Next Article in Journal
Transient Blockade of Type I Interferon Signalling Promotes Replication of Dengue Virus Strain D2Y98P in Adult Wild-Type Mice
Next Article in Special Issue
Recent Advances in Antivirals for Japanese Encephalitis Virus
Previous Article in Journal
Characterization of Human Norovirus Nonstructural Protein NS1.2 Involved in the Induction of the Filamentous Endoplasmic Reticulum, Enlarged Lipid Droplets, LC3 Recruitment, and Interaction with NTPase and NS4
Previous Article in Special Issue
Inhibitors of the Ubiquitin-Mediated Signaling Pathway Exhibit Broad-Spectrum Antiviral Activities against New World Alphaviruses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advancements in Mosquito-Borne Flavivirus Vaccine Development

Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
*
Authors to whom correspondence should be addressed.
Viruses 2023, 15(4), 813; https://doi.org/10.3390/v15040813
Submission received: 13 February 2023 / Revised: 21 March 2023 / Accepted: 21 March 2023 / Published: 23 March 2023
(This article belongs to the Special Issue Antivirals against Arboviruses)

Abstract

:
Lately, the global incidence of flavivirus infection has been increasing dramatically and presents formidable challenges for public health systems around the world. Most clinically significant flaviviruses are mosquito-borne, such as the four serotypes of dengue virus, Zika virus, West Nile virus, Japanese encephalitis virus and yellow fever virus. Until now, no effective antiflaviviral drugs are available to fight flaviviral infection; thus, a highly immunogenic vaccine would be the most effective weapon to control the diseases. In recent years, flavivirus vaccine research has made major breakthroughs with several vaccine candidates showing encouraging results in preclinical and clinical trials. This review summarizes the current advancement, safety, efficacy, advantages and disadvantages of vaccines against mosquito-borne flaviviruses posing significant threats to human health.

1. Introduction

Mosquito-borne flaviviruses, members of the genus Flavivirus, are small spherical particles transmitted by various species of mosquitoes [1]. The genome of these viruses is nearly 11 kb in length [2,3,4,5], encoding a single polyprotein precursor which includes three structural proteins, capsid (C), premembrane (prM) and envelope (E), and seven nonstructural (NS) proteins, NS1, NS2A, NS2B, NS3, NS4A, NS4B and NS5) [6,7]. Of these proteins, E protein is found to induce protective immune response, and the significance of antibodies targeting flaviviral E during antiviral protection has been verified in several animal models [8,9,10,11]. Dengue virus (DENV), Zika virus (ZIKV), West Nile virus (WNV), Japanese encephalitis virus (JEV) and yellow fever virus (YFV) are five typical mosquito-borne flaviviruses which could cause severe infectious diseases and, thus, show human medical significance [12]. The wide distribution of accordant mosquito vectors in the world results in the pandemic transmission of these flaviviruses and increases the infection risk of human populations [12,13].
Recently, epidemics of mosquito-borne flavivirus infections pose great concerns [6,14] and are tremendous burdens on global public health systems [15]. DENV affects approximately 130 countries. Nearly 390 million dengue cases are discovered annually [16], of which 96 million (25%) show significant clinical manifestations, with 500,000 cases requiring hospitalization [17,18,19]. DENV causes more human diseases than any other mosquito-borne virus [20], and the number of DENV infected cases has risen over 400% in recent decades [21,22]. ZIKV, first discovered in 1947 in Uganda [23,24,25,26], is a mosquito-transmitted pathogen and is distributed worldwide. It is estimated that about 2.17 billion people are living in regions at risk for ZIKV transmission [27,28,29]. ZIKV transmission can also occur through sexual contact and blood transfusion, or through placental transmission, which causes abnormal gestational development in the fetus [14,27,30,31,32]. Epidemiological investigations have shown that more than 40% of pregnant women infected with ZIKV give birth to fetuses with congenital neurological malformations such as postnatal neurodevelopmental deficits, ocular anomalies, and microcephaly [27,33]. These severe forms of illness place a heavy burden on public health systems [14]. West Nile virus (WNV), a zoonotic pathogen [34], is an important etiological mosquito-borne agent of encephalitis in human and equids [35,36]. Since its first discovery in the last century, WNV has rapidly spread in many countries of the world [37]. In humans, most WNV infected cases are asymptomatic [38,39] or are only present in flu-like symptoms; however, nearly 1% of the infections might develop into more severe neurological diseases, such as encephalitis, meningitis, or flaccid paralysis [38,40,41]. In addition, WNV can be transmitted vertically through the intrauterine route [42], or via breastfeeding milk [43,44] and blood transfusion [45,46,47]. JEV is the most important infectious agent of viral encephalitis in Asia [1,48,49], the endemic regions ranging from the China–Russia border region to northern Australia, and from the Western Pacific islands to the India–Pakistan border [50,51], where an estimated 50,000 clinical cases and 10,000 deaths occur annually [37], with a high incidence of neuropsychiatric sequelae among recovered patients [48,52]. YFV, circulating mainly in Africa and South America [53,54], causes a serious contagious disease, yellow fever (YF). According to the WHO, there are about 200,000 YF cases in clinics annually, with a high fatality rate of about 30% [37].
Although some antivirals, e.g., favipiravir [55], sofosbuvir [56] and TRIM56 [57,58] et al., have been demonstrated to be effective in suppressing some mosquito-borne flavivirus in vitro and in vivo, until now, specific antiviral interventions against these viral infections are not available in clinics [59]. Therefore, effective vaccines represent the optimal strategy to prevent and control these burgeoning diseases. Currently, conventional and experimental mosquito-borne flavivirus vaccines mainly include the following six major categories [60]: (1) live attenuated virus vaccines, (2) inactivated vaccines, (3) nucleic acid vaccines including RNA and DNA [61], (4) virus-like particle (VLP) vaccines, (5) subunit protein vaccines and (6) viral vector vaccines.
This paper will focus on the current status of mosquito-borne flavivirus vaccine development, clarify the composition and efficacy of these vaccines and discuss their advantages and disadvantages.

2. Live Attenuated Virus Vaccines

Due to significantly low manufacturing costs [15,62] and comprehensively long-lasting immune response after a single dose [15,59,61,62], live attenuated virus vaccines (LAVs) are the most successful vaccines at preventing infections. An estimated 63% of FDA-approved vaccines are LAVs [59,63]. LAVs against mosquito-transmitted flaviviruses are developing quickly in recent years. YFV-17D is considered to be the safest and most effective LAV against YFV infection [6]. SA14-14-2 is another effective LAV for JEV. One novel LAV technology, ChimeriVax (Chambers et al., St. Louis University Health Sciences Center, USA), has recently manifested considerable prospects in the development of vaccines against mosquito-borne flaviviruses [61]. ChimeriVax is composed of a backbone cDNA clone of a flavivirus, of which the prM-E segment is substituted with the corresponding one from the virus selected for the vaccine. Basically, the backbone comes directly from the virus of LAVs or engineered wild-type (WT) virus attenuated through mutations in vitro [6]. RepliVax (Widman et al., University of Texas Medical Branch, USA) is another emerging technology for the development of flavivirus vaccines based on deletion mutants [64]. These mutant viruses are not capable of assembling and releasing progeny virus particles (single-cycle virus) or replicating their viral genomes [15]. Generally, the flavivirus RepliVax vaccines are constructed by the deletion of C protein from single-cycle viruses, producing subviral particles [15,65] (SVPs, contain E and prM/M protein with antigenically significance, but delete C protein or the viral genome to contribute to the non-infectivity [25,66]). Similarly to ChimeriVax, RepliVax vaccines are relatively safe [15], with the ability to continuously stimulate immune responses [67], and do not require adjuvants, which are used in inactivated virus vaccines (INVs) [64]. However, the reversion of vaccine strains to increased virulence is the main problem related to live virus vaccines. In addition, LAVs are not recommended for the immunocompromised and pregnant women [62].

2.1. DENV

A tetravalent recombinant LAV called Dengvaxia® (Sanofi Pasteur, France) [18,68,69] is the only approved DENV vaccine [17,70,71,72,73]. The vaccine is produced by the chimeric technology, in which the prM and E genes of the YFV-17D backbone were substituted by those of DENV 1, 2, 3 and 4 [17,68,69,73,74,75,76]. Studies indicated that Dengvaxia® was safe and induced neutralizing antibodies against DENV of the existing four serotypes [77]. Although Dengvaxia® has been licensed for DENV prevention, global uptake is hampered due to the limitation of only administering it to seropositive persons who are aged above 9 years old, not to mention the sequential 3-dose schedule [21,78]. Takeda Pharmaceutical Company (Tokio, Japan) has developed a chimeric tetravalent LAV candidate (TDV) which contains the attenuated DENV-2-PDK53 strain with the prM and E genes of three other serotypes of DENV [18,71,75,79]. TDV has been confirmed to elicit both long-term humoral [80,81,82,83] and cellular immune response [83,84,85]. A recent phase III clinical trial has shown that TDV was well tolerated [71,86,87,88] and induced 62% seroconversion for four DENV serotypes [18,71]. The primary efficacy data indicated promising results, showing 80.2% overall vaccine efficacy, 95.4% preventive efficacy of severe dengue forms, and 74.9% efficacy in dengue seronegative patients [71]. LATV, another tetravalent LAV for dengue, was developed by the National Institute of Allergy and Infectious Diseases (NIAID) in the US [69,89]. It was composed of four attenuated DENV serotype strains. The phase I–III clinical trials verified that LATV was within an acceptable safety profile [69] and could elicit potent humoral immune responses against all DENV serotypes, with a long-lasting antibody persistence. In addition, the cross-reactive T cell-mediated immune responses are also activated comprehensively by the vaccine [79,88,90,91,92,93]. These data indicate that LATV may serve as an efficient and safe DENV vaccine for application in individuals covering all ages, regardless of DENV infection status before the vaccination [94]. The Walter Reed Army Institute of Research (WRAIR) and GlaxoSmithKline (GSK) have recently codeveloped a tetravalent dengue LAV called TDEN. The TDEN resulted in 100% seroconversion toward all the four DENV serotypes in flavivirus-primed subjects in phase II clinical trials. It has been shown that a two-dose administration regimen was safe and effective in volunteers ranging from 12 months to 50 years of age [69].

2.2. ZIKV

ChinZIKV (Beijing Institute of Microbiology and Epidemiology, China) is developed with the JEV SA14-14-2 strain by substituting its prM-E genes with the one from the ZIKV FSS 13025 strain. It has been shown that ChinZIKV protects mice and rhesus macaques against ZIKV challenges, and ZIKV intrauterine transmission was also blocked in pregnant mice [27,30,95,96]. ChimeriVax-Zika is also a chimeric vaccine which contains ZIKV prM-E antigens in the YFV-17D backbone. One dose of the vaccine induced robust ZIKV neutralizing antibodies which protected the mice against ZIKV challenges [27]. Recently, a ZIKV LAV called rZIKV/D4Δ30–713 has entered phase I clinical trial. It is also a chimeric vaccine, which introduces the DENV-4 as the backbone for the delivery of the ZIKV prM-E as the antigens. This chimeric recombinant vaccine was highly attenuated in A129 mice (type I interferon receptor knockout mice) and provided protective immunity against ZIKV challenges [27,96].

