COVID VACCINES

COVID-19 Vaccine Pioneers and Nanotechnology Design

 

Humanity is experiencing a catastrophic epidemic. SARS-CoV-2 has spread globally with significant morbidity and mortality, there are still unknowns about the biology and pathology of the virus. Even with testing, monitoring and social distancing, many countries are able to contain SARS-CoV-2 struggling to get it. COVID-19 can only be suppressed with an effective vaccine or when herd immunity has developed if the population is infected and resistant to re-infection. Until there is an effective vaccine, there is virtually no chance of returning to normal social behavior before COVID-19. The joint effort of doctors, academic labs and companies around the world has improved detection and treatment, and taking promising early steps, many vaccine candidates have been deployed at an unprecedented rate for previous diseases.As of August 11, 2020' 28 of these companies entered clinical trials with Moderna, CanSino, Oxford University, BioNTech, Sinovac, Sinopharm, Anhui Zhifei Longcom, Inovio, Novavax, Vaxine, Zydus Cadila, Institute of Medical Biology and Gamaleya Research Institute. This review analyzes pioneers in vaccine development and highlights published results while highlighting the role of nanotechnologies implemented by all vaccine developers. Gamaleya Research Institute has gone beyond initial safety and immunogenicity studies. It analyzes pioneers in vaccine development and highlights published results while highlighting the role of nanotechnologies implemented by all vaccine developers. Gamaleya Research Institute has gone beyond initial safety and immunogenicity studies. It analyzes pioneers in vaccine development and highlights published results while highlighting the role of nanotechnologies implemented by all vaccine developers. Gamaleya Research Institute has gone beyond initial safety and immunogenicity studies.

The cause of respiratory disease is a betacoronavirus class virus called coronavirus infectious disease-19 (COVID-19). The virus was named SARS-CoV-2 because of its genetic and structural similarity with SARS-CoV. He officially defined CoV-2 as a pandemic due to its rapid global spread. The continued rise of both cases and deaths requires the rapid development of an effective SARS-CoV-2 vaccine. A second wave in some countries that have reopened their economies further reinforces this need. While masking, social distancing and contact tracing can slow the spread of this virus, it seems too contagious to be eradicated by these strategies, and a vaccine is essential to bring a return to normal human social interaction.

Fortunately, in the relatively few months since SARS CoV-2 was identified as the cause of COVID-19, more than two hundred academic laboratories and companies have begun developing vaccines, and many are having a record time advancing to clinical trials Moderna, 63 days from sequence selection Then he started clinical vaccine studies. With an unestablished formulation of nanotechnology, established approaches ( i.e., inactivated and live-attenuated vaccines) reached clinical testing almost exactly a month before they entered clinical trials. This is an opportunity for less advanced technology platforms in vaccine development and, if proven successful, could respond more quickly to emerging infectious diseases in the future. It should also be noted that in previous severe coronavirus outbreaks of SARS-CoV and MERS-CoV, clinical trials were not reached 25 and 22 months after the onset of the outbreaks. Older outbreaks of severe infectious diseases, such as Dengue and Chikungunya, did not reach clinical trials until 52 and 19 years after the outbreak. The increasing pace of clinical trials is promising, but despite rapid progress, there is still cause for concern.

Vaccine development takes time, as vaccines need to be not only protective but also safe. Unlike other drugs given to patients, vaccines are administered to healthy individuals and require very high safety margins. Therefore, the population should be carefully monitored if vaccine candidates are widely administered according to the Emergency Use Authorization. It is vital as in past respiratory diseases such as SARS-CoV, MERS-CoV, respiratory syncytial virus and measles, and it has been shown that antibodies can increase disease severity through antibody-dependent augmentation. Many of the leading vaccines are preclinical nanotechnologies and have not been proven in clinical settings. For example, mRNA vaccines have been in development and clinical testing for the past 30 years, but the technology has not been previously validated. Platform technology offers speed and adaptability,


 

Vaccination Immunology

It is important to understand the concepts in vaccine immunology to better understand the clinical data. There is no “one size fits all” protective antiviral immune response. Each virus is different, with different routes of infection, different types of cells that can be infected, and different associated pathologies. Accordingly, the optimal immune response for protection against each virus will also be variable. Other factors such as gender, age, pregnancy, and route of infection can also affect the immune response. It is widely reported that some individuals are heavily infected with SARS-CoV-2, but remain asymptomatic, and some become critically ill and succumb to the disease. This extreme variability in response to infection highlights the variability of individual immune responses to this virus and shows that there may not be one perfect outcome that will provide a uniform long-lasting immunity in everyone. The specific immune responses that elicit the most rapid and reliable viral clearance need to be understood and replicated by vaccines. Whether humoral and/or cellular cytotoxic responses are required, which type of helper T cells are most effective (For example , Th1 versus Th2 versus Th17) as well as the isotype of the antibody response ( eg IgG and IgA) should be determined to most effectively protect against this virus.

Many of these questions are answered through analysis of serum and circulating cells from recovered patients, as well as laboratory studies.

Given the variability of host immune responses, unfortunately, there is no guarantee that a vaccine will protect against SARS-CoV-2, even if it has advanced towards further clinical trials. While a single vaccine can provide lifetime protection against chickenpox or polio virus Despite a worldwide effort to develop an effective HIV vaccine, HIV falls short of protection through vaccination. There are indications that respiratory viruses are particularly difficult to protect with vaccines. Respiratory syncytial virus is a prime example for which, despite significant efforts for development, approved vaccines have not been found.

