3,112
views
3
recommends
+1 Recommend
2 collections
    7
    shares

      Zoonoses now indexed by SCOPUS from December 2023. Interested in becoming a Zoonoses published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://zoonoses-journal.org/index.php/2023/04/26/zoonoses-call-for-papers-2/

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Variants of SARS Coronavirus-2 and Their Potential Impact on the Future of the COVID-19 Pandemic

      other
      1 , 2 , 1 , 2 , 1 , 2 , * ,
      Zoonoses
      Compuscript
      SARS-CoV-2, mutation, VOC, vaccination
      Bookmark

            Abstract

            The emergence of SARS-CoV-2 variants of concern (VOCs), especially the sweeping spread of the delta variant, and differing public health management strategies, have rendered global eradication of SARS-CoV-2 unlikely. The currently available COVID-19 vaccines, including the inactivated whole virus vaccines, mRNA vaccines, and adenovirus-vectored vaccines, are effective in protecting people from severe disease and death from COVID-19, but they may not confer good mucosal immunity to prevent the establishment of infection and subsequent viral shedding and transmission. Mucosal vaccines delivered via intranasal route may provide a promising direction, which, if given as a third dose after a two-dose series of intramuscular vaccination, likely promotes mucosal immunity in addition to boosting the systemic cell-mediated immunity and antibody response. However, immunity induced by vaccination, and natural infection as well, is likely to wane followed by re-infection as in the case of human coronaviruses OC43, 229E, NL63, and HKU1. It is a challenge to prevent and control COVID-19 worldwide with the increasing number of VOCs associated with increased transmissibility and changing antigenicity. Nevertheless, we may seek to end the current pandemic situation through mass vaccination and gradual relaxation of non-pharmaceutical measures, which would limit the incidence of severe COVID-19. Repeated doses of booster vaccine will likely be required, similar to influenza virus, especially for the elderly and the immunocompromised patients who are most vulnerable to infection.

            Main article text

            Coronavirus Disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has infected 214 million people and caused more than 4.4 million deaths as of 29 August, 2021 [1]. Since its first detection in humans in December of 2019, the rate of evolution of SARS-CoV-2 has proceeded at approximately two mutations per month in global human isolates [2]. These mutations have been found throughout the SARS-CoV-2 genome [3]. However, the phenotypic implications of many of these mutations have not been defined, except for those affecting the surface spike protein, which is responsible for cell entry through binding to the host receptor angiotensin converting enzyme 2 (ACE2) [4] and subsequent fusion of the viral lipid envelope with the host cell membrane [5].

            Although SARS-CoV-2 is suspected to have originated from bat SARS-related coronaviruses and to have passed through an unknown intermediate mammalian host before jumping to humans [6,7], definitive evidence remains lacking [8]. The mutation T372A in the spike receptor binding domain (RBD) has been proposed to be a key mutation in the adaption of SARS-CoV-2 to infect humans, because this mutation has been found only in SARS-CoV-2 but not in closely related viruses such as the bat SARS-related coronavirus RaTG13 [9].

            Another important example of spike mutation resulting in better adaptation of the virus to humans is the spike D614G, which was not isolated from the initial outbreak. SARS-CoV-2 strains with the D614G mutation were first found in late January 2020 [10] and are now found in nearly all strains globally. Phenotypic studies have shown that the D614G variant replicates more readily in primary human bronchial airway tissue, has greater transmissibility, and results in higher viral titer in nasal wash samples in a hamster model [11,12]. However, it does not affect disease severity or significantly change the neutralizing activity of convalescent sera [13,14]. A structural study has shown that the D614G mutation confers a more open conformation on the spike protein, thus enabling it to bind ACE2 more efficiently [15].

            Subsequently, another important spike mutation emerged, N501Y, which has been selected in viral adaptation experiments in Balb/c mice [16]. The structure of the complex between the ACE2 receptor and N501Y spike protein ectodomain has been determined through cryogenic electron microscopy, which showed that Y501 inserts into a cavity at the binding interface near Y41 of ACE2. This additional interaction confers greater ACE2 affinity on the N501Y mutant and is likely to contribute to its increased infectivity [17].

            Recently, the spike P681R mutation has been found to facilitate the cleavage of the spike protein and to enhance the fusion between the viral envelope and host cell membrane, thus leading to prominent syncytial formation in cell culture experiments. Moreover, the 681R mutant induces greater weight loss than the 681P wild type in hamster model [5].

            Although few genetic mutations at the receptor binding domain were observed in the early months of the pandemic, by late 2020, many SARS-CoV-2 variants of concern (VOCs) or variants of interest (VOIs) had emerged. The WHO defines SARS-CoV-2 VOCs as variants with higher transmissibility or detrimental changes in COVID-19 epidemiology; increased virulence or changes in clinical disease presentation; or decreased effectiveness of public health and social measures, or available diagnostics, vaccines, and therapeutics [18].

