Review Special Issues

The journey so far with SARS-CoV-2 variants: Pathogenesis, immunity and treatments

  • Received: 29 April 2023 Revised: 22 August 2023 Accepted: 24 August 2023 Published: 15 September 2023
  • The recruitment of therapeutics and most importantly COVID-19 vaccines has seen a measurable reduction in transmission, re-infection, severity, hospitalization and mortality associated with the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The development and approval of some vaccines and therapeutics undoubtedly signaled renewed hope for public health personnel, the government, the World Health Organization (WHO) and the entire world population. At present, most countries have progressed beyond administering first and second doses to administering COVID-19 vaccine updated boosters to prevent transmission and provide protection. Notably, a bivalent COVID-19 vaccine from Pfizer–BioNTech and Moderna, also called an “updated” COVID-19 vaccine booster dose, is a formulation that houses the original virus strain and omicron BA.1, which provides broad immunity against COVID-19 including the omicron variant (BA.1) and the Paxlovid drug (Nirmatrelvir-ritonavir) authorized for use by the Food and Drug Administration (FDA) and the European Medicines Agency. This current review outlines the variant of concern (VOC), viral cell entry and pathogenesis, host immunity and viral immune evasion. In addition, we discuss the therapeutic and vaccine treatment approach, WHO and FDA authorization, vaccine storage and vaccine efficacy. In conclusion, bearing in mind the trend of continued mutations as observed on the spike (S) glycoprotein and receptor binding domain (RBD) of SARS-CoV-2, which lead to more immune-evasive strains such as BQ.1, BQ.1.1, BF.7, XBB and XBB.1, researcher and clinician attention should be tailored toward the design and development of variant-specific vaccines for future interventions.

    Citation: Daniel Danladi Gaiya, Jonathan Danladi Gaiya, Richard Auta, Aliyu Muhammad, Bege Jonathan, Stella Kuyet Udu, Ekpa Emmanuel, Amina Shehu Bature. The journey so far with SARS-CoV-2 variants: Pathogenesis, immunity and treatments[J]. AIMS Allergy and Immunology, 2023, 7(4): 222-250. doi: 10.3934/Allergy.2023016

    Related Papers:

  • The recruitment of therapeutics and most importantly COVID-19 vaccines has seen a measurable reduction in transmission, re-infection, severity, hospitalization and mortality associated with the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The development and approval of some vaccines and therapeutics undoubtedly signaled renewed hope for public health personnel, the government, the World Health Organization (WHO) and the entire world population. At present, most countries have progressed beyond administering first and second doses to administering COVID-19 vaccine updated boosters to prevent transmission and provide protection. Notably, a bivalent COVID-19 vaccine from Pfizer–BioNTech and Moderna, also called an “updated” COVID-19 vaccine booster dose, is a formulation that houses the original virus strain and omicron BA.1, which provides broad immunity against COVID-19 including the omicron variant (BA.1) and the Paxlovid drug (Nirmatrelvir-ritonavir) authorized for use by the Food and Drug Administration (FDA) and the European Medicines Agency. This current review outlines the variant of concern (VOC), viral cell entry and pathogenesis, host immunity and viral immune evasion. In addition, we discuss the therapeutic and vaccine treatment approach, WHO and FDA authorization, vaccine storage and vaccine efficacy. In conclusion, bearing in mind the trend of continued mutations as observed on the spike (S) glycoprotein and receptor binding domain (RBD) of SARS-CoV-2, which lead to more immune-evasive strains such as BQ.1, BQ.1.1, BF.7, XBB and XBB.1, researcher and clinician attention should be tailored toward the design and development of variant-specific vaccines for future interventions.



    加载中


    Conflict of interest



    All authors declare no conflicts of interest in this paper.

    [1] Lauer SA, Grantz KH, Bi Q, et al. (2020) The incubation period of coronavirus disease 2019 (COVID-19) from publicly reported confirmed cases: Estimation and application. Ann Intern Med 172: 577-582. https://doi.org/10.7326/M20-0504
    [2] Dai L, Gao GF (2021) Viral targets for vaccines against COVID-19. Nat Rev Immunol 21: 73-82. https://doi.org/10.1038/s41577-020-00480-0
    [3] Townsend JP, Hassler HB, Wang Z, et al. (2021) The durability of immunity against reinfection by SARS-CoV-2: a comparative evolutionary study. Lancet Microbe 2: e666-e675. https://doi.org/10.1016/S2666-5247(21)00219-6
    [4] Choi JY, Smith DM (2021) SARS-CoV-2 variants of concern. Yonsei Med J 62: 961-968. https://doi.org/10.3349/ymj.2021.62.11.961
    [5] Petersen E, Ntoumi F, Hui DS, et al. (2022) Emergence of new SARS-CoV-2 variant of concern omicron (B.1.1.529)—highlights Africa's research capabilities, but exposes major knowledge gaps, inequities of vaccine distribution, inadequacies in global COVID-19 response and control efforts. Int J Infect Dis 114: 268-272. https://doi.org/10.1016/j.ijid.2021.11.040
