19. References
[1] F. Song, et al., Emerging 2019 Novel Coronavirus (2019-nCoV) Pneumonia, Radiology, 295 (2020) 210-217.
[2] D. Cucinotta, M. Vanelli, WHO Declares COVID-19 a Pandemic, Acta Biomed., 91 (2020) 157-160.
[3] B. Hu, et al., Characteristics of SARS-CoV-2 and COVID-19, Nat. Rev. Microbiol., 20 (2020) 1-14.
[4] R. Singla, et al., Human animal interface of SARS-CoV-2 (COVID-19) transmission: a critical appraisal of scientific evidence, Vet. Res. Commun., 44 (2020) 119-130.
[5] M. Pal, et al., Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2): An Update, Cureus, 12 (2020) e7423-e7423.
[6] Y.A. Malik, Properties of Coronavirus and SARS-CoV-2, Malays. J. Pathol., 42 (2020) 3-11.
[7] W. Ni, et al., Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19, Crit. Care, 24 (2020) 422-422.
[8] M. Jeyanathan, et al., Immunological considerations for COVID-19 vaccine strategies, Nat. Rev. Immunol., 20 (2020) 615-632.
[9] E. Prompetchara, et al., Immune responses in COVID-19 and potential vaccines: Lessons learned from SARS and MERS epidemic, Asian Pac. J. Allergy Immunol., 38 (2020) 1-9.
[10] M. Merad, J.C. Martin, Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages, Nat. Rev. Immunol., 20 (2020) 1-8.
[11] Y. Zhou, et al., Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients, Natl. Sci. Rev., 7 (2020) 998–1002.
[12] F. Zhou, et al., Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study, Lancet, 395 (2020) 1054-1062.
[13] D. Blanco-Melo, et al., Imbalanced host response to SARS-CoV-2 drives development of COVID-19, Cell, 181 (2020) 1036-1045.e1039.
[14] A. Grifoni, et al., Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals, Cell, 181 (2020) 1489-1501. e1415.
[15] J. Braun, et al., SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19, Nature, 587 (2020) 270-274.
[16] J. Mateus, et al., Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans, Science, 370 (2020) 89-94.
[17] N. Le Bert, et al., SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls, Nature, 584 (2020) 457-462.
[18] J. Zhao, et al., Antibody responses to SARS-CoV-2 in patients of novel coronavirus disease 2019, Clin. Infect. Dis., 71 (2020) 2027-2034.
[19] A. Grifoni, et al., Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals, Cell, (2020).
[20] J. Seow, et al., Longitudinal evaluation and decline of antibody responses in SARS-CoV-2 infection, MedRxiv, 20 (2020) 48429.
[21] A.C. Walls, et al., Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein, Cell, 181 (2020) 281-292.
[22] S. Jiang, et al., Neutralizing antibodies against SARS-CoV-2 and other human coronaviruses, Trends Immunol., 41 (2020) 355-359.
[23] J. Duan, et al., A human SARS-CoV neutralizing antibody against epitope on S2 protein, Biochem. Biophys. Res. Commun., 333 (2005) 186-193.
[24] K. Nakanaga, et al., Protective effect of monoclonal antibodies on lethal mouse hepatitis virus infection in mice, J. Virol., 59 (1986) 168-171.
[25] H.-q. Yu, et al., Distinct features of SARS-CoV-2-specific IgA response in COVID-19 patients, Eur. Respir. J., 56 (2020) 2001526.
[26] A. Sariol, S. Perlman, Lessons for COVID-19 immunity from other coronavirus infections, Immunity, 53 (2020) 248-263.
[27] M.S. Khuroo, et al., COVID-19 Vaccines: A Race Against Time in the Middle of Death and Devastation!, J. Clin. Exp. Hepatol., 10 (2020) 610-621.
[28] I.N. Abdullahi, et al., Implications of SARS-CoV-2 genetic diversity and mutations on pathogenicity of the COVID-19 and biomedical interventions, J. Taibah Univ. Med. Sci., 15 (2020) 258-264.
[29] L. van Dorp, et al., Emergence of genomic diversity and recurrent mutations in SARS-CoV-2, Infect. Genet. Evol., 83 (2020) 104351.
[30] I. Thevarajan, et al., Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19, Nat. Med., 26 (2020) 453-455.
