1,228
Views
0
CrossRef citations to date
0
Altmetric
Coronavirus

Clinical development of SpikoGen®, an Advax-CpG55.2 adjuvanted recombinant spike protein vaccine

ORCID Icon
Article: 2363016 | Received 01 Feb 2024, Accepted 29 May 2024, Published online: 05 Jun 2024

ABSTRACT

Recombinant protein vaccines represent a well-established, reliable and safe approach for pandemic vaccination. SpikoGen® is a recombinant spike protein trimer manufactured in insect cells and formulated with Advax-CpG55.2 adjuvant. In murine, hamster, ferret and non-human primate studies, SpikoGen® consistently provided protection against a range of SARS-CoV-2 variants. A pivotal Phase 3 placebo-controlled efficacy trial involving 16,876 participants confirmed the ability of SpikoGen® to prevent infection and severe disease caused by the virulent Delta strain. SpikoGen® subsequently received a marketing authorization from the Iranian FDA in early October 2021 for prevention of COVID-19 in adults. Following a successful pediatric study, its approval was extended to children 5 years and older. Eight million doses of SpikoGen® have been delivered, and a next-generation booster version is currently in development. This highlights the benefits of adjuvanted protein-based approaches which should not overlook when vaccine platforms are being selected for future pandemics.

KEYPOINTS

  • SpikoGen is a more traditional COVID-19 vaccine comprising SARS-CoV-2 spike protein extracellular domain formulated with Advax-CpG adjuvant

  • SpikoGen differs from the Novavax vaccine in major ways including its use of the soluble secreted spike protein ECD rather than nanoparticle formulation and the use of a different adjuvant

  • SpikoGen demonstrates robust protection against homologous and heterologous SARS-CoV-2 strains in hamster, ferret and non-human primate challenge models

  • SpikoGen induces broadly cross-neutralizing antibodies, but still protects even after these antibody levels wane

  • In a pivotal Phase 3 clinical trial, SpikoGen reduced the risk of severe infection by 77.5% and was not associated with myocarditis, thrombosis or any other adverse safety signals

  • SpikoGen received an Emergency Use Authorization in the Middle East on 6 October 2021, making it the first recombinant spike protein vaccine to achieve this milestone

  • Eight million doses of SpikoGen vaccine have been safely delivered to date

  • Protein-based vaccines have a long history of reliability and safety

Introduction

COVID-19 is caused by lung infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2).Citation1 Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a constellation of clinical outcomes from asymptomatic infection to respiratory failure and death.Citation2 The evolution of immune-escape SARS‑CoV‑2 variants continues to represent a global challengeCitation3 even though the pandemic phase has passed. While novel COVID-19 vaccines were rolled out with unprecedented speed, many challenges remain. New immune-escape variants continue to reinfect vaccinated as well as previously infected individuals. There remains a need for vaccines able to prevent SARS-CoV-2 infection and transmission, a weakness of most if not all current vaccines.Citation4

Now the pandemic phase has passed, it is a good time for reflection on what might have been done better with respect to the development and roll out of the COVID-19 vaccines.Citation5 This review specifically focuses on the experience with SpikoGen® vaccine as an example of one of the first successful protein-based COVID-19 vaccines. In particular, it seeks to answer the question of whether protein-based vaccines have been rendered obsolete by newer mRNA approaches. Certainly, during the early phase of the pandemic, protein-based vaccines were overlooked in favor of the newer mRNA and adenoviral vector approaches, but the reasons for this were financial and political rather than science based. Currently, large investments are being made in many countries for construction of mRNA manufacturing facilities but is this money well spent? How much of the mRNA euphoria is driven by reality and how much by hype?Citation6 Did protein-based vaccine approaches get a fair hearing in countries like the United States?

In addition to speed of development, an ideal pandemic vaccine technology should be low cost, scalable, temperature stable, well tolerated and safe, and provide robust and durable protection against both serious infection and transmission. Inactivated whole-virus COVID-19 vaccines were widely used in developing countries, with the main sources being China (Sinopharm) and India (Bharat Biotech).Citation7 These were shown to be safe and well tolerated, although their efficacy against infection proved to be modest, they did not prevent transmission, lacked durability and, required BSL3 manufacturing capability, which is extremely limited in most parts of the world. Other major COVID-19 vaccine approaches were mRNA or adenoviral vector based. However, this is not intended as a critical review of the mRNA or adenovirus vector platforms, which dominated Western markets during the early stages of the pandemic.Citation8 Unfortunately, adenovirus vector vaccines were associated with high reactogenicity and some lethal thrombotic events and have now been taken off the market.Citation9,Citation10 Similarly, mRNA vaccines were associated with high reactogenicity and inflammatory myocarditis,Citation11 provided relatively short-lived protection, required frequent boosters,Citation12 were expensive and were not stable except at very low temperatures. This indicates that these newer technologies are not ideal pandemic solutions.

Adjuvanted subunit vaccines presented an alternative well-established pandemic platform, but their potential was largely overlooked in Western countries during the pandemic. However, recombinant protein-based COVID-19 vaccines have now been licensed for human use in most countries. SpikoGen® vaccine was the first recombinant spike protein trimer-based vaccine to achieve regulatory licensure with emergency use approval by the Iranian FDA in early October 2021. This was just 18 months or so from the declaration of the pandemic by the World Health Organization (WHO). Other recombinant spike protein vaccines subsequently licensed include Novavax’s Nuvaxoid® vaccineCitation13 and Sanofi’s VidPrevtyn Beta vaccine.Citation14 Interestingly, all three of these recombinant vaccines are manufactured in insect cells using baculovirus transient infection, highlighting the strength of this platform for rapid and scalable recombinant protein production. A number of other protein-based receptor-binding domain (RBD) vaccines were licensed in Cuba, India and other countries.Citation15 These RBD-based vaccines induce antibodies that block RBD-ACE2 binding whereas the larger spike protein trimer vaccines, such as SpikoGen®, induce a much broader spectrum of antibodies against other important regions of the spike protein including the N-terminal domain (NTD), fusion peptide and stalk domain, all of which can help suppress virus infectivity.Citation16 This broader antibody repertoire induced by full-length spike protein versus the RBD could be of major importance for cross-protection against new variants as most immune-escape viral mutations are localized in the RBD region. The larger spike protein vaccines may also induce a broader diversity of T cell epitopes. During the pandemic, emergency use authorizations were expedited of a range of new vaccine technologies. This resulted in a massive global roll-out of novel mRNA and adenovirus vector vaccine technologies. Where does this leave the future of more traditional adjuvanted recombinant protein vaccine approaches? mRNA vaccines may be faster to produce than traditional protein-based vaccines,Citation17 but SpikoGen® generated its initial Phase 1 trial data in September 2020 just weeks behind the lead mRNA and adenoviral vector vaccines.Citation18 SpikoGen’s development was hampered by a lack of resources and funding, rather than any slowness of thetechnology itself. After completing its pivotal Phase 3 clinical trial, SpikoGen® received its first market authorization in Iran in early October 2021, just 10 months after the first mRNA and adenoviral vector vaccine approvals. This 10-month delay in time to market was driven by lack of resources and supply chain failures, and hence is not proof that protein expression being inherently slower than mRNA or adenoviral vector technologies. Arguably, given equivalent resources, recombinant protein vaccines could have been developed in a very similar time frame to the mRNA and adenoviral approaches.

Each vaccine technology presents its own challenges. Protein-based vaccines had additional challenges not faced by viral vector or mRNA vaccines, including the need to develop techniques for protein purification and to perform structural characterization of the expressed protein.Citation19 It is not possible to measure such parameters for mRNA or viral vectors which induce protein expression in vivo. Another challenge for recombinant protein-based approaches is low immunogenicity.Citation20 However, this can be overcome by addition of a suitable adjuvant. In recent years, multiple new adjuvants have been introduced alongside existing aluminum salt and oil emulsion adjuvants.Citation21 New adjuvants include ISS1018, a toll-like receptor (TLR)-9 agonist in Heplisav® hepatitis B vaccine,Citation22 Matrix M, a saponin-based adjuvant in Nuvaxoid COVID-19 vaccine,Citation23 the alum-CpG adjuvant in Corbevax® RBD vaccine,Citation24 Alhydroxyquim adjuvant in Bharat biotech’s inactivated COVID-19 vaccine approved in India,Citation25 ASO1B, an adjuvant containing a liposomal mixture of QS21 and monophosphoryl lipid A (MPL) in Shingrix® shingles vaccineCitation26 and Mosquirix® malaria vaccine,Citation27 AS04, an adjuvant comprising MPL absorbed to aluminum phosphate in Fendrix® hepatitis B vaccine,Citation28 and Vaxine’s own Advax-CpG55.2 adjuvant in SpikoGen® vaccine.