    [6] Alsaadi E, Jones IM (2019) Membrane binding proteins of coronaviruses. Future Virol 14: 275-286. https://doi.org/10.2217/fvl-2018-0144
    [7] Walls AC, Park Y, Tortorici MA, et al. (2020) Structure, function and antigenicity of the SAR-CoV-2 spike glycoprotein. Cell 180: 281-292. https://doi.org/10.1016/j.cell.2020.02.058
    [8] Chukwudozie OS, Gray CM, Fagbayi TA, et al. (2021) Immuno-informatics design of a multimeric epitope peptide based vaccine targeting SARS-CoV-2 spike glycoprotein. PloS One 16: e0248061. https://doi.org/10.1371/journal.pone.0248061
    [9] Amanat F, Krammer F (2020) SARS-CoV-2 vaccines: Status report. Immunity 52: 583-589. https://doi.org/10.1016/j.immuni.2020.03.007
    [10] Tosta E (2021) The protective immunity induced by SARS-CoV-2 infection and vaccination: a critical appraisal. Explor Immunol 1: 199-225. https://doi.org/10.37349/ei.2021.00014
    [11] Sumon TA, Hussain MA, Hasan MT, et al. (2021) A revisit to the research updates of drugs, vaccines, and bioinformatics approaches in combating COVID-19 pandemic. Front Mol Biosci 7: 585899. https://doi.org/10.3389/fmolb.2020.585899
    [12] Garg M, Maralakunte M, Kumar Y, et al. (2021) Vaccine-induced immune responses against SARS-CoV-2 infections. Explor Immunol 1: 356-373. https://doi.org/10.37349/ei.2021.00024
    [13] Sonani B, Aslam F, Goyal A, et al. (2021) COVID-19 vaccination in immunocompromised patients. Clin Rheumatol 40: 797-798. https://doi.org/10.1007/s10067-020-05547-w
    [14] Pollard AJ, Bijker EM (2021) Publisher correction: A guide to vaccinology: from basic principles to new developments. Nat Rev Immunol 21: 129. https://doi.org/10.1038/s41577-020-00497-5
    [15] Peng Y, Mentzer AJ, Liu G, et al. (2020) Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat Immunol 21: 1336-1345. https://doi.org/10.1038/s41590-020-0782-6
    [16] Kim YI, Casel MAB, Choi YK (2022) Transmissibility and pathogenicity of SARS-CoV-2 variants in animal models. J Microbiol 60: 255-267. https://doi.org/10.1007/s12275-022-2033-z
    [17] Chemaitelly H, Yassine HM, Benslimane FM, et al. (2021) mRNA-1273 COVID-19 vaccine effectiveness against the B.1.1.7 and B.1.351 variants and severe COVID-19 disease in Qatar. Nat Med 27: 1614-1621. https://doi.org/10.1038/s41591-021-01446-y
    [18] (2021) WHOTracking SARS-CoV-2 variants.World Health Organization. Available from: https://www.who.int/en/activities/tracking-SARS-CoV-2-variants/
    [19] Chaillon A, Smith DM, et al. (2021) Phylogenetic analyses of severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) B.1.1.7 lineage suggest a single origin followed by multiple exportation events versus convergent evolution. Clin Infect Dis 73: 2314-2317. https://doi.org/10.1093/cid/ciab265
    [20] Davies NG, Abbott S, Barnard RC, et al. (2021) Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science 372: eabg3055. https://doi.org/10.1126/science.abg3055
    [21] Subissi L, von Gottberg A, Thukral L, et al. (2022) An early warning system for emerging SARS-CoV-2 variants. Nat Med 28: 1110-1115. https://doi.org/10.1126/science.abg3055
    [22] Graham MS, Sudre CH, May A, 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: e335-e345. https://doi.org/10.1016/s2468-2667(21)00055-4
    [23] Chavda VP, Patel AB, Vaghasiya DD, et al. (2022) SARS-CoV-2 variants and vulnerability at the global level. J Med Virol 94: 2986-3005. https://doi.org/10.1002/jmv.27717
    [24] Meng B, Kemp SA, Papa G, et al. (2021) Recurrent emergence of SARS-CoV-2 spike deletion H69/V70 and its role in the Alpha variant B.1.1.7. Cell Rep 35: 109292. https://doi.org/10.1016/j.celrep.2021.109292
    [25] Starr TN, Greaney AJ, Hilton SK, et al. (2020) Deep mutational scanning of SARS-CoV-2 receptor binding domain reveals constraints on folding and ACE2 binding. Cell 182: 1295-1310. https://doi.org/10.1016/j.cell.2020.08.012
    [26] Collier DA, De Marco A, Ferreira IA, et al. (2022) Author correction: Sensitivity of SARS-CoV-2 B.1.1.7 to mRNA vaccine-elicited antibodies. Nature 608: E24. https://doi.org/10.1038/s41586-022-05103-3
    [27] Dagan N, Barda N, Kepten E, et al. (2021) BNT162b2 mRNA COVID-19 vaccine in a nationwide mass vaccination setting. N Engl J Med 384: 1412-1423. https://doi.org/10.1056/nejmoa2101765
    [28] Barbra AD, Gerlovin H, Madenci AL, et al. (2022) Comparative effectiveness of BNT162b2 and mRNA-1273 vaccines in U.S. veterans. N Engl J Med 386: 105-115. https://doi.org/10.1056/nejmoa2115463
    [29] McNamara LA, Wiegand RE, Burke RM, et al. (2022) Estimating the early impact of the US COVID-19 vaccination programme on COVID-19 cases, emergency department visits, hospital admissions, and deaths among adults aged 65 years and older: an ecological analysis of national surveillance data. Lancet 399: 152-160. https://doi.org/10.1016/s0140-6736(21)02226-1
    [30] George NI, Locke ER, O'Hare AM, et al. (2022) COVID-19 vaccination effectiveness against infection or death in a national US health care system: A target trial emulation study. Ann Intern Med 175: 352-361. https://doi.org/10.7326/m21-3256