[31] Y. Chen, L. Li, SARS-CoV-2: virus dynamics and host response, Lancet Infect. Dis., 20 (2020) 515-516.
[32] R.L. Tillett, et al., Genomic evidence for reinfection with SARS-CoV-2: a case study, Lancet Infect. Dis., 21 (2021) 52-58.
[33] C.Y. Yong, et al., Recent advances in the vaccine development against Middle East respiratory syndrome-coronavirus, Front. Microbiol., 10 (2019) 1781.
[34] R.L. Roper, K.E. Rehm, SARS vaccines: where are we?, Expert Rev. Vaccines, 8 (2009) 887-898.
[35] L. Peeples, News Feature: Avoiding pitfalls in the pursuit of a COVID-19 vaccine, PNAS, 117 (2020) 8218-8221.
[36] U.J. Buchholz, et al., Contributions of the structural proteins of severe acute respiratory syndrome coronavirus to protective immunity, PNAS, 101 (2004) 9804-9809.
[37] S.F. Ahmed, et al., Preliminary identification of potential vaccine targets for the COVID-19 coronavirus (SARS-CoV-2) based on SARS-CoV immunological studies, Viruses, 12 (2020) 254.
[38] Q. Gao, et al., Development of an inactivated vaccine candidate for SARS-CoV-2, Science, 369 (2020) 77.
[39] R. Sealy, et al., Preclinical and clinical development of a multi-envelope, DNA-virus-protein (DVP) HIV-1 vaccine, Int. Rev. Immunol., 28 (2009) 49-68.
[40] M. Enayatkhani, et al., Reverse vaccinology approach to design a novel multi-epitope vaccine candidate against COVID-19: an in silico study, J. Biomol. Struct. Dyn., (2020) 1-16.
[41] S. Rauch, et al., New vaccine technologies to combat outbreak situations, Front. Immunol., 9 (2018) 1963.
[42] T. Gupta, S.K. Gupta, Potential adjuvants for the development of a SARS-CoV-2 vaccine based on experimental results from similar coronaviruses, Int. Immunopharmacol., 86 (2020) 106717.
[43] Q. Gao, et al., Development of an inactivated vaccine candidate for SARS-CoV-2, Science (New York, N.Y.), 369 (2020) 77-81.
[44] M. Jeyanathan, et al., New tuberculosis vaccine strategies: taking aim at un-natural immunity, Trends Immunol., 39 (2018) 419-433.
[45] L. Moreno-Fierros, et al., Development of SARS-CoV-2 vaccines: should we focus on mucosal immunity?, Taylor & Francis, 2020.
[46] I.M. Belyakov, J.D. Ahlers, What role does the route of immunization play in the generation of protective immunity against mucosal pathogens?, J. Immunol., 183 (2009) 6883-6892.
[47] J. Zhao, et al., Airway memory CD4+ T cells mediate protective immunity against emerging respiratory coronaviruses, Immunity, 44 (2016) 1379-1391.
[48] Z. Xing, et al., Innate immune memory of tissue‐resident macrophages and trained innate immunity: Re‐vamping vaccine concept and strategies, J. Leukoc. Biol., 108 (2020) 825-834.
[49] S. Afkhami, et al., Methods and clinical development of adenovirus-vectored vaccines against mucosal pathogens, Mol. Ther. Methods Clin. Dev., 3 (2016) 16030.
[50] W.H. Organization, Guidelines on clinical evaluation of vaccines: regulatory expectations, World Health Organization WHO Expert Committee on Biological Standardization. Fifty-second report. Geneva, World Health Organization, (2004) 035-101.
[51] M.S. Khuroo, et al., COVID-19 vaccines: A race against time in the middle of death and devastation!, J. Clin. Exp. Hepatol., (2020).
[52] W.H. Organization, Draft landscape and tracker of COVID-19 candidate vaccines, World Health Organization, 2020.
[53] C.J. Zimmer Carl, Wee Sui-Lee, Coronavirus Vaccine Tracker, The New York Times, 2020
[54] H. Wang, et al., Development of an inactivated vaccine candidate, BBIBP-CorV, with potent protection against SARS-CoV-2, Cell, 182 (2020) 713-721. e719.