2.3. WNV

Researchers from the Beijing Institute of Microbiology and Epidemiology have engineered a chimeric JEV/WNV virus (ChinWNV) cDNA which used a subgenomic replicon SA14-14-2 as the genetic backbone, and the prM and E genes of JEV were substituted with the corresponding WNV genes. Studies have shown that one dose of ChinWNV rapidly induced strong humoral immune responses in mice, providing solid protection against the lethal WT-WNV challenge [97]. The US’s NIH has also developed a recombinant WNV LAV (WN/DEN4Δ30), which comprises the prM and E genes of WNV NY99 and the C and NS genes of rDEN4Δ30 [47,98,99]. The results of clinical trials revealed that WN/DEN4Δ30 was safe and immunogenic [98]. Another ChimeriVax WNV vaccine, WNV02 was generated based on the YFV 17D backbone, in which the prM/E genes of the YFV 17D were substituted with those of the WNV NY99 [59,100,101]. Monkeys vaccinated with WNV02 received potent protection against the lethal WT-WNV challenge [15,59] and with high safety [34] and this vaccine has entered phase II clinical trials [102]. A RepliVAX WNV vaccine (RepliVAX WN) containing a large deletion in the C protein gene [66] was demonstrated to provide complete protection against lethal WT-WNV challenges under a single inoculation at the lowest dose in mouse and hamster models [15,66,103].

2.4. JEV

SA14-14-2, developed in China [6,13,104], is based on the WT strain SA14, which was attenuated by 114 passages in primary hamster kidney cells [49,105]. Studies indicated that SA14-14-2 was highly immunogenic and a two-dose regimen induced almost 100% seroconversion and protection against the infection [106]. SA14-14-2 is the most widely applied LAV against JEV in the world [49,106] and has been successfully utilized in China with more than 100 million doses administered [105]. However, the theoretical risk for the attenuated SA14-14-2 virus to reverse into a highly virulent strain has, to some extent, limited its extensive application globally to prevent JEV infection [106]. ChimeriVax-JE, a chimeric JEV vaccine, employed YFV 17D as the backbone to express the prM and E of SA14-14-2 as the antigens [6,86,106]. Numerous preclinical studies have suggested that the ChimeriVax-JE vaccine was well tolerated and immunogenic, and that it provides protection against JEV infection in mice and non-human primate (NHP) models. The clinical trials indicated that a single dose of ChimeriVax-JE generated almost the same effect as three doses of JE-VAX (an inactivated JEV vaccine) to produce near-complete seroconversion in the subjects [107,108,109,110]. Ishikawa et al. developed a RepliVAX-JE.2 vaccine based on RepliVAX technology, which expresses the JEV prM and E genes in place of the WNV ones. It was indicated that RepliVAX JE.2 was well tolerated and provided full protection for mice suffered from lethal JEV challenges [65,111,112].

2.5. YFV

Yellow fever is the third human infectious disease (after smallpox and rabies) to be controlled by vaccination. A live attenuated YFV (17D), derived from a human isolate (Asibi), has been applied safely and effectively as an LAV for more than 80 years [113], and administered in more than 600 million people around the word [114]. One dose of the vaccine can generate long-lasting specific neutralizing antibodies even for 35 years in some vaccinees, explaining its long-term efficacy [14,114,115]. Another effective YFV LAV (FNV) was produced by attenuating the French viscerotropic strain of YFV in a mouse brain with 128 passages. It was applied in Francophone Africa and showed strong immunogenicity. Unfortunately, FNV was found to increase the neurotropic potential, making it unsafe to utilize in children [14]. FNV was terminated after the last doses were used in 1981 [105,114,116].

3. Inactivated Virus Vaccines

Inactivated virus vaccines (INVs) are chemically or physically inactivated whole virions or subunits of viruses [38,117]. The INVs are impossible to revert to a more pathogenic phenotype while containing the whole viral antigens [27], which can induce a balanced antibody response [62,118].The protection of INVs is mainly based on the ability of viral surface proteins, which can induce neutralizing antibodies. The immunogenicity of INVs is greatly enhanced when the antigen is presented in particulate form (virions or SVPs) [119,120]. However, two major disadvantages of INVs include high expense and the need for repeated vaccination [27], both of which make vaccination difficult to achieve in endemic areas [62].

3.1. DENV

A tetravalent dengue INV was developed by the WRAIR with the GlaxoSmithKline adjuvant systems. This vaccine resulted in robust humoral responses and balanced neutralizing antibody responses for all four DENV serotypes [121].

3.2. ZIKV

A purified inactivated ZIKV vaccine, ZPIV, was produced by inactivating the PRVABC59 strain with formalin and by being given an aluminum adjuvant [27,31,96,122]. One dose of ZPIV protected all mice from challenges with the ZIKV-BR strain. Two doses of ZPIV enabled monkeys to remain immune against ZIKV even after a year of vaccination [96,123]. Moreover, the antibodies from immunized monkeys could block ZIKV infection in mice and monkeys in a dose-dependent manner. Currently, ZIPV has entered a phase I clinical trial [27]. The formalin-inactivated MR766 strain was used to develop the ZIKV INV(PIV) [124]. It has been shown that PIV provided full protection against a lethal ZIKV challenge in AG129 mice models after two-dose administration. Importantly, the serum transferred from immunized rabbits also provided protection in mice models [27,96].

3.3. WNV

A formalin-inactivated whole-virus veterinary vaccine, WNVAX, (The Research Foundation for Microbial diseases of Osaka University, Japan) derived from the WNV-NY99 strain was licensed in 2003. The efficacy experiments showed WNVAX was a safe and effective vaccine [125,126], and even the 3.2 ng/dose WNVAX could provide 100% protection in the WNV NY99-infected C57BL/6N mice [125]. Another inactivated WNV vaccine, HydroVax-001 WNV, based on the Kunjin strain of WNV (WNV-KV, strain CH16532) [117], was inactivated by 3% H2O2 [38,47,117]. Vaccination with HydroVax-001 WNV could elicit robust virus-specific neutralizing antibody responses and reduce WNV-associated mortality in BALB/c mice or viremia in rhesus macaques. HydroVax-II, an advanced WNV INV, was inactivated by H2O2 with Cu2+. This INV induced higher (130-fold) WNV-specific neutralizing antibody responses than HydroVax-001 WNV and protected all BALB/c mice from lethal WNV infection [127].

3.4. JEV

JE-VAX, a licensed INV for JEV, was produced by inactivating the JEV Nakayama strain with formalin [50,128]. The original virus was replicated in a mouse brain, and the inactive virus was purified by ultracentrifugation [129]. This INV was confirmed to induce robust immune response [130,131,132,133]. However, as an INV, a high cost and the need for repeated administration are still limitations. Moreover, JE-VAX was also associated with some serious allergic and neurologic side effects [134,135,136,137,138]. Production of this vaccine ceased in 2006 due to the above drawbacks, and remaining stocks were depleted in 2011 [139,140]. IC51, an investigational INV against JEV, comprises 6 μg of purified, inactivated SA14-14-2 absorbed to 0.1% aluminum hydroxide [104]. It has been demonstrated that IC51 was considerably immunogenic, highly safe and well tolerated [141]. Two doses of the vaccine induced high protective antibody titers equivalent to three doses of JE-VAX [142]. CVI-JE, a freeze-dried INV, was based on the inactivated Beijing P-3 strain of JEV. The high safety and immunogenicity of CVI-JE has been demonstrated in clinical trials and two doses of the vaccine can induce 100% seroconversion rates [48]. More than seven million doses of CVI-JE have been administered in China since it was registered in 2008 [48,106].

4. Nucleic Acid Vaccines

Nucleic acid vaccines generally contain DNA and RNA vaccines [67,75]. DNA vaccines are commonly produced by cloning a promoter and the gene encoding the antigen of the vaccine into a plasmid [27] and they have several advantages over other types of vaccines, including the ability to induce intracellular antigen processing for adaptive immunity [75], the impossibility of the reversion to pathogenic phenotype, stability in extreme temperatures for long periods, and that they are easy to manufacture and low cost [6,62,75,143]. In addition, the DNA vaccines combination is not adversely affected by pre-existing antibodies or the replicative efficiency of each monovalent component [144,145]. However, the possibility of integrating it into the human genome and causing autoimmune diseases makes vaccination with a DNA vaccine a safety risk [6]. Furthermore, the immunogenicity of DNA vaccines is relatively weak in immunized human hosts [143]. Due to the minimal risk of integration into a host genome and a higher safety than DNA vaccines, mRNA technology has been used widely in the development of vaccine for mosquito-borne flavivirus [27,96]. mRNA vaccines utilize the biosynthesis process of host cells to express viral proteins of interest. Moreover, mRNA vaccines are stable in cells due to the untranslated regions at the 5′ and 3′ end, and they can also selectively activate innate immune responses by natural modifications [146,147].

4.1. DENV

The U.S. Army Medical Research and Material Command (AMRDCk, WRAIR, NMRC and Vical Inc., Fort Detrick, MD, USA) cloned four plasmids which encode the E and prM of DENV 1, 2, 3 and 4 into the VR1012 plasmid and produced a tetravalent DENV DNA vaccine (TVDV) [148]. The high safety and good tolerability of TVDV has been demonstrated in clinical trials and it induced dose-dependent anti-DENV IFN-γ responses [69]. Importantly, this DNA vaccine can eliminate some viral interference, which has been noted with some DENV LAVS vaccines [75].

4.2. ZIKV

The DNA vaccine candidate, termed GLS-5700, was the first Zika vaccine to enter clinical trials [27,31]. The GLS-5700 vaccine was developed by integrating the gene sequences of the prM and E of numerous ZIKV strains [96]. The study suggested that the vaccine could induce high levels of neutralizing antibodies and provide solid protection against ZIKV challenges in mice and NHPs [25]. GLS-5700 was well tolerated in clinical trials and three-dose regimens resulted in high-binding antibody titers to ZIKV-prM/E in all subjects. The antibodies of most volunteers could suppress ZIKV infection in vitro [25]. The other two DNA vaccines, VRC5283 [149] and VRC5288 [150], have been used in NHPs and have shown high ratios of seroconversion. The VRC5283 has been advanced into phase II studies in America [27,31,96].
The ZIKV mRNA vaccines, mRNA-1325 [151] and 1839 [152], were produced by enveloping synthetic RNA molecules with lipid nanoparticles. The experiments have shown that mRNA-1839 provided comparable protection, but higher levels of plasma and memory B cells associated with ZIKV-specific neutralizing antibodies, when compared to DNA vaccine VRC5283. These two vaccines have now entered phase I clinical trials [27]. Self-amplifying mRNA (SAM) is a novel technology for the development of mRNA vaccines [153,154]. The SAM vaccine contains an engineered genome of alphavirus responsible for the replication of vaccines and the genes encoding target antigens [155]. A fever dose of a SAM vaccine can induce stronger immune responses due to the replication characteristic and greater stimulation of the adaptive immune response by the double RNA generated during the replication process [156] than other mRNA vaccines [157].