    [31] Mahase E (2021) Covid-19: Novavax vaccine efficacy is 86% against UK variant and 60% against South African variant. BMJ 372: n296. https://doi.org/10.1136/bmj.n296
    [32] Mark WT, Self WH, Gaglani M, et al. (2022) Effectiveness of mRNA vaccination in preventing COVID-19-associated invasive mechanical ventilation and death—United States, March 2021–January 2022. MMWR Morb Mortal Wkly Rep 71: 459-465. https://doi.org/10.15585/mmwr.mm7112e1
    [33] Khan A, Zia T, Suleman M, et al. (2021) Higher infectivity of the SARS-CoV-2 new variants is associated with K417N/T, E484K, and N501Y mutants: An insight from structural data. J Cell Physiol 236: 7045-7057. https://doi.org/10.1002/jcp.30367
    [34] William NW, Ip S, Cooper JA, et al. (2022) Association of COVID-19 vaccines ChAdOx1 and BNT162b2 with major venous, arterial, or thrombocytopenic events: A population-based cohort study of 46 million adults in England. PLoS Med 19: e1003926. https://doi.org/10.1371/journal.pmed.1003926
    [35] Andeweg SP, Vennema H, Veldhuijzen I, et al. (2023) Elevated risk of infection with SARS-CoV-2 Beta, Gamma, and Delta variants compared with Alpha variant in vaccinated individuals. Sci Transl Med 15: eabn4338. https://doi.org/10.1126/scitranslmed.abn4338
    [36] Boehm E, Kronig I, Neher RA, et al. (2021) Novel SARS-CoV-2 variants: the pandemics within the pandemic. Clin Microbiol Infect 27: 1109-1117. https://doi.org/10.1016%2Fj.cmi.2021.05.022
    [37] Kustin T, Harel N, Finkel U, et al. (2021) Evidence for increased breakthrough rates of SARS-CoV-2 variants of concern in BNT162b2-mRNA-vaccinated individuals. Nat Med 27: 1379-1384. https://doi.org/10.1038/s41591-021-01413-7
    [38] Liu J, Liu Y, Xia H, et al. (2021) BNT162b2-elicited neutralization of B.1.617 and other SARS-CoV-2 variants. Nature 596: 273-275. https://doi.org/10.1038/s41586-021-03693-y
    [39] Xie X, Liu Y, Liu J, et al. (2021) Neutralization of SARS-CoV-2 spike 69/70 deletion, E484K and N501Y variants by BNT162b2 vaccine-elicited sera. Nat Med 27: 620-621. https://doi.org/10.1038/s41591-021-01270-4
    [40] Singh H, Dahiya N, Yadav M, et al. (2022) Emergence of SARS-CoV-2 new variants and their clinical significance. Can J Infect Dis Med Microbiol 2022: 7336309. https://doi.org/10.1155/2022%2F7336309
    [41] (2021) FDAFact sheet for health care providers emergency use authorization (EUA) of bamlanivimab and etesevimab.US Food and Drug Administration. Available from: https://www.fda.gov/media/145802/download
    [42] Wang Z, Schmidt F, Weisblum Y, et al. (2021) mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants. Nature 592: 616-622. https://doi.org/10.1038/s41586-021-03324-6
    [43] Prete CA, Buss LF, Buccheri R, et al. (2022) Reinfection by the SARS-CoV-2 Gamma variant in blood donors in Manaus, Brazil. BMC Infect Dis 22: 127. https://doi.org/10.1186/s12879-022-07094-y
    [44] Banho CA, Sacchetto L, Campos GRF, et al. (2022) Impact of SARS-CoV-2 Gamma lineage introduction and COVID-19 vaccination on the epidemiological landscape of a Brazilian city. Commun Med 2: 41. https://doi.org/10.1038/s43856-022-00108-5
    [45] Buss LF, Prete CA, Abrahim CMM, et al. (2021) Three-quarters attack rate of SARS-CoV-2 in the Brazilian Amazon during a largely unmitigated epidemic. Science 371: 288-292. https://doi.org/10.1126/science.abe9728
    [46] Sabino EC, Buss LF, Carvalho MPS, et al. (2021) Resurgence of COVID-19 in Manaus, Brazil, despite high seroprevalence. Lancet 397: 452-455. https://doi.org/10.1016/s0140-6736(21)00183-5
    [47] Volz E, Mishra S, Chand M, et al. (2021) Assessing transmissibility of SARS-CoV-2 lineage B.1.1.7 in England. Nature 593: 266-269. https://doi.org/10.1038/s41586-021-03470-x
    [48] Lucas C, Vogels CBF, Yildirim I, et al. (2021) Impact of circulating SARS-CoV-2 variants on mRNA vaccine-induced immunity. Nature 600: 523-529. https://doi.org/10.1038/s41586-021-04085-y
    [49] Collier DA, Ferreira IATM, Kotagiri P, et al. (2021) Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature 596: 417-422. https://doi.org/10.1038/s41586-021-03739-1
    [50] Wang P, Casner RG, Nair MS, et al. (2021) Increased resistance of SARS-CoV-2 variant P.1 to antibody neutralization. Cell Host Microbe 29: 747-751. https://doi.org/10.1101/2021.03.01.433466
    [51] Firouzabadi N, Ghasemiyeh P, Moradishooli F, et al. (2023) Update on the effectiveness of COVID-19 vaccines on different variants of SARS-CoV-2. Int Immunopharmacol 117: 109968. https://doi.org/10.1016/j.intimp.2023.109968
    [52] (2021) WHOClassification of omicron (B.1.1.529): SARS-CoV-2 variant of concern.World Health Organization. Available from: https://www.who.int/news/item/26-11-2021-classification-of-omicron-(b.1.1.529)-sars-cov-2-variant-of-concern
    [53] Lauring AS, Malani PN (2021) Variants of SARS-CoV-2. JAMA 326: 14181. https://doi.org/10.1001/jama.2021.14181
    [54] Alkhatib M, Svicher V, Salpini R, et al. (2021) SARS-CoV-2 variants and their relevant mutational profiles: Update summer 2021. Microbiol Spectr 9: e0109621. https://doi.org/10.1128/spectrum.01096-21
    [55] Cascella M, Rajnik M, Aleem A, et al. (2023) Features, evaluation, and treatment of coronavirus (COVID-19). StatPearls. Tampa: StatPearls Publishing.
