunity [5,6,7]. Furthermore, there are concerns that these variants may have significantly different clinical course or transmission dynamics compared to wild-type viruses [8, 9].

According to the latest published data, more than 100 candidate vaccines have been going through different phases of clinical trials with another 184 in pre-clinical phases of development [10]. Among them, five vaccines (BNT162b2 by Pfizer-BioNTech; mRNA-1273 by Moderna; ChAdOx1 nCoV-19 (AZD1222) by Oxford-AstraZeneca; Ad26.COV2-S by Janssen; and lastly Sinopharm BBIBP-CorV vaccine developed by Sinopharm, China) have received emergency use authorization (EUA) [11,12,13,14,15]. Several monoclonal antibodies (mAbs) have been developed and some of them have received EUA to treat COVID-19 cases [6, 16, 17]. To understand the extent these new variants are influencing the efficacy of currently available vaccines or mAbs, and to formulate an effective prevention and treatment strategy against the virus, a careful monitoring of variant-associated mutations, symptoms, clinical course, transmission dynamics, and other variables should be documented. Hence, in this review, we comprehensively summarize all the relevant literature documenting SARS-CoV-2 variants including their epidemiology, clinical symptoms, disease course, transmission dynamics, and neutralization by vaccines, convalescent sera, and mAbs.

Main text

Methodology

Publicly available databases and COVID-19 information repositories such as PubMed including Medline, Google Scholar, WHO, GISAID, CDC, Worldometers, Our World in Data, and several other sources were used to search for suitable articles. Peer-reviewed publications, as well as epidemiological updates and news pieces, from December 2019 to August 2021 were evaluated and if deemed suitable, authors downloaded full text. Search terms include, but are not limited to, SARS-CoV-2, COVID-19, symptoms, disease severity, variants, monoclonal antibody, vaccine, convalescent sera, epidemiology, B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma), B.1.617.2 (delta), and different combinations of listed search terms. Information was stored in Microsoft Excel and Word. A geographical heat map of COVID-19 variants was created using the Plotly in Python.

COVID-19 variants in circulation

SARS-CoV-2, a 29.9 Kb positive-sense single-stranded RNA virus, much like its predecessors and other viruses in the coronavirus family, such as MERS-CoV and SARS-CoV, mutates constantly, although the mutation rate is considerably slower than many other RNA viruses [18]. The more people the virus infects, the probability of acquiring genetic changes increases. Luckily, most of the genetic changes the virus acquires have little impact on the ability of the virus to cause severe disease or increased infection. But, at times, the virus accumulates genetic changes that confer competitive advantage and help the virus to transmit more easily or cause severe disease in infected individuals, through alteration of major proteins, such as that of spike protein in SARS-CoV-2 tasked with binding to a receptor through the receptor-binding domain (RBD) (spike protein residue 319-541) and entry of virus within host cells [18,19,20].

The original strain of SARS-CoV-2 that originated in Wuhan, China toward the end of 2019, mutated, albeit at a slower pace, and gave rise to other variants, defined as strains with significant phenotypic changes to the original strain [19], which now are predominant strains in different countries around the globe including UK, USA, India, South Africa, Brazil, and several other nations (Table 1). Some of these variants are described as variants of concern (VOCs), as classified by WHO, due to their ability to evade the host immune system, increased transmissibility, decreased efficacy of vaccination and therapeutic interventions, and the potential increase in disease severity. In addition to VOCs, several other variants of interest (VOI) are under observation with no variant of high consequence identified thus far [4, 5, 21]. In addition to VOC and VOI, several variants, with no clear-cut evidence on increased transmissibility or virulence, are under monitoring including P.2, P.3, R.1, R.2, B.1.466.2, B.1.621, B.1.427/B.1.429 (formerly known as epsilon and denoted as VOC in North America) etc. [21]. A geographical heatmap of major COVID-19 variants is provided in Fig. 1.

Table 1 Major SARS-CoV-2 Variants in circulation
Fig. 1
figure 1

Geographical distribution of major COVID-19 variants (a—Alpha, b—Beta, c—Gamma, and d—Delta) [22]

Clinical features, transmission, and disease course:

Classical clinical features

Earlier literature reporting COVID-19 disease in the Chinese population reported fever, dry cough, breathing difficulties, headache, and pneumonia as the most common presenting complaints [23]. However, most of the early cases had contacts with the Wuhan seafood market and hence, once the virus spread to other countries, diverse symptoms started to emerge and were reported in scientific literature. A recent meta-analysis of 152 publications from 23 countries including a total of 41409 participants demonstrated 26 symptoms experienced by COVID-19 infected patients. Among them, fever was the most frequently experienced symptom (58.66%) followed by cough (54.52%), dyspnea (30.82%), malaise (29.75%), fatigue (28.16%), and sputum secretion (25.33%) [24]. Other reported symptoms include neurological symptoms including loss of taste and smell, dermatological manifestations, myalgia, sneezing, sore throat, rhinitis, diarrhea, anorexia, chest pain, and headache. But in studies with participants more than 100, fever, cough, and dyspnea were the three most reported symptoms.

Infection with variants

The Office for National Statistics (ONS) in the UK surveyed people testing strong positive for COVID-19 in polymerase chain reaction (PCR) test and compare the symptoms with that of the original strain. The survey data did not reveal any unusual symptoms with the classic reported symptoms making up the bulk of the cases (Table 2) [25]. This was further reconfirmed in a recently published study in the UK that revealed patients affected with alpha reported similar symptoms to that of other widely circulating strains and there are no discernible differences between alpha variant and other strains [26]. Regarding the widely circulating delta variant, symptoms might differ from other circulating strains, as suggested by the Zoe COVID study in the UK. People infected with delta variant, both vaccinated and unvaccinated, primarily report symptoms such as runny nose, headache and sneezing, and sore throat [27]. While traditional symptoms such as anosmia and fever are still reported, the percentages are small compared to previous months.

Table 2 Comparison of symptoms between alpha variant and wild strain infected individuals in the UK

Similar comments were made by other researchers around the globe regarding beta and gamma variants and other variants of interest (VOI), as symptoms experienced by affected individuals were similar to that one of wild SARS-COV-2 strains widely prevalent during the early months of the pandemic.

Transmission dynamics

D614G mutation—wild strain

While SARS-CoV-2 sequences deposited in global repositories in February 2020 did not reveal this mutation to any of the sequences, the number of sequences with D614G mutation increased rapidly in the next months, reaching 70% in May, suggesting a transmission advantage over D614 from the original Wuhan strain [28]. It is postulated that the spike protein of SARS-CoV-2 is largely responsible for viral tropism and increased transmissibility. In previous researches, it was found out that sequences bearing D614G mutation were linked to higher viral loads in the upper respiratory tract [29, 30]. While the exact mechanism for that is yet to be understood but initial research suggests that D614G enhances viral cell entry as the G614 S-protein trimer being more suitable for ACE2 binding [29, 30].

Alpha variant

By analyzing the COG-UK datasets, researchers found out that the reproduction number (R) for the alpha variant is 43–90% more than other prevailing variants in the UK [31]. This finding was similar when analyzing data from other countries such as Denmark, Switzerland, and the USA [31]. Several hypotheses have already been proposed by researchers regarding increased transmissibility of alpha variant including a longer infectious period, ∆H69/∆V70 deletion-mediated immune escape, and high viral load as observed through low CT-values obtained during PCR testing [32,33,34].