4.3. WNV

A WNV DNA vaccine (pCBWN), containing the genes of viral prM and E, has been shown to result in a strong immune response in horses and full protection in mice when challenged with WNV [158]. Hall et al. developed a plasmid DNA vaccine (pKUN1 plasmid DNA), encoding the full-length RNA of the Kunjin strain. This vaccine was demonstrated to elicit cross-reactive antibodies that could suppress both the New York strain and the Kunjin strain of WNV. The previous results indicated that 0.1–1 μg of pKUN1 plasmid DNA provided solid protection against a lethal challenge with the New York strain of WNV or the Kunjin strain. These data suggest that pKUN1 plasmid DNA may be a promising vaccine candidate to control WNV infection [159].

5. Other Types of Vaccines

Other types of mosquito-borne flaviviral vaccines include viral vector vaccines, subunit protein vaccines and virus-like particle (VLP) vaccines. The live viruses, including replication-deficient adenovirus (AV), replication-competent measles virus (MV) and vaccinia virus [27,96], in viral vector vaccines, enable them to infect host cells and result in robust immune responses [160]. Similarly to LAVs, viral vector vaccines cannot be administered to pregnant women or immunocompromised people due to the risk of reversion to increased virulence [27,161]. Subunit protein vaccines are safer to produce and administer than LAVs and INVs since they do not contain any replicative virus [14,37]. Moreover, subunit protein vaccine technology is particularly required for DENV vaccine development as it can activate the immune responses against different serotypes of DENV [37]. Self-assembly properties of viral structural proteins were used to produce VLP vaccines. VLP vaccines contain multiple genes of target viral structure proteins, while lacking the ability of replication [27], which makes them more immunogenic and safe [27,162,163].

5.1. DENV

Two complex recombinant AV vector vaccines were produced to express the prM and E proteins of DENV-1, 2 (cAdVaxD12) or DENV-3, 4 (cAdVaxD34). The two vaccines could elicit humoral and cellular immune responses against DENV1-4 from 4 to 10 weeks following primary vaccination [75,143].
For the purpose of avoiding the cold chain and the risk of reversion to pathogenicity, researchers continue to be interested in the study of subunit vaccines [14]. A recombinant protein vaccine for DENV(V180), developed by Hawaii biotech (Merck & Co., Inc., Honolulu, HI, USA), comprised the ectodomains of the E protein of DENV1-4 [18,75]. The genes of viral E protein were amplified by RT-PCR methods and then cloned into pMtt1Xho vector. Studies have indicated that V180 induced neutralizing antibodies to all four DENV and protected rhesus macaques from viremia following the WT-viral challenge [75,164] and clinical trials confirmed that the V180 also induced neutralizing antibodies in volunteers [69,75].

5.2. ZIKV

Vesicular stomatitis virus (VSV) is particularly suitable to be developed into a multivalent vaccine due to its compatibility for foreign genes [165,166]. The nature hosts of VSV are livestock; thus, VSV can induce strong systemic immune responses in human populations due to the absence of pre-existing immunity [165,167]. Li et al. developed an attenuated recombinant VSV-based vaccine which generated the prM, E and NS1 of ZIKV. This vaccine candidate induced specific neutralizing antibodies, and T cells mediated immune responses in single-dose immunized mice, which offered full protection against a ZIKV challenge [166]. In addition, AV and MV vector systems are also introduced for ZIKV vaccine development, and are recently entering clinical trials. The AV 26 vector-based vaccine which carries ZIKV M and E (Ad26. ZIKV.001) was demonstrated to possess high immunogenicity. Antibodies from the immunized recipients protected mice against a lethal ZIKV challenge [27,168,169]. A chimpanzee AV system was also applied to generate a ChAdOx1 Zika vaccine, which expressed the prM and E antigen [170]. The efficacy of the vaccine was detected in mice and proved to be 100% protective against ZIKV infection, offering full immunity and reducing viremia and viral dissemination in target organs, such as the brain and ovaries [27]. The AV vectors provide an effective delivery platform due to their minimal immunity in human beings. MV, a live attenuated RNA virus, is among the most secure and efficient human vaccine vectors yet [171]. Two MV vector-based ZIKV vaccine candidates (MV-Zika and MV-ZIKA RSP), which express the prM and E as antigens, are under evaluation in clinical trials. MV-ZIKA RSP was found to be effective in a mouse model to protect the fetus by lowering the viral load during ZIKV infection [172]. A Modified Vaccinia Ankara system (GeoVax) was also utilized to produce a novel vaccine candidate, which delivered ZIKV NS1 as the antigen, perfectly reducing the cross-reacting antibodies and the incidence of antibody-dependent enhancement (ADE) [173]. This vaccine elicited protection against ZIKV challenges in a way that was different from antibody neutralization [27].
To et al. developed a recombinant protein vaccine, which expressed the prM and E of the French Polynesian strain (Accession # KJ776791). The vaccine, in combination with an adjuvant, induced neutralizing antibodies and protected the mice from viremia. [174]. Another recombinant protein vaccine was produced by fusing the E domain III region to the C terminal Fc region of human IgG. Studies have demonstrated that this vaccine could induce strong immune responses and provide solid protection in multiple animal models [175]. More importantly, this vaccine was effective in pregnant CD-1 (ICR) mice models with fetal protection [96,176]. Recently, a ZIKV vaccine expressing the prM, E and NS1 was shown to induce significantly strong immune responses in mice [177].
The first reported vaccine candidate, Zika-VLP, was composed of the constructs that expressed the structural C-prM-E to form the VLP and the nonstructural NS2B-NS3 to catalyze the cleavage of structure proteins, finally producing mature VLP to induce host immunity [178]. The efficacy of the vaccine was verified in mice to induce similar antibody response patterns to the ZIKV INV control. Nevertheless, the amount of specific neutralizing antibodies elicited by the Zika-VLP vaccine was much higher than the ones elicited by INV control. Moreover, no ADE was observed in DENV infection by challenging the Zika-VLP, indicating that dysfunctional responses, such as cross-reacting antibodies induction, were not triggered by the displayed epitopes of Zika-VLP [27]. Immune sera generated from immunized mice were found to protect immunodeficient AG129 mice against ZIKV infection. ZIKVLPs, produced by the University of Wisconsin, was also a vaccine candidate. It could generate potent neutralizing antibodies, reduce viremia in BALB/c mice and elevate the survival rate of AG129 mice after ZIKV challenges [179]. An optimized Zika-VLP vaccine was designated by expressing dimerized E to assemble the VLP of C-prM-E [180]. This optimization helped to develop the envelope dimer epitopes (EDE), which were key to eliciting higher neutralizing antibodies against ZIKV and DENV in mice compared to the VLP with WT sequences [181,182]. The modification was also demonstrated to decrease the ADE risk in in vitro studies [178,183,184]. It was believed that antibodies targeting EDE could prevent viral E from experiencing the conformational change of forming a trimer, thus suppressing viral membrane fusion [185]. VLP-based vaccines are relatively safe due to their inability to replicate in host cells, and will be an ideal option for immunocompromised people or pregnant women [186].

5.3. WNV

The recombinant ALVAC®-WNV vaccine, based on a modified live recombinant canarypox virus (vCP2017) backbone, expressed the prM/E genes of the WNV NY99 strain, and was formulated in a carbomer adjuvant [59,187]. With the absence of replication in humans and a high attenuation, the vaccine is fairly safe [188]. ALVAC®-WNV could elicit WNV-specific neutralizing antibodies in a variety of mammals and provide protection against viremia [189].
MVSchw-sEWNV, a recombinant MV vaccine, expressed the E protein of the WNV IS-98-ST1 strain [171,190]. Vaccination with MVSchw-sEWNV could result in high WNV-specific neutralizing antibody titer and provide protection against a lethal challenge with WNV in CD46-IFNAR mice models. Further experiments indicated that MVSchw-sEWNV was safe and could induce WNV-neutralizing antibodies in squirrel monkeys after a one-dose administration. [190]. Another viral vector vaccine for WNV was developed by cloning the gene sequence of E protein of the WNV LSU-AR01 strain into VSV [191].The vaccine provided 90% protection against the lethal challenge with LSU-AR01 virus in mice models [47].

5.4. YFV

With high safety and immunogenicity, YFV-17D is considered to be the most successful mosquito-borne flavivirus vaccine; however, it has been demonstrated that YFV-17D can cause viscerotropic disease and neurotropic disease, which are life-threatening adverse events [192,193,194,195]. Despite the fact that these side effects are rare, there is still a great need to develop safer and more effective vaccines due to the viruses’ high fatality rate [192,196]. The modified vaccinia virus, Ankara (MVA), is one of the most commonly used antigen delivery vectors that is highly attenuated and cannot replicate in humans. Ju lander et al. cloned the genes of YFV PrM and E into an MVA vector and developed YFV vaccine MVA-BN-YF. Studies indicated that MVA-BN-YF induced robust humoral immune response and provided solid protection comparable to that of YFV-17D in YFV-infected hamsters. Importantly, the sera of immunized hamsters can also protect naïve hamsters from lethal infection wth YFV, confirming that high-titer neutralizing antibodies were elicited by MVA-BN-YF. These data suggest MVA-BN YF may represent a safe alternative to YFV-17D [192].