    [56] Harvey WT, Carabelli AM, Jackson B, et al. (2021) SARS-CoV-2 variants, spike mutations and immune escape. Nat Rev Microbiol 19: 409-424. https://doi.org/10.1038/s41579-021-00573-0
    [57] Fisman DN, Tuite AR (2021) Progressive increase in virulence of novel SARS-CoV-2 variants in Ontario, Canada. CMAJ 193: E1619-E1625. https://doi.org/10.1503/cmaj.211248
    [58] King KL, Wilson S, Napolitano JM, et al. (2022) SARS-CoV-2 variants of concern Alpha and Delta show increased viral load in saliva. PloS One 17: e0267750. http://dx.doi.org/10.1371/journal.pone.0267750
    [59] (2021) CDC, SARS-CoV-2 variant classifications and definitions.Centers for Disease Control and Prevention. Available from: https://www.cdc.gov/coronavirus/2019-ncov/variants/variant-classifications.html
    [60] Kannan SR, Spratt AN, Cohen AR, et al. (2021) Evolutionary analysis of the Delta and Delta Plus variants of the SARS-CoV-2 viruses. J Autoimmun 124: 102715. https://doi.org/10.1016/j.jaut.2021.102715
    [61] Planas D, Veyer D, Baidaliuk A, et al. (2021) Reduced sensitivity of SARS-CoV-2 variant delta to antibody neutralization. Nature 596: 276-280. https://doi.org/10.1038/s41586-021-03777-9
    [62] Tchesnokova V, Kulasekara H, Larson L, et al. (2021) Acquisition of the L452R mutation in the ACE2-binding interface of spike protein triggers recent massive expansion of SARS-CoV-2 variants. J Clin Microbiol 59: e0092121. https://doi.org/10.1128/jcm.00921-21
    [63] Sheikh A, Mcmenamin J, Taylor B, et al. (2021) SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness. Lancet 397: 2461-2462. https://doi.org/10.1016/s0140-6736(21)01358-1
    [64] Bernal JL, Andrews N, Gower C, et al. (2021) Effectiveness of COVID-19 vaccines against the B.1.617.2 (delta) variant. N Engl J Med 385: 585-594. https://doi.org/10.1056/nejmoa2108891
    [65] Mohsin M, Mahmud S (2022) Omicron SARS-CoV-2 variant of concern: A review on its transmissibility, immune evasion, reinfection, and severity. Medicine 101: e29165. https://doi.org/10.1097/md.0000000000029165
    [66] Ntoumi FPE, Hui DS, Abubakar A, et al. (2022) Emergence of new SARS-CoV-2 variant of concern omicron (B.1.1.529)—highlights Africa's research capabilities, but exposes major knowledge gaps, inequities of vaccine distribution, inadequacies in global COVID-19 response and control efforts. Int J Infect Dis 114: 268-272. https://doi.org/10.1016/j.ijid.2021.11.040
    [67] Rio C, Malani PN (2022) COVID-19 in 2022—The beginning of the end or the end of the beginning?. JAMA 327: 2389-2390. https://doi.org/10.1001/jama.2022.9655
    [68] Yamasoba D, Kimura I, Nasser H, et al. (2022) Virological characteristics of the SARS-CoV-2 Omicron BA.2 spike. Cell 185: 2103-2115. https://doi.org/10.1016/j.cell.2022.04.035
    [69] Lassaunière R, Polacek C, Frische A, et al. (2022) Neutralizing antibodies against the SARS-CoV-2 omicron variant (BA.1) 1 to 18 weeks after the second and third doses of the BNT162b2 mRNA vaccine. JAMA Network Open 5: e2212073. https://doi.org/10.1001/jamanetworkopen.2022.12073
    [70] Cao Y, Yisimayi A, Jian F, et al. (2022) BA.2.12.1, BA.4 and BA.5 escape antibodies elicited by Omicron infection. Nature 608: 593-602. https://doi.org/10.1038/s41586-022-04980-y
    [71] Centers for Disease Control and Prevention.SARS-CoV-2 B.1.1.529 (Omicron) Variant United States. Morb Mortal Wkly Rep (2021) 70: 1731-1734. http://dx.doi.org/10.15585/mmwr.mm7050e1
    [72] Wolter N, Jassat W, Walaza S, et al. (2021) Early assessment of the clinical severity of the SARS-CoV-2 omicron variant in South Africa. Lancet 399: 437-446. https://doi.org/10.1016/s0140-6736(22)00017-4
    [73] Tegally H, Moir M, Everatt J, et al. (2022) Emergence of SARS-CoV-2 omicron lineages BA.4 and BA.5 in South Africa. Nat Med 28: 1785-1790. https://doi.org/10.1038/s41591-022-01911-2
    [74] Callaway E (2021) Heavily mutated omicron variant puts scientists on alert. Nature 600: 21. https://doi.org/10.1038/d41586-021-03552-w
    [75] Liu L, Iketani S, Guo Y, et al. (2022) Striking antibody evasion manifested by the omicron variant of SARS-CoV-2. Nature 602: 676-681. https://doi.org/10.1038/s41586-021-04388-0
    [76] Hoffmann M, Krüger N, Schulz S, et al. (2022) The Omicron variant is highly resistant against antibody-mediated neutralization: Implications for control of the COVID-19 pandemic. Cell 185: 447-456.e11. https://doi.org/10.1016/j.cell.2021.12.032
    [77] Jiang S, Hillyer C, Du L, et al. (2020) Neutralizing antibodies against SARS-CoV-2 and other human coronaviruses. Trends Immunol 41: 355-359. https://doi.org/10.1016/j.it.2020.03.007
    [78] Watanabe Y, Allen JD, Wrapp D, et al. (2020) Site-specific glycan analysis of the SARS-CoV-2 spike. Science 369: 330-333. https://doi.org/10.1126/science.abb9983
    [79] Zhou H, Ni WJ, Huang W, et al. (2022) Advances in pathogenesis, progression, potential targets and targeted therapeutic strategies in SARS-CoV-2-induced COVID-19. Front Immunol 13: 834942. https://doi.org/10.3389/fimmu.2022.834942