Beta variant

Compared to other strains, infection with the beta variant leads to increased viral load, much like the alpha variant, resulting in increased human-to-human transmission. This has been evidenced by the recent surge in beta variant cases in South Africa where, at the start of December 2020, close to 90% of new cases were infected by this variant. But at the start of October 2020, only 11% of case sequences were of this variant [8]. Similarly, in Bangladesh, the second wave was initiated with the rapid spread of beta variant, reaching up to 93% of total sequenced cases and increasing reproduction number (reaching up to 2 in March 2021 compared to 1 throughout 2020) [35, 36]. Another country, Zambia, did not report any cases of beta variant from March 2020 to early December 2020. But in mid-December, 22 out of 23 cases were detected as beta variant and this correlates with a 16-fold increase in infection within the country within a span of 1 month starting from December 2020 [8].

Gamma variant

Regarding gamma variants currently prevalent in Brazil, research results, based on data from Manaus, estimate that gamma variant cases are 1.7-2.4 times more transmissible than non-P.1 lineage despite higher seroprevalence among residents in Manaus compared to neighboring regions [37]. Another study conducted using publicly available data on hospitalized patients found the transmissibility of gamma variant, after adjusting for other variables, to be 2.5 times more than non-gamma variant [38].

Delta variant

The latest VOC declared by the WHO, the delta variant, currently spreading rapidly in India and neighboring Nepal and Sri Lanka, Bangladesh, and 100 other countries including UK and USA, is thought to have greater transmissibility than other lineages, prevalent in both UK and India, largely due to a double mutation in spike protein, E484Q, and L452R, that are responsible for higher viral load, increased transmissibility, and immune escape [39]. Recent data suggest that viral load in delta variant-infected individuals is 1000 times greater than people infected with the original strain. The variant is twice as much transmissible as the original strain [40].

Disease course

Alpha variant

A community-based study including a large British dataset, that depended on absence/presence of S-gene amplification in real-time PCR to determine B.1.1.7 lineage, of 2.2 million COVID-19 affected people from September 2020 to February 2021 revealed a 61% (CI: 42–82%) increased mortality among population infected with the alpha variant overpopulation infected with other strains [41]. This was further reconfirmed by The Office for National Statistics as patients with S-gene negative status were found to be appearing for a test earlier than the other group, suggesting a more rapid and severe disease course [25, 42]. Similar findings were obtained from a study conducted using data, comprising of 19995 cases, from seven European countries. Increased rate of hospitalization was observed in VOC cases (B.1.1.7; P.1; B.1.351) compared to non-VOC cases [43]. Furthermore, on unmatched analysis, B.1.1.7, B.1.351, and P.1 cases were 2.3, 3.3, and 2.2 times more likely to get admitted into ICU [43]. Other studies conducted by Challen et al. and Grint et al. revealed comparable findings with mortality hazard ratio after 28 days being two-thirds higher in VOC groups (1.64 and 1.67 respectively) compared to non-VOC cases [44, 45]. But these findings were challenged by a group of researchers who sequenced 341 samples and compared between sequences of B.1.1.7 lineages and non-B.1.1.7 lineages. While there is evidence that viral load and disease transmission is higher in B.1.1.7 lineages, no association could be established between severe disease and infection with alpha variant [34].

Beta variant

While no data regarding increased disease severity in individuals infected with the beta variant could be found as of writing this paper, in several countries such as South Africa, Zambia, and Asian countries such as Bangladesh, this variant has outcompeted other circulating variants and has led to increased transmission of the virus, and as a consequence, resulting in higher morbidity and mortality [8, 35]. Earlier reports suggest in admission mortality to be 20% higher in South Africa during the second wave of pandemic compared to the first wave. Again, no definitive answer could be provided regarding the association of the beta variant with increased mortality, as it could be down to a higher number of cases and strain on the healthcare system. In Bangladesh, despite an increase in case numbers, interestingly, the case fatality rate (CFR) has decreased compared to earlier months when others strains were prevalent within the country [46].

Gamma variant

While there is little research evidence on the increased mortality and morbidity caused by the gamma variant, recent evidence suggests that the variant may be responsible for increased case fatality rate (CFR) in all age groups with CFR increasing significantly in February, compared to data from January among residents in Parana state in Brazil [38]. Similar findings were observed among residents in Manaus where the mortality is estimated to rise 1.2 to 1.9 times since the emergence of gamma variant compared to previous periods [37]. But it remains to be elucidated whether the apparent rise in mortality is related to gamma variant rather than increased transmissibility and dwindling healthcare resources in Brazilian hospitals.

Delta variant

Actual data emanating from India reveal that, while there is a catastrophic rise in case of numbers in March and April with 0.4 million cases being detected each day, CFR has decreased and remained low throughout the duration [47, 48]. In a recent study conducted in Ontario Canada, delta variant-infected people were 108% more likely to get hospitalized compared to other strains. Regarding ICU admission and death, there was a 234% and 132% increase in risk compared to other strains [49]. Similarly, studies conducted in Scotland and Singapore revealed that people infected with the delta variant are more likely to get hospitalized, have higher odds of oxygen requirement, and suffer for a longer duration compared to other strains [50, 51].

Neutralization potency of post vaccinated sera

Alpha variant

Reduction of neutralization against full panel spike mutation (UKΔ8) of B.1.1.7 (alpha variant) pseudovirus and pseudoviruses carrying either single (N501Y) and triple mutations (N501Y, A570D, and the 69/70 deletion) have been tested with serum, vaccinated with BNT162b2 vaccine, mRNA-1273, and ChAdOx1 vaccine in several in vitro settings. For selected insertion, either single or triple, reduction in titers varied from no reduction to highest 3.3-fold decrease, whereas UKΔ8 resulted in highest fold change of 5.83 in 80–85 age group and lowest fold change of 1.78 in 60–65 age groups in cases of BNT162b2 vaccine, 1.8-fold for mRNA-1273 vaccine, and 9 times reduction in neutralization titer ChAdOx1 vaccine. Serum was tested against USA/WA-1/2020 background strain, wild-type SARS-CoV-2, or against Victoria virus strain and was taken after 2nd dose of vaccination. Despite this reduction in GMT, it is unlikely for the UK variant to escape vaccine-mediated protection because Smith et al. previously for influenza virus vaccine sufficed that, more than 20% reduction in neutralization titer was needed for vaccine immune evasion by newly emerging virus strains [52,53,54,55,56,57,58,59,60].

As for the clinical efficacy, the BNT162b2 vaccine showed effectiveness ranging from highest 95.3% in Israel and lowest 87% in Qatar against this VOC, assuming 94.5% and 44.5% prevalence rate of B.1.1.7 in those countries at the time of clinical trials. Most importantly, two doses of the BNT16b2 vaccine were capable of preventing hospitalization as a whole in 97.5% of cases and critical hospitalization in 96.7% of cases [61, 62]. As for the ChAdOx1 vaccine, in a clinical trial after receiving two doses of ChAdOx1 vaccine, they observed that the clinical efficacy against symptomatic COVID-19 is 70.4% [56].

Beta and gamma variant

These two variants, recently named as beta and gamma variants, share identical triplet mutations (E484K, K417N/T, and N501Y) in RBD and thus presumed to show the same extent of immune evasion capabilities, but surprisingly B.1.351 showed more resistance to antibody neutralization than P.1. BNT162b2 vaccinated sera showed a 2.6-fold and 6.7-fold reduction in GMT in two different studies, while ChAdOx1 and mRNA-1273 vaccinated sera showed 2.9- and 4.5-fold reduction in GMT against P.1 variant, compared to Victoria virus strain [60, 64]. When overall B.1.351 variant was considered, reduction in vitro study titer varied from 6.5- to 7.6-fold for sera vaccinated with BNT162b2 vaccine and 9-fold and 8.6-fold for sera vaccinated with ChAdOx1 and mRNA-1273 vaccines. Although, when all three strains (v1, v2, v3) of B.1.351 lineage were investigated separately, the highest reduction of titer was observed for v3 with a reduction of 42.4-fold for BNT162b2 and 19.2-fold for mRNA-1273 followed by v2 with 41.2-fold for BNT162b2 and 20.8-fold for mRNA-1273 and lastly of v1 with 34.5-fold for BNT162b2 and 27.7-fold for mRNA-1273 vaccine sera [55, 59, 63, 64]. Interestingly, Tada et al. and Wu et al. showed that, whether it is E484K, K417N-E484K-N501Y triple, or full panels of B.1.351 mutation, neutralization capacities of post vaccinated sere were reduced but not completely abolished, rather, were able to provide cross-protection with sufficiently high titer in in-vitro settings [58, 59, 65].

Fortunately, real-life scenarios for these in vitro studies have shown more promising results, especially in terms of preventing severe diseases. Effectiveness of BNT162b2 vaccine against beta variant among the residents of long-term care facilities in France, after two complete doses of vaccination, was 49% but against severe disease VE remained high with 86% [66], while, in Qatar, in the background of 50% beta variant prevalence, VE ranged from 72.1 to 75% based on cohort and case-control study design [61]. Similarly, AstraZeneca-Vaxzevria was found protective against infection, hospitalization, and death, among ≥ 60 years, in a setting of the high prevalence of gamma variant in Brazil, in 77.9%, 87.6%, and 93.6% vaccinated individuals respectively [67].

Delta variant

The most recent VOC, responsible for the 3rd wave of infection worldwide, the B.1.617.2 or delta variant has shown its potentials to increase transmissibility as well as enhanced capability of immune evasion. While numerous studies are on track to reveal the level of danger, Edara et al. compared neutralizing antibody response between B.1.617.1 and WA1/2020 variant, in sera vaccinated with mRNA-1273 and BNT162b2 vaccines. Differences in GMT were observed 1142 and 1012, for mRNA-1273 and BNT162b2 mRNA vaccinated sera respectively, against control strain. Despite 6.8-fold less susceptibility, all of the posts vaccinated sera were able to neutralize this VOC, which suggests some level of protective immunity against B.1.617.1 by existing mRNA vaccines [68], while Covaxin immune sera showed probable immune escape capacity with a 1.84 GMT ratio for B.1.617, but 1.06 for B.1.1.7 and D614G variants [69]. On the other hand, Ferreira et al. devoured to identify the most notorious mutation in this variant, and following other studies, they showed that E484K mutation conferred 10-fold reduction and E484Q conferred slightly milder yet significant reduction in neutralization by BNT162b2 vaccinated sera [70].

Some very recent preprint articles concluded that both BNT162b2 and Chadox1nCov vaccines, after two complete doses, are highly effective by conferring 88% and 67% protection respectively against symptomatic disease from B.1.617.2 variant [71, 72], with a single dose of ChAdOx1-19 vaccine is effective for preventing infection in 46.2% but in 79.2% for preventing moderate-severe Covid-19 [73].

A table has been provided with the article, representing the summarized results from the studies that have been exploring (both in-vitro/ex-vivo and clinical efficacy studies) for the impact of current VOCs against BNT162b2, mRNA-1273, Chadox1nCov, and Covaxin immune sera (Table 3).

Table 3 Overview of immune evasion capacities for current VOCs against BNT162b2, mRNA-1273, Chadox1nCov, and Covaxin immune sera

Other vaccines

Surprisingly, newly trialed vaccines from Johnson & Johnson and Novavax showed more promising results against these variants of concerns. Results from a phase III trial with 43,783 participants, conducted by Johnson & Johnson, across the US, Central, and South America and South Africa, showed only 468 symptomatic cases of COVID-19. Although, the full picture of this trial, including percentage of SARS-CoV-2 variants among the infected population or how many cases are among the vaccine recipients, etc. is yet to reveal. In contrast, vaccine trials from Novavax are published in a more detailed form. They showed vaccine efficacy after the post hoc analysis, against B.1.1.7 is 85.6% and against B.1.351 is 60% after two complete doses of vaccine. Further statistical analysis showed that overall vaccine efficacy in HIV-negative participants was 60.1% for the prevention of mild to moderate COVID with a reduction in efficacy against B.1.351 variant with 51% [74,75,76] (Table 3).

Neutralization potency of convalescent sera

Convalescent sera, being the last resort of treatment in some settings, for severe COVID, aggravated the need to evaluate its neutralization potential against several variants. More or less similar findings were observed for all of these studies, where they showed no single mutation is capable enough to cause immune escape except E484K, which is thought to be the major attribute for the reduction in antibody neutralization. Against single point mutation (Δ69-70) and N501Y, neutralization efficiency was the same as D614G, although K417N, E484K, and N501Y substitutions were observed 5-fold to 6.8-fold reduction in neutralization titer [54, 58, 81,82,83]. Likewise, a full mutation panel of B.1.351 showed a discernible negative impact on neutralization capacity of convalescent serum samples with 13.3-fold, 9.4-fold, and 11- to 33.1-fold reduction in titer in comparison to wild-type, D614G, or Victoria strain of SARS CoV-2. As for P.1 strain, a 3.1-fold reduction in GMT was observed when compared to Victoria [55, 63, 64]. Increased resistance of neutralization is also associated with an increase in frequency, as Wibmer et al. showed 70% serum samples with < 100 ID50 against B.1.351 lineage and in nearly half (48%) samples with no detectable neutralization activity, while 63% sample with < 100 ID50 for triple mutant containing K417N, E484K, and N501Y substitutions with only 27% with no detectable neutralization activity [82]. Interestingly, all of these studies contained some number of convalescent serum samples showing neutralizing activity against B.1.351 as efficient as the original strain, and one study showed six individuals with prior COVID 19 or with exposure history and with the highest antibody titer, exhibited significant cross-neutralization for the B.1.351 [59, 63]. A brief overview of all the studies have been summarized and given in a tabulated form in Table 4.

Table 4 Overview of immune evasion capacities for current VOCs against convalescent sera

Neutralization potency of monoclonal antibodies

Structural analysis with two monoclonal antibodies, REGN10933 and P2B-2F6, revealed that E484Q and L452R mutation may interrupt the binding of those two monoclonal antibodies with spike protein. The spike protein mutation L452R, found in both B.1.427/B.1.429 and B.1.617 lineages, is thought to be responsible for decreased neutralization efficacy [39, 69, 85, 86]. Regarding alpha and beta variants, 12 mAbs were directed against RBD and six against NTD of spike protein in Vero E6 cells and compared with wild-type strains. Results revealed a marked reduction in neutralization capacities of 910-302 and S30 mAbs against B.1.1.7 [87]. But results were much worse when tested against beta variant as 910-30, 2-15, LY-CoV555 (bamlanivimab), C121, and REGN10933 (casirivimab) showed markedly reduced capacity in virus neutralization, mediated by E484K and K417N mutations [87].

In addition, researchers evaluated the neutralizing efficacy of mAb therapies that are in clinical use. LY-CoV555 alone or in combination with CB6 failed to neutralize beta variant while some sort of activities was retained, though markedly reduced when tested against REGN10933+REGN10987 and COV2-2196+COV2-2130 combinations. In another study, COV2-2196+COV2-2130 retained their activity against all virus variants though there was a 4-fold reduction in neutralization efficacy [88]. But no significant alteration was observed when treated against S309 and Brii-196+Brii-198 combinations [88]. Interestingly, another VOC, the gamma variant, harbors three similar mutations in RBD residues to that of beta variant, namely E484K, K417T, and N501Y, and hence findings from B.1.351 testing should be relevant to P.1 variant also [88].

These findings including the role of E484K and K417N substitutions in mAb neutralization was pronounced in another study. K417N was responsible for a 27-fold reduction in virus neutralization by mAb COVOX-40 in in-vitro assay while no significant change was observed regarding other mAbs tested. But E484K mutation negatively impacted the efficacy of several mAbs including COV2-2196, COV2-3025, COV2-2381, and S2E12, where 4- to 5-fold reductions in efficacy were observed [88]. N501Y mutation observed in all three VOCs was responsible for a slight reduction in neutralization by mAbs and is consistent across studies in different settings [89]. Two other significant mutations P681H and 69-70del did not have a significant effect on mAb efficacy directed toward receptor binding motif (RBM) of RBD [39, 88] (Table 5).

Table 5 Neutralization efficacy of EUA approved mAbs against SARS-CoV-2 variants of concern

Conclusions

A major limitation of these studies is that almost all of them are performed using engineered pseudoviruses and without any serological correlations. Therefore, the role of humoral and cell-mediated immunity is mainly assumption-based. Secondly, complement-mediated cell lysis and antibody-dependent cell-mediated cytotoxicity or phagocytosis are overlooked, which can play either a protective or harmful role in-vivo situations. For convalescent serum samples, the timing of sample collection may not be optimal. If samples were collected early, titer may rise further or wane with time, thus, study results may become compromised. Another important point is there were some discrepancies among the results from pseudovirus and live virus neutralization assay, which should be resolved only with clinical data against all of the variant viruses. At last, laboratory-based results are only half-truth as they do not represent a complex mechanism of the human body. Therefore, more clinical trials are the demand of time to visualize the impact of emerging viruses on cross-protection either by vaccination or by the previous infection.

Availability of data and materials

Available from the corresponding author on request.

Abbreviations

SARS CoV-2:

Severe acute respiratory syndrome coronavirus-2

COVID-19:

Coronavirus disease-19

SARS CoV:

Severe acute respiratory syndrome coronavirus

MERS CoV:

Middle East respiratory syndrome coronavirus

VOC:

Variants of concern

VOI:

Variants of interest

RBD:

Receptor binding domain

RBM:

Receptor binding motif

mAb:

Monoclonal antibody

WHO:

World Health Organization

GISAID:

Global initiative on sharing all Influenza data

CDC:

Communicable disease control

EUA:

Emergency use authorization

ONS:

Office for National Statistics

PCR:

Polymerase chain reaction

NGS:

Next-generation sequencing

ACE2:

Angiotensin converting enzyme 2

CT value:

Cycle threshold level

CI:

Confidence interval

SGTF:

S gene target failure

GMT:

Geometric Mean Titre

References

  1. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J et al (2020) A novel coronavirus from patients with pneumonia in China, 2019. New England journal of medicine 382(8):727–733. https://doi.org/10.1056/NEJMoa2001017

    Article  CAS  Google Scholar 

  2. World Health Organization. WHO Director-General's opening remarks at the media briefing on COVID-19 - 11 March 2020 [Updated 11 Mar 2020; cited 26 May 2021]. Retrieved from https://www.who.int/director-general/speeches/detail/who-director-general-s-opening-remarks-at-the-media-briefing-on-covid-19%2D%2D-11-march-2020

  3. Worldometer. COVID-19 coronavirus pandemic: Worldometer; 2020 [updated 26 May 2021; cited 26 May 2021]. Retrieved from: https://www.worldometers.info/coronavirus/

  4. Center for Disease Control and Prevention. About variants of the virus that causes COVID-19​​ [Updated 20 May 2021; cited 26 May 2021]. Retrieved from https://www.cdc.gov/coronavirus/2019-ncov/variants/variant.html?CDC_AA_refVal=https%3A%2F%2Fwww.cdc.gov%2Fcoronavirus%2F2019-ncov%2Ftransmission%2Fvariant.html

  5. World Health Organization. Weekly epidemiological update on COVID-19 - 25 May 2021 [Updated 25 May 2021; cited 26 May 2021]. Retrieved from https://www.who.int/publications/m/item/weekly-epidemiological-update-on-covid-19%2D%2D-25-may-2021

  6. Deb P, Molla MMA, Rahman KSU (2021) An update to monoclonal antibody as therapeutic option against COVID-19. Biosafety and Health 3(2):87–91 https://doi.org/10.1016/j.bsheal.2021.02.001

    Article  Google Scholar 

  7. Kyriakidis NC, López-Cortés A, González EV, Grimaldos AB, Prado EO (2021) SARS-CoV-2 vaccines strategies: a comprehensive review of phase 3 candidates. npj. Vaccines 6(1):1–17 https://doi.org/10.1038/s41541-021-00292-w

    Google Scholar 

  8. Abdool Karim SS, de Oliveira T (2021) New SARS-CoV-2 variants—clinical, public health, and vaccine implications. New England Journal of Medicine 384(19):1866–1868 https://doi.org/10.1056/NEJMc2100362

    Article  Google Scholar 

  9. Horby P, Huntley C, Davies N, et al. NERVTAG paper on COVID-19 variant of concern B.1.1.7 [Updated 22 Jan 2021; cited 26 May 2021]. Retrieved from https://www.gov.uk/government/publications/nervtag-paper-on-covid-19-variant-of-concern-b117

  10. World Health Organization. The COVID-19 candidate vaccine landscape and tracker [Updated 25 May 2021; cited 26 May 2021]. Retrieved from https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines

  11. World Health Organization. WHO lists two additional COVID-19 vaccines for emergency use and COVAX roll-out [Updated 15 Feb 2021; cited 26 May 2021]. Retrieved from https://www.who.int/news/item/15-02-2021-who-lists-two-additional-covid-19-vaccines-for-emergency-use-and-covax-roll-out

  12. World Health Organization. WHO issues its first emergency use validation for a COVID-19 vaccine and emphasizes need for equitable global access [Updated 31 Dec 2020; cited 26 May 2021]. Retrieved from https://www.who.int/news/item/31-12-2020-who-issues-its-first-emergency-use-validation-for-a-covid-19-vaccine-and-emphasizes-need-for-equitable-global-access

  13. World Health Organization. WHO lists additional COVID-19 vaccine for emergency use and issues interim policy recommendations [Updated 7 May 2021; cited 26 May 2021]. Retrieved from https://www.who.int/news/item/07-05-2021-who-lists-additional-covid-19-vaccine-for-emergency-use-and-issues-interim-policy-recommendations

  14. World Health Organization. The Moderna COVID-19 (mRNA-1273) vaccine: what you need to know [Updated 26 Jan 2021; cited 26 May 2021]. Retrieved from https://www.who.int/news-room/feature-stories/detail/the-moderna-covid-19-mrna-1273-vaccine-what-you-need-to-know

  15. World Health Organization. WHO adds Janssen vaccine to list of safe and effective emergency tools against COVID-19 [Updated 12 Mar 2021; cited 26 May, 2021]. Retrieved from https://www.who.int/news/item/12-03-2021-who-adds-janssen-vaccine-to-list-of-safe-and-effective-emergency-tools-against-covid-19

  16. NIH. Anti-SARS-CoV-2 Monoclonal Antibodies [Updated 24 May 2021; cited 26 May 2021]. Retrieved from https://www.covid19treatmentguidelines.nih.gov/anti-sars-cov-2-antibody-products/anti-sars-cov-2-monoclonal-antibodies/

  17. FDA. Coronavirus (COVID-19) update: FDA authorizes additional monoclonal antibody for treatment of COVID-19 [Updated 26 May 2021; cited 26 May 2021]. REtrieved from https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-additional-monoclonal-antibody-treatment-covid-19

  18. Vilar S, Isom DG (2021) One year of SARS-CoV-2: How much has the virus changed? Biology 10(2):91 https://doi.org/10.3390/biology10020091

    Article  CAS  Google Scholar 

  19. Lauring AS, Hodcroft EB (2021) Genetic variants of SARS-CoV-2—what do they mean? JAMA 325(6):529–531 https://doi.org/10.1001/jama.2020.27124

    Article  CAS  Google Scholar 

  20. Grubaugh ND, Petrone ME, Holmes EC (2020) We shouldn’t worry when a virus mutates during disease outbreaks. Nature microbiology 5(4):529–530 https://doi.org/10.1038%2Fs41564-020-0690-4

    Article  CAS  Google Scholar 

  21. World Health Organization. Tracking SARS-CoV-2 variants [Updated 13 Aug, 2021; cited 15 Aug, 2021]. Retrieved from https://www.who.int/en/activities/tracking-SARS-CoV-2-variants/

  22. GISIAD. Tracking of variants [Updated 21 May 2021; cited 28 May 2021]. Retrieved from https://www.gisaid.org/hcov19-variants/

  23. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W et al (2020) A pneumonia outbreak associated with a new coronavirus of probable bat origin. nature 579(7798):270–273 https://doi.org/10.1038%2Fs41586-020-2012-7

    Article  CAS  Google Scholar 

  24. da Rosa Mesquita R, Junior LCFS, Santana FMS, de Oliveira TF, Alcântara RC, Arnozo GM et al (2020) Clinical manifestations of COVID-19 in the general population: systematic review. Wiener klinische Wochenschrift 133(7-8):377–382. https://doi.org/10.1007%2Fs00508-020-01760-4

    Article  Google Scholar 

  25. Office for National Statistics. Coronavirus (COVID-19) Infection survey: characteristics of people testing positive for COVID-19 in England, 27 January 2021 [Updated 27 Jan 2021; cited 26 May 2021]. Retrieved from https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsanddiseases/articles/coronaviruscovid19infectionsinthecommunityinengland/characteristicsofpeopletestingpositiveforcovid19inengland27january2021

  26. Graham MS, Sudre CH, May A, Antonelli M, Murray B, Varsavsky T et al (2021) Changes in symptomatology, reinfection, and transmissibility associated with the SARS-CoV-2 variant B. 1.1. 7: an ecological study. Lancet Public Health 6(5):e335–e345. https://doi.org/10.1016/S2468-2667(21)00055-4

    Article  Google Scholar 

  27. ZOE COVID Study. What are the new top 5 COVID symptoms? [Updated 23 Jun 2021; cited 15 Aug 2021]. Retrieved from https://covid.joinzoe.com/post/new-top-5-covid-symptoms

  28. Zhang L, Jackson CB, Mou H, Ojha A, Peng H, Quinlan BD, Rangarajan ES, Pan A, Vanderheiden A, Suthar MS, Li W, Izard T, Rader C, Farzan M, Choe H (2020). https://doi.org/10.1038/s41467-020-19808-4) SARS-CoV-2 spike-protein D614G mutation increases virion spike density and infectivity. Nature Communications 11(1):6013

    Article  CAS  Google Scholar 

  29. Plante JA, Liu Y, Liu J, Xia H, Johnson BA, Lokugamage KG, Shi PY (2021) Spike mutation D614G alters SARS-CoV-2 fitness. Nature 592(7852):116–121 https://doi.org/10.1038/s41586-020-2895-3

    Article  CAS  Google Scholar 

  30. Zhou B, Thao TTN, Hoffmann D, Taddeo A, Ebert N, Labroussaa F, Beer M (2021) SARS-CoV-2 spike D614G change enhances replication and transmission. Nature 592(7852):122–127 https://doi.org/10.1038/s41586-021-03361-1

    Article  CAS  Google Scholar 

  31. Davies, N. G., Abbott, S., Barnard, R. C., Jarvis, C. I., Kucharski, A. J., Munday, J. D., ... & Edmunds, W. J. (2021). Estimated transmissibility and impact of SARS-CoV-2 lineage B. 1.1. 7 in England. Science, 372(6538). https://doi.org/10.1126/science.abg3055

  32. Kemp SA, Collier DA, Datir RP, Ferreira IA, Gayed S, Jahun A, Gupta RK (2021) SARS-CoV-2 evolution during treatment of chronic infection. Nature 592(7853):277–282 https://doi.org/10.1038/s41586-021-03291-y

    Article  CAS  Google Scholar 

  33. Starr TN, Greaney AJ, Hilton SK, Ellis D, Crawford KH, Dingens AS et al (2020) Deep mutational scanning of SARS-CoV-2 receptor binding domain reveals constraints on folding and ACE2 binding. Cell 182(5):1295–1310 https://doi.org/10.1016/j.cell.2020.08.012pmid:32841599

    Article  CAS  Google Scholar 

  34. Frampton, D., Rampling, T., Cross, A., Bailey, H., Heaney, J., Byott, M., & Nastouli, E. (2021). Genomic characteristics and clinical effect of the emergent SARS-CoV-2 B. 1.1. 7 lineage in London, UK: a whole-genome sequencing and hospital-based cohort study. The Lancet Infectious Diseases, 1473-3099. https://doi.org/10.1016/S1473-3099(21)00170-5, 21, 9, 1246, 1256

  35. Brum, E., Saha, S., Sania, A., Tanmoy, A. M., Hooda, Y., Tanni, A., & Hampson, K. (2021). Surging COVID-19 in Bangladesh driven by B. 1.351 variant. medRxiv. https://doi.org/10.1101/2021.04.27.21255706

  36. Molla MMA, Disha JA, Yeasmin M, Ghosh AK, Nafisa T (2021) Decreasing transmission and initiation of countrywide vaccination: key challenges for future management of COVID-19 pandemic in Bangladesh. International Journal of Health Planning and Management 36(4):1014–1029 https://doi.org/10.1002/hpm.3156

    Article  Google Scholar 

  37. Faria NR, Mellan TA, Whittaker C, Claro IM, Candido DDS, Mishra S, Sabino EC (2021) Genomics and epidemiology of the P. 1 SARS-CoV-2 lineage in Manaus, Brazil. Science 372(6544):815–821 https://doi.org/10.1126/science.abh2644

    Article  CAS  Google Scholar 

  38. de Oliveira MHS, Lippi G, Henry BM (2021) Sudden rise in COVID-19 case fatality among young and middle-aged adults in the south of Brazil after identification of the novel B. 1.1. 28.1 (P. 1) SARS-CoV-2 strain: analysis of data from the state of Parana. medRxiv https://doi.org/10.1101/2021.03.24.21254046

  39. Cherian S, Potdar V, Jadhav S, Yadav P, Gupta N, Das M, Panda S (2021) Convergent evolution of SARS-CoV-2 spike mutations, L452R, E484Q and P681R, in the second wave of COVID-19 in Maharashtra, India. BioRxiv. https://doi.org/10.1101/2021.04.22.440932

  40. Hagen, A. (2021). How dangerous is the delta variant (B.1.617.2)? [Updated 30 Jul, 2021; cited 15 Aug, 2021]. Retrieved from https://asm.org/Articles/2021/July/How-Dangerous-is-the-Delta-Variant-B-1-617-2

  41. Davies NG, Jarvis CI, Edmunds WJ, Jewell NP, Diaz-Ordaz K, Keogh RH (2021) Increased mortality in community-tested cases of SARS-CoV-2 lineage B. 1.1. 7. Nature 593(7858):270–274 https://doi.org/10.1038/s41586-021-03426-1

    Article  CAS  Google Scholar 

  42. Office for National Statistics. Coronavirus (COVID-19) Infection Survey, UK: 21 May 2021 [Updated 21 May 2021; cited 26 May 2021]. Available from https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsanddiseases/bulletins/coronaviruscovid19infectionsurveypilot/21may2021

  43. Funk T, Pharris A, Spiteri G, Bundle N, Melidou A, Carr M, Adlhoch C (2021) Characteristics of SARS-CoV-2 variants of concern B. 1.1. 7, B. 1.351 or P. 1: data from seven EU/EEA countries, weeks 38/2020 to 10/2021. Eurosurveillance 26(16):2100348 https://doi.org/10.2807/1560-7917.ES.2021.26.16.2100348

    Article  CAS  Google Scholar 

  44. Challen, R., Brooks-Pollock, E., Read, J. M., Dyson, L., Tsaneva-Atanasova, K., & Danon, L. (2021). Risk of mortality in patients infected with SARS-CoV-2 variant of concern 202012/1: matched cohort study. bmj, 372(n579). https://doi.org/10.1136/bmj.n579

  45. Grint, D. J., Wing, K., Williamson, E., McDonald, H. I., Bhaskaran, K., Evans, D., ... & Eggo, R. M. (2021). Case fatality risk of the SARS-CoV-2 variant of concern B. 1.1. 7 in England, 16 November to 5 February. Eurosurveillance, 26(11), 2100256. https://doi.org/10.2807/1560-7917.ES.2021.26.11.2100256

  46. DGHS. COVID-19 press release [Updated 26 May 2021; cited 26 May 2021]. Retrieved from https://dghs.gov.bd/index.php/bd/component/content/article?layout=edit&id=5612

  47. Ranjan, R., Sharma, A., & Verma, M. K. (2021). Characterization of the Second Wave of COVID-19 in India. medRxiv. https://doi.org/10.1101/2021.04.17.21255665

  48. Shah MRT, Ahammed T, Anjum A, Chowdhury AA, Suchana AJ (2021) Finding the real COVID-19 case-fatality rates for SAARC countries. Biosafety and health. https://doi.org/10.1016/j.bsheal.2021.03.002 3(3):164–171

    Article  Google Scholar 

  49. Nasreen S, Chung H, He S, Brown KA, Gubbay JB, Buchan SA, Fell DB et al (2021) Effectiveness of COVID-19 vaccines against variants of concern in Ontario, Canada. medRxiv. https://doi.org/10.1101/2021.06.28.21259420

  50. Sheikh A, McMenamin J, Taylor B, Robertson C (2021) SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet 397(10293):P2461–P2462 https://doi.org/10.1016/S0140-6736(21)01358-1

    Article  Google Scholar 

  51. Chia, PY., Ong, SWX., Chiew, CJ., Ang, LW., Chavatte, JM., Mak, TM., et al. Virological and serological kinetics of SARS-CoV-2 Delta variant vaccine-breakthrough infections: a multi-center cohort study. medRxiv. https://doi.org/10.1101/2021.07.28.21261295

  52. Muik A, Wallisch AK, Sänger B, Swanson KA, Mühl J, Chen W, Şahin U (2021) Neutralization of SARS-CoV-2 lineage B. 1.1. 7 pseudovirus by BNT162b2 vaccine–elicited human sera. Science 371(6534):1152–1153, https://science.sciencemag.org/content/371/6534/1152. https://doi.org/10.1126/science.abg6105

  53. Smith DJ, Lapedes AS, De Jong JC, Bestebroer TM, Rimmelzwaan GF, Osterhaus AD, Fouchier RA (2004) Mapping the antigenic and genetic evolution of influenza virus. Science 305(5682):371–376.305 https://doi.org/10.1126/science.1097211

    Article  CAS  Google Scholar 

  54. Collier DA, De Marco A, Ferreira IA, Meng B, Datir RP, Walls AC et al (2021) Sensitivity of SARS-CoV-2 B. 1.1. 7 to mRNA vaccine-elicited antibodies. Nature 593(7857):136–141 https://doi.org/10.1038/s41586-021-03412-7

    Article  CAS  Google Scholar 

  55. Wang P, Casner RG, Nair MS, Wang M, Yu J, Cerutti G, Liu L, Kwong PD, Huang Y, Shapiro L, Ho DD (2021) Increased resistance of SARS-CoV-2 variant P. 1 to antibody neutralization. Cell host & microbe 29(5):747–751 https://doi.org/10.1016/j.chom.2021.04.007

    Article  CAS  Google Scholar 

  56. Emary, K. R., Golubchik, T., Aley, P. K., Ariani, C. V., Angus, B., Bibi, S., ... & Oxford COVID-19 Vaccine Trial Group. (2021). Efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B. 1.1. 7): an exploratory analysis of a randomised controlled trial. Lancet, 397(10282), 1351-1362. https://doi.org/10.1016/S0140-6736(21)00628-0

  57. Xie X, Liu Y, Liu J, Zhang X, Zou J, Fontes-Garfias CR, Xia H, Swanson KA, Cutler M, Cooper D, Menachery VD, Weaver SC, Dormitzer PR, Shi PY (2021) Neutralization of SARS-CoV-2 spike 69/70 deletion, E484K and N501Y variants by BNT162b2 vaccine-elicited sera. Nature Medicine 27(4):620–621. https://www.nature.com/articles/s41591-021-01270-4?s=08. https://doi.org/10.1038/s41591-021-01270-4

    Article  CAS  PubMed  Google Scholar 

  58. Tada T, Dcosta BM, Samanovic MI, Herati RS, Cornelius A, Zhou H, Vaill A, Kazmierski W, Mulligan MJ, Landau NR (2021) Convalescent-phase sera and vaccine-elicited antibodies largely maintain neutralizing titer against global SARS-CoV-2 variant spikes. Mbio 12(3):e00696–e00621 https://doi.org/10.1128/mBio.00696-21

    Article  CAS  Google Scholar 

  59. Garcia-Beltran WF, Lam EC, Denis KS, Nitido AD, Garcia ZH, Hauser BM, Balazs AB (2021) Multiple SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. Cell 184(9):2372–2383 https://doi.org/10.1016/j.cell.2021.03.013

    Article  CAS  Google Scholar 

  60. Supasa P, Zhou D, Dejnirattisai W, Liu C, Mentzer AJ, Ginn HM, Zhao Y, Duyvesteyn HME, Nutalai R, Tuekprakhon A, Wang B, Paesen GC, Slon-Campos J, López-Camacho C, Hallis B, Coombes N, Bewley KR, Charlton S, Walter TS, Barnes E, Dunachie SJ, Skelly D, Lumley SF, Baker N, Shaik I, Humphries HE, Godwin K, Gent N, Sienkiewicz A, Dold C, Levin R, Dong T, Pollard AJ, Knight JC, Klenerman P, Crook D, Lambe T, Clutterbuck E, Bibi S, Flaxman A, Bittaye M, Belij-Rammerstorfer S, Gilbert S, Hall DR, Williams MA, Paterson NG, James W, Carroll MW, Fry EE, Mongkolsapaya J, Ren J, Stuart DI, Screaton GR (2021) Reduced neutralization of SARS-CoV-2 B. 1.1. 7 variant by convalescent and vaccine sera. Cell 184(8):2201–2211 https://doi.org/10.1016/j.cell.2021.02.033

    Article  CAS  Google Scholar 

  61. Abu-Raddad LJ, Chemaitelly H, Butt AA (2021) Effectiveness of the BNT162b2 Covid-19 Vaccine against the B. 1.1. 7 and B. 1.351 Variants. New England Journal of Medicine. https://www.nejm.org/doi/full/10.1056/NEJMc2104974. 385(2):187–189

  62. Haas EJ, Angulo FJ, McLaughlin JM, Anis E, Singer SR, Khan F, Brooks N, Smaja M, Mircus G, Pan K, Southern J, Swerdlow DL, Jodar L, Levy Y, Alroy-Preis S (2021) Impact and effectiveness of mRNA BNT162b2 vaccine against SARS-CoV-2 infections and COVID-19 cases, hospitalisations, and deaths following a nationwide vaccination campaign in Israel: an observational study using national surveillance data. Lancet 397(10287):1819–1829 https://doi.org/10.1016/S0140-6736(21)00947-8

    Article  CAS  Google Scholar 

  63. Dejnirattisai, W., Zhou, D., Supasa, P., Liu, C., Mentzer, A. J., Ginn, H. M., ... & Screaton, G. R. (2021). Antibody evasion by the Brazilian P. 1 strain of SARS-CoV-2. BioRxiv. https://doi.org/10.1101/2021.03.12.435194

  64. Zhou D, Dejnirattisai W, Supasa P, Liu C, Mentzer AJ, Ginn HM, Zhao Y, Duyvesteyn HME, Tuekprakhon A, Nutalai R, Wang B, Paesen GC, Lopez-Camacho C, Slon-Campos J, Hallis B, Coombes N, Bewley K, Charlton S, Walter TS, Skelly D, Lumley SF, Dold C, Levin R, Dong T, Pollard AJ, Knight JC, Crook D, Lambe T, Clutterbuck E, Bibi S, Flaxman A, Bittaye M, Belij-Rammerstorfer S, Gilbert S, James W, Carroll MW, Klenerman P, Barnes E, Dunachie SJ, Fry EE, Mongkolsapaya J, Ren J, Stuart DI, Screaton GR (2021) Evidence of escape of SARS-CoV-2 variant B. 1.351 from natural and vaccine-induced sera. Cell 184(9):2348–2361 https://doi.org/10.1016/j.cell.2021.02.037

    Article  CAS  Google Scholar 

  65. Wu K, Werner AP, Moliva JI, Koch M, Choi A, Stewart-Jones GB et al (2021) mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants. BioRxiv. https://doi.org/10.1101/2021.01.25.427948

  66. Lefèvre B, Tondeur L, Madec Y, Grant R, Lina B, van der Werf S, Legoff A (2021) Impact of B. 1.351 (beta) SARS-CoV-2 variant on BNT162b2 mRNA vaccine effectiveness in long-term care facilities of eastern France: a retrospective cohort study. medRxiv https://doi.org/10.1101/2021.07.28.21261285

  67. Hitchings M, Ranzani OT, Dorion M, D'Agostini TL, de Paula RC, de Paula OFP, Croda J (2021) Effectiveness of the ChAdOx1 vaccine in the elderly during SARS-CoV-2 Gamma variant transmission in Brazil. medRxiv https://doi.org/10.1101/2021.07.19.21260802

  68. Edara VV, Lai L, Sahoo M, Floyd K, Sibai M, Solis D, Suthar M (2021) Infection and vaccine-induced neutralizing antibody responses to the SARS-CoV-2 B. 1.617. 1 variant. bioRxiv https://doi.org/10.1101/2021.05.09.443299

  69. Yadav P, Sapkal GN, Abraham P, Ella R, Deshpande G, Patil DY, Mohan VK (2021) Neutralization of variant under investigation B. 1.617 with sera of BBV152 vaccinees. bioRxiv https://doi.org/10.1101/2021.04.23.441101

  70. Ferreira I, Datir R, Papa G, Kemp S, Meng B, Rakshit P, Indian SARS-CoV-2 Genomics Consortium (2021) SARS-CoV-2 B. 1.617 emergence and sensitivity to vaccine-elicited antibodies. bioRxiv https://doi.org/10.1101/2021.05.08.443253

  71. Bernal JL, Andrews N, Gower C, Gallagher E, Simmons R, Thelwall S et al (2021) Effectiveness of COVID-19 vaccines against the B. 1.617. 2 variant. medRxiv https://doi.org/10.1101/2021.05.22.21257658

  72. Gov.UK Coronavirus (COVID-19) press release, [Updated 22 May 2021; cited 11 june 2021]. Retrieved from https://www.gov.uk/government/news/vaccines-highly-effective-against-b-1-617-2-variant-after-2-doses

  73. Thiruvengadam, R., Awasthi, A., Medigeshi, G., Bhattacharya, S., Mani, S., Sivasubbu, S., & Garg, P. K. (2021). Cellular immune responses are preserved and may contribute to Chadox1 ChAdOx1 nCoV-19 vaccine effectiveness against infection due to SARS-CoV-2 B· 1· 617· 2 delta variant despite reduced virus neutralisation. https://doi.org/10.2139/ssrn.3884946

  74. Shinde V, Bhikha S, Hoosain Z, Archary M, Bhorat Q, Fairlie L et al (2021) Efficacy of NVX-CoV2373 Covid-19 Vaccine against the B. 1.351 Variant. New England Journal of Medicine 384(20):1899–1909 https://www.nejm.org/doi/full/10.1056/NEJMoa2103055

    Article  CAS  Google Scholar 

  75. Novavax COVID-19 candidate update press release [Updated 28 January 2021; cited 26 May 2021]. Retrieved from https://ir.novavax.com/news-releases/news-release-details/novavax-covid-19-vaccine-demonstrates-893-efficacy-uk-phase-3

  76. Mahase E (2021) Covid-19: South Africa pauses use of Oxford vaccine after study casts doubt on efficacy against variant. BMJ 372(n372) https://doi.org/10.1136/bmj.n296

  77. Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, Gruber WC (2020). Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. New England Journal of Medicine 383(27):2603–2615. https://www.nejm.org/doi/full/10.1056/NEJMoa2034577

  78. Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, Zaks T (2021). Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. New England Journal of Medicine 384(5):403–416. https://www.nejm.org/doi/full/10.1056/nejmoa2035389

  79. Madhi SA, Baillie V, Cutland CL, Voysey M, Koen AL, Fairlie L, Izu A (2021). Efficacy of the ChAdOx1 nCoV-19 Covid-19 vaccine against the B. 1.351 variant. New England Journal of Medicine 384(20):1885–1898. https://www.nejm.org/doi/full/10.1056/NEJMoa2102214

  80. Voysey M, Clemens SAC, Madhi SA, Weckx LY, Folegatti PM, Aley PK, Bijker E (2021). Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. The Lancet 397(10269):99–111. https://doi.org/10.1016/S0140-6736(20)32661-1

  81. Kuzmina A, Khalaila Y, Voloshin O, Keren-Naus A, Boehm-Cohen L, Raviv Y, Shemer-Avni Y, Rosenberg E, Taube R (2021) SARS-CoV-2 spike variants exhibit differential infectivity and neutralization resistance to convalescent or post-vaccination sera. Cell host & microbe 29(4):522–528 https://doi.org/10.1016/j.chom.2021.03.008

    Article  CAS  Google Scholar 

  82. Wibmer CK, Ayres F, Hermanus T, Madzivhandila M, Kgagudi P, Oosthuysen B, Moore PL (2021) SARS-CoV-2 501Y. V2 escapes neutralization by South African COVID-19 donor plasma. Nature medicine 27(4):622–625. https://www.nature.com/articles/s41591-021-01285-x. https://doi.org/10.1038/s41591-021-01285-x

    Article  CAS  PubMed  Google Scholar 

  83. Diamond, M., Chen, R., Xie, X., Case, J., Zhang, X., VanBlargan, L., Liu, Y., & Gilchuk, P. (2021). SARS-CoV-2 variants show resistance to neutralization by many monoclonal and serum-derived polyclonal antibodies. Research square, rs.3.rs-228079. https://doi.org/10.21203/rs.3.rs-228079/v1

  84. Planas D, Veyer D, Baidaliuk A, Staropoli I, Guivel-Benhassine F, Rajah MM, Planchais C, Porrot F, Robillard N, Puech J, Prot M, Gallais F, Gantner P, Velay A, le Guen J, Kassis-Chikhani N, Edriss D, Belec L, Seve A, Courtellemont L, Péré H, Hocqueloux L, Fafi-Kremer S, Prazuck T, Mouquet H, Bruel T, Simon-Lorière E, Rey FA, Schwartz O (2021) Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature 596(7871):276–280. https://doi.org/10.1038/s41586-021-03777-9

    Article  CAS  Google Scholar 

  85. McCallum, M., Bassi, J., De Marco, A., Chen, A., Walls, A. C., Di Iulio, J., & Veesler, D. (2021). SARS-CoV-2 immune evasion by variant B. 1.427/B. 1.429. BioRxiv. https://doi.org/10.1101/2021.03.31.437925

  86. Deng X, Garcia-Knight MA, Khalid MM, Servellita V, Wang C, Morris MK, Chiu CY (2021) Transmission, infectivity, and neutralization of a spike L452R SARS-CoV-2 variant. Cell 184(13):1–12. https://doi.org/10.1016/j.cell.2021.04.025

    Article  Google Scholar 

  87. Westendorf K, Žentelis S, Foster D, Vaillancourt P, Wiggin M, Lovett E, Barnhart BC (2021) LY-CoV1404 potently neutralizes SARS-CoV-2 variants. bioRxiv https://doi.org/10.1101/2021.04.30.442182

  88. Chen RE, Zhang X, Case JB, Winkler ES, Liu Y, VanBlargan LA, Diamond MS (2021) Resistance of SARS-CoV-2 variants to neutralization by monoclonal and serum-derived polyclonal antibodies. Nature medicine 27(4):717–726. https://www.nature.com/articles/S41591-021-01294-w. https://doi.org/10.1038/s41591-021-01294-w

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Zost SJ, Gilchuk P, Chen RE, Case JB, Reidy JX, Trivette A, Crowe JE (2020) Rapid isolation and profiling of a diverse panel of human monoclonal antibodies targeting the SARS-CoV-2 spike protein. Nature medicine 26(9):1422–1427. https://www.nature.com/articles/s415910200998x?elqTrackId=8e85eb20005e47b387d146c2e1379c11. https://doi.org/10.1038/s41591-020-0998-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Wildera M, Wilhelm A, Hoehl S, Pallas C, Kohmer N, Wolf T et al (2021) Limited neutralization of authentic severe acute respiratory syndrome Coronavirus 2 variants carrying E484K in vitro. Journal of Infectious Diseases. https://doi.org/10.1093/infdis/jiab355

  91. Wilhelm A, Toptan T, Pallas C, Wolf T, Goetsch U, Gottschalk R et al (2021) Antibody-mediated neutralization of authentic SARS-CoV-2 B.1.617 variants harboring L452R and T478K/E484Q. medRxiv https://doi.org/10.1101/2021.08.09.21261704

  92. European Medicines Agency. (2021). EMA issues advice on use of sotrovimab (VIR-7831) for treating COVID-19 [Updated 21 May, 2021; cited 15 Aug 2021]. Retrieved from https://www.ema.europa.eu/en/documents/referral/sotrovimab-also-known-vir-7831-gsk4182136-covid19-article-53-procedure-conditions-use-conditions_en.pdf

Download references

Funding

There is no funding for this review.

Author information

Authors and Affiliations

Authors

Contributions

PD: data curation, formal analysis, project administration, software, validation, visualization, writing—original draft, writing—review and editing. MAM: conceptualization, data curation, formal analysis, software, validation, writing—original draft, writing—review and editing. KMSUR: conceptualization, methodology, supervision, validation, writing—review and editing. MCD: writing—review and editing. DD: conceptualization, supervision, validation, writing—review and editing. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to KM Saif-Ur-Rahman.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deb, P., Molla, M.M.A., Saif-Ur-Rahman, K. et al. A review of epidemiology, clinical features and disease course, transmission dynamics, and neutralization efficacy of SARS-CoV-2 variants. Egypt J Bronchol 15, 49 (2021). https://doi.org/10.1186/s43168-021-00090-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43168-021-00090-x

Keywords