One reason for vaccine failure against respiratory viruses is that the respiratory tract, including the lungs, is an external mucosal surface that is protected by the generation of secreted IgA antibodies; however, antibodies measured to determine whether an experimental subject "responds" to a vaccine usually focus on IgG, IgM, or total immunoglobulin in the blood. Most vaccines are administered intramuscularly and mucosal immunity and IgA secretion are therefore minimal. Also, IgA production from conventional vaccines is difficult to detect and there may be immunodeficiency. Nonetheless, there are efforts and reports of development of SARS-CoV-2 vaccine candidates that can elicit IgA responses. For example, Altimmune, an intranasally administered adenovirus (Ad)-based non-replicating viral vector vaccine, showed a 29-fold induction of IgA in mice. Other companies, such as Stabilitech Biopharma Limited and Quadram Institute Biosciences, are also developing mucosal vaccines. The value of IgA or other immunoglobulin isotypes in protection against SARS-CoV-2 has not been fully elucidated, but it is believed that IgA may prevent SARS-CoV-2 from binding to the airway epithelium, thereby helping to block both initial infections. It should be noted that it is not known what role, if any, IgA plays in protecting against SARS-CoV-2, and most current vaccines do not attempt to specifically activate IgA responses. Of course, as IgA production is negatively correlated with increased severity in COVID-19 patients, it is possible that IgA production is not important or even harmful for an effective vaccine. Other companies, such as Stabilitech Biopharma Limited and Quadram Institute Biosciences, are also developing mucosal vaccines. The value of IgA or other immunoglobulin isotypes in protection against SARS-CoV-2 has not been fully elucidated, but it is believed that IgA may prevent SARS-CoV-2 from binding to the airway epithelium, thereby helping to block both initial infections. It should be noted that it is not known what role, if any, IgA plays in protecting against SARS-CoV-2, and most current vaccines do not attempt to specifically activate IgA responses. Of course, as IgA production is negatively correlated with increased severity in COVID-19 patients, it is possible that IgA production is not important or even harmful for an effective vaccine. Other companies, such as Stabilitech Biopharma Limited and Quadram Institute Biosciences, are also developing mucosal vaccines. The value of IgA or other immunoglobulin isotypes in protection against SARS-CoV-2 has not been fully elucidated, but it is believed that IgA may prevent SARS-CoV-2 from binding to the airway epithelium, thereby helping to block both initial infections. It should be noted that it is not known what role, if any, IgA plays in protecting against SARS-CoV-2, and most current vaccines do not attempt to specifically activate IgA responses. Of course, as IgA production is negatively correlated with increased severity in COVID-19 patients, it is possible that IgA production is not important or even harmful for an effective vaccine.

SARS-CoV-2 is unusual for a respiratory virus in that it binds to a receptor, angiotensin converting enzyme 2 (ACE2), and is expressed in nearly all organs, but particularly in the lungs, brain, and gut. Therefore, unlike most respiratory viruses, SARS-CoV-2 has a wider biodistribution and can cause significant damage outside the respiratory tract. It adversely affects the digestive, urogenital, central nervous and circulatory systems, and the prevalence of the ACE2 receptor is why symptoms are highly variable and can range from shortness of breath, diarrhea, headaches, high blood pressure, venous thromboembolism, and more. Therefore, since most of the pathology is outside the airway due to systemic viral infection, a vaccine that elicits IgG antibodies may protect patients from systemic circulation of the virus.

Another hallmark of vaccine development is T-cell involvement, and differences in T-cell responses can affect the elimination of infected cells as well as the generation of high-affinity and neutralizing antibodies (NAbs). Immune memory and the production of high-affinity antibodies are highly dependent on T cells and do not normally develop without appropriate T-cell involvement. Immune memory is a key factor in long-term immune protection, and studies have shown that immune titers of patients first infected with SARS-CoV have increased up to 3 years after infection. showed that it can have significant antibody levels. Such antibody development would be extremely useful in the fight against SARS-CoV-2, and this long-term immune memory could potentially provide long-term protection with a vaccine.

It is currently unclear whether any of the tested vaccines will protect against SARS-CoV-2. Fortunately, as noted below, there are encouraging early results from multiple vaccines that are safe and immunogenic in limited patients. This early success warrants progress to Phase III clinical trials and expectations are that 20,000-40,000 subjects will be involved. The effort to develop effective vaccine(s) against SARS-CoV-2 is daunting, but there is no guarantee of success, but we are encouraged by the success of early testing and the rapid development of many candidate vaccines.


 

Nanotechnology Offers Opportunities in Vaccine Design

Nanoparticles and viruses operate on the same size scale; therefore, nanoparticles enter cells to induce expression of antigens from delivered nucleic acids (mRNA and DNA vaccines) and/or may have immune cells directly targeted for delivery of antigens (subunit vaccines). Many vaccine technologies use these direct pathways by encapsulating genomic material or protein/peptide antigens in nanoparticles such as lipid nanoparticles (LNPs) or other viruses such as Ads. BioNTech/Pfizer and Moderna encapsulate their mRNA vaccines in LNPs, while Oxford University/Astrazeneca (hereafter Oxford/Astrazeneca) and CanSino contain antigen-coding sequences within DNA. The nanoparticles are described in more detail in the following discussion.