    [80] De Abajo FJ, Rodríguez-Martín S, Lerma V, et al. (2020) Use of renin-angiotensin-aldosterone system inhibitors and risk of COVID-19requiring admission to hospital: a case-population study. Lancet 395: 1705-1714. https://doi.org/10.1016/s0140-6736(20)31030-8
    [81] Hoffmann M, Kleine-Weber H, Schroeder S, et al. (2020) SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181: 271-280. https://doi.org/10.1016/j.cell.2020.02.052
    [82] Xu H, Zhong L, Deng J, et al. (2020) High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int J Oral Sci 12: 8. https://doi.org/10.1038/s41368-020-0074-x
    [83] Lu S, Ye Q, Singh D, et al. (2021) The SARS-CoV-2 nucleocapsid phosphoprotein forms mutually exclusive condensates with RNA and the membrane-associated M protein. Nat Commun 12: 502. https://doi.org/10.1038/s41467-020-20768-y
    [84] Mandala VS, McKay MJ, Shcherbakov AA, et al. (2020) Structure and drug binding of the SARS-CoV-2 envelope protein transmembrane domain in lipid bilayers. Nat Struct Mol Biol 27: 1202-1208. https://doi.org/10.1038/s41594-020-00536-8
    [85] Peng Y, Du N, Lei Y, et al. (2020) Structures of the SARS-CoV-2 nucleocapsid and their perspectives for drug design. EMBO J 39: e105938. https://doi.org/10.15252/embj.2020105938
    [86] Ricci D, Etna MP, Rizzo F, et al. (2021) Innate immune response to SARS-CoV-2 infection: From cells to soluble mediators. Int J Mol Sci 22: 7017. https://doi.org/10.3390/ijms22137017
    [87] Schultze JL, Aschenbrenner AC (2021) COVID-19 and the human innate immune system. Cell 184: 1671-1692. https://doi.org/10.1016/j.cell.2021.02.029
    [88] Gu W, Gan H, Ma Y, et al. (2022) The molecular mechanism of SARS-CoV-2 evading host antiviral innate immunity. Virol J 19: 49. https://doi.org/10.1186/s12985-022-01783-5
    [89] Wauters E, Van Mol P, Garg AD, et al. (2021) Discriminating mild from critical COVID-19 by innate and adaptive immune single-cell profiling of bronchoalveolar lavages. Cell Res 31: 272-290. https://doi.org/10.1038/s41422-020-00455-9
    [90] Zhang S, Wang L, Cheng G, et al. (2022) The battle between host and SARS-CoV-2: Innate immunity and viral evasion strategies. Mol Ther 30: 1869-1884. https://doi.org/10.1016/j.ymthe.2022.02.014
    [91] Paludan SR, Mogensen TH (2022) Innate immunological pathways in COVID-19 pathogenesis. Sci Immunol 7: eabm5505. https://doi.org/10.1126/sciimmunol.abm5505
    [92] Thorne LG, Reuschl AK, Zuliani-Alvarez L, et al. (2021) SARS-CoV-2 sensing by RIG-I and MDA5 links epithelial infection to macrophage inflammation. EMBO J 40: e107826. https://doi.org/10.15252/embj.2021107826
    [93] Cheemarla NR, Watkins TA, Mihaylova VT, et al. (2021) Dynamic innate immune response determines susceptibility to SARS-CoV-2 infection and early replication kinetics. J Exp Med 218: e20210583. https://doi.org/10.1084/jem.20210583
    [94] Shah VK, Firmal P, Alam A, et al. (2020) Overview of immune response during SARS-CoV-2 infection: Lessons from the past. Front Immunol 11: 1949. https://doi.org/10.3389/fimmu.2020.01949
    [95] Li H, Liu L, Zhang D, et al. (2020) SARS-CoV-2 and viral sepsis: observations and hypotheses. Lancet 395: 1517-1520. https://doi.org/10.1016/S0140-6736(20)30920-X
    [96] Mehta P, McAuley DF, Brown M, et al. (2020) COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 395: 1033-1034. https://doi.org/10.1016/S0140-6736(20)30628-0
    [97] Karki R, Sharma BR, Tuladhar S, et al. (2021) Synergism of TNF-α and IFN-g triggers inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell 184: 149-168. https://doi.org/10.1016/j.cell.2020.11.025
    [98] Leisman DE, Ronner L, Pinotti R, et al. (2020) Cytokine elevation in severe and critical COVID-19: a rapid systematic review, meta-analysis, and comparison with other inflammatory syndromes. Lancet Respir Med 8: 1233-1244. https://doi.org/10.1016/s2213-2600(20)30404-5
    [99] Contoli M, Papi A, Tomassetti L, et al. (2021) Blood interferon-α levels and severity, outcomes, and inflammatory profiles in hospitalized COVID-19 patients. Front Immunol 12: 648004. https://doi.org/10.3389/fimmu.2021.648004
    [100] Chen G, Wu D, Guo W, et al. (2020) Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 130: 2620-2629. https://doi.org/10.1172/jci137244
    [101] Lau SKP, Lau CCY, Chan KH, et al. (2013) Delayed induction of proinflammatory cytokines and suppression of innate antiviral response by the novel Middle East respiratory syndrome coronavirus: implications for pathogenesis and treatment. J Gen Virol 94: 2679-2690. https://doi.org/10.1099/vir.0.055533-0
    [102] Pereda R, González D, Rivero HB, et al. (2020) Therapeutic effectiveness of interferon alpha 2b treatment for COVID-19 patient recovery. J Interferon Cytokine Res 40: 578-588. https://doi.org/10.1089/jir.2020.0188
    [103] Yang Y, Shen C, Li J, et al. (2020) Plasma IP-10 and MCP-3 levels are highly associated with disease severity and predict the progression of COVID-19. J Allergy Clin Immunol 146: 119-127. https://doi.org/10.1016/j.jaci.2020.04.027
    [104] Schreiber G (2020) The role of type I interferons in the pathogenesis and treatment of COVID-19. Front Immunol 11: 595739. https://doi.org/10.3389/fimmu.2020.595739
