key: cord-0690543-v0bumalz authors: Yang, Haiqiang; Cao, Jessica; Lin, Xiaoyang; Yue, Jingwen; Zieneldien, Tarek; Kim, Janice; Wang, Lianchun; Fang, Jianmin; Huang, Ruo-Pan; Bai, Yun; Sneed, Kevin; Cao, Chuanhai title: Developing an Effective Peptide-Based Vaccine for COVID-19: Preliminary Studies in Mice Models date: 2022-02-22 journal: Viruses DOI: 10.3390/v14030449 sha: ea74061ba4bbbde79de19036f329fa800815dc1d doc_id: 690543 cord_uid: v0bumalz Coronavirus disease 2019 (COVID-19) has caused massive health and economic disasters worldwide. Although several vaccines have effectively slowed the spread of the virus, their long-term protection and effectiveness against viral variants are still uncertain. To address these potential shortcomings, this study proposes a peptide-based vaccine to prevent COVID-19. A total of 15 B cell epitopes of the wild-type severe acute respiratory coronavirus 2 (SARS-CoV-2) spike (S) protein were selected, and their HLA affinities predicted in silico. Peptides were divided into two groups and tested in C57BL/6 mice with either QS21 or Al(OH)(3) as the adjuvant. Our results demonstrated that the peptide-based vaccine stimulated high and durable antibody responses in mice, with the T and B cell responses differing based on the type of adjuvant employed. Using epitope mapping, we showed that our peptide-based vaccine produced antibody patterns similar to those in COVID-19 convalescent individuals. Moreover, plasma from vaccinated mice and recovered COVID-19 humans had the same neutralizing activity when tested with a pseudo particle assay. Our data indicate that this adjuvant peptide-based vaccine can generate sustainable and effective B and T cell responses. Thus, we believe that our peptide-based vaccine can be a safe and effective vaccine against COVID-19, particularly because of the flexibility of including new peptides to prevent emerging SARS-CoV-2 variants and avoiding unwanted autoimmune responses. Coronavirus disease 2019 is caused by the severe acute respiratory coronavirus 2 (SARS-CoV-2), which is a single-strand positive RNA virus that enables the host cell to directly synthesize proteins upon viral entry and uncoating [1] . This enables viral replication and the rate of viral spread to occur much more rapidly than other types of viruses [2] . Given this alarming rate of transmission, many countries were unable to implement an effective and timely approach to curb the spread of the virus, and the worldwide economic impacts of COVID- 19 have been historically devastating [3] . Other viral outbreaks, such as SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV), have led to the development of several treatment strategies [4] [5] [6] . Drugs While DNA vaccines can elicit a good T cell response, the B cell response is relatively low compared to protein-based vaccines [53] [54] [55] [56] . Alternatively, mRNA-based vaccines are a novel approach for infectious diseases and are currently the dominant approach in fighting COVID-19 [57] . However, it remains unclear as to how effective they are in stimulating T cell and B cell responses in comparison to a more traditional protein-based vaccine or to natural infection. Here, we developed a peptide-based vaccine for COVID-19 prevention. There are four primary advantages of peptide-based vaccines: (1) large-scale peptide synthesis is relatively inexpensive and the technology is well-established, (2) peptides can fold into three-dimensional epitopes capable of inducing antibody responses to linear and conformational structures, (3) unique epitopes can be selected to avoid autoimmune responses that could be generated by the whole protein, and (4) new epitopes can be easily added to the peptide mixture as new viral mutations are identified [58] [59] [60] . The SARS-CoV-2 spike (S) protein of the wild-type "Wuhan" strain (Accession: YP_009724390.1) was submitted to DNAStar (Madison, WI, USA) for in silico antigen (peptide) identification. Nine epitopes were selected from the S protein's subunit 1 (S1) domain, and six sequences were selected