Beyond antigen delivery, nanoparticles can co-deliver with adjuvants to help prepare desired immune responses. Adjuvants are immunostimulatory molecules that are mainly administered with the vaccine to help strengthen immune responses by activating additional molecular receptors that predominantly recognize pathogens or danger signals. These pathways function primarily within the innate immune system, and each adjuvant usually has a different stimulation range of these pathogen or danger receptors. Encapsulation and/or conjugation of both adjuvant and antigen within the same nanoparticle enables targeted, synchronized delivery to the same antigen-presenting cell (APC). Many adjuvants have previously failed clinically due to toxicity issues, and this co-administration It can help direct antigen and adjuvant activity only in APCs that receive the antigen, thereby reducing off-target side effects. Targeted delivery of suitable adjuvants can also reduce the dose of antigen required for immune protection, thereby producing a protective effect with a dose. This effect will be practically and financially plentiful in the current pandemic due to the enormous number of doses required for global vaccination. Furthermore, when adjuvants and antigens are not administered together, they can rapidly decompose within the body, causing off-target effects and/or rapid degradation of the adjuvant and reducing the potency of the vaccine. Both Moderna and BioNTech encapsulate mRNA vaccines in LNPs to protect mRNA from nuclease degradation. It can help guide in the future and thus reduce off-target side effects. Targeted delivery of suitable adjuvants can also reduce the dose of antigen required for immune protection, thereby producing a protective effect with a dose. This effect will be practically and financially plentiful in the current pandemic due to the enormous number of doses required for global vaccination. Furthermore, when adjuvants and antigens are not administered together, they can rapidly decompose within the body, causing off-target effects and/or rapid degradation of the adjuvant and reducing the potency of the vaccine. Both Moderna and BioNTech encapsulate mRNA vaccines in LNPs to protect mRNA from nuclease degradation. It can help guide in the future and thus reduce off-target side effects. Targeted delivery of suitable adjuvants can also reduce the dose of antigen required for immune protection, thereby producing a protective effect with a dose. This effect will be practically and financially plentiful in the current pandemic due to the enormous number of doses required for global vaccination. Furthermore, when adjuvants and antigens are not administered together, they can rapidly decompose within the body, causing off-target effects and/or rapid degradation of the adjuvant and reducing the potency of the vaccine. Both Moderna and BioNTech encapsulate mRNA vaccines in LNPs to protect mRNA from nuclease degradation. it may also reduce the dose of antigen required for immune protection and thus produce a protective effect with one dose. This effect will be practically and financially plentiful in the current pandemic due to the enormous number of doses required for global vaccination. Furthermore, when adjuvants and antigens are not administered together, they can rapidly decompose within the body, causing off-target effects and/or rapid degradation of the adjuvant and reducing the potency of the vaccine. Both Moderna and BioNTech encapsulate mRNA vaccines in LNPs to protect mRNA from nuclease degradation. it may also reduce the dose of antigen required for immune protection and thus produce a protective effect with one dose. This effect will be practically and financially plentiful in the current pandemic due to the enormous number of doses required for global vaccination. Furthermore, when adjuvants and antigens are not administered together, they can rapidly decompose within the body, causing off-target effects and/or rapid degradation of the adjuvant and reducing the potency of the vaccine. Both Moderna and BioNTech encapsulate mRNA vaccines in LNPs to protect mRNA from nuclease degradation.that is, uptake of antigen and adjuvant by APCs at separate times can also lead to autoimmunity against host proteins, since the adjuvant can activate APCs that are not initiated against antigen but rather self-prepared. Therefore, nanotechnology presents an opportunity in vaccine design and SARS- There are several strategies for co-administration of CoV-2 antigens and adjuvants. The three main methods are (i) co-delivery by encapsulation or conjugation in a nanoparticle, (ii) direct antigen-adjuvant conjugation, and (iii) use of the delivery vehicle as an adjuvant.

Another benefit that nanoparticles can provide is multivalent antigen presentation and orientation of subunit antigens in their native form. For example, BioNTech/Pfizer, one of the leading companies producing the SARS-CoV-2 vaccine, formulates receptor binding domain (RBD) antigens on a T4 fibrite-derived "fold" trimerization base to better mimic the trimeric form of the spine (S) SARS-CoV -2 protein. Furthermore, the display of different RBD epitopes of influenza A on multivalent ferritin nanoparticles can increase the production of cross-reactive B cells against influenza A and produce a more diverse and effective antibody response than homotypic RBD-screened ferritin nanoparticles. heterotypic nanoparticles stimulate a broad NAb response, which is all-encompassing,

Finally, due to the "nano" scale and composition of nanomaterials, they can circulate in vivo unlike other materials. The lymphatic system, APCs and other lymphocytes are critical in initiating immune responses as they travel from peripheral organs to nearby lymph nodes using the lymphatic system. Access to the lymphatic system can be difficult, but nanomaterials can cross interstitial spaces and reach nearby lymph nodes. For example, inhaled radiolabeled solid lipid nanoparticles pass from the alveoli to nearby lymph nodes via the Lymphatic system, while free radiotracers travel through the systemic circulation. Lymphatic drainage to lymph nodes near the lungs, in particular, can be extremely useful in the fight against respiratory diseases such as SARS-CoV-2. Companies such as EtheRNA and Intravacc are developing intranasally administered vaccines that are delivered to the respiratory tract that can target such nearby lymph nodes.

For further reading on the opportunities of nanotechnology in SARS-CoV-2 vaccine design, we suggest the reader the following review. This review also discusses the challenges and opportunities of the manufacturing processes and distribution platforms required for global vaccination.


 

Status of COVID-19 Vaccine Candidates

According to the World Health Organization and the Milken Institute, as of August 11, 2020, there are 202 companies and universities working on a coronavirus vaccine worldwide, the vaccine types being established vaccines ( eg , inactivated and live attenuated), recently additionally clinically approved ( eg not yet routinely introduced). eg; mRNA , DNA, non-replicating viral vector, replicating viral vector).

Inactivated vaccines are similar to the native pathogen but are replication inefficient due to chemical or heat treatment. Live-attenuated vaccines are attenuated forms of the virus that can replicate in a limited manner. Subunit vaccines are generally less immunogenic and require an adjuvant to stimulate immune recognition of the antigens in the vaccine.

Nucleic acid-based vaccines can be mRNA or DNA-based and express the antigen in host cells using genomic material, rather than injecting it directly. Finally, viral vector vaccines contain genomes designed to encode the antigen of the target pathogen. When administered in vivo , viral vectors enter target cells and genomic material is copied and translated for in vivo antigen production. Replication-deficient vaccines may provide better safety, but immune memory is not long-lived. There are currently no viral vector vaccines used for humans in the clinic, but veterinary only a select few of the 202 companies have entered clinical trials;

As of 11 August 2020, WHO states that there are 29 vaccine candidates in clinical trials. Of these select vaccines, even fewer from completed Phase I and II studies, they have published data on their initial safety and immunogenicity. It is notable that each company that publishes the data reports positive results from early Phase clinical trials, allowing for progress involving larger and broader efficacy studies.