            Several notable variants that fit the VOC definition have emerged, including B.1.1.7 (alpha variant); B.1.351 (beta variant), P.1 (gamma variant), and B.1.617.2 (delta variant). They were first reported in the United Kingdom, South Africa, Brazil, and India, respectively. These variants usually demonstrate higher transmissibility and consequently have rapidly replaced the existing lineages. Some of these variants share critical mutations in the spike protein RBD among the constellation of nucleotide changes in their genomes [19]. For example, N501Y is present in B.1.1.7, B.1.351, and P.1, whereas E484K is present in B.1.351 and P.1 in addition to N501Y and D614G. E484K is a key mutation affecting the antigenicity of the spike protein [20]. The increased transmissibility of these viruses may be associated with better binding of their RBD to human ACE2 [4]. However, the delta variant has dominated the pandemic, as of August 2021. Unlike the original prototype virus, which has a mean basic reproductive number R0 of 2.79, the mean R0 of the delta variant is 5.08 [21]. The delta variant also shows a decrease in vaccine effectiveness, presumably because the spike mutations result in 2- to 10-fold lower neutralizing antibody titers [22,23].

            The emergence of the highly transmissible delta variant has sparked a debate regarding the correct strategy for pandemic control, ranging from a “zero COVID-19 policy” to a more relaxed mitigation approach. The former strategy has been successfully adopted in several places such as mainland China, Macau SAR, Hong Kong SAR, Australia, and New Zealand. In the Hong Kong SAR, the prevalence rate of COVID-19 is 0.16%, which is far lower than those in the United Kingdom (10%) and the United States (11%) at the time of writing [1,24]. The ability to achieve such a low incidence is largely attributed to the early implementation of border control and testing and quarantining of entrants, universal masking and social distancing, isolation of all confirmed cases in hospitals, rapid and comprehensive contact tracing, restriction of movement, and testing of all premises resided in or visited by confirmed cases [25,26]. With stringent measures, even a delta variant outbreak in mainland China, which started in July 2021, was successfully eliminated in August 2021. However, the social and economic effects of such drastic non-pharmaceutical measures for epidemic control are enormous and may not be sustainable in the long run. These measures are aimed not at eradicating SARS-CoV-2 but at buying time to achieve high levels of vaccine-induced immunity in the population. However, the arrival of the delta variant has dramatically changed the game. Approximately 40% and 60% of vaccinated individuals in Hong Kong SAR were inoculated with CoronaVac (Sinovac) and BNT162b2 (Comirnaty, BioNTech), respectively [27]. In test-negative case-control studies, the effectiveness of CoronVac and BNT162b2 vaccines against the delta variant is found to be 59% and 88%, respectively [28,29], compared with 65.9% and 95% respectively against the prototype virus [30,31]. On the basis of mathematical calculation, the critical vaccination level to achieve herd immunity would increase from between 67% and 98% when R0 was 2.79 previously, to between 91% and 136% when the R0 of the presently circulating delta variant is 5.08 [32]. These apparently absurd percentages indicate that although both the inactivated whole virus vaccine and mRNA vaccine are effective in providing personal protection against severe disease and death from lung infection, neither is highly effective against asymptomatic or mildly symptomatic upper respiratory tract infections, which may result in substantial viral shedding and transmission [33,34]. These findings are not unexpected, given the viral loads observed in the upper respiratory tracts of vaccinated non-human primates challenged with the prototype virus [35]. In Israel, which has a vaccination rate above 70%, the epidemic resurged after the border was reopened, and social distancing measures were relaxed [36,37]. The virus is likely to continue to circulate even among the vaccinated population and subsequently infect the unvaccinated population, which constitutes most hospital admissions and deaths.

            Although we have no crystal ball to predict whether another worse virus variant will come, the chances are high that SARS-CoV-2 will continue to circulate globally, similarly to the other coronaviruses causing common colds. Several precedents exist: the human coronavirus OC43 might have jumped to humans from bovine species in the 1890s; HKU1 might have jumped from rodents in the 1950s, 229E in the 1820s; and NL63 might have jumped from bats between 1200 and 1460 [38]. When most of the population has some degree of protection from neutralizing antibodies and cell mediated immunity, life may gradually return to normal with the reopening of borders and the relaxation of social distancing. However, as with the case of other human coronaviruses, frequent re-infection may occur following a predictable pattern, e.g., every 12–24 months due to waning immunity [39]. It is likely that the elderly and the immunosuppressed will require booster dose of vaccine when their level of protection goes down with time, perhaps as another annual vaccination similar to the case of seasonal influenza.

            Although a third booster dose of vaccine is likely to improve the systemic protection and, indirectly the mucosal protection, the existing vaccines confer poor mucosal immunity in the upper respiratory tract. Further studies must be performed to understand why the nasopharyngeal mucosa is sub-optimally protected despite high serum neutralizing antibody levels. Several mucosal vaccines administered by intranasal sprays are now in clinical trials, including two adenoviral vector COVID-19 vaccines and the influenza virus vector NS1 deleted vaccine carrying the spike RBD [4043]. These mucosal vaccines could easily be modified to carry the spike of the delta variant and are likely to constitute the second wave of COVID-19 vaccines for boosting the mucosal immunity of people who were already vaccinated with inactivated whole virus, mRNA, or adenoviral vector vaccines. Intranasal sprays are generally much more acceptable to the general population. Many people have been apprehensive about receiving injectable vaccines, because life-threatening adverse effects have been reported in rare cases, such as myopericarditis after mRNA vaccination, or dural venous sinus thrombosis with thrombocytopenia after adenoviral vector vaccination [4447].