from the subunit 2 (S2) domain. The selected peptide sequences were submitted to GenBank for homology analysis, which confirmed that the peptides were unique. Peptide antigenicity was confirmed by DNAStar. Human Leukocyte Antigen-I (HLA-I), Mouse Major Histocompatibility Complex-I (MHC-I), and HLA-II affinity analyses were conducted with the T Cell Epitope Prediction Tools, using the Immune Epitope Database (IEBD) Analysis Resource at http://tools.iedb. org/main/tcell/, accessed on 21 July 2021. The specificity of the peptides to SARS-CoV-2 were confirmed using the protein BLAST module at https://blast.ncbi.nlm.nih.gov/Blast. cgi, accessed on 21 July 2021. All peptides were provided by Biomer Technology Inc. (Pleasanton, CA, USA). The SARS-CoV-2 pseudovirus system was provided by Dr. Lianchun Wang's lab at USF. A SARS-CoV-2 spike open reading frame (ORF) mammalian expression plasmid was obtained from Addgene (Plasmid #145032; Watertown, MA, USA). NL4-3 mCherry Luciferase, a replication-defective HIV dual reporter vector expressing mCherry and firefly luciferase, was obtained from Addgene (Plasmid #44965). Lipofectamine™ 3000 Transfection Reagent was purchased from Invitrogen (Waltham, MA, USA). The human 293T embryonic kidney cell line was obtained from the American-Type Culture Collection (ATCC) (Manassas, VA, USA). The 293T-ACE2-GFP cell line was established by transfecting 293T cells with the ACE2-GFP expression vector (OriGene; Rockville, MD, USA). Both cell lines were maintained in Dulbecco's modified Eagle's medium (DMEM) with 2 mM L-glutamine, 10% FBS, 100 µg/mL streptomycin, and 100 IU/mL penicillin. Lentiviral vector particles concentration reagent Lenti-X™ Concentrator was purchased from TaKaRa (Mountain View, CA, USA). The Luciferase Assay System was purchased from Promega (Madison, WI, USA). For SARS-CoV-2 pseudovirus production, 4 × 10 6 293T cells were seeded in a 100 mm cell culture dish. After 48 h, 10 µg NL4-3 mCherry Luciferase plasmid and 10 µg SARS-CoV-2 spike plasmid were mixed in 1.5 mL Opti-MEM medium and incubated at RT for 5 min. Then, 65 µL of Lipo3000 was mixed in 1.5 mL Opti-MEM medium at RT for 5 min, and the plasmid mixture was then mixed with the Lipo3000 mixture and incubated at RT for 20 min. The mixture was added dropwise into a dish and incubated in the cell culture incubator. After 48 h, cells were centrifuged at 500× g for 10 min, cell supernatant was collected, and 9 mL of supernatant was transferred into a separate tube with a 3 mL Lenti-X concentrator. The supernatant was stored at 4 • C for 4 h and centrifuged at 1500× g for 45 min. The supernatant was aspirated, and the precipitate was resuspended with 90 µL cold DPBS and then frozen at −80 • C. C57BL/6 mice were used for this study. The mice were 6 months old, gender balanced in each group, and housed at the animal facility at the University of South Florida (USF) Health Center. All animal experiments were approved (IS00008370) by the Institutional Animal Care and Use Committee (IACUC) at USF and performed according to the National Institute of Health (NIH) guidelines. The peptides were split into two groups by S protein subdomain; each group contained peptides with high affinity to MHC (HLA) class I and class II to ensure that the peptide mixture would cover most MHC typing. HLA-I was predicted against all available allele types, and the top 1% was considered high affinity. HLA-II was also tested against all allele types in the database, and the selected standard was set at 10% rank as recommended. A total of 15 peptides were identified against the S protein. Each peptide was dissolved with DMSO at 50 mg/mL, grouped into two groups, and mixed at 50 µg each. The two most commonly utilized adjuvants-QS21 and Al(OH) 3 (0.4% Alhydrogel from InvivoGen (Cat no# vac-alu-50, San Diego, CA, USA)-were selected for the vaccine formula. QS21 was purchased from Desert King International (San Diego, CA, USA), and Al(OH) 3 was purchased from Invitrogen (Waltham, MA, USA). Common adjuvants include Al(OH) 3 , which can promote earlier antibody