In this review, we discuss vaccine nanotechnologies used by companies that publish their results. Two of the companies with early results are the Moderna and BioNTech / Pfizer partnerships. While Moderna has published its data in the New England Journal of Medicine , BioNTech/Pfizer's data has now been previously published on medRxiv and awaits peer review. Both using mRNA encoding subunits of the SARS-CoV-2 S protein. uses similar techniques for their vaccines. On the other hand, Oxford/Astrazeneca and CanSino are developing vaccines based on non-replicating viral vectors.

In the clinic, both viral vector vaccines and mRNA vaccines have had variable success with none of the vaccine types currently approved for a particular use. mRNA vaccines and viral vector vaccines both encode the target antigen(s) but differ in their vaccination approach Viral vector vaccines can provide high gene transduction capabilities due to their ability to enter cells using the virus's own receptor for infection, and efficient intracellular trafficking enables high production of target gene expression .However, the immunogenicity of viral vectors and other adverse effects pose barriers to their safe use. The immunogenicity of the viral vector may reduce the vaccine efficacy induced by NAbs against the viral vector in pre-existing patients developed during vaccination or due to previous exposure to the Ad vectors they use. Another safety issue for viral vectors is possible host genome integration that could cause cancer if integrated into oncogenes and other regulatory sequences.

mRNA vaccines are often encapsulated in nanoparticles. Both BioNTech/Pfizer and Moderna encapsulate RNA vaccines in LNPs through fusogenic mechanisms.However, although BioNTech/Pfizer mentions the use of cationic lipids, neither Moderna nor BioNTech/Pfizer specifically mention the use of fusogenic LNPs. Cytoplasmic administration can increase translation efficiency, but also reduce RNA immunostimulation. RNA stimulates the immune system and therefore, it acts as an adjuvant by activating specific toll-like receptors (TLRs), particularly TLRs 3, 7 and 8, all located within the endosomes of the cell. TLRs 7 and 8 degrade single-stranded RNA. It is especially important for mRNA vaccines as they recognize and enter virus recognition. Capsule in nanoparticles. With APCs with subsequent localization in endosomes, RNA develops phagocytosis. No endocytosis of RNA,It can lead to nuclease degradation and weak immune stimulation. While advances in nanoparticle design enable cytoplasmic delivery of mRNA, synthetic nanoparticles do not match the efficiency of machinery developed by viral vectors that enable traffic within the cell. Once inside the cell, the mRNA is translated directly within the cytoplasm; conversely, DNA plasmids from viral vectors need to be translocated to the nucleus, copied and returned to the cytoplasm. This means that mRNA vaccines can produce larger amounts of antigens from smaller doses, but one caveat is that DNA tends to be more stable than mRNA, so mRNA expression is often more The interaction between stability and translation efficiency can be a major determinant in effective antigen production.


 

Results – mRNA Vaccines: Moderna and BioNTech / Pfizer

As mentioned above, Moderna and BioNTech/Pfizer use mRNA vaccines that encode the S protein of SARS-CoV-2. The S protein is the viral protein that binds to ACE2 on cells to mediate infection, and antibodies that bind to the correct epitope on the S protein are neutralizing and thus it is a frequent vaccine target as it is expected to block viral spread between cells. The S protein has two subunits: S1 and S2.S1 subunit contains RBD and is responsible for initial binding to the host cell via the ACE2 receptor, while S2 subunit interacts with cells to initiate infection. promotes viral fusion. Moderna's vaccine, mRNA-1273,It was developed in conjunction with the National Institute of Allergy and Infectious Diseases and encodes a prefusion form of the S antigen (called S-2P) specifically containing a transmembrane anchor and an intact S1–S2 cleavage site. Vaccine mRNA at amino acids 986 and 987 located in the central helix of the S2 subunit. mRNA is encapsulated in an LNP of four lipids Full formulation not provided; however, the ionizable lipid, 1,2-distearoyl-Inferences can be made based on Moderna's previous LNP vaccines using sn formulations. -glycero-3-phosphocholine, cholesterol and polyethylene glycol-lipid. The exact lipids used are not specified. LNPs containing mRNA are dissolved and injected directly into the deltoid muscle. Moderna suggests the use of an adjuvant. does not specify explicitly, but the LNP transporter may be an adjuvant because other lipids have been reported to have adjuvant properties.

The mRNA used by BioNTech/Pfizer encodes for RBD. The mRNA, designated BNT162b1, is only available in vivoThe precise mechanism for enhanced translation is not fully elucidated, but one hypothesis is that nucleoside modification improves RNA stability by reducing rates of hydrolysis by phosphodiesterases. Proven to improve RNA secondary structure stability due to RNA stacking. In addition, as mentioned above, the formulated RBD antigen is constructed on a T4 fibrite-derived "foldon" trimerization base that helps guide the folding of the antigen into the native trimeric state. T4 trimerization is also essentially trimerized. It enhances immunogenicity due to the multivalent image presented in the precipitated state.It is important to note that BioNTech/Pfizer is testing at least four mRNA vaccines (BNT162a1, BNT162b1, BNT162b2 and BNT162c2) in parallel, but the previously published manuscript only contains data from the BNT162b1 candidate. encapsulated in an LNP and administered the vaccine by intramuscular injection.The LNP is available from a partnership with Acuitas Therapeutics. The exact formulation is not specified, but previous publications of Acuitas Therapeutics have shown that their LNPs are 0.05 RNA:lipid (w/) using ionizable cationic lipids, phosphatidylcholine, cholesterol, and polyethylene glycol-lipid. w).