[55] H.X. Lim, et al., Development of multi-epitope peptide-based vaccines against SARS-CoV-2, Biomed. J., (2020).
[56] S. Xia, et al., Effect of an Inactivated Vaccine Against SARS-CoV-2 on Safety and Immunogenicity Outcomes: Interim Analysis of 2 Randomized Clinical Trials, JAMA, 324 (2020) 951-960.
[57] W.-j. Guan, et al., Clinical characteristics of coronavirus disease 2019 in China, N. Engl. J. Med., 382 (2020) 1708-1720.
[58] S. Xia, et al., Safety and immunogenicity of an inactivated SARS-CoV-2 vaccine, BBIBP-CorV: a randomised, double-blind, placebo-controlled, phase 1/2 trial, Lancet Infect. Dis., 21 (2021) 39-51.
[59] N. van Doremalen, et al., ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques, Nature, 586 (2020) 578-582.
[60] P.M. Folegatti, et al., 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 (2020) 467-478.
[61] K. Rawat, et al., COVID-19 vaccine: A recent update in pipeline vaccines, their design and development strategies, Eur. J. Pharmacol., 892 (2021) 173751.
[62] D.Y. Logunov, et al., Safety and efficacy of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine: an interim analysis of a randomised controlled phase 3 trial in Russia, Lancet, 6736 (2021) 234-238.
[63] D.Y. Logunov, et al., Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia, Lancet, 396 (2020) 887-897.
[64] J. Sadoff, et al., Interim Results of a Phase 1–2a Trial of Ad26. COV2. S Covid-19 Vaccine, N. Engl. J. Med., 20 (2021) 201.
[65] L. Dai, G.F. Gao, Viral targets for vaccines against COVID-19, Nat. Rev. Immunol., 21 (2020) 73-82.
[66] N.i.o. health, Phase 3 trial of Novavax investigational COVID-19 vaccine opens, 2020.
[67] L. Mukhopadhyay, et al., Comparison of the immunogenicity & protective efficacy of various SARS-CoV-2 vaccine candidates in non-human primates, Indian J. Med. Res., (2020).
[68] L.A. Jackson, et al., An mRNA vaccine against SARS-CoV-2—preliminary report, N. Engl. J. Med., 383 (2020) 1920-1931.
[69] K.S. Corbett, et al., SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness, Nature, 586 (2020) 567-571.
[70] F.P. Polack, et al., Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine, N. Engl. J. Med., 383 (2020) 2603-2615.
[71] A. Cagigi, K. Loré, Immune Responses Induced by mRNA Vaccination in Mice, Monkeys and Humans, Vaccines, 9 (2021) 61.
[72] S. Rauch, et al., mRNA vaccine CVnCoV protects non-human primates from SARS-CoV-2 challenge infection, bioRxiv, 12 (2020) 424138.
[73] Baharatbiotech, COVAXIN™ (BBV152) Shows Demonstration of Protective Efficacy and Immunogenicity in Non-Human Primates, Baharatbiotech, 2020.
[74] J. Machhi, et al., Nanocarrier Vaccines for SARS-CoV-2, Adv. Drug Deliv. Rev., (2021) S0169-0409X(0121)00002-00008.
[75] P. Yadav, et al., Assessment of immunogenicity and protective efficacy of ZyCoV-D DNA vaccine candidates in Rhesus macaques against SARS-CoV-2 infection, bioRxiv, 2 (2021) 429480.
[76] B. Wire, COVAXX’s COVID-19 Vaccine, UB-612, Induced Neutralizing Antibodies in 100% of Participants During Phase 1 Clinical Trial, Business Wire, 2021.
[77] C.-E. Lien, et al., CpG-adjuvanted stable prefusion SARS-CoV-2 spike protein protected hamsters from SARS-CoV-2 challenge, BioRxiv, 1 (2021) 425674.
[78] P. Richmond, et al., Safety and immunogenicity of S-Trimer (SCB-2019), a protein subunit vaccine candidate for COVID-19 in healthy adults: a phase 1, randomised, double-blind, placebo-controlled trial, Lancet, 6736 (2021) 241-245.
[79] Precisionvaccinations, COVID-19 S-Trimer (SCB-2019) Vaccine, Precisionvaccinations, 2021.