production, and QS21, which promotes the release of Th1 cytokines participating in the elimination of intracellular pathogens [61] [62] [63] . The concentration of adjuvant is 25 µg per dose. Two total studies were conducted in this report, with peptide mixture groups 1 and 2 using different groups of mice. Group 1 mice (C57BL/6) were 6 months old and injected with A1-G1 (8 peptides); group 2 mice were 8 months old and received H1-E2 (7 peptides) at equal mixture amounts (50 µg/peptide) with either QS21 or Al(OH) 3 as the adjuvant via bi-weekly subcutaneous injections for 6 injections. Blood samples were obtained with EDTA vacutainer tubes 7 days after each injection, starting from the 2nd injection. Additionally, 50 µL of whole blood was taken for flow cytometry analysis and the rest of the blood was centrifuged at 300× g for 5 min. Plasma was then transferred into a new tube and stored at −80 • C for future application. The immunization and blood drawing schedule was conducted as shown in Scheme 1. The SARS-CoV-2 S protein's full length S1 (Cat#230-30161) and receptor binding domain (RBD) (Cat#230-30162) recombinant proteins expressed in human HEK293 cells were purchased from RayBiotech Life, Inc. (Peachtree Corners, GA, USA). The SARS-CoV-2 S protein's S1 (Cat#230-01101) and S2 (Cat#230-01103) subunit recombinant proteins expressed in E. coli were also purchased from RayBiotech Life, Inc. Immulon 4 HBX 96-well plates (Cat# 3855) were purchased from Thermo Fisher Scientific (Waltham, MA, USA). HRP conjugated anti-Mouse IgG antibodies were ordered from SouthernBiotech (Birmingham, AL, USA; cat no. 1030-05), and HRP-conjugated anti-Human IgG antibodies were ordered from SouthernBiotech (Cat: 2015-05). Human plasma was purchased from RayBiotech Life, Inc. An indirect ELISA was conducted to measure antibody response. First, 96-well plates were coated with 50 µL of peptides from 10 µg/mL S1 and S2 and 1 µg/mL recombinant protein. The plates were washed 4 times with PBST, 180 µL/well of 0.05% BSA buffer was added, and the plates were incubated at 37 • C for 1 h. The plates were then washed with 1× wash buffer, and diluted plasma was added to each well at 1:200, 1:800, and 1:3200-fold dilutions and incubated for 1 h at room temperature (RT). Anti-human or anti-mouse HRP-conjugated antibodies were then added at 1:10,000 dilutions, incubated for 1 h at RT, and washed with 1× wash buffer. Then, 100 µL of TMB substrate was added, followed by 0.4 M sulfuric acid to stop the reaction. Plates were read at OD450, with OD650 as the reference. For epitope mapping of mouse and human plasma, plasma was added to the antigencoated 96-well ELISA plate for indirect ELISA as described above at 1:200, 1:800, and 1:3200-fold dilutions and incubated for 1 h at RT. T cell population detection post vaccination was assessed using flow cytometry assay and 50 µL whole EDTA blood. Red blood cells were lysed with ACK buffer and washed twice with 1% (v/v) FBS in 1× PBS. The fluorophore-conjugated antibody mixture was added to the sample and incubated on ice for 20 min. FITC anti-mouse CD3ε Antibody (Cat#10036), APC/Cyanine7 anti-mouse CD8a Antibody (Cat#100714), APC anti-mouse CD4 Antibody (Cat#100412), and PE anti-mouse CD19 Antibody (Cat#115508) were bought from Biolegend (San Diego, CA, USA). Cells were then resuspended with 1% FBS in 1× PBS after washing 3 times. All samples were run on the flow cytometer. ACE2-293T-GFP cells were seeded into a poly-lysine-coated 96-well plate at a density of 1.2 × 10 4 cells/well. After 24 h, 6 µL of SARS-CoV-2 pseudovirus and diluted human plasma or mouse samples were added at the designated dilutions, and the medium was changed after 6 h of incubation. The plates were incubated in a CO 2 incubator for 48 h, the medium was removed, and the cells were washed with Hank's balanced salt solution (HBSS). HBSS was aspirated and 20 µL of Promega lysis buffer was added at RT for 5 min. Luminescence was measured after adding 50 µL of Luciferase substrate into 20 µL of lysis buffer. The mouse immunoglobulin isotyping (Cat#MGAMMAG-300K) and cytokine magnetic bead panel assay (Cat#MCYTOMAG-70K) were purchased from MilliporeSigma (Burlington, MA, USA). First, 100 µL of assay buffer was added to each well of the assay plate and mixed on a plate shaker for 10 min at RT. The assay buffer was then discarded, and the remaining residue was removed from the wells by inverting the plate and tapping it numerous times on an absorbent towel. Then, 50 µL of the assay buffer was added to the background wells, and 50 µL of the standard, control, and diluted samples were added to the appropriate wells. Afterwards, the MILLIPLEX MAP Anti-Mouse Multi-Immunoglobulin beads were vortexed at medium speed for 15 s and sonicated for 15 s using a sonication bath. Then, 25 µL of the bead solution was added into each well, and the plate was covered and incubated on a plate shaker for 15 min at RT. The plate was then washed twice with 200 µL/well of washing buffer. After washing, 25 µL/well of prepared Anti-Mouse κ Light Chain PE was added, covered, and incubated on a plate shaker at RT for 15 min. Finally, the fluid was aspirated, and beads were resuspended in 150 µL/well of sheath fluid. The plate was then read on the Bio-Rad Bio-Plex MAGPIX Multiplex Reader. First, 200 µL of washing buffer was added to each well, and the plate was sealed and incubated on the plate shaker for 10 min at RT. The wash buffer was then aspirated, and 25 µL of each standard or control was introduced into the appropriate wells, with the assay buffer utilized for the 0 pg/mL standard (background). Then, 25 µL of the assay buffer was added to the sample wells, and 25 µL of the serum matrix solution was added to the background, standard, and control wells. Afterwards, 25 µL of the diluted samples were introduced into the appropriate wells. Then, the mixing bottle was vortexed, and 25 µL of the mixed or premixed beads were added to each well and shaken for 10 min at RT. The plate was then sealed with a plate sealer, wrapped with aluminum foil, and incubated on a plate shaker overnight at 2-8 • C. The following day, well contents were gently removed, and the plate was washed twice. Then, 25 µL of the detection antibodies were added into each well and incubated on a plate shaker for 1 h at RT. The well contents were removed, the plates were washed twice, and 150 µL of sheath fluid was added to all wells. The beads were then resuspended on a plate shaker for 5 min, and the plate was read on the Bio-Rad Bio-Plex MAGPIX Multiplex Reader. Sixteen peptides were predicted to have high antigenicity with 100% homology to the SARS-CoV-2 virus were split across two peptide mixtures (Group 1, Group 2) that covered all Class I and II allele types of C57B/6 mice. The peptide sequences and T cell epitope results are listed in Table 1 and Supplementary Table S1 , respectively. First, 50 ug/peptide of peptides A-G of the S1 protein were mixed with either the QS21 or Al(OH) 3 adjuvant and injected into mice subcutaneously at two-week intervals. Blood was collected 10 days after each injection and used for antibody detection. It is evident that, after two injections, both groups could detect an antibody response, but the Al(OH) 3 group exhibited a higher antibody response than the QS21 group. With continuous boosting, antibody response to each peptide differed between the two adjuvants ( Figure 1 ). After the Group 1 peptide test, another group of peptides were injected into mice, with either QS21 or Al(OH) 3 adjuvants. There were a total seven peptides in this group-two in the S1 domain and five in the S2 domain. The ELISA results showed that antibodies were produced against five peptides in both QS21 and Al(OH) 3 groups but were relatively lower in the QS21 group than the Al(OH) 3 adjuvant group; Al(OH) 3 -2D produce higher antibodies than the QS21 adjuvant group (p < 0.001). No antibodies were induced against peptide 2A and 1J after two injections ( Figure 2 ). Figure 2 . Antibody response to peptide mixture after 2 vaccines. In total, 7 peptides were used in this injection, with either QS21 or Al(OH) 