6. Discussion

Mosquito-borne flaviviruses, some of the most critical human pathogenic arboviruses worldwide, have seriously affected public health in a number of endemic and/or epidemic regions [197]. These viruses cause a broad spectrum of diseases in humans including fever, encephalitis, meningitis and hemorrhage [198]. In recent years, mosquito-borne flavivirus infections have emerged at an alarming rate worldwide [52]. Effective vaccines are still the best approach to control these diseases due to the absence of effective anti-viral drugs.
During recent decades, breakthroughs have been made in the development of flavivirus vaccines (Table 1). The live attenuated YFV-17D and JEV-SA14-14-2 vaccines are regarded as available, safe and efficient mosquito-borne flavivirus preventatives. YFV-17D, in particular, is considered an excellent model to help develop effective vaccines against flaviviruses [199]. As for the other mosquito-borne flaviviruses, Dengvaxia® has been licensed as a dengue vaccine, and many vaccines (TDV, LATV, TDEV, DPIV and TVDV) for DENV have entered clinical trials (Table 1). There are also vaccines for ZIKV (rZIKV/D4Δ30–713, ZIPV, GLS-5700, MV-Zika, MV-Zika RSP and Ad26.ZIKV.001, VCR5283) and WNV (WN/DEN4Δ30), which have been evaluated in clinical trials (Table 1). Nevertheless, there are no effective vaccines against ZIKV and WNV [37] being used in clinics. YFV and JEV vaccines are already available; however, the ongoing vaccination efforts could hardly prevent infection in risk areas due to a lack of financial resources.
With the exception of smallpox (for which humans are the only hosts), the eradication of mosquito-borne flaviviruses might never be realized because of the wide distribution of and the difficulty inherent in controlling mosquitos, which means vaccination against mosquito-borne flaviviruses will be a long and continuous process. Moreover, the administration of some effective vaccines has been restricted because of many factors. For example, YFV-17D and SA14-14-2 are not suitable for immunocompromised people and pregnant women, and Dengvaxia® is only recommended to seropositive persons who are above 9 years old [21,78]. These limitations have led to a significant decrease in vaccine coverage. Furthermore, the development of vaccines against mosquito-borne flaviviruses is very difficult on account of some special requirements. For example, one of the most important target groups for the ZIKV vaccine is pregnant women, which creates a unique safety requirement for the vaccine. In addition, the DENV vaccine must focus on tetravalent formulations due to the absence of cross-protection between the four DENV serotypes. Thus, the successful research and development of a mosquito-borne flavivirus vaccine needs to balance various aspects, including the vaccine components, route of vaccination, target population, expense and social financial resources [6]. Some novel technologies, including ChimeriVax, RepliVax, SAM and subunit protein vaccines, have been used as models to construct more effective vaccines and have shown encouraging results in vitro and in vivo, even in clinical trials. These technologies provide promising prospects for the control of mosquito-borne flaviviruses.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lobigs, M.; Diamond, M.S. Feasibility of cross-protective vaccination against flaviviruses of the Japanese encephalitis serocomplex. Expert Rev. Vaccines 2012, 11, 177–187. [Google Scholar] [CrossRef] [PubMed]
  2. Arenas, M. Protein Evolution in the Flaviviruses. J. Mol. Evol. 2020, 88, 473–476. [Google Scholar] [CrossRef] [PubMed]
  3. Selisko, B.; Wang, C.; Harris, E.; Canard, B. Regulation of Flavivirus RNA synthesis and replication. Curr. Opin. Virol. 2014, 9, 74–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Liu, Z.Y.; Qin, C.F. Structure and function of cis-acting RNA elements of flavivirus. Rev. Med. Virol. 2020, 30, e2092. [Google Scholar] [CrossRef]
  5. Sharma, K.K.; Marzinek, J.K.; Tantirimudalige, S.N.; Bond, P.J.; Wohland, T. Single-molecule studies of flavivirus envelope dynamics: Experiment and computation. Prog. Biophys. Mol. Biol. 2019, 143, 38–51. [Google Scholar] [CrossRef]
  6. Chang, G.J.; Kuno, G.; Purdy, D.E.; Davis, B.S. Recent advancement in flavivirus vaccine development. Expert Rev. Vaccines 2004, 3, 199–220. [Google Scholar] [CrossRef]
  7. Chambers, T.J.; Hahn, C.S.; Galler, R.; Rice, C.M. Flavivirus genome organization, expression, and replication. Annu. Rev. Microbiol. 1990, 44, 649–688. [Google Scholar] [CrossRef]
  8. Brandriss, M.W.; Schlesinger, J.J.; Walsh, E.E.; Briselli, M. Lethal 17D yellow fever encephalitis in mice. I. Passive protection by monoclonal antibodies to the envelope proteins of 17D yellow fever and dengue 2 viruses. J. Gen. Virol. 1986, 67 Pt 2, 229–234. [Google Scholar] [CrossRef]
  9. Cecilia, D.; Gadkari, D.A.; Kedarnath, N.; Ghosh, S.N. Epitope mapping of Japanese encephalitis virus envelope protein using monoclonal antibodies against an Indian strain. J. Gen. Virol. 1988, 69 Pt 11, 2741–2747. [Google Scholar] [CrossRef]
  10. Zhang, M.J.; Wang, M.J.; Jiang, S.Z.; Ma, W.Y. Passive protection of mice, goats, and monkeys against Japanese encephalitis with monoclonal antibodies. J. Med. Virol. 1989, 29, 133–138. [Google Scholar] [CrossRef]
  11. Kreil, T.R.; Maier, E.; Fraiss, S.; Eibl, M.M. Neutralizing antibodies protect against lethal flavivirus challenge but allow for the development of active humoral immunity to a nonstructural virus protein. J. Virol. 1998, 72, 3076–3081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Qian, X.; Qi, Z. Mosquito-Borne Flaviviruses and Current Therapeutic Advances. Viruses 2022, 14, 1226. [Google Scholar] [CrossRef] [PubMed]
  13. Heinz, F.X.; Stiasny, K. Flaviviruses and flavivirus vaccines. Vaccine 2012, 30, 4301–4306. [Google Scholar] [CrossRef] [PubMed]
  14. Collins, M.H.; Metz, S.W. Progress and Works in Progress: Update on Flavivirus Vaccine Development. Clin. Ther. 2017, 39, 1519–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Widman, D.G.; Frolov, I.; Mason, P.W. Third-generation flavivirus vaccines based on single-cycle, encapsidation-defective viruses. Adv. Virus Res. 2008, 72, 77–126. [Google Scholar] [PubMed]
  16. Lazo, L.; Valdes, I.; Guillén, G.; Hermida, L.; Gil, L. Aiming at the heart: The capsid protein of dengue virus as a vaccine candidate. Expert Rev. Vaccines 2019, 18, 161–173. [Google Scholar] [CrossRef] [PubMed]
  17. Galula, J.U.; Salem, G.M.; Chang, G.J.; Chao, D.Y. Does structurally-mature dengue virion matter in vaccine preparation in post-Dengvaxia era? Hum. Vaccin. Immunother. 2019, 15, 2328–2336. [Google Scholar] [CrossRef] [Green Version]
  18. Murphy, D.; Reche, P.; Flower, D.R. Selection-based design of in silico dengue epitope ensemble vaccines. Chem. Biol. Drug. Des. 2019, 93, 21–28. [Google Scholar] [CrossRef]
  19. Bhatt, S.; Gething, P.W.; Brady, O.J.; Messina, J.P.; Farlow, A.W.; Moyes, C.L.; Drake, J.M.; Brownstein, J.S.; Hoen, A.G.; Sankoh, O.; et al. The global distribution and burden of dengue. Nature 2013, 496, 504–507. [Google Scholar] [CrossRef] [Green Version]
  20. Blaney, J.E., Jr.; Matro, J.M.; Murphy, B.R.; Whitehead, S.S. Recombinant, live-attenuated tetravalent dengue virus vaccine formulations induce a balanced, broad, and protective neutralizing antibody response against each of the four serotypes in rhesus monkeys. J. Virol. 2005, 79, 5516–5528. [Google Scholar] [CrossRef] [Green Version]
  21. Fongwen, N.; Delrieu, I.; Ham, L.H.; Gubler, D.J.; Durbin, A.; Ooi, E.E.; Peeling, R.W.; Flasche, S.; Hartigan-Go, K.; Clifford, S.; et al. Implementation strategies for the first licensed dengue vaccine: A meeting report. Vaccine 2021, 39, 4759–4765. [Google Scholar] [CrossRef] [PubMed]
  22. Stanaway, J.D.; Shepard, D.S.; Undurraga, E.A.; Halasa, Y.A.; Coffeng, L.E.; Brady, O.J.; Hay, S.I.; Bedi, N.; Bensenor, I.M.; Castañeda-Orjuela, C.A.; et al. The global burden of dengue: An analysis from the Global Burden of Disease Study 2013. Lancet Infect. Dis. 2016, 16, 712–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Burgess, C.; Nelis, L.; Huang, C. Modeling Zika Vaccination Combined With Vector Interventions in DoD Populations. Mil. Med. 2021, 186, 82–90. [Google Scholar] [CrossRef] [PubMed]
  24. Lima, T.M.; Souza, M.O.; Castilho, L.R. Purification of flavivirus VLPs by a two-step chomatographic process. Vaccine 2019, 37, 7061–7069. [Google Scholar] [CrossRef]
  25. Kudchodkar, S.B.; Choi, H.; Reuschel, E.L.; Esquivel, R.; Jin-Ah Kwon, J.; Jeong, M.; Maslow, J.N.; Reed, C.C.; White, S.; Kim, J.J.; et al. Rapid response to an emerging infectious disease—Lessons learned from development of a synthetic DNA vaccine targeting Zika virus. Microbes Infect. 2018, 20, 676–684. [Google Scholar] [CrossRef]