    [105] Sui L, Zhao Y, Wang W, et al. (2021) SARS-CoV-2 membrane protein inhibits type I interferon production through ubiquitin-mediated degradation of TBK1. Front Immunol 12: 662989. https://doi.org/10.3389/fimmu.2021.662989
    [106] Li A, Zhao K, Zhang B, Hua R, et al. (2021) SARS-CoV-2 NSP12 protein is not an interferon-β antagonist. J Virol 95: e0074721. https://doi.org/10.1128/jvi.00747-21
    [107] Konno Y, Kimura I, Uriu K, et al. (2020) SARS-CoV-2 ORF3b is a potent interferon antagonist whose activity is increased by a naturally occurring elongation variant. Cell Rep 32: 108185. https://doi.org/10.1016/j.celrep.2020.108185
    [108] Geng H, Subramanian S, Wu L, et al. (2021) SARS-CoV-2 ORF8 forms intracellular aggregates and inhibits IFNγ-induced antiviral gene expression in human lung epithelial cells. Front Immunol 12: 679482. https://doi.org/10.3389/fimmu.2021.679482
    [109] Wang W, Zhou Z, Xiao X, et al. (2021) SARS-CoV-2 nsp12 attenuates type I interferon production by inhibiting IRF3 nuclear translocation. Cell Mol Immunol 18: 945-953. https://doi.org/10.1038/s41423-020-00619-y
    [110] Lei J, Hilgenfeld R (2017) RNA-virus proteases counteracting host innate immunity. FEBS Lett 591: 3190-3210. https://doi.org/10.1002/1873-3468.12827
    [111] Prescott L (2022) SARS-CoV-2 3CLpro whole human proteome cleavage prediction and enrichment/depletion analysis. Comput Biol Chem 98: 107671. https://doi.org/10.1016/j.compbiolchem.2022.107671
    [112] Wang D, Fang L, Shi Y (2016) Porcine epidemic diarrhea virus 3C-like protease regulates its interferon antagonism by cleaving NEMO. J Virol 90: 2090-2101. https://doi.org/10.1128/jvi.02514-15
    [113] Gaglia MM, Covarrubias S, Wong W, et al. (2012) A common strategy for host RNA degradation by divergent viruses. J Virol 86: 9527-9530. https://doi.org/10.1128/jvi.01230-12
    [114] Zhang K, Miorin L, Makio T, et al. (2021) Nsp1 protein of SARS-CoV-2 disrupts the mRNA export machinery to inhibit host gene expression. Sci Adv 7: eabe7386. https://doi.org/10.1126/sciadv.abe7386
    [115] Finkel Y, Gluck A, Nachshon A, et al. (2021) SARS-CoV-2 uses a multipronged strategy to impede host protein synthesis. Nature 594: 240-245. https://doi.org/10.1038/s41586-021-03610-3
    [116] Lo MW, Woodruff TM (2020) Complement: Bridging the innate and adaptive immune systems in sterile inflammation. J Leukoc Biol 108: 339-351. https://doi.org/10.1002/JLB.3MIR0220-270R
    [117] Hsu JCC, Laurent-Rolle M, Pawlak JB, et al. (2021) Translational shutdown and evasion of the innate immune response by SARS-CoV-2 NSP14 protein. Proc Natl Acad Sci USA 118: e2101161118. https://doi.org/10.1073/pnas.2101161118
    [118] Noris M, Remuzzi G (2013) Overview of complement activation and regulation. Sem Nephr 33: 479-492. https://doi.org/10.1016/j.semnephrol.2013.08.001
    [119] Ali YM, Ferrari M, Lynch NJ, et al. (2021) Lectin pathway mediates complement activation by SARS-CoV-2 proteins. Front Immunol 12: 714511. https://doi.org/10.3389/fimmu.2021.714511
    [120] Holter JC, Pischke SE, de Boer E, et al. (2020) Systemic complement activation isassociated with respiratory failure in COVID-19 hospitalized patients. Proc Natl Acad Sci USA 117: 25018-25025. https://doi.org/10.1073/pnas.2010540117
    [121] Gao T, Hu M, Zhang X, et al. (2020) Highly pathogenic coronavirus N protein aggravates lung injury by MASP-2-mediated complement over-activation. medRxiv . Preprint. http://dx.doi.org/10.1101/2020.03.29.20041962
    [122] Yu J, Yuan X, Chen H, et al. (2020) Direct activation of the alternative complement pathway by SARS-CoV-2 spike proteins is blocked by factor D inhibition. Blood 136: 2080-2089. https://doi.org/10.1182/blood.2020008248
    [123] Wen W, Chen C, Tang J, et al. (2022b) Efficacy and safety of three new oral antiviral treatment (molnupiravir, fluvoxamine and Paxlovid) for COVID-19: a meta-analysis. Ann Med 54: 516-523. https://doi.org/10.1080/07853890.2022.2034936
    [124] Bernal AJ, da Silva MMG, Musungaie DB, et al. (2022) Molnupiravir for oral treatment of Covid-19 in nonhospitalized patients. N Engl J Med 386: 509-520. https://doi.org/10.1056/nejmoa2116044
    [125] Gandhi RT, Malani PN, Del Rio C (2022) COVID-19 therapeutics for nonhospitalized patients. JAMA 327: 617-618. https://doi.org/10.1001/jama.2022.0335
    [126] (2021) Pfizer Inc.Pfizer's novel COVID-19 oral antiviral treatment candidate reduced risk of hospitalization or death by 89% in interim analysis of phase 2/3 EPIC-HR study.Pfizer Inc.. Available from: https://www.pfizer.com/news/press-release/press-release-detail/pfizers-novel-covid-19-oral-antiviral-treatment-candidate
    [127] Mahase E (2021b) Covid-19: Pfizer's paxlovid is 89% effective in patients at risk of serious illness, company reports. BMJ 375: n2713. https://doi.org/10.1136/bmj.n2713
    [128] Seftel D, Boulware DR (2021) Prospective cohort of fluvoxamine for early treatment of Coronavirus disease 19. Open Forum Infect Dis 8: ofab050. https://doi.org/10.1093/ofid/ofab050