            Novel SARS-CoV-2 variants will continue to emerge. Recently, genomic recombination has been found in the B.1.1.7 lineage viruses, thus suggesting the possibility of a sudden emergence of novel variants that dramatically differ from existing strains [48]. Global efforts in continual genomic surveillance are required for the early detection of novel variants. Antigenic characterization, similar to that performed for influenza virus, will be required to detect strains with antigenic drift. Furthermore, animal surveillance will also be important, because reverse zoonosis of SARS-CoV-2 is not an isolated event. Novel variants may arise in animals, as observed in mink-associated human COVID-19 cases [49,50].

            REFERENCES

            1. World Health Organization. WHO Coronavirus (COVID-19) Dashboard. cited 2021 Aug 29 https://covid19.who.int

            2. Bedford T, Greninger AL, Roychoudhury P, Starita LM, Famulare M, Huang ML, et al.. Cryptic transmission of SARS-CoV-2 in Washington state. Science. 2020. Vol. 370(6516):571–575

            3. Day T, Gandon S, Lion S, Otto SP. On the evolutionary epidemiology of SARS-CoV-2. Curr Biol. 2020. Vol. 30(15):R849–r857

            4. 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. Nat Microbiol. 2021. Vol. 6(9):1188–1198

            5. Saito A, Irie T, Suzuki R, Maemura T, Nasser H, Uriu K, et al.. SARS-CoV-2 spike P681R mutation, a hallmark of the Delta variant, enhances viral fusogenicity and pathogenicity. bioRxiv. 2021.

            6. Boni MF, Lemey P, Jiang X, Lam TT, Perry BW, Castoe TA, et al.. Evolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemic. Nat Microbiol. 2020. Vol. 5(11):1408–1417

            7. Lam TT, Jia N, Zhang YW, Shum MH, Jiang JF, Zhu HC, et al.. Identifying SARS-CoV-2-related coronaviruses in Malayan pangolins. Nature. 2020. Vol. 583(7815):282–285

            8. Ye Z-W, Yuan S, Yuen KS, Fung SY, Chan CP, Jin DY. Zoonotic origins of human coronaviruses. Int J Biol Sci. 2020. Vol. 16(10):1686–1697

            9. Kang L, Xiao P, Zhou B, Chen Y, Kang L, Wang Q, et al.. A selective sweep in the Spike gene has driven SARS-CoV-2 human adaptation. Cell. 2021. Vol. 184(17):4392–4400.e4

            10. Callaway E. The coronavirus is mutating – does it matter? Nature. 2020. Vol. 585(7824):174–177

            11. Hou YJ, Chiba S, Halfmann P, Ehre C, Kuroda M, Dinnon KH, et al.. SARS-CoV-2 D614G variant exhibits efficient replication ex vivo and transmission in vivo. Science. 2020. Vol. 370(6523):1464–1468

            12. Plante JA, Liu Y, Liu J, Xia H, Johnson BA, Lokugamage KG, et al.. Spike mutation D614G alters SARS-CoV-2 fitness. Nature. 2021. Vol. 592(7852):116–121

            13. Volz E, Hill V, McCrone JT, Price A, Jorgensen D, O’Toole Á, et al.. Evaluating the effects of SARS-CoV-2 spike mutation D614G on transmissibility and pathogenicity. Cell. 2021. Vol. 184(1):64–75.e11

            14. Beaudoin-Bussières G, Laumaea A, Anand SP, Prévost J, Gasser R, Goyette G, et al.. Decline of humoral responses against SARS-CoV-2 spike in convalescent individuals. mBio. 2020. Vol. 11(5):e02590–e02520

            15. Yurkovetskiy L, Wang X, Pascal KE, Tomkins-Tinch C, Nyalile TP, Wang Y, et al.. Structural and functional analysis of the D614G SARS-CoV-2 spike protein variant. Cell. 2020. Vol. 183(3):739–751.e8

            16. Gu H, Chen Q, Yang G, He L, Fan H, Deng YQ, et al.. Adaptation of SARS-CoV-2 in BALB/c mice for testing vaccine efficacy. Science. 2020. Vol. 369(6511):1603–1607

            17. Zhu X, Mannar D, Srivastava SS, Berezuk AM, Demers JP, Saville JW, et al.. Cryo-electron microscopy structures of the N501Y SARS-CoV-2 spike protein in complex with ACE2 and 2 potent neutralizing antibodies. PLoS Biol. 2021. Vol. 19(4):e3001237

            18. World Health Organization. Tracking SARS-CoV-2 variants. 25–August. 2021 cited 2021 Aug 29 https://www.who.int/en/activities/tracking-SARS-CoV-2-variants/

            19. Gobeil SMC, Janowska K, McDowell S, Mansouri K, Parks R, Stalls V, et al.. Effect of natural mutations of SARS-CoV-2 on spike structure, conformation, and antigenicity. Science. 2021. Vol. 373(6555):eabi6226

            20. Chen LL, Lu L, Choi CY, Cai JP, Tsoi HW, Chu AW, et al.. Impact of SARS-CoV-2 variant-associated RBD mutations on the susceptibility to serum antibodies elicited by COVID-19 infection or vaccination. Clin Infect Dis. 2021. ciab656