produced higher geometric mean titers (GMTs) of S-2P at day 36 (163,449 versus 142,140 arbitrary units (AU)) while the 25 and 100 μg groups produced higher GMTs at day 36 (respectively) 7 days after a second increase. BioNTech/Pfizer recorded neutralizing anti-RBD titers much higher than recovery serum levels.21. Up to day 28 (for the 10 μg group (4.813 U/mL)) on day 28 (day of the second dose or day 21), the 30 μg group convalescent serums (602 U/mL versus 1,536) had a higher geometric mean concentration (GMC). a second dose). The 100 μg group using only one dose had higher GMC levels (1,778 U/mL) at day 21. H1 skewed T-cell responses with detectable CD4+ and CD8+ response to their respective antigens. Both enhancers He did not mention the production of antibodies other than IgG. The vaccination schedule in both Moderna and BioNTech / Pfizer's Phase III trials will not deviate from their Phase II setup. Moderna will continue to increase at day 29 after the first injection and BioNTech / Pfizer will increase by day 21. However, Phase III trials will only evaluate one dose. In Moderna's case, the intermediate dose resulted in higher immunogenicity than the highest dose, while BioNTech/Pfizer showed no significant difference between the intermediate and high-level doses. Therefore, Moderna and BioNTech/ Pfizer has chosen to move forward with intermediate doses (100 μg and 30 μg, respectively). For Phase III, Moderna and BioNTech / Pfizer will also vaccinate much larger populations of 30,000 participants each.


 

Results-Non-repeat Viral Vector Vaccines: Oxford / Astrazeneca and CanSino

One of the most researched viral vector options is Ads, currently used by both CanSino and Oxford/Astrazenca. Ads are common cold-causing viruses with a double-stranded DNA genome. Specifically, CanSino named the vaccine Ad5-nCoV type 5'. i (Ad5). Oxford/Astrazeneca uses a different viral vector, a chimpanzee-derived Ad (use of the chimpanzee vector minimizes possible interaction with common antibodies against Ads) and was later named AZD1222. Ad5-nCoV, both S In contrast to Moderna and BioNTech/Pfizer, which encode subunits of the protein, it specifically encodes the full-length S protein of SARS-CoV-2. The gene was derived from the Wuhan-Hu-1 sequence for SARS-CoV-2 and was combined with a tissue plasminogen activator signal peptide. together,E1 and E3 were cloned into a deleted Ad5 vector. Deletion of E1 inactivates the replication potential of the vaccine, while deletion of E3 allows insertion of larger genes up to 8 kb. CanSino vectors were resolved and administered intramuscularly to patients' arms. Each shot is 5 x 10 It contains 5 x 10 particles per viral dose, and patients tested at high doses will receive multiple shots, allowing for administration in multiples of 10 particles.Each shot contains 5 x 10 particles per viral dose, and patients tested at high doses will receive multiple shots, allowing for administration in multiples of 5 x 10 particles.Each shot contains 5 x 10 particles per viral dose, and patients tested at high doses will receive multiple shots, allowing for administration in multiples of 5 x 10 particles.

AZD1222 was designed similarly to Ad5-nCoV with the deletion of E1 to inhibit replication and deletion of E3 to allow incorporation of greater genetic load into the viral vector. The inserted sequence encodes the full-length S protein with a tissue plasminogen activator leader sequence and the S protein The sequence is codon-optimized. The main differences between the two viral vector platforms are discussed in more detail in the discussion section. None of the vaccine manufacturers mention the use of an adjuvant, so these vectors are most likely due to the DNA they carry of non-replicating virus, perhaps via the DNA they carry that can activate TLR9 within endosomes. Another possibility is recognition of the viral capsid, which can occur through both TLR-dependent and TLR-independent mechanisms.The intracellular adapter protein MyD88 has been shown to play an important role in initiating TLR-mediated immunogenicity by the ability of viral vectors to engage multiple MyD88 signaling pathways.

Data from Phase I/II trials are summarized. CanSino has completed and published results for both Phase I and II vaccines; the following will only present data from Phase II trials. The major difference between the two trials is that CanSino has severe fever, fatigue, and muscle and arthralgia. particles.28. The group saw RBD-specific antibody GMT levels peak around 11656.5 AU at day 1 × 10, while GMT levels peak around 5 × 10 10. RBD-specific antibody seroconversion occurred in 96% and 97% patients within 1 × 10, 11 and 5 × 10 10, respectively. group.measured in vitro .

Oxford/AstraZeneca tested at a dose of only 5 x 10 viral particles per dose of 10, but in a few patients ( n = 533 )= 10), also tested a booster at the first same dose 28 days later. Some of the patients were given acetaminophen, a common anti-inflammatory drug, prophylactically, and these patients experienced fewer adverse events. Concentrations of antibodies to the SARS-CoV-2 S protein, In single-dose patients (157 AU), the extra dose further improved the response (639 AU over 56 days), while peaking at day 28. Acetaminophen did not prevent the formation of the antibody response. It induced neutralizing titers in 100% and up to 100% in double-dose patients. Both studies documented moderate increases in T-cell responses as measured by ELISpot tests.