[80] P. Tebas, et al., Safety and immunogenicity of INO-4800 DNA vaccine against SARS-CoV-2: A preliminary report of an open-label, Phase 1 clinical trial, EClinicalMedicine, 31 (2021) 100689.
[81] T.R. Smith, et al., Immunogenicity of a DNA vaccine candidate for COVID-19, Nat. Commun., 11 (2020) 1-13.
[82] S. Plotkin, History of vaccination, PNAS, 111 (2014) 12283-12287.
[83] F. Almazán, et al., Engineering a replication-competent, propagation-defective Middle East respiratory syndrome coronavirus as a vaccine candidate, MBio, 4 (2013).
[84] J. Netland, et al., Immunization with an attenuated severe acute respiratory syndrome coronavirus deleted in E protein protects against lethal respiratory disease, Virology, 399 (2010) 120-128.
[85] Y. Hou, et al., Deletion of both the tyrosine-based endocytosis signal and the endoplasmic reticulum retrieval signal in the cytoplasmic tail of spike protein attenuates porcine epidemic diarrhea virus in pigs, J. Virol., 93 (2019) e01758-01718.
[86] J.M. Jimenez-Guardeño, et al., Identification of the mechanisms causing reversion to virulence in an attenuated SARS-CoV for the design of a genetically stable vaccine, Plos Pathog., 11 (2015) e1005215.
[87] B.Y. Cheng, et al., Development of live-attenuated arenavirus vaccines based on codon deoptimization, J. Virol., 89 (2015) 3523-3533.
[88] S. Mueller, et al., A codon-pair deoptimized live-attenuated vaccine against respiratory syncytial virus is immunogenic and efficacious in non-human primates, Vaccine, 38 (2020) 2943-2948.
[89] Y. Tao, et al., Surveillance of Bat Coronaviruses in Kenya Identifies Relatives of Human Coronaviruses NL63 and 229E and Their Recombination History, J. Virol., 91 (2017) e01953-01916.
[90] W.H. Organization, Draft landscape of COVID-19 candidate vaccines., WHO.
[91] A.D. Murdin, et al., Inactivated poliovirus vaccine: past and present experience, Vaccine, 14 (1996) 735-746.
[92] J.M. Wood, J.S. Robertson, From lethal virus to life-saving vaccine: developing inactivated vaccines for pandemic influenza, Nat. Rev. Microbiol., 2 (2004) 842-847.
[93] M.T. Ul Qamar, et al., Epitope‐based peptide vaccine design and target site depiction against Middle East Respiratory Syndrome Coronavirus: an immune-informatics study, J. Transl. Med., 17 (2019) 362.
[94] Y. Watanabe, et al., Site-specific glycan analysis of the SARS-CoV-2 spike, Science, 369 (2020) 330-333.
[95] Q. Gao, et al., Development of an inactivated vaccine candidate for SARS-CoV-2, Science, 369 (2020) 77-81.
[96] M. Bolles, et al., A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge, J. Virol., 85 (2011) 12201-12215.
[97] N. Iwata-Yoshikawa, et al., Effects of Toll-like receptor stimulation on eosinophilic infiltration in lungs of BALB/c mice immunized with UV-inactivated severe acute respiratory syndrome-related coronavirus vaccine, J. Virol., 88 (2014) 8597-8614.
[98] G. Del Giudice, et al., Correlates of adjuvanticity: a review on adjuvants in licensed vaccines, Seminars in immunology, Elsevier, 2018, pp. 14-21.
[99] B. Ganneru, et al., Evaluation of Safety and Immunogenicity of an Adjuvanted, TH-1 Skewed, Whole Virion InactivatedSARS-CoV-2 Vaccine - BBV152, BioRxiv, 9 (2020) 285445.
[100] K.J. Ewer, et al., Viral vectors as vaccine platforms: from immunogenicity to impact, Curr. Opin. Immunol., 41 (2016) 47-54.
[101] F.-C. Zhu, et al., Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial, Lancet, 396 (2020) 479-488.
[102] S.P. Graham, et al., Evaluation of the immunogenicity of prime-boost vaccination with the replication-deficient viral vectored COVID-19 vaccine candidate ChAdOx1 nCoV-19, NPJ vaccines, 5 (2020) 1-6.