Advax-CpG55.2 is a proprietary adjuvant formulation combining Advax, particles of plant-derived delta-inulin polysaccharide, with CpG55.2 oligonucleotide, a potent human TLR-9 agonist. CpG55.2 is the first licensed human medicinal product designed by artificial intelligence.Citation29,Citation30 Advax-CpG adjuvant has been shown to enhance vaccine immunogenicity and protection across a broad range of animal modelsCitation31–34 including in vaccines against severe acute respiratory syndrome (SARS) coronavirus and the Middle East respiratory syndrome (MERS) coronavirus.Citation35 Advax-CpG adjuvanted SARS vaccine not only provided robust protection but also prevented vaccine-enhanced disease seen with other SARS vaccines.Citation36

SpikoGen® vaccine design

The key antigen component in SpikoGen® vaccine is a spike protein extracellular domain (ECD) antigen derived from the original Wuhan strain sequence. Despite the absence of the transmembrane domain it still self-assembles into a native trimer structure.Citation37 The SARS-CoV-2 genome sequence became available in NCBI (accession number: NC 045512) in mid-January 2020, allowing the structure of SARS-CoV to be used as a template to model the SARS-CoV-2 spike protein. We identified SARS-CoV-2 as having significant homology to SARS-CoV. We then performed structural homology modeling using the SARS-CoV structure (PDB-ID 6ACC) to obtain a 3D structure of SARS-CoV-2 spike protein. To help identify the putative cellular receptor for SARS-CoV-2, the crystal structure of human ACE2 was retrieved, and docked against human ACE2 protein. This predicted human ACE2 as the entry receptor for SARS-CoV-2 spike protein and confirmed the spike protein as an ideal target for vaccine design based on our experience with developing an effective vaccine against SARS CoV. Molecular dynamic simulations were performed on various modified spike protein ECD vaccine constructs to help select a suitable vaccine candidate while also being used to predict species susceptibility to SARS-CoV-2 infection.Citation38

SpikoGen® formulation and analytical testing

An insect cell codon-optimized expression cassette encoding the spike protein ECD with a His tag and stabilizing mutations was designed and cloned into pFASTBac1 to generate baculovirus following standard procedures.Citation32 Recombinant baculovirus was expanded in Sf9 insect cells to P3 and then used to infect Trichoplusia ni cells for protein expression. The recombinant spike protein ECD was purified by nickel affinity chromatography. Prior to the COVID-19 pandemic, Advax-CpG adjuvant had already been shown to be safe and effective, including in clinical trials of influenza [Clinicaltrials.gov; NCT03945825; NCT03038776] and hepatitis B [Clinicaltrials.gov; NCT01951677] vaccines. This made Advax-CpG a strong candidate for inclusion in our recombinant protein-based COVID-19 vaccine. Importantly, Advax-CpG adjuvant previously showed beneficial effects in vaccines against SARS and MERS coronaviruses.Citation35,Citation36 This animal immunogenicity data supported the decision to proceed with development of the recombinant spike protein ECD vaccine formulated with Advax-CpG55.2 adjuvant.Citation35,Citation36

In addition to a suitable adjuvant, many other challenges had to be solved when developing a recombinant protein-based vaccine. This included maximizing upstream protein yields while developing a downstream purification process that yielded a pure, stable and consistent protein product. Alongside optimization of the protein expression system, a large suite of analytical methods had to be rapidly developed for in-process testing and final product characterization and release.Citation39 Many methods for structure characterization, purification and quantification including ELISAs, and Western blots rely on availability of monoclonal antibodies. Such tools and reagents were not available at the start of the pandemic. Hence, the team needed to source and in some cases internally generate polyclonal and monoclonal antibodies needed for assay development. Another key reagent that needed to be internally generated was recombinant human ACE2 protein for use in developing a spike protein binding ELISA assay and a surrogate virus neutralization assay.Citation40 Spike protein pseudotyped lentivirus virus had to be produced for each of the emerging variants of concern for use in pseudotype virus neutralization assays for vaccine immunogenicity assessment as part of in vivo potency assays. Finally, a wide range of protein characterization tests including SDS gel electrophoresis, Western blots, size-exclusion chromatography, glycan composition analysis, glycan linkage-type analysis, peptide sequencing by mass spectroscopy, HDX-MS, negative stain EM, monoclonal antibody mapping, ACE2 binding, and accelerated stability studies, were needed to better understand the structure and behavior of the recombinant spike protein making up SpikoGen® vaccine.Citation39

SpikoGen® pre-clinical studies

The safety and immunogenicity of SpikoGen® vaccine was first assessed in BL6 and BALB/c mice. Immunization with Advax-CpG55.2-adjuvanted recombinant spike protein ECD induced spike-specific IgG and IgM antibodies with potent virus neutralization activity. Neutralization activity was measured by either surrogate virus neutralization assays based on inhibition of RBD binding to human ACE2, lentivirus pseudotype neutralization assays, or wildtype virus neutralization assays.Citation32 These assays confirmed that the recombinant spike ECD produced in T.ni insect cells when combined with Advax-CpG55.2 adjuvant was a good vaccine immunogen. The insect cell expression system enabled eukaryotic-like glycosylation of the spike protein thereby ensuring its structural integrity, whilst mitigating the risk of adventitious virus contamination associated with mammalian cell systems. It also provided good yields of protein at 10–40 mg/L thereby ensuring a competitive cost per dose and was able to be rapidly scaled.Citation41 Immunization with spike protein alone or mixed with alum adjuvant produced a Th2-biased immune response whereas spike protein formulated with Advax-CpG adjuvant induced a balanced T cell response characterized by both Th1 and Th2 cytokines.Citation42 This was considered important as both SARS CoV and SARS-CoV-2 viruses inhibit Th1 interferon pathways as a mechanism of immune escape.Citation43 Countering these viral effects by priming a strong memory Th1 and interferon response should be beneficial, with CD8+ T cells playing a critical role in silencing virus-infected cells.Citation44 Advax-CpG adjuvant induced a mixed Th1/Th2 response to SARS-CoV-2 recombinant spike protein in mice resulting in increased IgG1, IgG2 and IgG3 production and an increased frequency of IL-2, IFN-γ and TNF-α secreting T cells.Citation32 Sera from SpikoGen®-immunized mice cross-neutralized the B.1.1.7 (Alpha or UK) variant. The CpG55.2 component in SpikoGen® also induced cytotoxic T cells able to kill spike peptide-labeled target cells, in vivo.Citation32

SpikoGen® vaccine was next assessed for its ability to protect against SARS-CoV-2 infection in relevant animal models. The ferret challenge model was initially used as there was a high degree of uncertainty what might be the best animal model for the new SARS-CoV-2 virus. In the ferret model, two doses of SpikoGen® vaccine given intramuscularly several weeks apart provided complete lung protection and also completely stopped day 3 nasal virus shedding, an exciting finding as it suggested a possible impact on virus transmission.Citation32 Ferrets turned out to not be a good model of severe infection as they did not develop major clinical symptoms or weight loss when infected with SARS-CoV-2. SpikoGen® vaccine was next tested in the Golden Syrian hamster model which had advantages over ferrets as the SARS-CoV-2 virus replicates efficiently in naïve hamster lungs and the animals develop signs of clinical disease including weight loss and lung pathology.Citation45 To assess protection, hamsters were immunized intramuscularly with SpikoGen® vaccine, either once or twice 3 weeks apart. Sera were obtained 2 weeks after the final vaccination for measurement of neutralizing antibodies, and the hamsters were then challenged intranasally with a homologous wild-type SARS-CoV-2 virus isolate, USAWA1/2020. SpikoGen®-immunized hamsters achieved high serum neutralizing titers against the homologous SARS-CoV-2 virus. Naive control hamsters lost an average of 6.4–6.8% body weight during the first two days post-challenge, whereas SpikoGen®-immunized animals exhibited significantly reduced weight loss. There was a strong negative correlation (R = −0.8) between the serum neutralization titer, pre-challenge, and cumulative weight loss, post challenge. Both the dual- and single-dose vaccine regimens protected against lung infection and pathology, with a high correlation between serum neutralizing antibody levels and lung protection. Other studies have similarly shown a strong correlation between serum neutralizing antibody levels and protection against systemic disease.Citation46,Citation47 The viral load in lungs and nasal turbinate strongly correlated with weight-loss and lung pathology scores and serum neutralizing antibody levels correlated with reduced viral load in lung and nasal turbinates.Citation45 Interestingly, throat swab viral loads did not predict disease severity and showed a poor correlation with weight loss or lung scores. There was also a poor correlation between serum neutralizing antibody levels and throat viral loads. This suggests that serum antibody do not mediate mucosal protection but instead largely act to prevent the virus entering the lower respiratory tract.

SpikoGen® vaccine was also tested for safety and efficacy in the macaque non-human primate (NHP) model. As seen in hamster and ferret models, two doses of Advax-CpG adjuvanted spike protein vaccine induced robust serum neutralizing antibody levels against the homologous virus in the NHPs and protected against lung virus replication and lung pathology.Citation48 An interesting feature of this study was that the virus challenges were performed approximately 3 months after the second vaccine dose at a time when serum neutralizing antibody levels had fallen to undetectable levels in most of the immunized animals. Nevertheless, all the immunized animals, bar one, were completely protected against any lung virus replication. The one exception was an elderly female who was clinically well but showed a low level of lung virus day 3 post-challenge.

Preclinical studies in hamsters and NHP were also undertaken to assess the capacity of SpikoGen® vaccine, which was based on the original Wuhan spike protein, to protect against heterologous virus variants. The hamster study showed that two doses of SpikoGen® vaccine could protect hamsters against both the beta or delta variants, with the best protection been seen with a trivalent version of SpikoGen® that contained Wuhan, beta and gamma spike proteins.Citation49 This study also confirmed for the first time that SpikoGen® vaccine could prevent transmission from immunized animals to naïve co-housed sentinels. A more recent NHP study also showed that a bivalent SpikoGen® version containing Wuhan and Mu spike proteins, after three intramuscular doses, protected against the Omicron BA.5 variant, despite the animals having no detectable serum neutralizing activity against BA.5 pre-challenge.Citation50 The same study also showed that the original monovalent SpikoGen®vaccine when given as a course of two intramuscular doses followed by two oral doses protected against Omicron BA.5 infection. These animal data highlight that the original monovalent SpikoGen® vaccine, despite containing Wuhan spike protein, continues to provide cross-protection against newer Omicron strains such as BA.5. Given this protection was seen in the absence of BA.5 neutralizing antibodies, this suggests either that SpikoGen® vaccine is inducing protective non-neutralizing antibodies, or protecting by another mechanism such as induction of protective T cell responses.Citation51

A consistent finding across the animal studies was that SpikoGen® vaccine reduced nasal virus shedding and in hamster studies this was confirmed to translate to prevention of virus transmission from an infected host to a naïve co-housed sentinel.Citation49 SpikoGen® vaccine was extremely well tolerated in all tested species, with no major adverse events. Given its strong safety and efficacy profile in multiple species, SpikoGen® technology was selected by the Australian Zoos Association to immunize susceptible zoo species including lions, tigers, and non-human primates against COVID-19.Citation52 A bivalent version called SpikeVet™ that contained Wuhan and Mu spike protein CD with Advax-CpG55.2 adjuvant was administered as a course of two or three immunizations, with 867 vaccine doses being administered to 354 zoo animals, drawn from 38 species in total, thereby potentially representing perhaps the largest OneHealth pandemic vaccine studies ever attempted. SpikeVet™ was found to have an excellent safety profile in the zoo species tested with the low rate of minor adverse effects and the absence of major adverse events. The types of local and systemic minor adverse effects observed in zoo animals were broadly comparable to those reported for other protein-based vaccines in humans.Citation53

SpikoGen® human clinical trials

An Australian first-in-man Phase 1 clinical trial of Covax-19®, the earliest version of SpikoGen®, was conducted in 40 adult participants commencing in mid-2020, mere months into the pandemic (clinicaltrials.gov; NCT04453852). This confirmed the safety of the adjuvanted recombinant spike protein vaccine platform. Mainly mild local and systemic adverse events were short-lived and commensurate with the side effects of existing licensed vaccines.Citation54 Next, a randomized, placebo-controlled, double-blind Phase 2 trial was conducted in the Middle East on 400 adult participants randomized 3:1 to receive two doses of 25 µg SpikoGen® 3 weeks apart or saline placebo (clinicaltrials.gov; NCT04944368). SpikoGen® vaccine induced humoral and T cell responses against spike protein in the majority of participants and was not associated with any adverse safety signals.Citation18 The most common solicited adverse events were injection site pain, fatigue and headache, and these were predominantly graded as mild and transient. Two weeks post the second vaccine dose, 87% of immunized participant sera neutralized SARS-CoV-2 virus at titers > 1:32. Baseline seronegative subjects achieved neutralizing antibody levels post 2-doses that were 3.3-fold higher than levels in convalescent infected individuals. The SpikoGen® group, 2 weeks post the second dose, exhibited increased antigen-stimulated T cell proliferation and interferon gamma production. Baseline seropositive participants showed a large amnestic SARS-CoV-2 antibody response after just a single SpikoGen® dose, consistent with having B cell memory due to previous infection. Based on the positive Phase 2 safety and immunogenicity results, SpikoGen® vaccine was advanced into a pivotal Phase 3 trial conducted as a placebo-controlled, double-blind trial in participants aged 18 to 50 years.Citation18 The primary endpoint was efficacy in preventing symptomatic PCR-confirmed SARS-CoV-2 infection, with a total of 16,876 participants undergoing randomization on 3:1 basis to receive two intramuscular doses 3 weeks apart of SpikoGen® (25 mcg) or saline placebo. A single interim analysis was pre-specified upon accrual of a minimum of 74 primary endpoints, representing 50% of the 147 endpoints planned for a final analysis. The vaccine safety data in the Phase 3 trial mimicked that of the Phase 1 and 2 trials and was characterized by transient mild-to-moderate injection site pain, fatigue, and headache. No myocarditis, thrombosis or other vaccine-related safety concerns were identified. A total of 5 study participants in the vaccine group reported pregnancy during the study period with no adverse outcomes being reported. At the time of the pe-specified interim analysis, in the combined per-protocol plus nuclear antibody positive populations, 96 infections had occurred at least 14 days after the second dose, with 52/9246 (0.56%) in the SpikoGen® group and 44/3014 (1.46%) in the placebo group, translating to a vaccine efficacy (VE) against infection of 64.36% [95% CI 46.54 to 76.11]. In the per-protocol participants (serum nuclear antibody negative at baseline), infection occurred in 50/8100 (0.62%) of the SpikoGen® group and 37/2512 (1.47%) in the placebo group for a VE of 59.69%, [95% CI; 37.95 to 73.57]. This exceeded the benchmark minimum criteria set by the WHO which stipulated a clear demonstration of efficacy, ideally with a VE point estimate around 50% and the lower bounds of the 95% confidence interval exceeding 30%.Citation55 The final trial design was performed in consultation with the Iranian FDA who did not request inclusion of an adjusted confidence interval for VE according to the alpha spending function.Citation56 At the time of the single interim analysis, 96 infections had already occurred, which was 65.3% of the target total, thereby reducing the risk of a type 1 error despite lack of adjustment for this interim analysis. Of 50 trial infection virus isolates sequenced, all were confirmed as the B.1.617.2 (delta) variant. On the basis of this data, SpikoGen® received an emergency use authorization (EUA) from the Iranian FDA on 6 October 2021 for use in prevention of COVID-19 in adults. Following the EUA, a subsequent post-hoc final analysis was performed with censoring of data at 100 days post-randomization. Efficacy against severe COVID-19 disease remained high at 77.5%, although the efficacy against symptomatic infection in the per-protocol population had reduced to 43.99% (95% CI: 30.30–55.00) at the time of this second analysis. This waning over time of vaccine protection against symptomatic infection was also seen with the other COVID-19 vaccines.Citation57 Other potential confounders may have contributed to the lower VE against symptomatic infection seen at the time of the second posthoc analysis. As other approved vaccines were available in the latter stages of the Phase 3 trial, a higher proportion of participants in the placebo control arm requested unblinding and dropped out to receive other vaccines, thereby creating imbalances between the two groups. We speculate that the use of a saline placebo may have led to a degree of unblinding due to its lack of injection site reactogenicity, thereby enabling participants to distinguish whether they had had a placebo. The Oxford adenoviral vector vaccine trials were amongst the few to use an active vaccine comparator, meningococcal vaccine.Citation58 Another factor potentially contributing to the lower efficacy on the second post hoc analysis was that VE is liable to be underestimated during periods of extremely high community infection ratesCitation59 when individuals are repeatedly exposed to high levels of virus. Notably, the delta variant infection rates in the SpikoGen® Phase 3 trial were exceptionally high at 313.35 per 1000 person/yr in the placebo group as compared to much lower infection rates reported in Phase 3 studies of the mRNACitation60 and adenoviral vector vaccines,Citation58 which were less than half this rate. Hence, high community infection rates with delta virus could have reduced estimates of VE during SpikoGen’s Phase 3 trial, by making vaccine breakthrough infections more likely. As delta was the only strain found present during the conduct of the SpikoGen® Phase 3 trial, data on SpikoGen® efficacy against the ancestral Wuhan and alpha strains were not able to be assessed, although animal challenge data had shown strong protection against the ancestral strains such as Wuhan.Citation32 Furthermore, algorithms based on spike-binding IgG and virus neutralization antibody levels were used to predict SpikoGen® efficacy against ancestral strains.Citation61 These predicted SpikoGen’s efficacy at protecting against the ancestral strain in the range of 85–95%Citation18 Elderly subjects were excluded from the Phase 3 trial as the IFDA did not agree to enrolling participants older than 50 years into a placebo-controlled study at a time when other COVID-19 vaccines were becoming available. This meant that no direct efficacy data is available for SpikoGen® in the elderly. However, SpikoGen® trials that have included older subjects have found no significant correlation between age and serum neutralizing antibody levels (manuscript in preparation). This suggests VE should be preserved in the elderly, even although direct data on this are not available.

Study limitations

It would be particularly interesting to have trial data comparing SpikoGen® vaccine with other protein-based vaccines such as those from Novavax from the United States, Sanofi in France and ZF2001 RBD vaccine from China. Unfortunately, it has not been possible to collect real-world data on SpikoGen’s effectiveness outside of clinical trials, due to the challenges of collecting population data in Iran, the only country where SpikoGen has been in widespread population use. It is not possible to directly compare SpikoGen’s efficacy results with those from other vaccine trials, given differences in populations, endpoints, and circulating virus strains, amongst other differences. Notably, SpikoGen® vaccine efficacy was measured against a major wave of delta variant in the Middle East whereas the Phase 3 trials for the mRNA and adenoviral vector vaccines were mostly conducted prior to arrival of the delta variant. Almost all vaccines showed reduced effectiveness against the delta variant as compared to ancestral strains. For example, a test negative case-control study showed 2-dose effectiveness of AstraZeneca adenoviral vaccine to be only 59.8% against B.1.617.2 (delta) versus 66.1% for the earlier B.1.1.7 (alpha) strain.Citation62 Similarly, a US Veterans Affairs study showed VE against delta of just 13.1% for the Janssen single-dose vaccine, 43.3% for Pfizer, and 58% for Moderna, versus effectiveness against earlier strains (predominantly alpha) of 86.4%, 86.9% and 89.2%, respectively.Citation62

A primary course of SpikoGen® vaccine initially comprised two doses given 3 weeks apart. In retrospect, even greater protection might have been achieved with a 3-dose course. This was shown in a recent NHP study where three doses of SpikoGen® vaccine protected against a widely heterologous Omicron BA.5 infection. At the time of the SpikoGen® phase 3 trial, regulatory bodies including WHO had published target product profiles for COVID-19 vaccines that specified a maximum of two doses as the minimal acceptable criteria. This created a major bias against protein-based vaccines as it is not unusual for three or even more primary doses of protein vaccines to be needed to establish maximal protection. For example, pediatric hepatitis B or hemophilus influenza B vaccines recommend three or four doses as the primary course.Citation63 In retrospect, these minimum acceptable criteria for COVID-19 were overly restrictive, as protection against infection by two doses of any of the COVID-19 vaccines, including adenovirus and mRNA vaccines, waned rapidly and was inadequate to protect against the newly emerging variants such as Omicron. This led to the introduction of 3rd and then 4th dose boosters within months of each other to try and restore vaccine protection against the Omicron strains. Given this extremely short-time interval between doses, these 3rd and 4th doses should in retrospect be considered part of a primary series, meaning that none of the vaccines that received EUA’s would have met WHO’s minimum acceptable criteria. This is an important point as it would be a grave mistake if similar restrictive minimum acceptability criteria were set for future pandemic vaccines.

Booster doses

A 3rd dose booster study was undertaken with SpikoGen® vaccine in adult subjects aged 18–90 years who had received a primary immunization with a primary course of any COVID-19 vaccine, at least four or more months previously (clinicaltrials.com; NCT05175625). This double-blind and randomized placebo-controlled study was initiated in December 2021 and enrolled 300 previously immunized adult participants, drawn from groups who had previously received either inactivated whole virus, viral vector or recombinant protein (SpikoGen®) COVID-19 vaccines. Participants were randomized on a 3:1 basis to receive a single intramuscular booster dose of SpikoGen® vaccine or saline and then had serum antibody levels measured 14 days after the booster dose. Interestingly, despite the participants having been vaccinated just 4 or more months previously, regardless of what type of vaccine they had received, ~50% already had very low spike antibody levels consistent with waning immunity.Citation64 In response to the SpikoGen® booster dose, 76% of participants seroconverted by surrogate virus neutralization test (sVNT) against the ancestral Wuhan strain, versus just 3% of the placebo group, where seroconversion was defined as a change in the status of antibody levels from negative to positive based on the prespecified commercial ELISA kit threshold. The inactivated virus-primed group had the highest 25.8-fold rise in sVNT with a 14.3-fold rise in the viral vector-primed group and an 11.3-fold rise in the SpikoGen®-primed group. There were no significant differences in mean sVNT titers between the different primary vaccine groups post the booster dose. Notably, the SpikoGen® booster dose induced broad cross-neutralization of the major SARS-CoV-2 variants of concern, including a 13- to 18-fold rise in serum neutralization of the Omicron BA.1 variant. This indicated a 3-dose primary course of SpikoGen® was needed for optimal for maximum protection against Omicron strains. Just why a 3rd SpikoGen® dose was so critical to the cross-neutralizing antibody response is not known, but similar effects of broader responses after a 3rd dose were reported for other vaccines, suggesting a generalized phenomena.Citation65 We theorize that three rounds of vaccine stimulation are required to generate immune responses against subdominant epitopes in the spike protein that are highly conserved amongst the variants. This would then explain why three doses of COVID-19 vaccine are so important to get broad cross-neutralizing responses.

The SpikoGen® booster dose was licensed by the Iranian FDA in January 2022. The optimal timing of the 3rd dose is still not known, particularly for new vaccine-resistant variants such as Omicron, to which vaccine immunity wanes faster than against the ancestral strains.Citation66 A recently completed Australian clinical trial tested the effect of different dose intervals and the results are still being analyzed (ClinicalTrials.gov: NCT05279456). As mentioned above, the response to the 3rd dose was particularly robust for those who had had an inactivated vaccine-prime with this group having a 25.8-fold increase in sVNT from baseline levels. Inactivated vaccines typically present only low and variable amounts of spike protein to the immune system due to the low ratio of spike protein to total virus protein content in the inactivated vaccine, and because some cleavage of S1 from S2 occurs during the beta-propiolactone inactivation process.Citation67 Hence, we hypothesize that in individuals primed with inactivated or adenoviral vector vaccines, exposure to the much larger dose of spike protein in the SpikoGen® 3rd dose may help stimulate a strong recall response in spike-specific memory B cells that were only weakly induced by the primary course of inactivated vaccine. In addition, boosting with a heterologous vaccine often gives a stronger response than boosting with a homologous vaccine.Citation68 The Advax-CpG adjuvant in the SpikoGen® booster may also contribute to the strong 3rd dose response by stimulating more broadly cross-neutralizing antibodies. For example, mice immunized with inactivated Japanese encephalitis virus (JEV) adjuvanted with Advax-CpG were cross-protected against lethal infection with West Nile virus infection.Citation69 Sera from these JEV+Advax-CpG immunized mice was able to cross-neutralize a broad repertoire of flaviviruses, including West Nile virus, Murray Valley encephalitis virus, St Louis encephalitis virus and dengue viruses.Citation70

Special populations

Another important population who need protection during a pandemic is immune-suppressed patients. A single-arm trial of SpikoGen® as a booster dose was performed with 43 kidney transplant patients who had had primary vaccination with 2 doses of Sinopharm inactivated COVID-19 vaccine 1–3 months previously.Citation71 SpikoGen® induced positive humoral and cellular responses regardless of prior seropositive or seronegative status of the kidney transplant patients, with seroconversion rates 30 days after the booster dose of 35.3% to anti-S1 binding IgG and 29.4% to surrogate neutralizing antibodies. The most common solicited adverse events were injection site pain and fatigue, which were predominantly mild and transient. No serious adverse events were recorded. These results supported the suitability of SpikoGen® for immune suppressed patients.

Another recently completed pediatric clinical trial compared the immunogenicity of SpikoGen® vaccine in children 5 years and older to young adults (ClinicalTrials.gov: NCT05231590). This showed that a half-antigen dose of SpikoGen® vaccine in children aged 5–11 years gave equivalent immunogenicity and safety to a full antigen dose in children aged 12–18 years and young adults. SpikoGen® vaccine was subsequently licensed by the Iranian FDA for use in children 5 years and older in Jan 2022.Citation72

Ongoing development of SpikoGen® platform

Since the first launch of SpikoGen® vaccine 8 million doses have been delivered in the Middle East. The clinical trials and ongoing pharmacovigilance have not revealed any unexpected serious vaccine-associated adverse events. In particular, SpikoGen® vaccine has not been associated with myocarditis/pericarditisCitation73 or central venous thrombosis,Citation74 as have occurred after other COVID-19 vaccines. For convenience and speed of protection, the initial 2-dose primary course of SpikoGen® was administered just 3 weeks apart, which may not be optimal for maximum vaccine immunogenicity. For example, the adenoviral vector vaccines were shown to be more protective if the second dose was given 12 weeks after the first dose.Citation75 In retrospect, a 3rd dose could have been built into the primary vaccine course for maximal efficacy against variants. The effect of changing the dosing interval of SpikoGen® is being studied in two Australian clinical trials, the first looking at the effects of varying the dose interval between the first 2 doses from 3 to 6 weeks (Clinicaltrial.gov; NCT05148871). A second trial is comparing administration of the 3rd dose either 1 or 3 months after the 2nd dose (ClinicalTrials.gov; NCT05279456). Yet a further Australian clinical trial is examining the effect of using SpikoGen® as a booster dose for individuals who have received two or more prior doses of any COVID-19 vaccine type, including mRNA or adenoviral vector vaccines (ClinicalTrials.gov: NCT05542862).

Since the initial clinical trials and licensure of SpikoGen® vaccine, SARS-CoV-2 has continued to mutate at a rapid pace, first into the original Omicron BA.1 to BA.5 strains and then into recombinant lineages such as XBB.1 and more recently into variants such as BA.2.86 (Pirola).Citation76 Each new variant has been more highly transmissible and more vaccine-resistant than the previous generation. Bivalent versions of the mRNA vaccines were released to the market, containing a mix of mRNA encoding for the original Wuhan and BA.1 (Australian market) or BA.5 (US market) spike proteins to try and compensate for poor efficacy of the original mRNA vaccines. While these showed modest short-term efficacy against the homologous BA.4/5 strains, they were not shown to be effective against the XBB lineage.Citation77 The bivalent mRNA vaccines have already been replaced by monovalent Omicron XBB.1.5 vaccines, although the effectiveness of these against new Pirola subvariants such as JN.1 is unknown.

The continual generation of SARS-CoV-2 immune-escape variants thereby presents an ongoing major challenge for all COVID-19 vaccines. As described above, multivalent (bi-, tri-, quadra- and even penta-valent) formulations of SpikoGen® vaccine that include multiple-spike proteins have been produced and shown immunogenic and protective in animal studies. In addition, updated monovalent SpikoGen® boosters targeting BA.2.86 are currently being developed. Parenterally administered vaccines while effective in preventing lung virus replication and severe disease, have proved less effective at preventing initial infection and have not stopped virus transmission,Citation78 SpikoGen® is a possible exception, having been shown to block virus transmission in the hamster model.Citation49 SpikoGen® has recently been tested as a mucosal booster delivered via either oral or intrapulmonary routes in NHP.Citation50 While nasal administration is another alternative route for mucosal immunization, we chose oral administration because of its convenience (it does not require a specialized delivery device), and because we speculate that priming of gut associated lymphoid tissue (GALT) may be beneficial in control of SARS-CoV-2 infection. The results suggested that the oral booster might indeed provide enhanced protection against a heterologous Omicron BA.5 challenge. The effect of an oral SpikoGen® booster alone or in combination with a quadrivalent seasonal influenza vaccine is to be tested in an Australian clinical trial (clinicaltrial.gov NCT06355232).

Conclusions

The adenoviral vector vaccines have been largely discontinued from use due to major safety concerns including deaths from thromboembolic disease, as well as weak efficacy. While most attention remains on the mRNA vaccines, adjuvanted recombinant protein vaccines such as SpikoGen® offer a suitable alternative, with lower reactogenicity and better safety. As yet, there is very little head-to-head data comparing the efficacy of booster doses of mRNA and recombinant protein COVID-19 vaccines, with most comparison studies just based on serum antibody levels. Notably, absence of serum neutralizing antibody in SpikoGen®-vaccinated animals did not mean they were not protected.Citation50 Conversely, we have seen individuals with high serum antibody levels after mRNA vaccination still develop SARS-CoV-2 infection. A Danish registry study concluded “quantitative levels of anti-spike IgG have limited impact on the risk of breakthrough infection with Omicron.”Citation79 Hence, any head-to-head comparative efficacy studies of different COVID-19 vaccines would need to compare clinical endpoints such as disease severity rather than just compare serum antibody surrogates. Furthermore, there is the possibility that the kinetics and patterns of protection may be different between recombinant protein and mRNA vaccines. mRNA vaccines appear to provide a short sharp burst in antibodies with associated short-term reductions in infection risk that then rapidly fade away. By contrast, adjuvanted protein vaccines may provide more durable protection despite inducing lower antibody titers, particularly if they facilitate early mild infections that could then provide more robust hybrid immunity going forward.Citation80 This could present a paradox where the apparently weaker vaccine that allows an early mild infection might provide better long-term outcomes than a initially more potent vaccine that prevents infection short-term but which leaves the individual vulnerable to repeated infections when the initial protection rapidly wanes. This could explain recent data showing that the more doses of mRNA vaccine an individual received, the greater their risk of experiencing a SARS-CoV-2 infection.Citation81

Long-term safety is another consideration when assessing the relative merits of recombinant protein versus mRNA vaccines, as the long-term effects of repeated doses of mRNA remain unknown. By contrast, repeated doses of protein-based vaccines such as influenza vaccines are known to be able to be given annually for many years with no cumulative adverse effects.Citation82 The last four years have seen a battle between reliable protein-based technologies with well-understood safety and mechanism of action versus newer mRNA and viral vector approaches that continue to have uncertainty around their mechanisms of action and long-term effects. The vaccine industry has traditionally been extremely conservative. In a twist of fate, the inability of large vaccine companies to commit to ultrashort pandemic vaccine delivery timelines demanded by politicians meant that the US WarpSpeed and UK government funding initiatives instead directed vast sums to accelerate development of nascent technologies, in particular mRNA and adenoviral vectors, based on their promises of speedy delivery. Only time will tell whether this was the right move. One might ask whether regulators are now prepared to routinely accept any new vaccine technology with minimal animal testing before permitting its use in billions of people or whether this was just a pandemic anomaly.Citation73 We still do not know the significance of mRNA vaccines increasing IgG4 levelsCitation83 and what might this mean for the effectiveness of mRNA in the longer-term.Citation84 Only time will ultimately tell. SpikoGen® represented a remarkable achievement given the extremely limited resources available to the project team. SpikoGen® may be the only COVID-19 vaccine that was successfully developed without major government or instirutional support evertheless, it was delivered in world record time for a recombinant protein vaccine. Traditional protein-based vaccines have stood the test of time and have saved countless hundreds of millions of lives. Whether mRNA vaccines can achieve the same going forward is still not clear. In the meantime, it would be wise for funders to continue to support protein-based vaccine development and not neglect this area due to excess mRNA vaccine euphoria.

Institutional Review Board Statement

All described clinical studies were conducted according to the guidelines of the Declaration of Helsinki and approved by the appropriate Institutional Review Boards. Informed consent was obtained from all subjects involved in clinical studies.

Acknowledgments

Thanks are due to the many clinical trial participants whose contributions were crucial. Thanks are also due to the CinnaGen and Vaxine staff working on the project. At Vaxine, we particularly thank Lei Li, Yoshikazu Honda-Okubo, Isaac Sakala and Greiciely Andre. These staff all worked tirelessly under difficult circumstances during the pandemic to make SpikoGen® a reality. Development of Advax-CpG55.2 adjuvant and animal studies was supported by funding from National Institute of Allergy and Infectious Diseases of the National Institutes of Health under Contracts HHS-N272201400053C, HHSN272201800044C, and HHSN272201800024C, the MTPConnect Biomedical Translation Bridge Program, a Fast Grant administered by George Mason University, Macrovax Trust and Bridgenorth Pastoral Co. Funding of clinical trials was provided by Vaxine Pty Ltd and Cinnagen Co.

Disclosure statement

The author is affiliated with Vaxine Pty Ltd which holds the rights to COVAX-19®/Spikogen® vaccine and Advax™ and CpG55.2™ adjuvants. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Additional information

Funding

The work was supported by the National Institute of Allergy and Infectious Diseases, National Institutes of Health [5U01AI061142]; [HHS272200800039C], [HHS-N272201400053C], [HHSN272201800044C], [HHSN272201800024C], MTPConnect Biomedical Translation Bridge Program, George Mason University Fast Grant, Macrovax Trust, Bridgenorth Pastoral Co, Vaxine Pty Ltd and Cinnagen Co.

References

  • Hu B, Guo H, Zhou P, Shi Z-L. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol. 2020;19(3):1–11. doi:10.1038/s41579-020-00459-7.
  • Gu J, Korteweg C. Pathology and pathogenesis of severe acute respiratory syndrome. Am J Pathol. 2007;170(4):1136–47. doi:10.2353/ajpath.2007.061088.
  • Jeworowski LM, Muhlemann B, Walper F, Schmidt ML, Jansen J, Krumbholz A, Simon-Lorière E, Jones TC, Corman VM, Drosten C. Humoral immune escape by current SARS-CoV-2 variants BA.2.86 and JN.1, December 2023. Euro Surveill. 2024;29(2). doi:10.2807/1560-7917.ES.2024.29.2.2300740.
  • Morens DM, Taubenberger JK, Fauci AS. Rethinking next-generation vaccines for coronaviruses, influenzaviruses, and other respiratory viruses. Cell Host Microbe. 2023;31(1):146–57. doi:10.1016/j.chom.2022.11.016.
  • Fauci AS, Folkers GK. Pandemic preparedness and response: lessons from COVID-19. J Infect Dis. 2023;228(4):422–5. doi:10.1093/infdis/jiad095.
  • Brown BD, Fauci AS, Belkaid Y, Merad M. RNA vaccines: a transformational advance. Immunity. 2023;56(12):2665–9. doi:10.1016/j.immuni.2023.11.009.
  • Hu L, Sun J, Wang Y, Tan D, Cao Z, Gao L, Guan Y, Jia X, Mao J. A review of inactivated COVID-19 vaccine development in China: focusing on safety and efficacy in special populations. Vaccines. 2023;11(6):11. doi:10.3390/vaccines11061045.
  • Heinz FX, Stiasny K. Distinguishing features of current COVID-19 vaccines: knowns and unknowns of antigen presentation and modes of action. NPJ Vaccines. 2021;6(1):104. doi:10.1038/s41541-021-00369-6.
  • Cervantes-Torres J, Cabello-Gutierrez C, Ayon-Nunez DA, Soldevila G, Olguin-Alor R, Diaz G, Acero G, Segura-Velázquez R, Huerta L, Gracia-Mora I. Caveats of chimpanzee ChAdOx1 adenovirus-vectored vaccines to boost anti-SARS-CoV-2 protective immunity in mice. Appl Microbiol Biotechnol. 2024;108(1):179. doi:10.1007/s00253-023-12927-0.
  • Clerici B, Pontisso E, Aloise C, Peroni B, Perricone R, Pisetta C, Scavone M, Birocchi S, Podda GM. Thrombosis and bleeding in patients with vaccine-induced immune thrombotic thrombocytopenia: a systematic review of published cases. Thromb Haemost. 2023;124(5):423–31. doi:10.1055/s-0043-1777134.
  • Jaiswal V, Mukherjee D, Peng Ang S, Kainth T, Naz S, Babu Shrestha A, Agrawal V, Mitra S, Ee Chia J, Jilma B. COVID-19 vaccine-associated myocarditis: analysis of the suspected cases reported to the EudraVigilance and a systematic review of the published literature. Int J Cardiol Heart Vasc. 2023;49:101280. doi:10.1016/j.ijcha.2023.101280.
  • Echaide M, Chocarro de Erauso L, Bocanegra A, Blanco E, Kochan G, Escors D. Escors D. mRNA vaccines against SARS-CoV-2: advantages and caveats. Int J Mol Sci. 2023;24(6):24. doi:10.3390/ijms24065944.
  • Mateo-Urdiales A, Sacco C, Petrone D, Bella A, Riccardo F, Del Manso M, Bressi M, Siddu A, Brusaferro S, Palamara AT. Estimated effectiveness of a primary cycle of protein recombinant vaccine NVX-CoV2373 against COVID-19. JAMA Netw Open. 2023;6(10):e2336854. doi:10.1001/jamanetworkopen.2023.36854.
  • Dayan GH, Rouphael N, Walsh SR, Chen A, Grunenberg N, Allen M, Antony J, Asante KP, Bhate AS, Beresnev T. Efficacy of a bivalent (D614 + B.1.351) SARS-CoV-2 recombinant protein vaccine with AS03 adjuvant in adults: a phase 3, parallel, randomised, modified double-blind, placebo-controlled trial. Lancet Respir Med. 2023;11(11):975–90. doi:10.1016/S2213-2600(23)00263-1.
  • Yang J, Wang W, Chen Z, Lu S, Yang F, Bi Z, Bao L, Mo F, Li X, Huang Y. A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity. Nature. 2020;586(7830):572–7. doi:10.1038/s41586-020-2599-8.
  • Xia X. Domains and functions of spike protein in Sars-Cov-2 in the context of vaccine design. Viruses. 2021;13(1):109. doi:10.3390/v13010109.
  • Hogan MJ, Pardi N. mRNA vaccines in the COVID-19 pandemic and beyond. Annu Rev Med. 2022;73(1):17–39. doi:10.1146/annurev-med-042420-112725.
  • Tabarsi P, Anjidani N, Shahpari R, Mardani M, Sabzvari A, Yazdani B, Roshanzamir K, Bayatani B, Taheri A, Petrovsky N. Safety and immunogenicity of SpikoGen®, an Advax-CpG55.2-adjuvanted SARS-CoV-2 spike protein vaccine: a phase 2 randomized placebo-controlled trial in both seropositive and seronegative populations. Clin Microbiol Infect. 2022;28(9):1263–71. doi:10.1016/j.cmi.2022.04.004.
  • Bruch EM, Zhu S, Szymkowicz L, Blake T, Kiss T, James DA, Rak A, Narayan K, Balmer MT, Chicz RM. Structural and biochemical rationale for Beta variant protein booster vaccine broad cross-neutralization of SARS-CoV-2. Sci Rep. 2024;14(1):2038. doi:10.1038/s41598-024-52499-1.
  • Pollet J, Chen W-H, Strych U. Recombinant protein vaccines, a proven approach against coronavirus pandemics. Adv Drug Deliv Rev. 2021;170:71–82. doi:10.1016/j.addr.2021.01.001.
  • Petrovsky N, Aguilar JC. Vaccine adjuvants: current state and future trends. Immunol Cell Biol. 2004;82(5):488–96. doi:10.1111/j.0818-9641.2004.01272.x.
  • Girndt M, Houser P, Manllo-Karim R, Ervin JE, Charytan C, Chow S, Symonian-Silver M, Lehrner L, Linfert D, Shemin D. Long-term immunogenicity and safety of the hepatitis B vaccine HepB-CpG (HEPLISAV-B) compared with HepB-Eng (Engerix-B) in adults with chronic kidney disease. Vaccine. 2023;41(20):3224–32. doi:10.1016/j.vaccine.2023.04.028.
  • Stertman L, Palm AE, Zarnegar B, Carow B, Lunderius Andersson C, Magnusson SE, Carnrot C, Shinde V, Smith G, Glenn G. The matrix-M™ adjuvant: a critical component of vaccines for the 21 st century. Hum Vaccin Immunother. 2023;19(1):2189885. doi:10.1080/21645515.2023.2189885.
  • Thuluva S, Paradkar V, Gunneri S, Yerroju V, Mogulla R, Suneetha PV, Turaga K, Kyasani M, Manoharan SK, Adabala S. Immunogenicity and safety of Biological E’s CORBEVAX™ vaccine compared to COVISHIELD™ (ChAdox1 nCoV-19) vaccine studied in a phase-3, single blind, multicentre, randomized clinical trial. Hum Vaccin Immunother. 2023;19(1):2203632. doi:10.1080/21645515.2023.2203632.
  • Counoupas C, Pino P, Stella AO, Ashley C, Lukeman H, Bhattacharyya ND, Tada T, Anchisi S, Metayer C, Martinis J. High-titer neutralizing antibodies against the SARS-CoV-2 delta variant induced by alhydroxyquim-II-adjuvanted trimeric spike antigens. Microbiol Spectr. 2022;10(1):e0169521. doi:10.1128/spectrum.01695-21.
  • Chlibek R, Bayas JM, Collins H, de la Pinta ML, Ledent E, Mols JF, Heineman TC. Safety and immunogenicity of an AS01-adjuvanted varicella-zoster virus subunit candidate vaccine against herpes zoster in adults ≥50 years of age. J Infect Dis. 2013;208(12):1953–61. doi:10.1093/infdis/jit365.
  • Rts SY. RTS,S/AS01 malaria vaccine (Mosquirix®): a profile of its use. Drugs Ther Perspect. 2022;38(9):373–81. doi:10.1007/s40267-022-00937-3.
  • Girndt M, Pluer M, Dellanna F, Michelsen AK, Beige J, Toussaint K, Wehweck HJ, Koch M, Hafezi Rachti S, Faust J. Immunogenicity and safety of a booster dose of the hepatitis B vaccine HepB-CpG (HEPLISAV-B®) compared with HepB-Eng (Engerix-B®) and HepB-AS04 (Fendrix®) in adults receiving hemodialysis who previously received hepatitis B vaccination and are not seroprotected: results of a randomized, multicenter phase 3 study. Hum Vaccin Immunother. 2022;18(6):2136912. doi:10.1080/21645515.2022.2136912.
  • Petrovsky N, Cooper PD. Advax™, a novel microcrystalline polysaccharide particle engineered from delta inulin, provides robust adjuvant potency together with tolerability and safety. Vaccine. 2015;33(44):5920–6. doi:10.1016/j.vaccine.2015.09.030.
  • Thomas S, Abraham A, Baldwin J, Piplani S, Petrovsky N. Artificial Intelligence in Vaccine and Drug Design. Vaccine Design: Springer; 2022. p. 131–46.
  • Görander S, Honda-Okubo Y, Bäckström M, Baldwin J, Bergström T, Petrovsky N, Liljeqvist J-Å. A truncated glycoprotein G vaccine formulated with Advax-CpG adjuvant provides protection of mice against genital herpes simplex virus 2 infection. Vaccine. 2021;39(40):5866–75. doi:10.1016/j.vaccine.2021.08.050.
  • Sakala IG, Honda-Okubo Y, Li L, Baldwin J, Petrovsky N. A M2 protein-based universal influenza vaccine containing Advax-SM adjuvant provides newborn protection via maternal or neonatal immunization. Vaccine. 2021;39(36):5162–72. doi:10.1016/j.vaccine.2021.07.037.
  • Honda-Okubo Y, Baldwin J, Petrovsky N. Advax-CpG adjuvant provides antigen dose-sparing and enhanced immunogenicity for inactivated poliomyelitis virus vaccines. Pathogens. 2021;10(5):10. doi:10.3390/pathogens10050500.
  • Eichinger KM, Kosanovich JL, Gidwani SV, Zomback A, Lipp MA, Perkins TN, Oury TD, Petrovsky N, Marshall CP, Yondola MA. Prefusion RSV F immunization elicits Th2-mediated lung pathology in mice when formulated with a Th2 (but not a Th1/Th2-balanced) adjuvant despite complete viral protection. Front Immunol. 2020;11:1673. doi:10.3389/fimmu.2020.01673.
  • Adney DR, Wang L, Van Doremalen N, Shi W, Zhang Y, Kong W-P, Miller M, Bushmaker T, Scott D, de Wit E. Efficacy of an adjuvanted middle east respiratory syndrome coronavirus spike protein vaccine in dromedary camels and alpacas. Viruses. 2019;11(3):212. doi:10.3390/v11030212.
  • Honda-Okubo Y, Barnard D, Ong CH, Peng B-H, Tseng C-T, Petrovsky N, Perlman S. Severe acute respiratory syndrome-associated coronavirus vaccines formulated with delta inulin adjuvants provide enhanced protection while ameliorating lung eosinophilic immunopathology. J Virol. 2015;89(6):2995–3007. doi:10.1128/JVI.02980-14.
  • Li L, Honda-Okubo Y, Huang Y, Jang H, Carlock MA, Baldwin J, Piplani S, Bebin-Blackwell AG, Forgacs D, Sakamoto K. Immunisation of ferrets and mice with recombinant SARS-CoV-2 spike protein formulated with Advax-SM adjuvant protects against COVID-19 infection. Vaccine. 2021;39(40):5940–53. doi:10.1016/j.vaccine.2021.07.087.
  • Piplani S, Singh PK, Winkler DA, Petrovsky N. In silico comparison of SARS-CoV-2 spike protein-ACE2 binding affinities across species and implications for virus origin. Sci Rep. 2021;11(1):13063. doi:10.1038/s41598-021-92388-5.
  • Baldwin J, Piplani S, Sakala IG, Honda-Okubo Y, Li L, Petrovsky N. Rapid development of analytical methods for evaluating pandemic vaccines: a COVID-19 perspective. Bioanalysis. 2021;13(24):1805–26. doi:10.4155/bio-2021-0096.
  • Schmidt F, Weisblum Y, Muecksch F, Hoffmann HH, Michailidis E, Lorenzi JCC, Mendoza P, Rutkowska M, Bednarski E, Gaebler C. Measuring SARS-CoV-2 neutralizing antibody activity using pseudotyped and chimeric viruses. J Exp Med. 2020;217(11). doi:10.1084/jem.20201181.
  • McPherson C, Chubet R, Holtz K, Honda-Okubo Y, Barnard D, Cox M, Petrovsky N. Development of a SARS coronavirus vaccine from recombinant spike protein plus delta inulin adjuvant. Methods Mol Biol. 2016;1403:269–84.
  • Silva DG, Cooper PD, Petrovsky N. Inulin-derived adjuvants efficiently promote both Th1 and Th2 immune responses. Immunol Cell Biol. 2004;82(6):611–6. doi:10.1111/j.1440-1711.2004.01290.x.
  • Lei X, Dong X, Ma R, Wang W, Xiao X, Tian Z, Wang C, Wang Y, Li L, Ren L. Activation and evasion of type I interferon responses by SARS-CoV-2. Nat Commun. 2020;11(1):3810. doi:10.1038/s41467-020-17665-9.
  • Appay V, Douek DC, Price DA. CD8+ T cell efficacy in vaccination and disease. Nat Med. 2008;14(6):623–8. doi:10.1038/nm.f.1774.
  • Li L, Honda-Okubo Y, Baldwin J, Bowen R, Bielefeldt-Ohmann H, Petrovsky N. Covax-19/Spikogen® vaccine based on recombinant spike protein extracellular domain with Advax-CpG55.2 adjuvant provides single dose protection against SARS-CoV-2 infection in hamsters. Vaccine. 2022;40(23):3182–92. doi:10.1016/j.vaccine.2022.04.041.
  • Rogers TF, Zhao F, Huang D, Beutler N, Burns A, He WT, Limbo O, Smith C, Song G, Woehl J. Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model. Science. 2020;369(6506):956–63. doi:10.1126/science.abc7520.
  • Garcia-Beltran WF, Lam EC, Astudillo MG, Yang D, Miller TE, Feldman J, Hauser BM, Caradonna TM, Clayton KL, Nitido AD. COVID-19-neutralizing antibodies predict disease severity and survival. Cell. 2021;184(2):476–88 e11. doi:10.1016/j.cell.2020.12.015.
  • Honda-Okubo Y, Li L, Andre G, Leong KH, Howerth EW, Bebin-Blackwell AG, Ross TM, Petrovsky N. An Advax-CpG55.2™ adjuvanted recombinant spike protein vaccine protects cynomolgus macaques from a homologous SARS-CoV-2 virus challenge. Vaccine. 2023;41(32):4710–8. doi:10.1016/j.vaccine.2023.06.063.
  • Honda-Okubo Y, Bowen R, Barker M, Bielefeldt-Ohmann H, Petrovsky N. Advax-CpG55.2-adjuvanted monovalent or trivalent SARS-CoV-2 recombinant spike protein vaccine protects hamsters against heterologous infection with beta or delta variants. Vaccine. 2023;41(48):7116–28. doi:10.1016/j.vaccine.2023.10.018.
  • Pal R, Ferrari MG, Honda-Okubo Y, Wattay L, Caple J, Navarrete J, Andersen H, Petrovsky N. Study of immunogenicity and efficacy against omicron BA.5 of recombinant protein-based COVID-19 vaccine delivered by intramuscular and mucosal routes in nonhuman primates. Vaccine. 2024;42(5):1122–35. doi:10.1016/j.vaccine.2024.01.034.
  • Moss P. The T cell immune response against SARS-CoV-2. Nat Immunol. 2022;23(2):186–93. doi:10.1038/s41590-021-01122-w.
  • McLelland DJ, Lynch M, Vogelnest L, Eden P, Wallace A, Weller J, Young S, Vaughan‐Higgins R, Antipov A, Honda‐Okubo Y. Safety and immunogenicity of an adjuvanted recombinant spike protein-based severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine,SpikeVet ™, in selected carnivora, primates and Artiodactyla in Australian zoos. J Vet Pharmacol Ther. 2024; doi:10.1111/jvp.13429.
  • Rabail R, Ahmed W, Ilyas M, Rajoka MSR, Hassoun A, Khalid AR, Khan MR, Aadil RM. The side effects and adverse clinical cases reported after COVID-19 immunization. Vaccines. 2022;10(4):10. doi:10.3390/vaccines10040488.
  • Pollard AJ, Bijker EM. A guide to vaccinology: from basic principles to new developments. Nat Rev Immunol. 2021;21(2):83–100. doi:10.1038/s41577-020-00479-7.
  • WHO. World Health Organization Target product profiles for COVID-19 vaccines. https://wwwwhoint/who-documents-detail/who-target-product-profiles-for-covid-19-vaccines2020.
  • Ciolino JD, Kaizer AM, Bonner LB. Guidance on interim analysis methods in clinical trials. J Clin Transl Sci. 2023;7(1):e124. doi:10.1017/cts.2023.552.
  • Levin EG, Lustig Y, Cohen C, Fluss R, Indenbaum V, Amit S, Doolman R, Asraf K, Mendelson E, Ziv A. Waning immune humoral response to BNT162b2 Covid-19 vaccine over 6 months. N Engl J Med. 2021;385(24):e84. doi:10.1056/NEJMoa2114583.
  • Voysey M, Clemens SAC, Madhi SA, Weckx LY, Folegatti PM, Aley PK, Angus B, Baillie VL, Barnabas SL, Bhorat QE. 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. Lancet. 2021;397(10269):99–111. doi:10.1016/S0140-6736(20)32661-1.
  • Scala A, Cavallo P, Pereira T. Measuring the efficacy of a vaccine during an epidemic. PLOS ONE. 2023;18(9):e0290652. doi:10.1371/journal.pone.0290652.
  • El Sahly HM, Baden LR, Essink B, Doblecki-Lewis S, Martin JM, Anderson EJ, Campbell TB, Clark J, Jackson LA, Fichtenbaum CJ. Efficacy of the mRNA-1273 SARS-CoV-2 vaccine at completion of blinded phase. N Engl J Med. 2021;385(19):1774–85. doi:10.1056/NEJMoa2113017.
  • Goldblatt D, Fiore-Gartland A, Johnson M, Hunt A, Bengt C, Zavadska D, Snipe HD, Brown JS, Workman L, Zar HJ. Towards a population-based threshold of protection for COVID-19 vaccines. Vaccine. 2022;40(2):306–15. doi:10.1016/j.vaccine.2021.12.006.
  • Lopez Bernal J, Andrews N, Gower C, Gallagher E, Simmons R, Thelwall S, Stowe J, Tessier E, Groves N, Dabrera G. Effectiveness of Covid-19 vaccines against the B.1.617.2 (Delta) variant. N Engl J Med. 2021;385(7):585–94. doi:10.1056/NEJMoa2108891.
  • World Health O. Hepatitis B vaccines: WHO position paper, July 2017 – recommendations. Vaccine. 2019;37(2):223–5. doi:10.1016/j.vaccine.2017.07.046.
  • Tabarsi P, Anjidani N, Shahpari R, Roshanzamir K, Fallah N, Andre G, Petrovsky N, Barati S. Immunogenicity and safety of SpikoGen®, an adjuvanted recombinant SARS-CoV-2 spike protein vaccine as a homologous and heterologous booster vaccination: a randomized placebo-controlled trial. Immunology. 2022;167(3):340–53. doi:10.1111/imm.13540.
  • Fendler A, Shepherd STC, Au L, Wu M, Harvey R, Schmitt AM, Tippu Z, Shum B, Farag S, Rogiers A. Omicron neutralising antibodies after third COVID-19 vaccine dose in patients with cancer. Lancet. 2022;399(10328):905–7. doi:10.1016/S0140-6736(22)00147-7.
  • Cohn BA, Cirillo PM, Murphy CC, Krigbaum NY, Wallace AW. SARS-CoV-2 vaccine protection and deaths among US veterans during 2021. Science. 2022;375(6578):331–6. doi:10.1126/science.abm0620.
  • Gupta D, Parthasarathy H, Sah V, Tandel D, Vedagiri D, Reddy S, Harshan KH. Inactivation of SARS-CoV-2 by beta-propiolactone causes aggregation of viral particles and loss of antigenic potential. Virus Res. 2021;305:198555. doi:10.1016/j.virusres.2021.198555.
  • Natarajan K, Prasad N, Dascomb K, Irving SA, Yang DH, Gaglani M, Klein NP, DeSilva MB, Ong TC, Grannis SJ. Effectiveness of homologous and heterologous COVID-19 booster doses following 1 Ad.26.COV2.S (Janssen [Johnson & Johnson]) vaccine dose against COVID-19-associated emergency department and urgent care encounters and hospitalizations among Adults - VISION network, 10 states, December 2021-March 2022. Morb Mortal Wkly Rep. 2022;71(13):495–502. doi:10.15585/mmwr.mm7113e2.
  • Petrovsky N, Larena M, Siddharthan V, Prow NA, Hall RA, Lobigs M, Morrey J. An inactivated cell culture Japanese encephalitis vaccine (JE-ADVAX) formulated with delta inulin adjuvant provides robust heterologous protection against West Nile encephalitis via cross-protective memory B cells and neutralizing antibody. J Virol. 2013;87(18):10324–33. doi:10.1128/JVI.00480-13.
  • Komiya T, Honda-Okubo Y, Baldwin J, Petrovsky N. An Advax-adjuvanted inactivated cell-culture derived Japanese encephalitis vaccine induces broadly neutralising anti-flavivirus antibodies, robust cellular immunity and provides single dose protection. Vaccines. 2021;9:1235.
  • Nafar M, Mostafaloo N, Firouzan A, Poorrezagholi F, Samadian F, Dalili N, Barati S, Anjidani N, Kafi H, Shahpari R. Immunogenicity and safety of SpikoGen, an adjuvanted recombinant SARS-CoV-2 Spike protein, as a heterologous third booster dose in kidney transplant patients: a Single-arm clinical trial. Clin Ther. 2022;44(12):1566–76. doi:10.1016/j.clinthera.2022.10.002.
  • Tabarsi P, Mamishi S, Anjidani N, Shahpari R, Kafi H, Fallah N, Yazdani B, Ebrahimi A, Roshanzamir K, Ebrahimi H. Comparative immunogenicity and safety of SpikoGen®, a recombinant SARS-CoV-2 spike protein vaccine in children and young adults: an immuno-bridging clinical trial. Int Immunopharmacol. 2024;127:111436. doi:10.1016/j.intimp.2023.111436.
  • Lee YK, Kwon Y, Lim D, Seo SY, Kim EK, Kim SY, Kim S, Ko M, Lim D, Seo S-Y. COVID-19 vaccine safety profile in Republic of Korea, February 26, 2021 through April 30, 2022. Clin Exp Pediatr. 2023;66(10):415–23. doi:10.3345/cep.2022.00815.
  • Ostrowski SR, Sogaard OS, Tolstrup M, Staerke NB, Lundgren J, Ostergaard L, Hvas AM. Inflammation and platelet activation after COVID-19 vaccines - possible mechanisms behind vaccine-induced immune thrombocytopenia and thrombosis. Front Immunol. 2021;12:779453. doi:10.3389/fimmu.2021.779453.
  • Sallard E, Zhang W, Aydin M, Schroer K, Ehrhardt A. The adenovirus vector platform: novel Insights into rational vector design and lessons learned from the COVID-19 vaccine. Viruses. 2023;15(1):15. doi:10.3390/v15010204.
  • Satapathy P, Kumar P, Gupta JK, Rabaan AA, Al Kaabi NA, Mohanty D, Naveen P, Khatib MN, Gaidhane S, Zahiruddin QS. The emergence and implications of SARS-CoV-2 omicron subvariant BA.2.86 on global health. Int J Surg. 2024;110(4):2498–501. doi:10.1097/JS9.0000000000001070.
  • Shrestha NK, Burke PC, Nowacki AS, Simon JF, Hagen A, Gordon SM. Effectiveness of the coronavirus disease 2019 bivalent vaccine. Open Forum Infect Dis. 2023;10(6):ofad209. doi:10.1093/ofid/ofad209.
  • Mostaghimi D, Valdez CN, Larson HT, Kalinich CC, Iwasaki A. Prevention of host-to-host transmission by SARS-CoV-2 vaccines. Lancet Infect Dis. 2022;22(2):52–8. doi:10.1016/S1473-3099(21)00472-2.
  • Staerke NB, Reekie J, Nielsen H, Benfield T, Wiese L, Knudsen LS, Iversen MB, Iversen K, Fogh K, Bodilsen J. Levels of SARS-CoV-2 antibodies among fully vaccinated individuals with delta or omicron variant breakthrough infections. Nat Commun. 2022;13(1):4466. doi:10.1038/s41467-022-32254-8.
  • Hui DS. Hybrid immunity and strategies for COVID-19 vaccination. Lancet Infect Dis. 2023;23(1):2–3. doi:10.1016/S1473-3099(22)00640-5.
  • Shrestha NK, Burke PC, Nowacki AS, Gordon SM, Acuti Martellucci C. Risk of coronavirus disease 2019 (COVID-19) among those up-to-date and not up-to-date on COVID-19 vaccination by US CDC criteria. PLOS ONE. 2023;18(11):e0293449. doi:10.1371/journal.pone.0293449.
  • Sajkov D, Woodman R, Honda-Okubo Y, Barbara J, Chew D, Toson B, Petrovsky N. A multi-season randomised controlled trial of Advax-adjuvanted seasonal influenza vaccine in participants with chronic disease or older age. J Infect Dis. 2023; doi:10.1093/infdis/jiad589.
  • Akhtar M, Islam MR, Khaton F, Soltana UH, Jafrin SA, Rahman SIA, Tauheed I, Ahmed T, Khan II, Akter A. Appearance of tolerance-induction and non-inflammatory SARS-CoV-2 spike-specific IgG4 antibodies after COVID-19 booster vaccinations. Front Immunol. 2023;14:1309997. doi:10.3389/fimmu.2023.1309997.
  • Boretti A. mRNA vaccine boosters and impaired immune system response in immune compromised individuals: a narrative review. Clin Exp Med. 2024;24(1):23. doi:10.1007/s10238-023-01264-1.