  26. Whitehead, S.S.; Subbarao, K. Which Dengue Vaccine Approach Is the Most Promising, and Should We Be Concerned about Enhanced Disease after Vaccination? The Risks of Incomplete Immunity to Dengue Virus Revealed by Vaccination. Cold Spring Harb. Perspect Biol. 2018, 10, a028811. [Google Scholar] [CrossRef] [Green Version]
  27. Cimica, V.; Galarza, J.M.; Rashid, S.; Stedman, T.T. Current development of Zika virus vaccines with special emphasis on virus-like particle technology. Expert Rev. Vaccines 2021, 20, 1483–1498. [Google Scholar] [CrossRef]
  28. Messina, J.P.; Kraemer, M.U.; Brady, O.J.; Pigott, D.M.; Shearer, F.M.; Weiss, D.J.; Golding, N.; Ruktanonchai, C.W.; Gething, P.W.; Cohn, E.; et al. Mapping global environmental suitability for Zika virus. eLife 2016, 5, e15272. [Google Scholar] [CrossRef]
  29. Kraemer, M.U.; Sinka, M.E.; Duda, K.A.; Mylne, A.Q.; Shearer, F.M.; Barker, C.M.; Moore, C.G.; Carvalho, R.G.; Coelho, G.E.; Van Bortel, W.; et al. The global distribution of the arbovirus vectors Aedes aegypti and Ae. albopictus. eLife 2015, 4, e08347. [Google Scholar] [CrossRef]
  30. Thomas, S.J.; Barrett, A. Zika vaccine pre-clinical and clinical data review with perspectives on the future development. Hum. Vaccin. Immunother. 2020, 16, 2524–2536. [Google Scholar] [CrossRef]
  31. Vress, D. Future vaccines in pregnancy. Best Pract. Res. Clin. Obstet. Gynaecol. 2021, 76, 96–106. [Google Scholar] [CrossRef] [PubMed]
  32. In, H.J.; Lee, Y.H.; Jang, S.; Lim, H.J.; Kim, M.Y.; Kim, J.A.; Yoo, J.S.; Chung, G.T.; Kim, Y.J. Enhanced effect of modified Zika virus E antigen on the immunogenicity of DNA vaccine. Virology 2020, 549, 25–31. [Google Scholar] [CrossRef] [PubMed]
  33. Brasil, P.; Pereira, J.P., Jr.; Moreira, M.E.; Ribeiro Nogueira, R.M.; Damasceno, L.; Wakimoto, M.; Rabello, R.S.; Valderramos, S.G.; Halai, U.A.; Salles, T.S.; et al. Zika Virus Infection in Pregnant Women in Rio de Janeiro. N. Engl. J. Med. 2016, 375, 2321–2334. [Google Scholar] [CrossRef] [PubMed]
  34. Guy, B.; Guirakhoo, F.; Barban, V.; Higgs, S.; Monath, T.P.; Lang, J. Preclinical and clinical development of YFV 17D-based chimeric vaccines against dengue, West Nile and Japanese encephalitis viruses. Vaccine 2010, 28, 632–649. [Google Scholar] [CrossRef]
  35. Minke, J.M.; Siger, L.; Cupillard, L.; Powers, B.; Bakonyi, T.; Boyum, S.; Nowotny, N.; Bowen, R. Protection provided by a recombinant ALVAC(®)-WNV vaccine expressing the prM/E genes of a lineage 1 strain of WNV against a virulent challenge with a lineage 2 strain. Vaccine 2011, 29, 4608–4612. [Google Scholar] [CrossRef]
  36. Long, M.T.; Gibbs, E.P.; Mellencamp, M.W.; Bowen, R.A.; Seino, K.K.; Zhang, S.; Beachboard, S.E.; Humphrey, P.P. Efficacy, duration, and onset of immunogenicity of a West Nile virus vaccine, live Flavivirus chimera, in horses with a clinical disease challenge model. Equine Vet. J. 2007, 39, 491–497. [Google Scholar] [CrossRef]
  37. Martínez, C.A.; Giulietti, A.M.; Talou, J.R. Research advances in plant-made flavivirus antigens. Biotechnol. Adv. 2012, 30, 1493–1505. [Google Scholar] [CrossRef]
  38. Ulbert, S.; Magnusson, S.E. Technologies for the development of West Nile virus vaccines. Future Microbiol. 2014, 9, 1221–1232. [Google Scholar] [CrossRef] [Green Version]
  39. Widman, D.G.; Ishikawa, T.; Fayzulin, R.; Bourne, N.; Mason, P.W. Construction and characterization of a second-generation pseudoinfectious West Nile virus vaccine propagated using a new cultivation system. Vaccine 2008, 26, 2762–2771. [Google Scholar] [CrossRef]
  40. Hayes, E.B.; Sejvar, J.J.; Zaki, S.R.; Lanciotti, R.S.; Bode, A.V.; Campbell, G.L. Virology, pathology, and clinical manifestations of West Nile virus disease. Emerg. Infect. Dis. 2005, 11, 1174–1179. [Google Scholar] [CrossRef]
  41. Papa, A. West Nile virus infections in humans–focus on Greece. J. Clin. Virol. 2013, 58, 351–353. [Google Scholar] [CrossRef] [PubMed]
  42. Intrauterine West Nile virus infection–New York, 2002. MMWR Morb. Mortal. Wkly. Rep. 2002, 51, 1135–1136.
  43. Possible West Nile virus transmission to an infant through breast-feeding--Michigan, 2002. MMWR Morb. Mortal. Wkly. Rep. 2002, 51, 877–878.
  44. Hayes, E.B.; O’Leary, D.R. West Nile virus infection: A pediatric perspective. Pediatrics 2004, 113, 1375–1381. [Google Scholar] [CrossRef] [PubMed]
  45. Investigations of West Nile virus infections in recipients of blood transfusions. MMWR Morb. Mortal. Wkly. Rep. 2002, 51, 973–974.
  46. Detection of West Nile virus in blood donations--United States, 2003. MMWR Morb. Mortal. Wkly. Rep. 2003, 52, 769–772.
  47. Iyer, A.V.; Kousoulas, K.G. A review of vaccine approaches for West Nile virus. Int. J. Environ. Res. Public Health 2013, 10, 4200–4223. [Google Scholar] [CrossRef]
  48. Chanthavanich, P.; Limkittikul, K.; Sirivichayakul, C.; Chokejindachai, W.; Hattasingh, W.; Pengsaa, K.; Surangsrirat, S.; Srisuwannaporn, T.; Kaewma, B.; Yoksan, S.; et al. Immunogenicity and safety of inactivated chromatographically purified Vero cell-derived Japanese encephalitis vaccine in Thai children. Hum. Vaccin. Immunother. 2018, 14, 900–905. [Google Scholar] [CrossRef]
  49. Liu, X.; Zhao, D.; Jia, L.; Xu, H.; Na, R.; Ge, Y.; Liu, S.; Yu, Y.; Li, Y. Genetic and neuroattenuation phenotypic characteristics and their stabilities of SA14-14-2 vaccine seed virus. Vaccine 2018, 36, 4650–4656. [Google Scholar] [CrossRef]
  50. Endy, T.P.; Nisalak, A. Japanese encephalitis virus: Ecology and epidemiology. Curr. Top. Microbiol. Immunol. 2002, 267, 11–48. [Google Scholar]
  51. Mackenzie, J.S.; Gubler, D.J.; Petersen, L.R. Emerging flaviviruses: The spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nat. Med. 2004, 10, S98–S109. [Google Scholar] [CrossRef] [PubMed]
  52. Pugachev, K.V.; Guirakhoo, F.; Monath, T.P. New developments in flavivirus vaccines with special attention to yellow fever. Curr. Opin. Infect. Dis. 2005, 18, 387–394. [Google Scholar] [CrossRef] [PubMed]
  53. Qian, X.; Wu, B.; Tang, H.; Luo, Z.; Xu, Z.; Ouyang, S.; Li, X.; Xie, J.; Yi, Z.; Leng, Q.; et al. Rifapentine is an entry and replication inhibitor against yellow fever virus both in vitro and in vivo. Emerg. Microbes Infect. 2022, 11, 873–884. [Google Scholar] [CrossRef] [PubMed]
  54. Paules, C.I.; Fauci, A.S. Yellow Fever—Once Again on the Radar Screen in the Americas. N. Engl. J. Med. 2017, 376, 1397–1399. [Google Scholar] [CrossRef]
  55. Furuta, Y.; Takahashi, K.; Shiraki, K.; Sakamoto, K.; Smee, D.F.; Barnard, D.L.; Gowen, B.B.; Julander, J.G.; Morrey, J.D. T-705 (favipiravir) and related compounds: Novel broad-spectrum inhibitors of RNA viral infections. Antiviral. Res. 2009, 82, 95–102. [Google Scholar] [CrossRef]
  56. de Freitas, C.S.; Higa, L.M.; Sacramento, C.Q.; Ferreira, A.C.; Reis, P.A.; Delvecchio, R.; Monteiro, F.L.; Barbosa-Lima, G.; James Westgarth, H.; Vieira, Y.R.; et al. Yellow fever virus is susceptible to sofosbuvir both in vitro and in vivo. PLoS Negl. Trop. Dis. 2019, 13, e0007072. [Google Scholar] [CrossRef] [Green Version]
  57. Liu, B.; Li, N.L.; Wang, J.; Shi, P.Y.; Wang, T.; Miller, M.A.; Li, K. Overlapping and distinct molecular determinants dictating the antiviral activities of TRIM56 against flaviviruses and coronavirus. J. Virol. 2014, 88, 13821–13835. [Google Scholar] [CrossRef] [Green Version]
  58. Yang, D.; Li, N.L.; Wei, D.; Liu, B.; Guo, F.; Elbahesh, H.; Zhang, Y.; Zhou, Z.; Chen, G.Y.; Li, K. The E3 ligase TRIM56 is a host restriction factor of Zika virus and depends on its RNA-binding activity but not miRNA regulation, for antiviral function. PLoS Negl. Trop. Dis. 2019, 13, e0007537. [Google Scholar] [CrossRef] [Green Version]
  59. Ray, D.; Shi, P.Y. Recent advances in flavivirus antiviral drug discovery and vaccine development. Recent Pat. AntiInfect. Drug Discov. 2006, 1, 45–55. [Google Scholar] [CrossRef]
  60. Pugachev, K.V.; Guirakhoo, F.; Trent, D.W.; Monath, T.P. Traditional and novel approaches to flavivirus vaccines. Int. J. Parasitol. 2003, 33, 567–582. [Google Scholar] [CrossRef]
  61. Kofler, R.M.; Aberle, J.H.; Aberle, S.W.; Allison, S.L.; Heinz, F.X.; Mandl, C.W. Mimicking live flavivirus immunization with a noninfectious RNA vaccine. Proc. Natl. Acad. Sci. USA 2004, 101, 1951–1956. [Google Scholar] [CrossRef] [Green Version]
  62. Wilder-Smith, A.; Deen, J.L. Dengue vaccines for travelers. Expert Rev. Vaccines 2008, 7, 569–578. [Google Scholar] [CrossRef] [PubMed]
  63. Monath, T.P.; Kanesa-Thasan, N.; Guirakhoo, F.; Pugachev, K.; Almond, J.; Lang, J.; Quentin-Millet, M.J.; Barrett, A.D.; Brinton, M.A.; Cetron, M.S.; et al. Recombination and flavivirus vaccines: A commentary. Vaccine 2005, 23, 2956–2958. [Google Scholar] [CrossRef] [PubMed]
  64. Rumyantsev, A.A.; Giel-Moloney, M.; Liu, Y.; Gao, Q.S.; Zhang, Z.X.; Catalan, J.; Frolov, I.; Almond, J.; Kleanthous, H.; Pugachev, K.V. Characterization of the RepliVax platform for replication-defective flavivirus vaccines. Vaccine 2011, 29, 5184–5194. [Google Scholar] [CrossRef] [PubMed]
  65. Ishikawa, T.; Widman, D.G.; Bourne, N.; Konishi, E.; Mason, P.W. Construction and evaluation of a chimeric pseudoinfectious virus vaccine to prevent Japanese encephalitis. Vaccine 2008, 26, 2772–2781. [Google Scholar] [CrossRef]