Adverse events from both the Oxford/Astrazeneca and CanSino vaccines ranged from mild to moderate and did not warrant discontinuation of either study. The most prominent adverse events in both the Oxford/Astrazeneca and CanSino trials were pain, fatigue, and headache. Oxford/Astrazeneca, other than IgG did not test for antibodies. CanSino tested for IgM antibodies, but only against the nucleocapsid of SARS-CoV-2 to ensure participants were not previously exposed to the virus. For phase III clinical trials progress, Cansino recorded the lowest dose (5 x 10 10 particles) ) showed similar immunogenicity (1 x 10 11 particles) compared to its intermediate dose, while also reducing adverse events. Thus, Phase III trials will proceed at the lowest dose.5.They will begin testing in countries outside of China, such as Saudi Arabia, where they already have an agreement to vaccinate 000 participants. The company wants to increase the number of participants by holding clinical trials in other countries, such as Russia, Brazil and Chile. Oxford / Astrazeneca will continue to test at a dose of 5 × 10. , but will not test a main increase in Brazilian trials. This is most likely due to the similar immunogenicity and neutralization capacity observed between the single-dose and double-dose groups. They will test in multiple countries with Phase III trials ongoing in Brazil, South Africa and the United Kingdom .Oxford/Astrazeneca will continue to provide prophylactic acetaminophen for pain management.It wants to increase the number of participants by conducting clinical trials in other countries such as Brazil and Chile. Oxford/Astrazeneca will continue to test at the 5 × 10 dose, but will not test a major increase in the Brazilian trials. This is most likely the case observed between the single-dose and double-dose groups. due to similar immunogenicity and neutralizing capacity. They will test in multiple countries with ongoing Phase III trials in Brazil, South Africa and the UK. Oxford/Astrazeneca will continue to provide prophylactic acetaminophen for pain management.It wants to increase the number of participants by conducting clinical trials in other countries such as Brazil and Chile. Oxford/Astrazeneca will continue to test at the 5 × 10 dose, but will not test a major increase in the Brazilian trials. This is most likely the case observed between the single-dose and double-dose groups. due to similar immunogenicity and neutralizing capacity. They will test in multiple countries with ongoing Phase III trials in Brazil, South Africa and the UK. Oxford/Astrazeneca will continue to provide prophylactic acetaminophen for pain management.This is due to the similar immunogenicity and neutralizing capacity observed between the single-dose and double-dose groups. They will test in multiple countries with ongoing Phase III trials in Brazil, South Africa and the United Kingdom. Oxford/Astrazeneca will continue to provide prophylactic acetaminophen for pain management.This is due to the similar immunogenicity and neutralizing capacity observed between the single-dose and double-dose groups. They will test in multiple countries with ongoing Phase III trials in Brazil, South Africa and the United Kingdom. Oxford/Astrazeneca will continue to provide prophylactic acetaminophen for pain management.


 

Argument

SARS-CoV-2 has required competition as well as global cooperation and teamwork in combating the disease. Current vaccination efforts exemplify that the discovery and manufacture of these vaccines is a truly worldwide effort that takes place in the context of commercial competition. BioNTech is headquartered in Germany, Moderna and Pfizer are headquartered in the United States, CanSino is headquartered in China, Oxford is headquartered in the United Kingdom, and Astrazeneca is headquartered in both the United Kingdom and Sweden. Bharat is headquartered in India, South Korea, and Japan, respectively. There are other companies involved in clinical trials such as Biotech, Genexine, and Anges. This is not an exhaustive list of all countries working on a coronavirus vaccine, as there are other vaccine development efforts spanning the globe.

The need for vaccines against COVID-19 is so urgent that unless one vaccine is much more effective than the others, multiple vaccine candidates will receive approval in different locations. The creation of a large number of required doses will also support the production and sale of multiple companies. Multiple countries from multiple countries more vaccine candidates will allow for wider and faster distribution to build global "herd immunity". Most or all vaccines will initially be purchased by their country's governments for distribution. While there may be places for vaccine testing if COVID-19 is widespread, less developed countries may contribute to the vaccine effort. not included. This has already been proven in Phase III clinical testing of CanSino and Oxford/Astrazeneca in Brazil and other countries. Distribution to developing countries, from vaccine design,For example, vaccines that require cold chain distribution or administration by healthcare professionals present greater logistical challenges, especially in countries with limited resources.

The vaccine race has also been enhanced by the fact that there are many different vaccination nanotechnologies at play. Both BioNTech/Pfizer and Moderna use vaccination technologies in nucleic acid-based vaccines that have not gone beyond clinical trials for previous diseases. A key advantage of mRNA vaccines, however, is their rapid delivery. In fact, Moderna was the first company in the United States to enter clinical trials for COVID-19 vaccines and dosed the first patient in a Phase I study within 63 days of sequence selection. The first batch of the vaccine was created 25 days after an impressive array selection. In principle mRNA vaccines allow for an indefinite number of booster regimens because the vaccine does not carry the antigen that can be blocked by an established neutralization response in seropositive individuals. Also, due to antigen expression in the host,any occurringlivepost-translational modification represents the natural antigen in the body. This is particularly important for the S protein, which has up to 22 glycosylation sites within each protomer. The glycosylation domains can negate the neutralizing abilities of antibodies that render vaccines ineffective, even if high antibody titers are measured. , glycosylation is an important feature to consider when producing subunit vaccines via heterologous expression. Nucleic acid-based vaccines also activate the host's immune system using multiple mechanisms that elicit both B- and T-cell responses. RNA is both TLR3 for CD8+ T-cell activation. can induce immune responses by activating both 7/8 and can also promote antibody production and CD4+ T cell activation.BioNTech/Pfizer,however, RNA modifications also reduce the immunogenicity of their RNA, which may reduce the adjuvant qualities of RNA. It remains to be seen whether the increased translational abilities from RNA modification outweigh the reduction in immunogenicity. Compared to DNA vaccines, RNA vaccines RNA does not require nuclear localization for antigen production, which allows for activity once RNA enters the cytoplasm of the cell. On the other hand, this also means that expression is generally shorter-lived and may require supplementation while DNA vaccine immunogenicity may persist and not require multiple dosing. A potential way to enhance the immune response is to vectors. using the encoded replication mechanismRNA modifications also reduce the immunogenicity of their RNA, which may reduce the adjuvant qualities of RNA. It remains to be seen whether the increased translational abilities from RNA modification outweigh the reduction in immunogenicity. Compared to DNA vaccines, RNA vaccines allow antigen activity when the RNA enters the cytoplasm of the cell. On the other hand, this also means that expression is generally shorter-lived and may require supplementation while DNA vaccine immunogenicity may persist and may not require multiple dosing. A potential way to enhance the immune response is by using the replication mechanism encoded into vectors.RNA modifications also reduce the immunogenicity of their RNA, which may reduce the adjuvant qualities of RNA. It remains to be seen whether the increased translational abilities from RNA modification outweigh the reduction in immunogenicity. Compared to DNA vaccines, RNA vaccines allow antigen activity when the RNA enters the cytoplasm of the cell. On the other hand, this also means that expression is generally shorter-lived and may require supplementation while DNA vaccine immunogenicity may persist and may not require multiple dosing. A potential way to enhance the immune response is by using the replication mechanism encoded into vectors.It remains to be seen whether the increased translational abilities from RNA modification outweigh the reduction in immunogenicity. Compared to DNA vaccines, RNA vaccines do not require nuclear localization for antigen production, which allows for activity when RNA enters the cytoplasm of the cell. On the other hand, this is also where expression is generally shorter-lived. and DNA vaccine immunogenicity means that while it may continue and may not require multiple dosing, it may require supplementation. One potential way to increase the immune response is by using the replication mechanism encoded into vectors.It remains to be seen whether the increased translational abilities from RNA modification outweigh the reduction in immunogenicity. Compared to DNA vaccines, RNA vaccines do not require nuclear localization for antigen production, which allows for activity when RNA enters the cytoplasm of the cell. On the other hand, this is also where expression is generally shorter-lived. and DNA vaccine immunogenicity means that while it may continue and may not require multiple dosing, it may require supplementation. One potential way to increase the immune response is by using the replication mechanism encoded into vectors.RNA does not require nuclear localization for antigen production, which allows for activity once it enters the cytoplasm of the cell. On the other hand, this also means that expression is generally shorter-lived and may require supplementation while DNA vaccine immunogenicity may persist and not require multiple dosing. A potential way to increase the immune response is to encode vectors using the replication mechanismRNA does not require nuclear localization for antigen production, which allows for activity once it enters the cytoplasm of the cell. On the other hand, this also means that expression is generally shorter-lived and may require supplementation while DNA vaccine immunogenicity may persist and not require multiple dosing. A potential way to enhance the immune response is to encode vectors using the replication mechanismIn viva will be the use of self-amplifying RNA that can replicate. In fact, Imperial College London in collaboration with Morningside Ventures has used this approach for SARS-CoV-2 and is currently in phase I/II testing. Also, half of the unmodified RNA lifespan is very short due to rapid degradation within the body. To help meet this challenge, there are nucleotide modifications that can increase stability. Complexing RNA with protamine improves TLR-mediated adjuvant activity while also reducing RNase degradation. Both Moderna and BioNTech, RNA encapsulates their RNA in LNPs to help protect against degradation as well as chemical modifications to improve stability.