3 as adjuvants. Peptides S1I and S1J are selected from S1 domain. Peptide S2A, S2B, S2D, S2E and S2F are selected from S2 domain. (N = 3, p < 0.001). Four mice plasma (two from each peptide mixture group) and four human plasma samples (two recovered patient blood and two from our tissue bank collected in 2008 as negative controls) were selected for epitope mapping. A total of 16 peptides and recombinant S1 and S2 proteins were predicted in this assay. JK2 and JK3 (recovered patients) display a pattern similar to our peptide vaccine, indicating that the vaccine has included all major epitopes (binding to C, D, H1, I1) ( Figure 3) . Coating peptides A-J are peptides from the S1 protein, A2-F2 are peptides from the S2 protein. S1 is the recombinant S1 of the S protein and S2 is the recombinant S2 of the S protein. Ctr is a control peptide of human alpha-synuclein protein, which served as a negative control. Plasma was diluted at a 1:800 dilution with the sample, and ELISA results were shown in the graphs. When examining the duration of the antibody response post-vaccination, Al(OH) 3 appears to generate a longer antibody response than the QS21 adjuvant. Nonetheless, QS21 induced a higher antibody response to some antigens, such as the ones seen in peptide S1-B (Figure 4 ). . Antibody duration between two adjuvants. The graphs above are the results of antibody testing via ELISA on blood from two time points (1 and 2): 1 is 10 days after the 6th vaccination and 2 is three months after 1. Al(OH) 3 can generate a longer antibody response than QS21 (B,C). However, QS21 can induce a higher antibody response to some antigens, such as that seen in S1-B (A). Mice blood samples were collected after the third vaccination of peptide mixture group 1 with different adjuvants to compare the B and T cell population difference. Flow cytometry assay was used to analyze the T and B cell population for each vaccine adjuvant. CD3+ and CD8+ T cells ( Figure 5A ,C) were significantly higher in Al(OH) 3 groups than QS21 groups (p < 0.05; N = 3 per group). CD4+ T cells ( Figure 5B ) were also significantly higher in the Al(OH) 3 adjuvant groups compared to both the QS21 and control groups (p < 0.05; n = 3 per group). Interestingly, the B cell population in the Al(OH) 3 adjuvant group was significantly decreased when compared to the QS21 adjuvant group (N = 3 per group, α = 0.05). These results imply that the Al(OH) 3 adjuvant is ideal for obtaining the desired cellular response ( Figure 5 ). The pseudovirus system in vitro was compared with recovered patient plasma to determine the neutralization potential of post-vaccinated mice plasma. The results showed that mice plasma had better neutralization function than human plasma in this system ( Figure 6 ). There are no differences in IgG levels among all three groups. IgM levels are higher in the vaccine group than control mice, indicating that the adjuvant-based peptide vaccine could not change the immunotype of the mice (Figure 7 ). Vaccines are undeniably the most effective solution to stopping the spread of COVID-19, and several vaccines have already been distributed to contribute to this effort. Pfizer and Moderna have developed nanolipid-based mRNA particle vaccines, while Astrazeneca and Johnson & Johnson have produced adenoviral vector-based DNA vaccines [26, 64] . While these have become the most widely used vaccines to prevent COVID-19, other types of vaccines have also been developed, such as an inactivated whole viral particle vaccine (Kexing) and recombinant subunit vaccine (Novavax) [65] [66] [67] . Although the formulation of each vaccines varies, they all aim to block the binding of the S protein to ACE2 and inhibit viral entry into the host cell [1] . Virologists have proved that the COVID-19 virus surface has spike (S) proteins that can specifically bind to ACE2 through a receptor binding domain (RBD) [39] . The RBD on the S protein of COVID-19 serves as the ligand for ACE2 on the cell surface. It is important to note that most human organs produce the ACE2 protein [40] , which may have contributed to the many deaths caused by COVID-19 that were also attributed to multiple organ failure [68] . The COVID-19 vaccines that are currently approved have been reported to have high rates of effectiveness, ranging from 66.3% to 95%, and both B and T cell responses have been observed [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] . Moreover, no large-scale and severe adverse effects have been reported. However, long term monitoring is still necessary before an affirmative assessment can be made regarding vaccine safety. The human immune system works diligently to protect the body from viruses [79] . During viral infection, immune cells produce large amounts of cytokines, which can result in cytokine storm syndrome [80] . Normally, a virus has a specific tissue receptor, meaning the virus can only infect certain tissues, while causing no harm to other tissues [81] . For instance, HIV targets the CD4 receptor, only infecting and killing CD4 T cells [82, 83] . However, SARS-CoV-2 is markedly different from other viruses because it utilizes ACE2 receptors, and ACE2 is widely expressed by many types of cells across different organs [40] . ACE2 bound by the SARS-CoV-2 S protein can be considered a viral particle and will trigger an immune response [84] . This scenario complicates vaccine development because the abundance of ACE2 increases the likelihood of potential adverse responses caused by the vaccine and must therefore be carefully considered. Most of the current vaccines use the S protein as the antigen, and target for blocking viral entry by sealing RBD on S1 of COVID-19, which induces our immune system to attack cells that bind to the S protein [21] . To induce an immune response, antigen-presenting cells (APCs) must uptake the S protein, process it, and then present it to CD4 or CD8 T cells [85] . Although this process is highly effective with other vaccines, it can be problematic for S gene-or S protein-based vaccines, since the S protein will bind to the ACE2 of the host cell. To combat the potential issues of current COVID-19 vaccines, our study uses peptides as opposed to whole S protein to develop a vaccine for COVID-19. We selected 15 peptides for the whole S protein utilizing B and T cell epitope prediction methods. We have also used two of the most common adjuvants in our vaccine formula-QS21 and Al(OH) 3 -to enhance the immune response and tested this vaccine on a mouse model. The immunization results indicate that antibody production against each peptide varies with time and adjuvant used. In the QS21 adjuvant group, peptide B was the first antibody produced, followed by antibodies against peptides C, F, and D. In the Al(OH) 3 adjuvant group, antibodies against B, C, and F were detected at the same time after the second immunization, which was quicker than in the QS21 adjuvant group (Figure 1 ). Another major difference between the two adjuvants is that the Al(OH) 3 group produced antibodies against peptide A much earlier than the QS21 group. Since it is imperative for the body to generate a sufficient T cell response to combat infectious diseases [86] , the T and B cell population of each adjuvant was further analyzed. The results indicate that Al(OH) 3 is the ideal adjuvant because it can induce a more robust T cell response than QS21 ( Figure 5 ), although both adjuvants were able to induce an antibody response when tested using the ELISA assay. Since a high antibody titer does not always signify satisfactory protection, we also tested the protection of plasma with the pseudoviral system [87] . This showed that the plasma from the vaccinated mice can effectively neutralize viral entry ( Figure 6 ). Another issue related to the anti-viral activity of plasma is determining how many epitopes are adequate to completely neutralize the viral entry that can be used as target sites for vaccine development [88] . To collect this information, epitope mapping with plasma from vaccinated mice and from infected patients was conducted. Epitope mapping revealed a similar antibody pattern from vaccinated mice to human samples, which confirmed that our epitope covered most B cell epitopes of the S protein ( Figure 2) . Thus, our results demonstrated that development of a S protein peptide-based vaccine is likely feasible to prevent COVID-19. The authors show that a peptide-based COVID-19 vaccine using S protein peptides with the Al(OH) 3 adjuvant generates a high antibody and T cell response. We also demonstrate that this peptide-based vaccine includes most major B cell epitopes of the S protein by comparing the serological antibody profiles of mice injected with our peptide-based vaccine with humans who were previously infected with COVID-19. Further investigation into peptide-based COVID-19 vaccines is warranted to evaluate important factors such as long-term efficacy. Supplementary Materials: The following supporting information can be downloaded at: https: //www.mdpi.com/article/10.3390/v14030449/s1, Table S1 : Selection of SARS-CoV-2 peptides. The data presented in this article are available on request from the corresponding author. The authors declare no conflict of interest. COVID-19 Vaccines: Current Conditions and Future Prospects The Perspective of Coronavirus Disease Outbreak: Epidemiology, Transmission, and Possible Treatment. Vector Borne Zoonotic Dis COVID-19 and Its Global Economic Impact Rapid and Impressive Response to a Combined Treatment with Single-Dose Tocilizumab and NIV in a Patient with COVID-19 Pneumonia/ARDS A Review of Remdesivir for COVID-19: Data to Date Controversial treatments: An updated understanding of the coronavirus disease 2019 SARS-CoV-2-neutralising monoclonal antibodies for treatment of COVID-19 Convalescent plasma treatment of severe COVID-19: A propensity score-matched control study Convalescent plasma transfusion therapy in severe COVID-19 patients-A safety, efficacy and dose response study: A structured summary of a study protocol of a phase II randomized controlled trial Neutralizing Antibody Responses in COVID-19 Convalescent Sera Compassionate use of convalescent plasma for treatment of moderate and severe pneumonia in COVID-19 patients and association with IgG antibody levels in donated plasma Effectiveness of convalescent plasma therapy in severe COVID-19 patients Monoclonal antibody as a potential anti-COVID-19 Eradication of vaccine-preventable diseases Prevention of hepatitis B mother-to-child transmission in Namibia: A cost-effectiveness analysis Efficacy of Birth Dose Vaccination in Preventing Mother-to-Child Transmission of Hepatitis B: A Randomized Controlled Trial Comparing Engerix-B and Sci-B-Vac. Vaccines Impact of HPV vaccination and cervical screening on cervical cancer elimination: A comparative modelling analysis in 78 low-income and lower-middle-income countries Toward global prevention of sexually transmitted infections (STIs): The need for STI vaccines Vaccines: Status Report. Immunity The SARS-CoV-2 Vaccine Pipeline: An Overview Vaccines for COVID-19: Perspectives, Prospects, and Challenges Based on Candidate Sars, Mers, and Animal Coronavirus Vaccines Preliminary Identification of Potential Vaccine Targets for the COVID-19 Coronavirus (SARS-CoV-2) Based on SARS-CoV Immunological Studies Possible Tools to Combat SARS-CoV-2 Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: An interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK Oxford-AstraZeneca COVID-19 vaccine efficacy Sputnik V COVID-19 vaccine candidate appears safe and effective The Russian vaccine for COVID-19 The arrival of Sputnik V Double-Blind, Placebo-Controlled Phase 3 Study to Assess the Efficacy and Safety of Ad26.COV2.S for the Prevention of SARS-CoV-2-Mediated COVID-19 in Adults Aged 18 Years and Older Phase 1-2 Trial of a SARS-CoV-2 Recombinant Spike Protein Nanoparticle Vaccine Designing spike protein (S-Protein) based multi-epitope peptide vaccine against SARS COVID-19 by immunoinformatics SARS-CoV-2 vaccines strategies: A comprehensive review of phase 3 candidates Structure, Function, and Evolution of Coronavirus Spike Proteins Mechanisms of SARS-CoV-2 entry