  66. Widman, D.G.; Ishikawa, T.; Giavedoni, L.D.; Hodara, V.L.; Garza Mde, L.; Montalbo, J.A.; Travassos Da Rosa, A.P.; Tesh, R.B.; Patterson, J.L.; Carrion, R., Jr.; et al. Evaluation of RepliVAX WN, a single-cycle flavivirus vaccine, in a non-human primate model of West Nile virus infection. Am. J. Trop. Med. Hyg. 2010, 82, 1160–1167. [Google Scholar] [CrossRef] [Green Version]
  67. Mandl, C.W. Flavivirus immunization with capsid-deletion mutants: Basics, benefits, and barriers. Viral. Immunol. 2004, 17, 461–472. [Google Scholar] [CrossRef]
  68. Wilder-Smith, A.; Hombach, J.; Ferguson, N.; Selgelid, M.; O’Brien, K.; Vannice, K.; Barrett, A.; Ferdinand, E.; Flasche, S.; Guzman, M.; et al. Deliberations of the Strategic Advisory Group of Experts on Immunization on the use of CYD-TDV dengue vaccine. Lancet Infect. Dis. 2019, 19, e31–e38. [Google Scholar] [CrossRef]
  69. Pinheiro-Michelsen, J.R.; Souza, R.; Santana, I.V.R.; da Silva, P.S.; Mendez, E.C.; Luiz, W.B.; Amorim, J.H. Anti-dengue Vaccines: From Development to Clinical Trials. Front. Immunol. 2020, 11, 1252. [Google Scholar] [CrossRef]
  70. Michlmayr, D.; Andrade, P.; Nascimento, E.J.M.; Parker, A.; Narvekar, P.; Dean, H.J.; Harris, E. Characterization of the Type-Specific and Cross-Reactive B-Cell Responses Elicited by a Live-Attenuated Tetravalent Dengue Vaccine. J. Infect. Dis. 2021, 223, 247–257. [Google Scholar] [CrossRef]
  71. Redoni, M.; Yacoub, S.; Rivino, L.; Giacobbe, D.R.; Luzzati, R.; Di Bella, S. Dengue: Status of current and under-development vaccines. Rev. Med. Virol. 2020, 30, e2101. [Google Scholar] [CrossRef] [PubMed]
  72. Robinson, M.L.; Durbin, A.P. Dengue vaccines: Implications for dengue control. Curr. Opin. Infect. Dis. 2017, 30, 449–454. [Google Scholar] [CrossRef] [PubMed]
  73. Wilder-Smith, A.; Ooi, E.E.; Horstick, O.; Wills, B. Dengue. Lancet 2019, 393, 350–363. [Google Scholar] [CrossRef] [PubMed]
  74. Guirakhoo, F.; Pugachev, K.; Zhang, Z.; Myers, G.; Levenbook, I.; Draper, K.; Lang, J.; Ocran, S.; Mitchell, F.; Parsons, M.; et al. Safety and efficacy of chimeric yellow Fever-dengue virus tetravalent vaccine formulations in nonhuman primates. J. Virol. 2004, 78, 4761–4775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Durbin, A.P.; Whitehead, S.S. Dengue vaccine candidates in development. Curr. Top. Microbiol. Immunol. 2010, 338, 129–143. [Google Scholar] [PubMed]
  76. Mantel, N.; Girerd, Y.; Geny, C.; Bernard, I.; Pontvianne, J.; Lang, J.; Barban, V. Genetic stability of a dengue vaccine based on chimeric yellow fever/dengue viruses. Vaccine 2011, 29, 6629–6635. [Google Scholar] [CrossRef]
  77. Poo, J.; Galan, F.; Forrat, R.; Zambrano, B.; Lang, J.; Dayan, G. Live-attenuated Tetravalent Dengue Vaccine in Dengue-naïve Children, Adolescents, and Adults in Mexico City: Randomized Controlled Phase 1 Trial of Safety and Immunogenicity. Pediatr. Infect. Dis. J. 2011, 30, e9–e17. [Google Scholar] [CrossRef]
  78. López-Medina, E.; Biswal, S.; Saez-Llorens, X.; Borja-Tabora, C.; Bravo, L.; Sirivichayakul, C.; Vargas, L.M.; Alera, M.T.; Velásquez, H.; Reynales, H.; et al. Efficacy of a Dengue Vaccine Candidate (TAK-003) in Healthy Children and Adolescents 2 Years after Vaccination. J. Infect. Dis. 2022, 225, 1521–1532. [Google Scholar] [CrossRef]
  79. Chu, H.; George, S.L.; Stinchcomb, D.T.; Osorio, J.E.; Partidos, C.D. CD8+ T-cell Responses in Flavivirus-Naive Individuals Following Immunization with a Live-Attenuated Tetravalent Dengue Vaccine Candidate. J. Infect. Dis. 2015, 212, 1618–1628. [Google Scholar] [CrossRef] [Green Version]
  80. Bhamarapravati, N.; Yoksan, S.; Chayaniyayothin, T.; Angsubphakorn, S.; Bunyaratvej, A. Immunization with a live attenuated dengue-2-virus candidate vaccine (16681-PDK 53): Clinical, immunological and biological responses in adult volunteers. Bull. World Health Organ. 1987, 65, 189–195. [Google Scholar]
  81. Kanesa-thasan, N.; Sun, W.; Kim-Ahn, G.; Van Albert, S.; Putnak, J.R.; King, A.; Raengsakulsrach, B.; Christ-Schmidt, H.; Gilson, K.; Zahradnik, J.M.; et al. Safety and immunogenicity of attenuated dengue virus vaccines (Aventis Pasteur) in human volunteers. Vaccine 2001, 19, 3179–3188. [Google Scholar] [CrossRef] [PubMed]
  82. Vaughn, D.W.; Hoke, C.H., Jr.; Yoksan, S.; LaChance, R.; Innis, B.L.; Rice, R.M.; Bhamarapravati, N. Testing of a dengue 2 live-attenuated vaccine (strain 16681 PDK 53) in ten American volunteers. Vaccine 1996, 14, 329–336. [Google Scholar] [CrossRef] [PubMed]
  83. Wiggan, O.; Livengood, J.A.; Silengo, S.J.; Kinney, R.M.; Osorio, J.E.; Huang, C.Y.; Stinchcomb, D.T. Novel formulations enhance the thermal stability of live-attenuated flavivirus vaccines. Vaccine 2011, 29, 7456–7462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Dharakul, T.; Kurane, I.; Bhamarapravati, N.; Yoksan, S.; Vaughn, D.W.; Hoke, C.H.; Ennis, F.A. Dengue virus-specific memory T cell responses in human volunteers receiving a live attenuated dengue virus type 2 candidate vaccine. J. Infect. Dis. 1994, 170, 27–33. [Google Scholar] [CrossRef] [PubMed]
  85. Rothman, A.L.; Kanesa-thasan, N.; West, K.; Janus, J.; Saluzzo, J.F.; Ennis, F.A. Induction of T lymphocyte responses to dengue virus by a candidate tetravalent live attenuated dengue virus vaccine. Vaccine 2001, 19, 4694–4699. [Google Scholar] [CrossRef]
  86. Hombach, J.; Barrett, A.D.; Cardosa, M.J.; Deubel, V.; Guzman, M.; Kurane, I.; Roehrig, J.T.; Sabchareon, A.; Kieny, M.P. Review on flavivirus vaccine development. Proceedings of a meeting jointly organised by the World Health Organization and the Thai Ministry of Public Health, 26-27 April 2004, Bangkok, Thailand. Vaccine 2005, 23, 2689–2695. [Google Scholar] [CrossRef]
  87. Sáez-Llorens, X.; Tricou, V.; Yu, D.; Rivera, L.; Tuboi, S.; Garbes, P.; Borkowski, A.; Wallace, D. Safety and immunogenicity of one versus two doses of Takeda’s tetravalent dengue vaccine in children in Asia and Latin America: Interim results from a phase 2, randomised, placebo-controlled study. Lancet Infect. Dis. 2017, 17, 615–625. [Google Scholar] [CrossRef]
  88. Osorio, J.E.; Velez, I.D.; Thomson, C.; Lopez, L.; Jimenez, A.; Haller, A.A.; Silengo, S.; Scott, J.; Boroughs, K.L.; Stovall, J.L.; et al. Safety and immunogenicity of a recombinant live attenuated tetravalent dengue vaccine (DENVax) in flavivirus-naive healthy adults in Colombia: A randomised, placebo-controlled, phase 1 study. Lancet Infect. Dis. 2014, 14, 830–838. [Google Scholar] [CrossRef] [Green Version]
  89. Silveira, C.G.T.; Magnani, D.M.; Costa, P.R.; Avelino-Silva, V.I.; Ricciardi, M.J.; Timenetsky, M.; Goulart, R.; Correia, C.A.; Marmorato, M.P.; Ferrari, L.; et al. Plasmablast Expansion Following the Tetravalent, Live-Attenuated Dengue Vaccine Butantan-DV in DENV-Naïve and DENV-Exposed Individuals in a Brazilian Cohort. Front. Immunol. 2022, 13, 908398. [Google Scholar] [CrossRef]
  90. George, S.L.; Wong, M.A.; Dube, T.J.; Boroughs, K.L.; Stovall, J.L.; Luy, B.E.; Haller, A.A.; Osorio, J.E.; Eggemeyer, L.M.; Irby-Moore, S.; et al. Safety and Immunogenicity of a Live Attenuated Tetravalent Dengue Vaccine Candidate in Flavivirus-Naive Adults: A Randomized, Double-Blinded Phase 1 Clinical Trial. J. Infect. Dis. 2015, 212, 1032–1041. [Google Scholar] [CrossRef] [Green Version]
  91. Jackson, L.A.; Rupp, R.; Papadimitriou, A.; Wallace, D.; Raanan, M.; Moss, K.J. A phase 1 study of safety and immunogenicity following intradermal administration of a tetravalent dengue vaccine candidate. Vaccine 2018, 36, 3976–3983. [Google Scholar] [CrossRef]
  92. Sirivichayakul, C.; Barranco-Santana, E.A.; Esquilin-Rivera, I.; Oh, H.M.; Raanan, M.; Sariol, C.A.; Shek, L.P.; Simasathien, S.; Smith, M.K.; Velez, I.D.; et al. Safety and Immunogenicity of a Tetravalent Dengue Vaccine Candidate in Healthy Children and Adults in Dengue-Endemic Regions: A Randomized, Placebo-Controlled Phase 2 Study. J. Infect. Dis. 2016, 213, 1562–1572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Turner, M.; Papadimitriou, A.; Winkle, P.; Segall, N.; Levin, M.; Doust, M.; Johnson, C.; Lucksinger, G.; Fierro, C.; Pickrell, P.; et al. Immunogenicity and safety of lyophilized and liquid dengue tetravalent vaccine candidate formulations in healthy adults: A randomized, phase 2 clinical trial. Hum. Vaccin. Immunother. 2020, 16, 2456–2464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Biswal, S.; Borja-Tabora, C.; Martinez Vargas, L.; Velásquez, H.; Theresa Alera, M.; Sierra, V.; Johana Rodriguez-Arenales, E.; Yu, D.; Wickramasinghe, V.P.; Duarte Moreira, E., Jr.; et al. Efficacy of a tetravalent dengue vaccine in healthy children aged 4-16 years: A randomised, placebo-controlled, phase 3 trial. Lancet 2020, 395, 1423–1433. [Google Scholar] [CrossRef]