Nonreplicative viral vector vaccines have also made rapid progress in SARS-CoV-2 vaccination efforts. Both Oxford/Astrazeneca and CanSino have demonstrated safe yet immunogenic vaccines that warrant progress to Phase III trials. These viral vectors have been researched for years and technological advances have enabled vector production and large-scale Viral vector vaccines also have innate immunostimulatory profiles via both TLR-dependent and TLR-independent mechanisms. In some cases, they can elicit humoral responses and also enhance CD4+ and CD8+ T-cell responses even without the use of another adjuvant; ELISpot assays showed significant T-cell response activation in both the Oxford/Astrazeneca and CanSino trials. Another benefit is that viral vectors can be transferred to dendritic cells (DC's), leading to increased antigen presentation and immune cell activation. 

Humans have been widely exposed to human Ads that lead to pre-existing immunity to certain Ad vectors that may affect the efficacy of vaccines. Indeed, CanSino noted that 52% of its participants had pre-existing immunity to Ad5.% of respondents with low pre-existing immunity. 48 produced twice the levels of NAb and RBD-specific antibodies than the higher immune group, suggesting that pre-existing immunity to the viral vector impairs the response to the vaccine. solves the problem. Oxford/Astrazenca detected only NAb against the chimpanzee viral vector in one of 98 patients. There are other ways to address the pre-existing immune problem. For example, Ad'Most of the pre-existing immunity generated from the hexone protein is derived from hypervariable regions of the hexon protein. Besides the hypervariable regions, another structural protein, genetic modifications on the fiber knob domain can reduce neutralization. Finally, Ads infect a wide variety of animals, from birds and reptiles to bats, and the Oxford / Similar to Astrazeneca it uses less common chimpanzee Ad vectors, reuse of other Ad viral vectors is conceivable. Other non-human Ad vectors investigated in the past include swine, bovine, canine, ovine, and poultry viral vectors. All of the above-mentioned viral vectors , not pathogenic to humans,however, it can infect mammalian cells and some - such as the bovine viral vector - are inherently replication-free and offer high safety.

Other Companies in Advanced Clinical Trials

Apart from the four major companies discussed above, there are few other companies that have gone beyond the initial clinical trials and are currently investigating more extensive clinical trials in Phase II or III. These companies have not published their data but have announced positive results in their trials with press releases and statements. Some companies are currently is preparing the data for publication. The following information is summarized for both. Sinovac Biotech and Sinopharm are two of these companies and both use an inactivated form of the virus. Inactivated vaccines are solidly built and are produced through chemical or heat treatment that leads to replication-deficient vaccines .Sinopharm's vaccine has already received emergency clearance in China for those working in state-owned enterprises that need global travel.It has two products in clinical testing developed by the Wuhan Institute of Biological Products or the Beijing Institute of Biological Products. The Institute of Medical Biology of the Chinese Academy of Medical Sciences is developing another inactivated vaccine and has progressed to Phase II clinical trials, but there is no clinical trials or public information about the vaccine. Other two companies, Inovio Pharmaceuticals and Zydus Cadila, have recently completed Phase I testing, citing positive safety results. Inovio also announced the induction of both T-cell and humoral responses. Inovio and Zydus Cadila produce nucleic acid-based vaccines, but Moderna and BioNTech/Pfizer' Anhui Zhifei Longcom Biopharmaceutical and Institute of Microbiology of the Chinese Academy of Sciences are developing a subunit vaccine that is moved to Phase II testing, unlike RNA.however, no public announcement has been made regarding the trial. The vaccine consists of an RBD dimer from SARS-CoV-2 administered with an adjuvant. Novavax is a vaccine that has recently published positive data from Phase I/II trials and also has a reassuring safety profile. is another subunit vaccine developer. They use a full-length S protein subunit vaccine administered with patented saponin-based Matrix-M adjuvants. Recombinant S protein with mutations at the S1 and S2 cleavage site to protect against protease degradation and heptad repeat to keep the protein in a pre-fusion conformation / is produced by additional proline substitutions in the central helix. The S protein and Matrix-M adjuvant are mixed just prior to injection.led to antibody neutralization titers against both S protein (after one dose) and wild-type virus (after two doses) in all patients. Vaxine announced positive safety data as the latest subunit vaccine to enter advanced clinical trials. It was developed using computer modeling to identify epitopes that may interfere with binding.