    [129] Calusic M, Marcec R, Luksa L, et al. (2022) Safety and efficacy of fluvoxamine in COVID-19 ICU patients: An open label, prospective cohort trial with matched controls. Br J Clin Pharmacol 88: 2065-2073. https://doi.org/10.1111/bcp.15126
    [130] Gottlieb RL, Vaca CE, Paredes R, et al. (2022) Early Remdesivir to Prevent Progression to Severe Covid-19 in Outpatients. N Engl J Med 386: 305-315. https://doi.org/10.1056/nejmoa2116846
    [131] Jason DG, Lye DCB, Hui DS, et al. (2020) Remdesivir for 5 or 10 days in patients with severe COVID-19. N Engl J Med 383: 1827-1837. https://doi.org/10.1056/nejmoa2015301
    [132] Spinner CD, Gottlieb RL, Criner GJ, et al. (2020) Effect of remdesivir vs standard care on clinical status at 11 days in patients with moderate COVID-19: A randomized clinical trial. JAMA 324: 1048-1057. https://doi.org/10.1001/jama.2020.16349
    [133] Vegivinti CTR, Evanson KW, Lyons H, et al. (2022) Efficacy of antiviral therapies for COVID-19: a systematic review of randomized controlled trials. BMC Infect Dis 22: 107. https://doi.org/10.1186/s12879-022-07068-0
    [134] Wang L, Zhou T, Zhang Y, et al. (2021) Antibodies with potent and broad neutralizing activity against antigenically diverse and highly transmissible SARS-CoV-2 variants. BioRxiv . Preprint. https://doi.org/10.1101/2021.02.25.432969
    [135] Wang Q, Bowen A, Valdez R, et al. (2023) Antibody response to omicron BA.4-BA.5 bivalent booster. N Engl J Med 388: 567-569. https://doi.org/10.1056/NEJMc2213907
    [136] Eric JH, Angulo FJ, McLaughlin JM, et al. (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: 1819-1829. https://doi.org/10.1016/s0140-6736(21)00947-8
    [137] Thompson MG, Stenehjem E, Grannis S, et al. (2021) Effectiveness of Covid-19 vaccines in ambulatory and inpatient care settings. N Engl J Med 385: 1355-1371. https://doi.org/10.1056/NEJMoa2110362
    [138] Vasileiou E, Simpson CR, Shi T, et al. (2021) Interim findings from first-dose mass COVID-19 vaccination roll-out and COVID-19 hospital admissions in Scotland: a national prospective cohort study. Lancet 397: 1646-1657. https://doi.org/10.1016/s0140-6736(21)00677-2
    [139] Al Kaabi N, Zhang Y, Xia S, et al. (2021) Effect of 2 inactivated SARS-CoV-2 vaccines on symptomatic COVID-19 infection in adults: A randomized clinical trial: A randomized clinical trial. JAMA 326: 35-45. https://doi.org/10.1001/jama.2021.8565
    [140] Offit PA (2023) Bivalent COVID-19 vaccines—A cautionary tale. N Engl J Med 388: 481-483. https://doi.org/10.1056/nejmp2215780
    [141] Link-Gelles R, Ciesla AA, Fleming-Dutra KE, et al. (2022) Effectiveness of bivalent mRNA vaccines in preventing symptomatic SARS-CoV-2 infection—Increasing Community Access to testing program, United States, September-November 2022. MMWR Morb Mortal Wkly Rep 71: 1526-1530. http://dx.doi.org/10.15585/mmwr.mm7148e1
    [142] Zeng B, Gao L, Zhou Q, et al. (2022) Effectiveness of COVID-19 vaccines against SARS-CoV-2 variants of concern: a systematic review and meta-analysis. BMC Med 20: 200. https://doi.org/10.1186/s12916-022-02397-y
    [143] Fiolet T, Kherabi Y, MacDonald CJ, et al. (2022) Comparing COVID-19 vaccines fortheir characteristics, efficacy and effectiveness against SARS-CoV-2 and variants of concern: a narrative review. Clin Microbiol Infect 28: 202-221. https://doi.org/10.1016/j.cmi.2021.10.005
    [144] Graña C, Ghosn L, Evrenoglou T, et al. (2022) Efficacy and safety of COVID-19 vaccines. Cochrane Database Syst Rev 12: CD015477. https://doi.org/10.1002/14651858.CD015477
    [145] Scovino AM, Dahab EC, Vieira GF, et al. (2022) SARS-CoV-2's variants of concern: A brief characterization. Front Immunol 13: 834098. https://doi.org/10.3389/fimmu.2022.834098
    [146] Xu S, Sun M (2022) COVID-19 vaccine effectiveness during Omicron BA.2 pandemic in Shanghai: A cross-sectional study based on EMR. Medicine 101: e31763. https://doi.org/10.1097/MD.0000000000031763
    [147] Shkoda AS, Gushchin VA, Ogarkova DA, et al. (2022) Sputnik V effectiveness against hospitalization with COVID-19 during Omicron dominance. Vaccines 10: 938. https://doi.org/10.3390/vaccines10060938
    [148] Zheng C, Shao W, Chen X, et al. (2022) Effectiveness of COVID-19 vaccines against SARS-CoV-2 variants of concern in real-world: a literature review and meta-analysis. Int J Infect Dis 11: 2383-2392. https://doi.org/10.1016/j.ijid.2021.11.009
    [149] Cerqueira-Silva T, Andrews JR, Boaventura VS, et al. (2022) Effectiveness of CoronaVac, ChAdOx1 nCoV-19, BNT162b2, and Ad26.COV2.S among individuals with previous SARS-CoV-2 infection in Brazil: a test-negative, case-control study. Lancet Infect Dis 22: 791-801. https://doi.org/10.1016/S1473-3099(22)00140-2
    [150] Negahdaripour M, Shafiekhani M, Moezzi SMI, et al. (2021) Administration of COVID-19 vaccines in immunocompromised patients. Int Immunopharmacol 99: 108021. https://doi.org/10.1016/j.intimp.2021.108021
    [151] Zhao J, Zhao S, Ou J, et al. (2020) COVID-19: Coronavirus vaccine development updates. Front Immunol 11: 602256. https://doi.org/10.3389/fimmu.2020.602256