  95. Li, X.F.; Dong, H.L.; Wang, H.J.; Huang, X.Y.; Qiu, Y.F.; Ji, X.; Ye, Q.; Li, C.; Liu, Y.; Deng, Y.Q.; et al. Development of a chimeric Zika vaccine using a licensed live-attenuated flavivirus vaccine as backbone. Nat. Commun. 2018, 9, 673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Garg, H.; Mehmetoglu-Gurbuz, T.; Joshi, A. Recent Advances in Zika Virus Vaccines. Viruses 2018, 10, 631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Li, X.F.; Zhao, W.; Lin, F.; Ye, Q.; Wang, H.J.; Yang, D.; Li, S.H.; Zhao, H.; Xu, Y.P.; Ma, J.; et al. Development of chimaeric West Nile virus attenuated vaccine candidate based on the Japanese encephalitis vaccine strain SA14-14-2. J. Gen. Virol. 2013, 94, 2700–2709. [Google Scholar] [CrossRef] [Green Version]
  98. Durbin, A.P.; Wright, P.F.; Cox, A.; Kagucia, W.; Elwood, D.; Henderson, S.; Wanionek, K.; Speicher, J.; Whitehead, S.S.; Pletnev, A.G. The live attenuated chimeric vaccine rWN/DEN4Δ30 is well-tolerated and immunogenic in healthy flavivirus-naïve adult volunteers. Vaccine 2013, 31, 5772–5777. [Google Scholar] [CrossRef] [Green Version]
  99. Pletnev, A.G.; Swayne, D.E.; Speicher, J.; Rumyantsev, A.A.; Murphy, B.R. Chimeric West Nile/dengue virus vaccine candidate: Preclinical evaluation in mice, geese and monkeys for safety and immunogenicity. Vaccine 2006, 24, 6392–6404. [Google Scholar] [CrossRef]
  100. Monath, T.P.; Liu, J.; Kanesa-Thasan, N.; Myers, G.A.; Nichols, R.; Deary, A.; McCarthy, K.; Johnson, C.; Ermak, T.; Shin, S.; et al. A live, attenuated recombinant West Nile virus vaccine. Proc. Natl. Acad. Sci. USA 2006, 103, 6694–6699. [Google Scholar] [CrossRef] [Green Version]
  101. Arroyo, J.; Miller, C.; Catalan, J.; Myers, G.A.; Ratterree, M.S.; Trent, D.W.; Monath, T.P. ChimeriVax-West Nile virus live-attenuated vaccine: Preclinical evaluation of safety, immunogenicity, and efficacy. J. Virol. 2004, 78, 12497–12507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Biedenbender, R.; Bevilacqua, J.; Gregg, A.M.; Watson, M.; Dayan, G. Phase II, randomized, double-blind, placebo-controlled, multicenter study to investigate the immunogenicity and safety of a West Nile virus vaccine in healthy adults. J. Infect. Dis. 2011, 203, 75–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Widman, D.G.; Ishikawa, T.; Winkelmann, E.R.; Infante, E.; Bourne, N.; Mason, P.W. RepliVAX WN, a single-cycle flavivirus vaccine to prevent West Nile disease, elicits durable protective immunity in hamsters. Vaccine 2009, 27, 5550–5553. [Google Scholar] [CrossRef]
  104. Schuller, E.; Klade, C.S.; Wölfl, G.; Kaltenböck, A.; Dewasthaly, S.; Tauber, E. Comparison of a single, high-dose vaccination regimen to the standard regimen for the investigational Japanese encephalitis vaccine, IC51: A randomized, observer-blind, controlled Phase 3 study. Vaccine 2009, 27, 2188–2193. [Google Scholar] [CrossRef]
  105. Barrett, A.D. Current status of flavivirus vaccines. Ann. N. Y. Acad. Sci. 2001, 951, 262–271. [Google Scholar] [CrossRef] [PubMed]
  106. Yun, S.I.; Lee, Y.M. Japanese encephalitis: The virus and vaccines. Hum. Vaccin. Immunother. 2014, 10, 263–279. [Google Scholar] [CrossRef] [Green Version]
  107. Monath, T.P.; Myers, G.A.; Beck, R.A.; Knauber, M.; Scappaticci, K.; Pullano, T.; Archambault, W.T.; Catalan, J.; Miller, C.; Zhang, Z.X.; et al. Safety testing for neurovirulence of novel live, attenuated flavivirus vaccines: Infant mice provide an accurate surrogate for the test in monkeys. Biologicals 2005, 33, 131–144. [Google Scholar] [CrossRef]
  108. Arroyo, J.; Guirakhoo, F.; Fenner, S.; Zhang, Z.X.; Monath, T.P.; Chambers, T.J. Molecular basis for attenuation of neurovirulence of a yellow fever Virus/Japanese encephalitis virus chimera vaccine (ChimeriVax-JE). J. Virol. 2001, 75, 934–942. [Google Scholar] [CrossRef] [Green Version]
  109. Monath, T.P.; Levenbook, I.; Soike, K.; Zhang, Z.X.; Ratterree, M.; Draper, K.; Barrett, A.D.; Nichols, R.; Weltzin, R.; Arroyo, J.; et al. Chimeric yellow fever virus 17D-Japanese encephalitis virus vaccine: Dose-response effectiveness and extended safety testing in rhesus monkeys. J. Virol. 2000, 74, 1742–1751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Torresi, J.; McCarthy, K.; Feroldi, E.; Méric, C. Immunogenicity, safety and tolerability in adults of a new single-dose, live-attenuated vaccine against Japanese encephalitis: Randomised controlled phase 3 trials. Vaccine 2010, 28, 7993–8000. [Google Scholar] [CrossRef]
  111. Mason, P.W.; Pincus, S.; Fournier, M.J.; Mason, T.L.; Shope, R.E.; Paoletti, E. Japanese encephalitis virus-vaccinia recombinants produce particulate forms of the structural membrane proteins and induce high levels of protection against lethal JEV infection. Virology 1991, 180, 294–305. [Google Scholar] [CrossRef] [PubMed]
  112. Mason, P.W.; Shustov, A.V.; Frolov, I. Production and characterization of vaccines based on flaviviruses defective in replication. Virology 2006, 351, 432–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Lang, J.; Zuckerman, J.; Clarke, P.; Barrett, P.; Kirkpatrick, C.; Blondeau, C. Comparison of the immunogenicity and safety of two 17D yellow fever vaccines. Am. J. Trop. Med. Hyg. 1999, 60, 1045–1050. [Google Scholar] [CrossRef] [Green Version]
  114. Liang, H.; Lee, M.; Jin, X. Guiding dengue vaccine development using knowledge gained from the success of the yellow fever vaccine. Cell. Mol. Immunol. 2016, 13, 36–46. [Google Scholar] [CrossRef] [Green Version]
  115. Burkhard, J.; Ciurea, A.; Gabay, C.; Hasler, P.; Müller, R.; Niedrig, M.; Fehr, J.; Villiger, P.; Visser, L.G.; de Visser, A.W.; et al. Long-term immunogenicity after yellow fever vaccination in immunosuppressed and healthy individuals. Vaccine 2020, 38, 3610–3617. [Google Scholar] [CrossRef]
  116. Roukens, A.H.; Gelinck, L.B.; Visser, L.G. Intradermal vaccination to protect against yellow fever and influenza. Curr. Top. Microbiol. Immunol. 2012, 351, 159–179. [Google Scholar]
  117. Poore, E.A.; Slifka, D.K.; Raué, H.P.; Thomas, A.; Hammarlund, E.; Quintel, B.K.; Torrey, L.L.; Slifka, A.M.; Richner, J.M.; Dubois, M.E.; et al. Pre-clinical development of a hydrogen peroxide-inactivated West Nile virus vaccine. Vaccine 2017, 35, 283–292. [Google Scholar] [CrossRef] [Green Version]
  118. Whitehead, S.S.; Blaney, J.E.; Durbin, A.P.; Murphy, B.R. Prospects for a dengue virus vaccine. Nat. Rev. Microbiol. 2007, 5, 518–528. [Google Scholar] [CrossRef]
  119. Heinz, F.X.; Allison, S.L.; Stiasny, K.; Schalich, J.; Holzmann, H.; Mandl, C.W.; Kunz, C. Recombinant and virion-derived soluble and particulate immunogens for vaccination against tick-borne encephalitis. Vaccine 1995, 13, 1636–1642. [Google Scholar] [CrossRef]
  120. Aberle, J.H.; Aberle, S.W.; Allison, S.L.; Stiasny, K.; Ecker, M.; Mandl, C.W.; Berger, R.; Heinz, F.X. A DNA immunization model study with constructs expressing the tick-borne encephalitis virus envelope protein E in different physical forms. J. Immunol. 1999, 163, 6756–6761. [Google Scholar] [CrossRef]
  121. Schmidt, A.C.; Lin, L.; Martinez, L.J.; Ruck, R.C.; Eckels, K.H.; Collard, A.; De La Barrera, R.; Paolino, K.M.; Toussaint, J.F.; Lepine, E.; et al. Phase 1 Randomized Study of a Tetravalent Dengue Purified Inactivated Vaccine in Healthy Adults in the United States. Am. J. Trop. Med. Hyg. 2017, 96, 1325–1337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Han, H.H.; Diaz, C.; Acosta, C.J.; Liu, M.; Borkowski, A. Safety and immunogenicity of a purified inactivated Zika virus vaccine candidate in healthy adults: An observer-blind, randomised, phase 1 trial. Lancet Infect. Dis. 2021, 21, 1282–1292. [Google Scholar] [CrossRef] [PubMed]
  123. Abbink, P.; Larocca, R.A.; Visitsunthorn, K.; Boyd, M.; De La Barrera, R.A.; Gromowski, G.D.; Kirilova, M.; Peterson, R.; Li, Z.; Nanayakkara, O.; et al. Durability and correlates of vaccine protection against Zika virus in rhesus monkeys. Sci. Transl. Med. 2017, 9, eaao4163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Sumathy, K.; Kulkarni, B.; Gondu, R.K.; Ponnuru, S.K.; Bonguram, N.; Eligeti, R.; Gadiyaram, S.; Praturi, U.; Chougule, B.; Karunakaran, L.; et al. Protective efficacy of Zika vaccine in AG129 mouse model. Sci. Rep. 2017, 7, 46375. [Google Scholar] [CrossRef] [Green Version]
  125. Posadas-Herrera, G.; Inoue, S.; Fuke, I.; Muraki, Y.; Mapua, C.A.; Khan, A.H.; Parquet Mdel, C.; Manabe, S.; Tanishita, O.; Ishikawa, T.; et al. Development and evaluation of a formalin-inactivated West Nile Virus vaccine (WN-VAX) for a human vaccine candidate. Vaccine 2010, 28, 7939–7946. [Google Scholar] [CrossRef] [Green Version]
  126. Muraki, Y.; Fujita, T.; Matsuura, M.; Fuke, I.; Manabe, S.; Ishikawa, T.; Okuno, Y.; Morita, K. The efficacy of inactivated West Nile vaccine (WN-VAX) in mice and monkeys. Virol. J. 2015, 12, 54. [Google Scholar] [CrossRef] [Green Version]
  127. Quintel, B.K.; Thomas, A.; Poer DeRaad, D.E.; Slifka, M.K.; Amanna, I.J. Advanced oxidation technology for the development of a next-generation inactivated West Nile virus vaccine. Vaccine 2019, 37, 4214–4221. [Google Scholar] [CrossRef]