[103] F. Krammer, SARS-CoV-2 vaccines in development, Nature, 586 (2020) 516-527.
[104] I.R. Humphreys, S. Sebastian, Novel viral vectors in infectious diseases, Immunology, 153 (2018) 1-9.
[105] S.J. Draper, J.L. Heeney, Viruses as vaccine vectors for infectious diseases and cancer, Nat. Rev. Microbiol., 8 (2010) 62-73.
[106] J. Cohen, Top US scientists left out of White House selection of COVID-19 vaccine short list, Science, (2020).
[107] H. Mou, et al., The receptor binding domain of the new Middle East respiratory syndrome coronavirus maps to a 231-residue region in the spike protein that efficiently elicits neutralizing antibodies, J. Virol., 87 (2013) 9379-9383.
[108] Y. Guo, et al., Elicitation of immunity in mice after immunization with the S2 subunit of the severe acute respiratory syndrome coronavirus, DNA Cell Biol., 24 (2005) 510-515.
[109] J. Oscherwitz, The promise and challenge of epitope-focused vaccines, Hum. Vaccines Immunother., 12 (2016) 2113-2116.
[110] L. Du, et al., Recombinant receptor-binding domain of SARS-CoV spike protein expressed in mammalian, insect and E. coli cells elicits potent neutralizing antibody and protective immunity, Virology, 393 (2009) 144-150.
[111] M.S. Diamond, T.C. Pierson, The challenges of vaccine development against a new virus during a pandemic, Cell Host Microbe, 27 (2020) 699-703.
[112] W. Ren, et al., Recombinant SARS-CoV-2 spike S1-Fc fusion protein induced high levels of neutralizing responses in nonhuman primates, Vaccine, 38 (2020) 5653-5658.
[113] B. Donaldson, et al., Virus-like particle vaccines: immunology and formulation for clinical translation, Expert Rev. Vaccines, 17 (2018) 833-849.
[114] X. Lu, et al., Immune responses against severe acute respiratory syndrome coronavirus induced by virus‐like particles in mice, Immunology, 122 (2007) 496-502.
[115] A. Naskalska, et al., Novel coronavirus-like particles targeting cells lining the respiratory tract, Plos One, 13 (2018) e0203489.
[116] B. Donaldson, et al., Virus-like particle vaccines: immunology and formulation for clinical translation, Expert Rev. Vaccines, 17 (2018) 833-849.
[117] DrugBank, Coronavirus-Like Particle COVID-19 Vaccine, 2020.
[118] B. Wire, Medicago announces positive results in animal trials for its vaccine candidate against COVID-19. , STT, 2020.
[119] N.A. Jackson, et al., The promise of mRNA vaccines: A biotech and industrial perspective, NPJ vaccines, 5 (2020) 1-6.
[120] B. Ferraro, et al., Clinical applications of DNA vaccines: current progress, Clin. Infect. Dis., 53 (2011) 296-302.
[121] C. Liu, et al., Research and development on therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases, ACS Publications, 2020.
[122] K.J. Hassett, et al., Optimization of lipid nanoparticles for intramuscular administration of mRNA vaccines, Mol. Ther. Nucleic Acids, 15 (2019) 1-11.
[123] N. Pardi, et al., mRNA vaccines—a new era in vaccinology, Nat. Rev. Drug Discov., 17 (2018) 261.
[124] D.H. Fuller, P. Berglund, Amplifying RNA vaccine development, N. Engl. J. Med., 382 (2020) 2469-2471.
[125] S. Rauch, et al., mRNA based SARS-CoV-2 vaccine candidate CVnCoV induces high levels of virus neutralizing antibodies and mediates protection in rodents, BioRxiv, 10 (2020) 351775.
[126] R.M. de Alwis, et al., A Single Dose of Self-Transcribing and Replicating RNA Based SARS-CoV-2 Vaccine Produces Protective Adaptive Immunity In Mice, BioRxiv, (2020).
[127] P.F. McKay, et al., Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice, Nat. Commun., 11 (2020) 1-7.
[128] S. Andrew, Imperial social enterprise to accelerate low-cost COVID-19 vaccine, 2020.
[129] A. Haque, A.B. Pant, Efforts at COVID-19 Vaccine Development: Challenges and Successes, Vacc. nes., 8 739.