            21. Liu Y, Rocklöv J. The reproductive number of the Delta variant of SARS-CoV-2 is far higher compared to the ancestral SARS-CoV-2 virus. J Travel Med. 2021. taab124[Cross Ref]

            22. Burki TK. Lifting of COVID-19 restrictions in the UK and the Delta variant. Lancet Respir Med. 2021. Vol. 9(8):e85

            23. World Health Organization. Coronavirus disease (COVID-19) Weekly Epidemiological Update and Weekly Operational Update. cited 2021 Aug 29 https://www.who.int/emergencies/diseases/novel-coronavirus-2019/situation-reports

            24. The Government of the Hong Kong Special Administrative Region. Coronavirus Disease (COVID-19) in HK. cited 2021 Aug 29 https://chp-dashboard.geodata.gov.hk/covid-19/en.html

            25. Cheng VC, Wong SC, Chuang VW, So SY, Chen JH, Sridhar S, et al.. The role of community-wide wearing of face mask for control of coronavirus disease 2019 (COVID-19) epidemic due to SARS-CoV-2. J Infect. 2020. Vol. 81(1):107–114

            26. To KK, Sridhar S, Chiu KH, Hung DL, Li X, Hung IF, et al.. Lessons learned 1 year after SARS-CoV-2 emergence leading to COVID-19 pandemic. Emerg Microbes Infect. 2021. Vol. 10(1):507–535

            27. The HKSAR Government. Hong Kong Vaccination Dashboard. cited 2021 Aug 30 https://www.covidvaccine.gov.hk/en/dashboard

            28. Li XN, Huang Y, Wang W, Jing QL, Zhang CH, Qin PZ, et al.. Efficacy of inactivated SARS-CoV-2 vaccines against the Delta variant infection in Guangzhou: A test-negative case-control real-world study. Emerg Microbes Infect. 2021. 1–32. [Cross Ref]

            29. Lopez Bernal J, Andrews N, Gower C, Gallagher E, Simmons R, Thelwall S, et al.. Effectiveness of Covid-19 Vaccines against the B.1.617.2 (Delta) Variant. N Engl J Med. 2021. Vol. 385(7):585–594. [Cross Ref]

            30. Jara A, Undurraga EA, González C, Paredes F, Fontecilla T, Jara G, et al.. Effectiveness of an Inactivated SARS-CoV-2 Vaccine in Chile. N Engl J Med. 2021. Vol. 385(10):875–884

            31. Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al.. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med. 2020. Vol. 383(27):2603–2615

            32. Fine P, Eames K, Heymann DL. “Herd Immunity”: a rough guide. Clin Infect Dis. 2011. Vol. 52(7):911–916

            33. Brown CM, Vostok J, Johnson H, Burns M, Gharpure R, Sami S, et al.. Outbreak of SARS-CoV-2 infections, including COVID-19 vaccine breakthrough infections, associated with large public gatherings – Barnstable County, Massachusetts, July 2021. MMWR Morb Mortal Wkly Rep. 2021. Vol. 70(31):1059–1062

            34. Chia PY, Ong SWX, Chiew CJ, Ang LW, Chavatte J-M, Mak T-M, et al.. Virological and serological kinetics of SARS-CoV-2 Delta variant vaccine-breakthrough infections: a multi-center cohort study. medRxiv. 2021.

            35. Corbett KS, Flynn B, Foulds KE, Francica JR, Boyoglu-Barnum S, Werner AP, et al.. Evaluation of the mRNA-1273 Vaccine against SARS-CoV-2 in Nonhuman Primates. N Engl J Med. 2020. Vol. 383(16):1544–1555

            36. Wadman M. Israel’s grim warning: Delta can overwhelm shots. Science. 2021. Vol. 373(6557):838

            37. Kershner I. Isreal, Once the Model for Beating Covid, Faces New Surge of Infections. The New York Times. 2021.

            38. Telenti A, Arvin A, Corey L, Corti D, Diamond MS, García-Sastre A, et al.. After the pandemic: perspectives on the future trajectory of COVID-19. Nature. 2021. Vol. 596(7873):495–504

            39. Edridge AWD, Kaczorowska J, Hoste ACR, Bakker M, Klein M, Loens K, et al.. Seasonal coronavirus protective immunity is short-lasting. Nature Medicine. 2020. Vol. 26(11):1691–1693

            40. Hassan AO, Kafai NM, Dmitriev IP, Fox JM, Smith BK, Harvey IB, et al.. A single-dose intranasal ChAd vaccine protects upper and lower respiratory tracts against SARS-CoV-2. Cell. 2020. Vol. 183(1):169–184.e13

            41. van Doremalen N, Purushotham JN, Schulz JE, Holbrook MG, Bushmaker T, Carmody A, et al.. Intranasal ChAdOx1 nCoV-19/AZD1222 vaccination reduces viral shedding after SARS-CoV-2 D614G challenge in preclinical models. Sci Transl Med. 2021. Vol. 13(607):eabh0755

            42. Wu S, Huang J, Zhang Z, Wu J, Zhang J, Hu H, et al.. Safety, tolerability, and immunogenicity of an aerosolised adenovirus type-5 vector-based COVID-19 vaccine (Ad5-nCoV) in adults: preliminary report of an open-label and randomised phase 1 clinical trial. Lancet Infect Dis. 2021. [Cross Ref]

            43. The University of Hong Kong, HKU’s COVID-19 intranasal vaccine updates (Q&A). 2021

            44. Li C, Chen Y, Zhao Y, Lung DC, Ye Z, Song W, et al.. Intravenous injection of COVID-19 mRNA vaccine can induce acute myopericarditis in mouse model. Clin Infect Dis. 2021. ciab707

            45. Verma AK, Lavine KJ, Lin C-Y. Myocarditis after Covid-19 mRNA vaccination. New Eng J Med. 2021.

            46. Montgomery J, Ryan M, Engler R, Hoffman D, McClenathan B, Collins L, et al.. Myocarditis following immunization with mRNA COVID-19 vaccines in members of the US military. J Am Med Assoc Cardiol. 2021. e212833

            47. Wolf ME, Luz B, Niehaus L, Bhogal P, Bäzner H, Henkes H. Thrombocytopenia and intracranial venous sinus thrombosis after “COVID-19 Vaccine AstraZeneca” exposure. J Clin Med. 2021. Vol. 10(8):1599

            48. Jackson B, Boni MF, Bull MJ, Colleran A, Colquhoun RM, Darby A, et al.. Generation and transmission of inter-lineage recombinants in the SARS-CoV-2 pandemic. Cell. 2021.

            49. Oude Munnink BB, Sikkema RS, Nieuwenhuijse DF, Molenaar RJ, Munger E, Molenkamp R, et al.. Transmission of SARS-CoV-2 on mink farms between humans and mink and back to humans. Science. 2021. Vol. 371(6525):172

            50. Burkholz S, Pokhrel S, Kraemer BR, Mochly-Rosen D, Carback RT 3rd, Hodge T, et al.. Paired SARS-CoV-2 spike protein mutations observed during ongoing SARS-CoV-2 viral transfer from humans to minks and back to humans. Infect Genet Evol. 2021. Vol. 93:104897

            Author and article information

            Journal
            Zoonoses
            Zoonoses
            Zoonoses
            Compuscript (Shannon, Ireland )
            2737-7466
            2737-7474
            10 September 2021
            : 1
            : 1
            : e993
            Affiliations
            [1 ]State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People’s Republic of China
            [2 ]Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, People’s Republic of China
            Author notes
            *Corresponding author: E-mail: kyyuen@ 123456hku.hk (K-YY)

            Edited by: Cao Chen, Chinese Center for Disease Control and Prevention, China

            Reviewed by: Xiaoping Dong, Chinese Center for Disease Control and Prevention, China

            Article
            10.15212/ZOONOSES-2021-1003
            d6cdcf5c-0667-4b15-9fe3-edeb6834e73f
            Copyright © 2021 The Authors.

            This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY) 4.0, which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.

            History
            : 30 August 2021
            : 31 August 2021
            : 31 August 2021
            Page count
            References: 48, Pages: 4
            Categories
            Invited Commentary

            Parasitology,Animal science & Zoology,Molecular biology,Public health,Microbiology & Virology,Infectious disease & Microbiology
            VOC,vaccination,mutation,SARS-CoV-2

            Comments

            Title of comment 

            Enhancing nasal antibody responses, mucosal vaccines may prevent SARS COV19 Omicron VOC infection and transmission more effectively, enabling greater control of the pandemic and limiting the emergence of variants.

            Author

            Pranab Kumar Bhattacharya MD ( University of Calcutta ) Ficpath, WBMES ( Retired) : Affiliation -: Retired professor and Head of Pathology department , Calcutta School of Tropical medicine 108 Chittaranjan Avenue Kolkata 700073 West Bengal, India and ex Principal JMN Medical College Chakdha Nadia West 3

            Intramuscular (i.m.) ChAdOx1 nCOV19 , trade name "covishield " vaccine of company astrazeneca and sub licensed with Serum Institute of India ( mRNA 1273 vaccine for Spike Protein /RBD) and of Bharat Biotech company's whole virion inactivated Vero cell "co vaccine" which were given to Indian population in two minimum doses( free of cost) , in India ,were remarkably effective in preventing severe COVID-19 pneumonia, ARDS and hospitalisation with all sub variants( VOC) of omicron variants of concern of SARS COV-2 ,those were circulated so far in India since 2021 March and their full dose ( two/ or precautionary 3rd doses in 27% Indians ) use being associated with declining hospitalisation , ARDS, ventilation and COVID 19 related death . However, current vaccines those are given by the Indian government's health department can provide only transient protection against respiratory SARS- COV -2 viral replications , onward transmission and continuing emergence of variants of concern, by contrast, respiratory infection with SARS-CoV-2 could induces more potent serum IGg and IGa antibody mediated ( against spike protein and RBD protein) immunity against breakthrough infections ( re- infection even after vaccination or after clinical or subclinical infection) , mucosal immune defences that could inhibit viral replication and transmission, but did not happened it so in real world . Although covishield/ or co vaccine i.m. vaccinations transiently could reduce viral transmission, vaccines with if breakthrough infections occurred showed peak nasopharyngeal viral loads similar to those in unvaccinated individuals (1,2 ).  

            One study, showed that viral loads declined more rapidly in vaccines with neutralising plasma antibodies ( 2 ) but it is not yet clear whether this effect is also mediated by passive transudation of plasma antibody IGa into the nasal or other naso pharyngial respiratory mucosa, or whether intra muscular COVID- 19 vaccination can also elicit nasal mucosal responses primed by infection (as it is observed after i.m. influenza vaccination following an intranasal priming).

             Vaccine with covisheld or covaccine that induces serum IGa and IGg are mostly monomeric in nature and these antibodies are produced in the bone marrow, whereas nasal IGg and IGa are polymeric and they are produced locally by mucosal plasma cells. We all know that the polymeric nasal IGa , the antibody that plays very important role for neutralisation of any virus in the upper respiratory tract, and so passive transudation of plasma antibody if at all into the nasal mucosa is unlikely to provide long durable sterilising immunity (3).So understanding whether i.m. vaccination after COVID-19 can at all recall nasal IGa responses is an important step towards market development of nasal vaccines for SARS- COV-2 which can prevent infection and transmission of the virus .

            During worldwide circulation of SARS-CoV-2,from 2019 onward to till date multiple successive variants of concern (VOC) has genetically mutated,and emerged with high and higher Ro in the community transmission as well as immune evasion or vaccine escape phenomenon, resulting in breakthrough infection even to vaccinated people worldwide including in India also . Some Omicron sub variants appeared that are less susceptible to vaccine-induced immunity and showed high reinfection rates like B 5.2.1.7 VOC as in China, Japan , USA.but not in India ( The real data from china usually not found ) . In India, immunity is obtained to a large percentage of people by successive subclinical or clinical omicron infections and minimum two doses vaccination programs possibly provided and will provide superior protection against Omicron compared with either alone . vaccination regimes which combined both intra nasal and i.m. administration in mice induced enhanced mucosal protection against SARS-CoV-2 variants (4) .This suggests that first priming the nasal mucosa with infection or by i.m vaccine is required to induce effective intra nasal local antibody responses that may provide enhanced immunity against current and future variants. However, the cross-reactivity of nasal antibodies after infection with pre-Omicron virus is unknown.

            It is seen that commonly nasal virus-specific IGA levels also fell back to pre-COVID levels after 3 to 9 months and Omicron-binding nasal responses were particularly short-lived one .Nasal IGa responses are compartmentalised from systemic responses after vaccination, which boosted nasal and plasma IGg but had limited effects on nasal IGA.The durability of nasal antibody responses has hitherto been unclear. A Dutch study with healthcare workers found that nasal antibodies lasted maximum for 9 months after mild infection, others demonstrated rapid waning even after 3 months ( 5,6) . Neither study examined a large cohort of hospitalised patients, and findings confirm that COVID-19 can induce durable mucosal immunity. Average, nasal IGa responses disappear after 9 months and Omicron-binding IGa is particularly short-lived. Nasal IGa is the most abundant mucosal antibody and provides an important first-line defence against respiratory infection. What this author want to say is that i.m. vaccination can also boost nasal IgG, but it has limited effects on IgA, that i.m vaccination only transiently boosts nasal IgA. mRNA vaccines tend to induce stronger circulating antibody responses than those using adenoviral vectors, but this may not apply to nasal responses. Taken together this author suggest that i.m. vaccination after COVID-19 is unlikely to recall mucosal responses, and nasal IgA responses, especially those to Omicron, are more short-lived and are not substantially affected by vaccination. There is lack of long-term sterilising immunity after previous infection and/or vaccination and it highlight the need for mucosal vaccines that target nasal IgA responses. By enhancing nasal antibody responses, mucosal vaccines might prevent infection and transmission more effectively, enabling greater control of the pandemic and limiting the emergence of variants.

            Bharat Biotech's intranasal Covid vaccine named "Incovacc " received Indian government approval for use as a booster dose in India the country-wide mass inoculation program. It will be administered to those above 18 years old who have previous two doses of covishield or co vaccine as a booster dose and will be first made available in all private hospitals with purchase price by minimum money Rs 800/ per dose in private hospitals of India ( i.e. Rs 1600/ for 8 nasal drop doses 4 drops in each nostril ) and Rs 300/ ( 8 nasal drops costs government 600/ ) in government rate- A good method of huge profitable business in the name of covid- 19 omicron sub VOC . It will also be available on CoWIN app . Few weeks back on 1st December 2022, it had got approval from the Central Drugs Standard Control Organisation( CDSCO) for restricted use in emergency situations in the age group of 18 and above. The government in a release on December 1 had called it "World’s first intranasal covid vaccine. Bharat Biotech ,"Incovacc " is a "recombinant replication deficient adenovirus vectored vaccine with a pre-fusion stabilised spike protein." It stimulates a broad immune response – neutralising IgG, mucosal IgA, and T cell responses( ??), it says. It also triggers immune responses at the site of infection (in the nasal mucosa) – essential for blocking both infection and transmission of Covid-19. Phase 3 clinical trial done in 3000 participants, Incovacc has been shown to generate good immunity following 2 doses given 4 weeks apart.Headache, Fever, Running nose, Sneezing were reported as likely side effects, while severe allergic reaction was said to be "rare."

            References

            1).Eyre D.W. Taylor D. Purver M. et al.

            Effect of covid-19 vaccination on transmission of alpha and delta variants.

            N Engl J Med. 2022; 386: 744-756

             

            2).Singanayagam A. Hakki S. Dunning J. et al.

            Community transmission and viral load kinetics of the SARS-CoV-2 delta (B.1.617.2) variant in vaccinated and unvaccinated individuals in the UK: a prospective, longitudinal, cohort study.

            Lancet Infect Dis. 2022; 22: 183-195

             

            3)Wang Z. Lorenzi J.C.C. Muecksch F. et al.Enhanced SARS-CoV-2 neutralization by dimeric IgA.Sci Transl Med [Internet]. 2020; (Available from:)eabf1555

            https://stm.sciencemag.org/lookup/doi/10.1126/scitranslmed.abf1555

             

            4) .Mao T. Israelow B. Peña-Hernández M.A. et al.Unadjuvanted intranasal spike vaccine elicits protective mucosal immunity against sarbecoviruses.

            Science. 2022;

             

            5) Fröberg J. Gillard J. Philipsen R. et al.

            SARS-CoV-2 mucosal antibody development and persistence and their relation to viral load and COVID-19 symptoms. Nat Commun. 2021; 12: 5621

             

            6)Cagigi A. Yu M. Österberg B. et al.

            Airway antibodies emerge according to COVID-19 severity and wane rapidly but reappear after SARS-CoV-2 vaccination.

            JCI Insight [Internet]. 2021; 6 (Available from:)

            https://insight.jci.org/articles/view/151463

             

            2023-01-21 17:27 UTC
            +1
            One person recommends this

            Title of comment 

            Enhancing nasal antibody responses, mucosal vaccines may prevent SARS COV19 Omicron VOC infection and transmission more effectively, enabling greater control of the pandemic and limiting the emergence of variants.

            Author

            Pranab Kumar Bhattacharya MD ( University of Calcutta ) Ficpath, WBMES ( Retired) : Affiliation -: Retired professor and Head of Pathology department , Calcutta School of Tropical medicine 108 Chittaranjan Avenue Kolkata 700073 West Bengal, India and ex Principal JMN Medical College Chakdha Nadia West 3

            Intramuscular (i.m.) ChAdOx1 nCOV19 , trade name "covishield " vaccine of company astrazeneca and sub licensed with Serum Institute of India ( mRNA 1273 vaccine for Spike Protein /RBD) and of Bharat Biotech company's whole virion inactivated Vero cell "co vaccine" which were given to Indian population in two minimum doses( free of cost) , in India ,were remarkably effective in preventing severe COVID-19 pneumonia, ARDS and hospitalisation with all sub variants( VOC) of omicron variants of concern of SARS COV-2 ,those were circulated so far in India since 2021 March and their full dose ( two/ or precautionary 3rd doses in 27% Indians ) use being associated with declining hospitalisation , ARDS, ventilation and COVID 19 related death . However, current vaccines those are given by the Indian government's health department can provide only transient protection against respiratory SARS- COV -2 viral replications , onward transmission and continuing emergence of variants of concern, by contrast, respiratory infection with SARS-CoV-2 could induces more potent serum IGg and IGa antibody mediated ( against spike protein and RBD protein) immunity against breakthrough infections ( re- infection even after vaccination or after clinical or subclinical infection) , mucosal immune defences that could inhibit viral replication and transmission, but did not happened it so in real world . Although covishield/ or co vaccine i.m. vaccinations transiently could reduce viral transmission, vaccines with if breakthrough infections occurred showed peak nasopharyngeal viral loads similar to those in unvaccinated individuals (1,2 ).  

            One study, showed that viral loads declined more rapidly in vaccines with neutralising plasma antibodies ( 2 ) but it is not yet clear whether this effect is also mediated by passive transudation of plasma antibody IGa into the nasal or other naso pharyngial respiratory mucosa, or whether intra muscular COVID- 19 vaccination can also elicit nasal mucosal responses primed by infection (as it is observed after i.m. influenza vaccination following an intranasal priming).

             Vaccine with covisheld or covaccine that induces serum IGa and IGg are mostly monomeric in nature and these antibodies are produced in the bone marrow, whereas nasal IGg and IGa are polymeric and they are produced locally by mucosal plasma cells. We all know that the polymeric nasal IGa , the antibody that plays very important role for neutralisation of any virus in the upper respiratory tract, and so passive transudation of plasma antibody if at all into the nasal mucosa is unlikely to provide long durable sterilising immunity (3).So understanding whether i.m. vaccination after COVID-19 can at all recall nasal IGa responses is an important step towards market development of nasal vaccines for SARS- COV-2 which can prevent infection and transmission of the virus .

            During worldwide circulation of SARS-CoV-2,from 2019 onward to till date multiple successive variants of concern (VOC) has genetically mutated,and emerged with high and higher Ro in the community transmission as well as immune evasion or vaccine escape phenomenon, resulting in breakthrough infection even to vaccinated people worldwide including in India also . Some Omicron sub variants appeared that are less susceptible to vaccine-induced immunity and showed high reinfection rates like B 5.2.1.7 VOC as in China, Japan , USA.but not in India ( The real data from china usually not found ) . In India, immunity is obtained to a large percentage of people by successive subclinical or clinical omicron infections and minimum two doses vaccination programs possibly provided and will provide superior protection against Omicron compared with either alone . vaccination regimes which combined both intra nasal and i.m. administration in mice induced enhanced mucosal protection against SARS-CoV-2 variants (4) .This suggests that first priming the nasal mucosa with infection or by i.m vaccine is required to induce effective intra nasal local antibody responses that may provide enhanced immunity against current and future variants. However, the cross-reactivity of nasal antibodies after infection with pre-Omicron virus is unknown.