Russia-based Gamaleya Research Institute is developing non-replicating Ad vector vaccines. The vaccine consists of two vectors, Ad5 and Ad26. The vaccine was the first “proprietary” vaccine in the world, but reports indicate that only Phase II clinical trials have been completed on several hundred participants. The company has standard phase III clinical trials. worked in coordination with the Russian government to bypass the trials, and instead, phase III trials will be conducted in parallel with broad vaccination efforts. To the best of our knowledge, results from phase II clinical trials have not been made public. They announced that they have garnered attention from 20 foreign countries with over-dose pre-orders.Russia,plans to mass-produce the vaccine, followed by public vaccination in the fall, to immediately vaccinate frontline health workers and teachers nationwide.

Adjuvan Königung

Most of the vaccines mentioned above may require the use of an adjuvant to induce the antigen response; however, the adjuvant discussion is often overlooked. Of the companies covered in this review, only Anhui Zhifei Longcom, Novavax, Sinovac, and Vaxine explicitly mention the use of an adjuvant. Novavax's adjuvant Matrix-M increases vaccine immunogenicity by recruiting APCs to the injection site, thereby increasing antigen presentation to T cells within draining lymph nodes. Advax adjuvant used by Vaxine is polyfructofuranosyl- d-In an influenza split vaccine with a Th2 antigen, Advax acts as a Th2 adjuvant, while in an influenza inactivated antigen with a Th1 antigen, it acts as a Thl adjuvant. As far as we know, there is no public information about the adjuvant used by Anhui Zhifei Longcom.

BioNTech/Pfizer and Moderna do not explicitly state the use of an adjuvant in their vaccines, but RNA already contains immunostimulatory properties and signals through pathogen recognition receptors. Whether immunostimulation from RNA is potent enough to provide complete protection against SARS-CoV-2 remains to be seen. .In addition, the LNP transporters they use are also likely to provide adjuvant properties. The discovery that liposomes containing diethyl phosphate provide greater protection against diphtheria toxoid than unmodified liposomes, and studies on the use of lipid transporters as adjuvants have increased significantly. Influenza and hepatitis A'Single-layered phospholipid membrane nanoparticles encapsulating viral antigens (otherwise known as virosomes) with adjuvant properties against pylori are currently in clinical use.

Other companies such as GlaxoSmithKline (GSK) and Dynavax have offered their own adjuvants for testing in combination with different vaccines. GSK's adjuvant system (AS03) consists of α-tocopherol, squalene and polysorbate 80 in an oil-in-water emulsion and antigen in lymph nodes. It helps increase antigen-specific antibody production by increasing uptake and presentation. Its adjuvant was previously used in the 2009 H1N1 outbreak. Currently, GSK offers AS03 in collaboration with Clover Biopharmaceuticals and Medicago, and Sanofi and Innovax, both of which are in Phase I clinical testing. GSK also presented its assistive technology to the Epidemic Preparedness Innovation Coalition, which helps fund companies like Moderna and Inovio. Dynavax adjuvant (CpG 1018),It consists of a 22-mer oligonucleotide sequence capable of stimulating the TLR9 pathogen recognition receptor for enhanced CD4 + and CD8 + responses as well as formation of B and T cell memory. This adjuvant has been used with the hepatitis B vaccine for enhanced antibody response. Dynavax has also been used with Clover and Medicago and Medigen It has partnered with Vaccine. It has just entered phase I clinical testing. Additionally, Dynavax has offered its adjuvant to Sinovac and Valneva, who remain in the preclinical stage.Sinovac and Valneva, who remained in the preclinical stage, presented the adjuvant.Sinovac and Valneva, who remained in the preclinical stage, presented the adjuvant.

As discussed in a previous section, nanoparticles offer opportunities for co-delivery of antigen and adjuvant to target lymph nodes and APCs, and a growing body of data shows that co-delivery of antigen and adjuvant improves the potency of vaccines at lower doses and reduces side effects. Pfizer claims that the mRNA in its vaccines acts as its own adjuvant, enabling co-delivery of antigen and adjuvant. Viral DNA, as well as viral vectors produced by Oxford/Astrazeneca and CanSino, can act as adjuvants. However, neither mRNA nor viral vector vaccines have not been successfully translated into the clinic, and it is clear that more research is needed to fully understand the potential and limitations of various nanotechnology platforms in vaccine development.


 

Conclusion

There are more than a hundred vaccines developed worldwide, and the race to become the first effective vaccine has fueled the rapid development of both preclinical and currently used vaccine approaches. There is no "one size fits all solution" as each vaccine strategy has both advantages and disadvantages. Leading company Moderna, BioNTech / Pfizer and Inovio produce nucleic acid-based vaccines, and early studies by Moderna and BioNTech / Pfizer have generated potent antibodies. Oxford / Astrazeneca and CanSino have similarly published appropriate early data, while ad-based vaccines continue to rank high. Gamaleya Research Institute and other state-run agencies and companies such as Sinopharm have further advanced into their respective Phases of clinical trials and have been granted emergency use authorization by their respective countries.Because so many vaccines are produced in so many different countries, there will potentially be more than one effective vaccine, and it is crucial that effective vaccines be distributed around the world to create a global herd immunity. It is really significant that the global effort to be made so quickly is of great importance. Success in this effort will not only lead to the end of the epidemic.It is truly remarkable that the global effort to produce and distribute it so quickly is done. Success in this effort will not only lead to an end to the epidemic.It is truly remarkable that the global effort to produce and distribute it so quickly is done. Success in this effort will not only lead to an end to the epidemic.