into cells Stabilizing the closed SARS-CoV-2 spike trimer ACE2: The Major Cell Entry Receptor for SARS-CoV-2. Lung Structural and functional properties of SARS-CoV-2 spike protein: Potential antivirus drug development for COVID-19 Cell entry mechanisms of SARS-CoV-2 Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19) COVID-19: What have we learnt about the new variant in the UK? Different mutations in SARS-CoV-2 associate with severe and mild outcome COVID-19 reinfection-An enigmatic public health threat. Monaldi Arch Analysis of Indian SARS-CoV-2 Genomes Reveals Prevalence of D614G Mutation in Spike Protein Predicting an Increase in Interaction with TMPRSS2 and Virus Infectivity A case of COVID-19 reinfection in the UK What reinfections mean for COVID-19 SARS-CoV-2 mutations, vaccines, and immunity: Implication of variants of concern Immunostimulatory CpG oligonucleotides: Effect on gene expression and utility as vaccine adjuvants The safety evaluation of adjuvants during vaccine development: The AS04 experience Vaccine adjuvants: Current state and future trends Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization The search for a potent DNA vaccine against AIDS: The enhancement of immunogenicity by chemical and genetic adjuvants Malaria DNA vaccine gp96NTD-CSP elicits both CSP-specific antibody and CD8(+) T cell response DNA gag/adenovirus type 5 (Ad5) gag and Ad5 gag/Ad5 gag vaccines induce distinct T-cell response profiles DNA prime-adenovirus boost immunization induces a vigorous and multifunctional T-cell response against hepadnaviral proteins in the mouse and woodchuck model Safety and Efficacy of the BNT162b2 mRNA COVID-19 Vaccine Peptide-based therapeutic vaccines for allergic and autoimmune diseases Peptide Vaccine: Progress and Challenges Peptide-Based Immunotherapeutics and Vaccines Aluminum hydroxide adjuvant differentially activates the three complement pathways with major involvement of the alternative pathway On the effect of Al(OH) 3 as an immunological adjuvant Updated insights into the mechanism of action and clinical profile of the immunoadjuvant QS-21: A review The Johnson & Johnson Vaccine for COVID-19 A Review of the Progress and Challenges of Developing a Vaccine for COVID-19 Novavax vaccine delivers 89% efficacy against COVID-19 in UK-But is less potent in South Africa Efficacy of NVX-CoV2373 COVID-19 Vaccine against the B.1.351 Variant COVID vaccines and safety: What the research says The BNT162b2 (BioNTech/Pfizer) vaccine had 95% efficacy against COVID-19 ≥7 days after the 2nd dose COVID-19 Vaccine Decisions: Considering the Choices and Opportunities. Microbes Infect COVID-19: Oxford vaccine is up to 90% effective, interim analysis indicates COVID-19: Russian vaccine efficacy is 91.6%, show phase III trial results Coronavirus vaccines leap through safety trials-But which will work is anybody's guess Vaccine Candidates against Coronavirus Infections. Where Does COVID-19 Stand? Viruses COVID-19 vaccines: Comparison of biological, pharmacological characteristics and adverse effects of Pfizer/BioNTech and Moderna Vaccines Prior SARS-CoV-2 infection rescues B and T cell responses to variants after first vaccine dose COVID-19 vaccine BNT162b1 elicits human antibody and T H 1T cell responses Innate immune response to viral infection How COVID-19 induces cytokine storm with high mortality The University of Texas Medical Branch at Galveston Immunity and immunopathology to viruses: What decides the outcome? Human Immunodeficiency Virus Immune Cell Receptors, Coreceptors, and Cofactors: Implications for Prevention and Treatment Overview of Immune Response During SARS-CoV-2 Infection: Lessons From the Past Distinct pathways of antigen uptake and intracellular routing in CD4 and CD8 T cell activation How 'killer' T cells could boost COVID immunity in face of new variants Correlates of antibody-mediated protection against HIV infection Evolution of cross-neutralizing antibodies and mapping epitope specificity in plasma of chronic HIV-1-infected antiretroviral therapy-naïve children from India