            It is seen that commonly nasal virus-specific IGA levels also fell back to pre-COVID levels after 3 to 9 months and Omicron-binding nasal responses were particularly short-lived one .Nasal IGa responses are compartmentalised from systemic responses after vaccination, which boosted nasal and plasma IGg but had limited effects on nasal IGA.The durability of nasal antibody responses has hitherto been unclear. A Dutch study with healthcare workers found that nasal antibodies lasted maximum for 9 months after mild infection, others demonstrated rapid waning even after 3 months ( 5,6) . Neither study examined a large cohort of hospitalised patients, and findings confirm that COVID-19 can induce durable mucosal immunity. Average, nasal IGa responses disappear after 9 months and Omicron-binding IGa is particularly short-lived. Nasal IGa is the most abundant mucosal antibody and provides an important first-line defence against respiratory infection. What this author want to say is that i.m. vaccination can also boost nasal IgG, but it has limited effects on IgA, that i.m vaccination only transiently boosts nasal IgA. mRNA vaccines tend to induce stronger circulating antibody responses than those using adenoviral vectors, but this may not apply to nasal responses. Taken together this author suggest that i.m. vaccination after COVID-19 is unlikely to recall mucosal responses, and nasal IgA responses, especially those to Omicron, are more short-lived and are not substantially affected by vaccination. There is lack of long-term sterilising immunity after previous infection and/or vaccination and it highlight the need for mucosal vaccines that target nasal IgA responses. By enhancing nasal antibody responses, mucosal vaccines might prevent infection and transmission more effectively, enabling greater control of the pandemic and limiting the emergence of variants.

            Bharat Biotech's intranasal Covid vaccine named "Incovacc " received Indian government approval for use as a booster dose in India the country-wide mass inoculation program. It will be administered to those above 18 years old who have previous two doses of covishield or co vaccine as a booster dose and will be first made available in all private hospitals with purchase price by minimum money Rs 800/ per dose in private hospitals of India ( i.e. Rs 1600/ for 8 nasal drop doses 4 drops in each nostril ) and Rs 300/ ( 8 nasal drops costs government 600/ ) in government rate- A good method of huge profitable business in the name of covid- 19 omicron sub VOC . It will also be available on CoWIN app . Few weeks back on 1st December 2022, it had got approval from the Central Drugs Standard Control Organisation( CDSCO) for restricted use in emergency situations in the age group of 18 and above. The government in a release on December 1 had called it "World’s first intranasal covid vaccine. Bharat Biotech ,"Incovacc " is a "recombinant replication deficient adenovirus vectored vaccine with a pre-fusion stabilised spike protein." It stimulates a broad immune response – neutralising IgG, mucosal IgA, and T cell responses( ??), it says. It also triggers immune responses at the site of infection (in the nasal mucosa) – essential for blocking both infection and transmission of Covid-19. Phase 3 clinical trial done in 3000 participants, Incovacc has been shown to generate good immunity following 2 doses given 4 weeks apart.Headache, Fever, Running nose, Sneezing were reported as likely side effects, while severe allergic reaction was said to be "rare."

            References

            1).Eyre D.W. Taylor D. Purver M. et al.

            Effect of covid-19 vaccination on transmission of alpha and delta variants.

            N Engl J Med. 2022; 386: 744-756

             

            2).Singanayagam A. Hakki S. Dunning J. et al.

            Community transmission and viral load kinetics of the SARS-CoV-2 delta (B.1.617.2) variant in vaccinated and unvaccinated individuals in the UK: a prospective, longitudinal, cohort study.

            Lancet Infect Dis. 2022; 22: 183-195

             

            3)Wang Z. Lorenzi J.C.C. Muecksch F. et al.Enhanced SARS-CoV-2 neutralization by dimeric IgA.Sci Transl Med [Internet]. 2020; (Available from:)eabf1555

            https://stm.sciencemag.org/lookup/doi/10.1126/scitranslmed.abf1555

             

            4) .Mao T. Israelow B. Peña-Hernández M.A. et al.Unadjuvanted intranasal spike vaccine elicits protective mucosal immunity against sarbecoviruses.

            Science. 2022;

             

            5) Fröberg J. Gillard J. Philipsen R. et al.

            SARS-CoV-2 mucosal antibody development and persistence and their relation to viral load and COVID-19 symptoms. Nat Commun. 2021; 12: 5621

             

            6)Cagigi A. Yu M. Österberg B. et al.

            Airway antibodies emerge according to COVID-19 severity and wane rapidly but reappear after SARS-CoV-2 vaccination.

            JCI Insight [Internet]. 2021; 6 (Available from:)

            https://insight.jci.org/articles/view/151463

             

            2023-01-21 17:27 UTC
            +1
            One person recommends this

            Comment on this article