    [152] Chen Y, Shen H, Huang R, et al. (2021) Serum neutralising activity against SARS-CoV-2 variants elicited by CoronaVac. Lancet Infect Dis 21: 1071-1072. https://doi.org/10.1016/S1473-3099(21)00287-5
    [153] Olu-Abiodun O, Abiodun O, Okafor N, et al. (2022) COVID-19 vaccination in Nigeria: A rapid review of vaccine acceptance rate and the associated factors. PloS One 17: e0267691. https://doi.org/10.1371/journal.pone.0267691
    [154] Wu Z, Hu Y, Xu M, et al. (2021) Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine (CoronaVac) in healthy adults aged 60 years and older: a randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. Lancet Infect Dis 21: 803-812. https://doi.org/10.1016/s1473-3099(20)30987-7
    [155] Jafari A, Pouya FD, Niknam Z, et al. (2022) Current advances and challenges in COVID-19 vaccine development: from conventional vaccines to next-generation vaccine platforms. Mol Biol Rep 49: 4943-4957. https://doi.org/10.1007/s11033-022-07132-7
    [156] Trimpert J, Dietert K, Firsching TC, et al. (2021) Development of safe and highly protective live-attenuated SARS-CoV-2 vaccine candidates by genome recoding. Cell Rep 36: 109493. https://doi.org/10.1016/j.celrep.2021.109493
    [157] Jeyanathan M, Afkhami S, Smaill F, et al. (2020) Immunological considerations for COVID-19 vaccine strategies. Nat Rev Immunol 20: 615-632. https://doi.org/10.1038/s41577-020-00434-6
    [158] Li M, Wang H, Tian L, et al. (2022) COVID-19 vaccine development: milestones, lessons and prospects. Signal Transduct Target Ther 7: 146. https://doi.org/10.1038/s41392-022-00996-y
    [159] Folegatti PM, Ewer KJ, Aley PK, et al. (2020) Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet 396: 467-478. https://doi.org/10.1016/s0140-6736(20)31604-4
    [160] Zhang Z, Shen Q, Chang H, et al. (2022) Vaccines for COVID-19: A systematic review of immunogenicity, current development, and future prospects. Front Immunol 13: 843928. https://doi.org/10.3389/fimmu.2022.843928
    [161] Sah R, Shrestha S, Mehta R (2021) AZD1222 (Covishield) vaccination for COVID-19: Experiences, challenges, and solutions in Nepal. Travel Med Infect Dis 40: 101989. https://doi.org/10.1016/j.tmaid.2021.101989
    [162] Naskalska A, Dabrowska A, Nowak P, et al. (2018) Novel coronavirus-like particles targeting cells lining the respiratory tract. PloS One 13: e0203489. https://doi.org/10.1371/journal.pone.0203489
    [163] Ward BJ, Gobeil P, Séguin A, et al. (2021) Phase 1 randomized trial of a plant-derived virus-like particle vaccine for COVID-19. Nat Med 27: 1071-1078. https://doi.org/10.1038/s41591-021-01370-1
    [164] Fang E, Liu X, Li M, et al. (2022) Advances in COVID-19 mRNA vaccinedevelopment. Signal Transduct Target Ther 7: 94. https://doi.org/10.1038/s41392-022-00950-y
    [165] Collier ARY, Miller J, Hachmann NP, et al. (2023) Immunogenicity of the BA.5 bivalent mRNA vaccine boosters. New Engl J Med 388: 565-567. https://doi.org/10.1056/NEJMc2213948
    [166] Vogel AB, Kanevsky I, Che Y, et al. (2021) BNT162b vaccines protect rhesus macaques from SARS-CoV-2. Nature 592: 283-289. https://doi.org/10.1038/s41586-021-03275-y
    [167] Li M, Li Y, Li S, et al. (2022) The nano delivery systems and applications of mRNA. Eur J Med Chem 227: 113910. https://doi.org/10.1016/j.ejmech.2021.113910
    [168] Gooch KE, Smith TRF, Salguero FJ, et al. (2021) One or two dose regimen of the SARS-CoV-2 synthetic DNA vaccine INO-4800 protects against respiratory tract disease burden in nonhuman primate challenge model. Vaccine 39: 4885-4894. https://doi.org/10.1016/j.vaccine.2021.06.057
    [169] Kaur SP, Gupta V (2020) COVID-19 vaccine: a comprehensive status report. Virus Res 288: 198114. https://doi.org/10.1016/j.virusres.2020.198114
    [170] Tebas P, Yang S, Boyer JD, et al. (2021) Safety and immunogenicity of INO-4800 DNAvaccine against SARS-CoV-2: A preliminary report of an open-label, Phase 1 clinical trial. EClinical Medicine 31: 100689. https://doi.org/10.1016/j.eclinm.2020.100689
    [171] Brocato RL, Kwilas SA, Kim RK, et al. (2021) Protective efficacy of a SARS-CoV-2 DNA vaccine in wild-type and immunosuppressed Syrian hamsters. NPJ Vaccines 6: 16. https://doi.org/10.1038/s41541-020-00279-z
    [172] Mallory RM, Formica N, Pfeiffer S, et al. (2022) Safety and immunogenicity following a homologous booster dose of a SARS-CoV-2 recombinant spike protein vaccine (NVX-CoV2373): a secondary analysis of a randomised, placebo-controlled, phase 2 trial. Lancet Infect Dis 22: 1565-1576. https://doi.org/10.1016/s1473-3099(22)00420-0
    [173] Krammer F (2020) SARS-CoV-2 vaccines in development. Nature 586: 516-527. https://doi.org/10.1038/s41586-020-2798-3
    [174] Sinclair AH, Taylor MK, Weitz JS, et al. (2023) Reasons for receiving or not receiving bivalent COVID-19 booster vaccinations among adults—United States, November 1–December 10, 2022. MMWR Morb Mortal Wkly Rep 72: 73-75. https://doi.org/10.15585/mmwr.mm7203a5
  • Reader Comments
  • © 2023 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1169) PDF downloads(182) Cited by(0)

Article outline

Figures and Tables

Figures(1)  /  Tables(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog