5
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Current Flavivirus Research Important for Vaccine Development

      editorial
      , , , *
      Vaccines
      MDPI

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The Flaviviridae family of RNA viruses includes numerous human disease-causing pathogens that largely are increasing in prevalence due to continual climate change, rising population sizes and improved ease of global travel. Escalating circulation of these emerging and re-emerging pathogens draws attention to the need for vaccines to protect against the severe diseases they cause, and this need is further exacerbated by their transmission that occurs primarily through arthropod vectors. When constructing new, efficacious vaccine candidates, several goals are targeted including safety, protective capacity, ability to confer sustained protection, induction of neutralizing antibody and protective T cell responses, as well as practicality. Prophylactic vaccination as a means to protect against flavivirus infection has become an intense area of research in the last several decades due to the inherent nature of this family of viruses to cause explosive outbreaks [1]. However, even with the enormous prevalence and impact on the human population, there are minimal flaviviruses with approved human vaccines to date: yellow fever (YF), Japanese encephalitis virus (JEV), tick-borne encephalitis virus (TBEV) and dengue virus (DENV) [2]. This Special Issue highlights current studies that delve into research expanding the range of flavivirus candidate vaccines. These findings are essential to advance the field and further protect vulnerable populations from these disease-causing pathogens. Although the members of the flavivirus family have been shown to be antigenically similar [3], these viruses are indeed unique enough to often warrant varying vaccines. This concept is highlighted by the four distinct DENV serotypes. DENV is widespread and endemic in greater than 125 countries and currently accounts for an estimated 100–400,000,000 infections each year, with half of the world’s population being at risk of infection [4]. As the four serotypes of DENV are distinct, infection with one serotype results in only transient protection against the other three serotypes [5], and each subsequent heterologous infection increases the risk for an infected person to develop severe dengue. Although there is a currently approved DENV vaccine, safety and efficacy data support the need for alternative prophylactic vaccines to protect a broader population of individuals. Strides toward this goal include virus-like particles (VLPs), which are outlined in the review written by Wong et al. which summarizes VLP-based DENV vaccines currently being tested. As noted by Wong et al., many of the current VLP-based DENV vaccines lack the viral pre-Membrane (prM) protein; thus, it is believed these vaccines will circumvent the induction of cross-reactive non-neutralizing antibodies that may mediate antibody dependent enhancement. Importantly, these constructs still maintain key structural components that induce T cell and antibody responses, like envelope domain III (E DIII) against which most neutralizing antibodies are targeted [6]. Additionally, such VLP vaccines have greater safety profiles than live-attenuated vaccines, which have a higher risk of reverting to infectious virus in the elderly, a population that has been shown to be at high risk for infection and difficult to vaccinate [7,8]. The case report presented by Domingo et al. features an instance where an elderly individual received the YF vaccine prior to traveling to Brazil, then became ill and experienced septic shock and multiorgan failure. It was soon determined that he already had discernable YF antibodies a week after vaccination, in addition to high YF viral load in serum, plasma, respiratory secretions and urine [9]. Thus, the benefits of VLP vaccines include eliminating mutations to infectious virus in these critical, vulnerable populations while maintaining antigenic structural proteins in a native conformation. Further, deeper understanding and advances in flavivirus genome structure and replication cycles have been able to aid the development of other kinds of unique DENV vaccine candidates. In this Issue, Park et al. outlines a candidate subunit vaccine based on eliciting a strong neutralizing antibody response against E DIII. This vaccine was developed to combat all four DENV serotypes by combining two subunits: partial envelope domain II and consensus envelope domain III (cEDIII) [10]. The authors found that an antibody purified from mice following immunization with this combined candidate vaccine was strongly neutralizing compared to their single subunit vaccine containing only cEDIII, with limited non-neutralizing antigen-specific antibody [10]. In addition, this combined subunit vaccine was able to reduce DENV titers in several tissues when compared to the single subunit vaccine, and conferred protection against DENV1, 2 and 4 in murine challenge models, thus showing promise as a candidate DENV subunit vaccine [10]. Similarly, Tremblay et al. highlights how the discovery that flavivirus vRNA 2′-O-methylation enables mimicry of cellular mRNAs has been instrumental in reverse genetics studies to improve DENV vaccine design [11]. Specifically, when mutant DENV clones are modified to encode a single point mutation in the viral nonstructural gene 5 (NS5) methyltransferase catalytic site, sensitivity to interferon treatment is enhanced, and pre-treatment with such mutant viruses protects mice and monkeys from lethal DENV infection [12]; this same approach has also proven successful in laboratory studies seeking to formulate an alternative JEV vaccine construct [13]. In addition to the previously mentioned challenges of developing flavivirus vaccines, another strong impediment to their development are practical considerations. As endemic regions of flavivirus circulation tend to be rural, access to trained medical professionals and refrigeration is often sparse. To counteract these obstacles, some vaccine constructs are aimed at achieving formulations that are biologically stable over time and do not require a medical professional to administer. In Muller et al., a tetravalent E DENV subunit vaccine was administered via a microarray patch and, when compared to traditional vaccination methods, led to enhanced neutralizing antibody titers to all four DENV serotypes potentially due to the delivery of antigens in close proximity to antigen presenting cells located beneath the skin. Inclusion of an adjuvant to enhance immunogenicity led to complete protection in mice following DENV challenge [14]. In addition to being efficacious, this form of vaccination would also be beneficial to the population since a nanopatch can be administered by someone who is not medically trained, and it does not require refrigeration. This Special Issue also focuses its attention on Zika virus (ZIKV), a re-emerging pathogen with a history of causing epidemics: 2007 (Yap islands), 2013 (French Polynesia) and 2015 (South America) [15]. The 2015 ZIKV epidemic was rife with enhanced viral spread due to travelers, further highlighting the need for a prophylactic vaccine that can be administered to residents of ZIKV endemic areas, as well as to travelers. Similar to DENV vaccination efforts, many trial ZIKV vaccines are formulated to elicit strongly neutralizing antibodies, with several purified inactivated, live-attenuated and DNA-based vaccines correlating with protection in mice, rhesus macaque animal models and phase I human clinical trials [16,17,18]. In a study presented by Frumence et al., a chimeric ZIKV vaccine clone was generated where the glycan loop of the viral E protein of African strain MR766 was modified to encode three counterpart E glycan loop amino acids of the epidemic strain, BeH819015, formulating a chimeric clone referred to as ZIKVBeHMR-2. Following these substitutions and subsequent vaccination with ZIKVBeHMR-2, neutralizing ZIKV antibodies were rapidly detected and effective at combatting MR766 challenge in mice [19]. In addition to the need for development and maintenance of strongly neutralizing antibodies following vaccination, cellular responses have also proven to be essential in combatting ZIKV, as it has been shown that CD4 and CD8+ T cells are critical in reducing viral load and mortality in mouse models of ZIKV infection [20,21,22]. To this end, the review written by Wong et al. highlights pre-Membrane:Envelope (prME) and Capsid:pre-Membrane:Envelope (CprME) VLP-based vaccines that elicit T cell responses and stronger neutralizing antibody responses than other DNA and formalin inactivated preparations [23,24]. As previously mentioned, VLP-based vaccines have proven to be safer than live-attenuated formulations, which will be key in ZIKV vaccine development since pregnant women are classified as one of the most vulnerable populations. Nonetheless, in 2018, a live-attenuated ZIKV vaccine that was protective in animal challenge models was tested in a phase I clinical trial with support of the National Institutes of Health. The possibility of reversion that comes with a live attenuated vaccine denotes the need for a metric to examine mutation rates of such vaccines. In the study presented by Collins et al., next generation sequencing (NGS) was utilized to measure genetic diversity of live attenuated candidate ZIKV vaccines and revealed that viral adaptation occurred throughout cellular passages [25]. Thus, this study highlights a role for NGS in studying the genetic stability of live attenuated vaccines. Another article, co-authored by our group, also focused on ZIKV vaccine development, highlighting another promising vaccine design avenue: adenovirus-vectors. These vectors have increased in popularity over the last several years as vaccine platforms primarily due to excellent safety profiles, the ability to be grown to high titers in cell culture, capacity to induce strong inflammatory immune response, etc. [26]. Steffen et al. developed an Early region 1A/Early region 3 (E1A/E3)-deleted adenovirus-vector ZIKV vaccine that included an amino acid mutation to improve prM-E polypeptide processing, in addition to including both B and T cell epitopes. Further highlighting the advantages of vaccines that elicit both humoral and cell-mediated immune responses, we showed that the vaccine construct was both immunogenic and efficacious in producing ZIKV-specific CD4+, CD8+ and antibody responses that were protective in a murine ZIKV challenge model [27]. Thus, this study provided strong evidence that an adenovirus-vector vaccine against ZIKV has the ability to induce a potent immune response against this re-emerging pathogen. As this Special Issue highlights, developing vaccines for numerous members of the Flaviviridae family has proven to be challenging for a variety of reasons, including virus interplay with the immune system, a concept highlighted in the review written by Tremblay et al. [11]. One vaccine approach which would alleviate the concerns of flavivirus immune suppression on vaccine efficacy was highlighted by Wang et al. [28], who sought to focus vaccine efforts on the vector rather than the pathogen. Since many flaviviruses are often transmitted by the same species of arthropod vectors, some research efforts have focused on generating vaccines against antigens produced by the transmitting vector. Of particular note, it has been shown that an Aedes aegypti salivary gland protein, AgBR1, shapes innate immune responses in murine models following mosquito bites [29]. Based on this discovery, Wang et al. developed an AgBR1 adjuvanted active immunization strategy in mice that led to the delay of lethal mosquito-transmitted ZIKV infection [28]. This paper introduces an interesting concept where immunization with a mosquito-derived protein could protect populations from numerous mosquito-spread pathogens by targeting the innate immune response. Hand in hand with vaccine design, methods of evaluation for vaccine efficacy are also under intense research. In a study by Frumence et al., a flow cytometry-based neutralization test (FNT) was employed as an alternative to the conventional plaque-reduction neutralization test (PRNT) as a means to enable a high throughput screening strategy that does not rely on methods requiring cell lines capable of fostering plaque formation. Through the development of a GFP reporter of ZIKV, this study highlights the ability to measure ZIKV neutralizing antibodies via flow cytometry with the antibody titers detected being equivalent to those done by PRNTs in tandem [30]. Another promising way to evaluate and enhance vaccine efficacy is highlighted in the study presented by Salat et al. where mass spectrometry was used to analyze the content of two TBEV vaccines. Upon confirming the presence of nonstructural protein 1 (NS1) in one vaccine tested, it appeared to be highly immunogenic when administered to mice and was able to enhance their survival following lethal virus challenge [31]. Thus, this study highlights another viral protein, NS1, in addition to E that can enhance the protective capacity of TBEV vaccines [31]. Overall, this Special Issue highlights numerous studies, reviews and a case report that outline current research being done to further flavivirus vaccine development. These studies address both the challenges of developing flavivirus vaccines and the diverse and novel approaches to vaccine development that are currently underway. In addition, this Special Issue also features work being done to improve pre-existing vaccines, as well as techniques to streamline the process of determining vaccine efficacy while confirming safety. While the rates of flavivirus infections continue to rise globally, research efforts like the ones outlined in this Issue continually increase our knowledge of flaviviruses, thus informing the development of novel vaccines which will meet the growing need for protection against these pathogens.

          Related collections

          Most cited references30

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Antibody-dependent enhancement of severe dengue disease in humans

          For dengue viruses 1 to 4 (DENV1-4), a specific range of antibody titer has been shown to enhance viral replication in vitro and severe disease in animal models. Although suspected, such antibody-dependent enhancement of severe disease has not been shown to occur in humans. Using multiple statistical approaches to study a long-term pediatric cohort in Nicaragua, we show that risk of severe dengue disease is highest within a narrow range of preexisting anti-DENV antibody titers. By contrast, we observe protection from all symptomatic dengue disease at high antibody titers. Thus, immune correlates of severe dengue must be evaluated separately from correlates of protection against symptomatic disease. These results have implications for studies of dengue pathogenesis and for vaccine development, because enhancement, not just lack of protection, is of concern.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Safety, tolerability, and immunogenicity of two Zika virus DNA vaccine candidates in healthy adults: randomised, open-label, phase 1 clinical trials

            Summary Background The Zika virus epidemic and associated congenital infections have prompted rapid vaccine development. We assessed two new DNA vaccines expressing premembrane and envelope Zika virus structural proteins. Methods We did two phase 1, randomised, open-label trials involving healthy adult volunteers. The VRC 319 trial, done in three centres, assessed plasmid VRC5288 (Zika virus and Japanese encephalitis virus chimera), and the VRC 320, done in one centre, assessed plasmid VRC5283 (wild-type Zika virus). Eligible participants were aged 18–35 years in VRC19 and 18–50 years in VRC 320. Participants were randomly assigned 1:1 by a computer-generated randomisation schedule prepared by the study statistician. All participants received intramuscular injection of 4 mg vaccine. In VRC 319 participants were assigned to receive vaccinations via needle and syringe at 0 and 8 weeks, 0 and 12 weeks, 0, 4, and 8 weeks, or 0, 4, and 20 weeks. In VRC 320 participants were assigned to receive vaccinations at 0, 4, and 8 weeks via single-dose needle and syringe injection in one deltoid or split-dose needle and syringe or needle-free injection with the Stratis device (Pharmajet, Golden, CO, USA) in each deltoid. Both trials followed up volunteers for 24 months for the primary endpoint of safety, assessed as local and systemic reactogenicity in the 7 days after each vaccination and all adverse events in the 28 days after each vaccination. The secondary endpoint in both trials was immunogenicity 4 weeks after last vaccination. These trials are registered with ClinicalTrials.gov, numbers NCT02840487 and NCT02996461. Findings VRC 319 enrolled 80 participants (20 in each group), and VRC 320 enrolled 45 participants (15 in each group). One participant in VRC 319 and two in VRC 320 withdrew after one dose of vaccine, but were included in the safety analyses. Both vaccines were safe and well tolerated. All local and systemic symptoms were mild to moderate. In both studies, pain and tenderness at the injection site was the most frequent local symptoms (37 [46%] of 80 participants in VRC 319 and 36 [80%] of 45 in VRC 320) and malaise and headache were the most frequent systemic symptoms (22 [27%] and 18 [22%], respectively, in VRC 319 and 17 [38%] and 15 [33%], respectively, in VRC 320). For VRC5283, 14 of 14 (100%) participants who received split-dose vaccinations by needle-free injection had detectable positive antibody responses, and the geometric mean titre of 304 was the highest across all groups in both trials. Interpretation VRC5283 was well tolerated and has advanced to phase 2 efficacy testing. Funding Intramural Research Program of the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Zika virus: a previously slow pandemic spreads rapidly through the Americas.

              Zika virus (family Flaviviridae) is an emerging arbovirus. Spread by Aedes mosquitoes, it was first discovered in Uganda in 1947, and later in humans elsewhere in sub-Saharan Africa, arriving in south-east Asia at latest by the mid-twentieth century. In the twenty-first century, it spread across the Pacific islands reaching South America around 2014. Since then it has spread rapidly northwards reaching Mexico in November 2015. Its clinical profile is that of a dengue-like febrile illness, but associations with Guillain-Barré syndrome and microcephaly have appeared recently. The final geographical range and ultimate clinical impact of Zika virus are still a matter for speculation.
                Bookmark

                Author and article information

                Journal
                Vaccines (Basel)
                Vaccines (Basel)
                vaccines
                Vaccines
                MDPI
                2076-393X
                27 August 2020
                September 2020
                : 8
                : 3
                : 477
                Affiliations
                Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 63103, USA; lizzie.geerling@ 123456slu.edu (E.G.); tara.steffen@ 123456slu.edu (T.L.S.); james.brien@ 123456health.slu.edu (J.D.B.)
                Author notes
                Author information
                https://orcid.org/0000-0002-5104-499X
                https://orcid.org/0000-0002-1670-8041
                Article
                vaccines-08-00477
                10.3390/vaccines8030477
                7563144
                32867038
                9879a834-65a1-4087-84cb-91bac3e241ae
                © 2020 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 21 August 2020
                : 23 August 2020
                Categories
                Editorial

                Comments

                Comment on this article