  128. Monath, T.P. Japanese encephalitis vaccines: Current vaccines and future prospects. Curr. Top. Microbiol. Immunol. 2002, 267, 105–138. [Google Scholar]
  129. Fan, Y.C.; Chen, J.M.; Chiu, H.C.; Chen, Y.Y.; Lin, J.W.; Shih, C.C.; Chen, C.M.; Chang, C.C.; Chang, G.J.; Chiou, S.S. Partially neutralizing potency against emerging genotype I virus among children received formalin-inactivated Japanese encephalitis virus vaccine. PLoS Negl. Trop. Dis. 2012, 6, e1834. [Google Scholar] [CrossRef] [Green Version]
  130. Hoke, C.H.; Nisalak, A.; Sangawhipa, N.; Jatanasen, S.; Laorakapongse, T.; Innis, B.L.; Kotchasenee, S.; Gingrich, J.B.; Latendresse, J.; Fukai, K.; et al. Protection against Japanese encephalitis by inactivated vaccines. N. Engl. J. Med. 1988, 319, 608–614. [Google Scholar] [CrossRef]
  131. Poland, J.D.; Cropp, C.B.; Craven, R.B.; Monath, T.P. Evaluation of the potency and safety of inactivated Japanese encephalitis vaccine in US inhabitants. J. Infect. Dis. 1990, 161, 878–882. [Google Scholar] [CrossRef] [PubMed]
  132. Defraites, R.F.; Gambel, J.M.; Hoke, C.H., Jr.; Sanchez, J.L.; Withers, B.G.; Karabatsos, N.; Shope, R.E.; Tirrell, S.; Yoshida, I.; Takagi, M.; et al. Japanese encephalitis vaccine (inactivated, BIKEN) in U.S. soldiers: Immunogenicity and safety of vaccine administered in two dosing regimens. Am. J. Trop. Med. Hyg. 1999, 61, 288–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Sanchez, J.L.; Hoke, C.H.; McCown, J.; DeFraites, R.F.; Takafuji, E.T.; Diniega, B.M.; Pang, L.W. Further experience with Japanese encephalitis vaccine. Lancet 1990, 335, 972–973. [Google Scholar] [CrossRef] [PubMed]
  134. Plesner, A.M.; Arlien-Soborg, P.; Herning, M. Neurological complications to vaccination against Japanese encephalitis. Eur. J. Neurol. 1998, 5, 479–485. [Google Scholar] [CrossRef]
  135. Lindsey, N.P.; Staples, J.E.; Jones, J.F.; Sejvar, J.J.; Griggs, A.; Iskander, J.; Miller, E.R.; Fischer, M. Adverse event reports following Japanese encephalitis vaccination in the United States, 1999–2009. Vaccine 2010, 29, 58–64. [Google Scholar] [CrossRef]
  136. Plesner, A.; Rønne, T.; Wachmann, H. Case-control study of allergic reactions to Japanese encephalitis vaccine. Vaccine 2000, 18, 1830–1836. [Google Scholar] [CrossRef]
  137. Andersen, M.M.; Rønne, T. Side-effects with Japanese encephalitis vaccine. Lancet 1991, 337, 1044. [Google Scholar] [CrossRef]
  138. Ruff, T.A.; Eisen, D.; Fuller, A.; Kass, R. Adverse reactions to Japanese encephalitis vaccine. Lancet 1991, 338, 881–882. [Google Scholar] [CrossRef]
  139. Halstead, S.B.; Thomas, S.J. New Japanese encephalitis vaccines: Alternatives to production in mouse brain. Expert Rev. Vaccines 2011, 10, 355–364. [Google Scholar] [CrossRef]
  140. Fischer, M.; Lindsey, N.; Staples, J.E.; Hills, S. Japanese encephalitis vaccines: Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm. Rep. 2010, 59, 1–27. [Google Scholar]
  141. Tauber, E.; Kollaritsch, H.; von Sonnenburg, F.; Lademann, M.; Jilma, B.; Firbas, C.; Jelinek, T.; Beckett, C.; Knobloch, J.; McBride, W.J.; et al. Randomized, double-blind, placebo-controlled phase 3 trial of the safety and tolerability of IC51, an inactivated Japanese encephalitis vaccine. J. Infect. Dis. 2008, 198, 493–499. [Google Scholar] [CrossRef] [PubMed]
  142. Tauber, E.; Kollaritsch, H.; Korinek, M.; Rendi-Wagner, P.; Jilma, B.; Firbas, C.; Schranz, S.; Jong, E.; Klingler, A.; Dewasthaly, S.; et al. Safety and immunogenicity of a Vero-cell-derived, inactivated Japanese encephalitis vaccine: A non-inferiority, phase III, randomised controlled trial. Lancet 2007, 370, 1847–1853. [Google Scholar] [CrossRef] [PubMed]
  143. Murrell, S.; Wu, S.C.; Butler, M. Review of dengue virus and the development of a vaccine. Biotechnol. Adv. 2011, 29, 239–247. [Google Scholar] [CrossRef] [PubMed]
  144. Chang, G.J.; Davis, B.S.; Hunt, A.R.; Holmes, D.A.; Kuno, G. Flavivirus DNA vaccines: Current status and potential. Ann. N. Y. Acad. Sci. 2001, 951, 272–285. [Google Scholar] [CrossRef]
  145. Schmaljohn, C.; Custer, D.; VanderZanden, L.; Spik, K.; Rossi, C.; Bray, M. Evaluation of tick-borne encephalitis DNA vaccines in monkeys. Virology 1999, 263, 166–174. [Google Scholar] [CrossRef] [Green Version]
  146. Anderson, B.R.; Muramatsu, H.; Jha, B.K.; Silverman, R.H.; Weissman, D.; Karikó, K. Nucleoside modifications in RNA limit activation of 2’-5’-oligoadenylate synthetase and increase resistance to cleavage by RNase L. Nucleic Acids Res. 2011, 39, 9329–9338. [Google Scholar] [CrossRef] [Green Version]
  147. Karikó, K.; Ni, H.; Capodici, J.; Lamphier, M.; Weissman, D. mRNA is an endogenous ligand for Toll-like receptor 3. J. Biol. Chem. 2004, 279, 12542–12550. [Google Scholar] [CrossRef] [Green Version]
  148. Porter, K.R.; Ewing, D.; Chen, L.; Wu, S.J.; Hayes, C.G.; Ferrari, M.; Teneza-Mora, N.; Raviprakash, K. Immunogenicity and protective efficacy of a vaxfectin-adjuvanted tetravalent dengue DNA vaccine. Vaccine 2012, 30, 336–341. [Google Scholar] [CrossRef]
  149. Dowd, K.A.; Ko, S.Y.; Morabito, K.M.; Yang, E.S.; Pelc, R.S.; DeMaso, C.R.; Castilho, L.R.; Abbink, P.; Boyd, M.; Nityanandam, R.; et al. Rapid development of a DNA vaccine for Zika virus. Science 2016, 354, 237–240. [Google Scholar] [CrossRef] [Green Version]
  150. Tebas, P.; Roberts, C.C.; Muthumani, K.; Reuschel, E.L.; Kudchodkar, S.B.; Zaidi, F.I.; White, S.; Khan, A.S.; Racine, T.; Choi, H.; et al. Safety and Immunogenicity of an Anti-Zika Virus DNA Vaccine. N. Engl. J. Med. 2021, 385, e35. [Google Scholar] [CrossRef]
  151. Richner, J.M.; Himansu, S.; Dowd, K.A.; Butler, S.L.; Salazar, V.; Fox, J.M.; Julander, J.G.; Tang, W.W.; Shresta, S.; Pierson, T.C.; et al. Modified mRNA Vaccines Protect against Zika Virus Infection. Cell 2017, 168, 1114–1125.e1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Jagger, B.W.; Dowd, K.A.; Chen, R.E.; Desai, P.; Foreman, B.; Burgomaster, K.E.; Himansu, S.; Kong, W.P.; Graham, B.S.; Pierson, T.C.; et al. Protective Efficacy of Nucleic Acid Vaccines Against Transmission of Zika Virus During Pregnancy in Mice. J. Infect. Dis. 2019, 220, 1577–1588. [Google Scholar] [CrossRef] [PubMed]
  153. Brito, L.A.; Kommareddy, S.; Maione, D.; Uematsu, Y.; Giovani, C.; Berlanda Scorza, F.; Otten, G.R.; Yu, D.; Mandl, C.W.; Mason, P.W.; et al. Self-amplifying mRNA vaccines. Adv. Genet. 2015, 89, 179–233. [Google Scholar] [PubMed]
  154. Luisi, K.; Morabito, K.M.; Burgomaster, K.E.; Sharma, M.; Kong, W.P.; Foreman, B.M.; Patel, S.; Fisher, B.; Aleshnick, M.A.; Laliberte, J.; et al. Development of a potent Zika virus vaccine using self-amplifying messenger RNA. Sci. Adv. 2020, 6, eaba5068. [Google Scholar] [CrossRef] [PubMed]
  155. Iavarone, C.; O’Hagan, D.T.; Yu, D.; Delahaye, N.F.; Ulmer, J.B. Mechanism of action of mRNA-based vaccines. Expert Rev. Vaccines 2017, 16, 871–881. [Google Scholar] [CrossRef] [PubMed]
  156. Vogel, A.B.; Lambert, L.; Kinnear, E.; Busse, D.; Erbar, S.; Reuter, K.C.; Wicke, L.; Perkovic, M.; Beissert, T.; Haas, H.; et al. Self-Amplifying RNA Vaccines Give Equivalent Protection against Influenza to mRNA Vaccines but at Much Lower Doses. Mol. Ther. 2018, 26, 446–455. [Google Scholar] [CrossRef] [Green Version]
  157. Pepini, T.; Pulichino, A.M.; Carsillo, T.; Carlson, A.L.; Sari-Sarraf, F.; Ramsauer, K.; Debasitis, J.C.; Maruggi, G.; Otten, G.R.; Geall, A.J.; et al. Induction of an IFN-Mediated Antiviral Response by a Self-Amplifying RNA Vaccine: Implications for Vaccine Design. J. Immunol. 2017, 198, 4012–4024. [Google Scholar] [CrossRef] [Green Version]
  158. Davis, B.S.; Chang, G.J.; Cropp, B.; Roehrig, J.T.; Martin, D.A.; Mitchell, C.J.; Bowen, R.; Bunning, M.L. West Nile virus recombinant DNA vaccine protects mouse and horse from virus challenge and expresses in vitro a noninfectious recombinant antigen that can be used in enzyme-linked immunosorbent assays. J. Virol. 2001, 75, 4040–4047. [Google Scholar] [CrossRef] [Green Version]
  159. Hall, R.A.; Nisbet, D.J.; Pham, K.B.; Pyke, A.T.; Smith, G.A.; Khromykh, A.A. DNA vaccine coding for the full-length infectious Kunjin virus RNA protects mice against the New York strain of West Nile virus. Proc. Natl. Acad. Sci. USA 2003, 100, 10460–10464. [Google Scholar] [CrossRef] [Green Version]
  160. Mühlebach, M.D. Vaccine platform recombinant measles virus. Virus Genes 2017, 53, 733–740. [Google Scholar] [CrossRef]
  161. Swamy, G.K.; Heine, R.P. Vaccinations for pregnant women. Obstet. Gynecol. 2015, 125, 212–226. [Google Scholar] [CrossRef] [PubMed]
  162. Zabel, F.; Kündig, T.M.; Bachmann, M.F. Virus-induced humoral immunity: On how B cell responses are initiated. Curr. Opin. Virol. 2013, 3, 357–362. [Google Scholar] [CrossRef] [PubMed]
  163. Schiller, J.T.; Müller, M. Next generation prophylactic human papillomavirus vaccines. Lancet Oncol. 2015, 16, e217–e225. [Google Scholar] [CrossRef]
  164. Torresi, J.; Ebert, G.; Pellegrini, M. Vaccines licensed and in clinical trials for the prevention of dengue. Hum. Vaccin. Immunother. 2017, 13, 1059–1072. [Google Scholar] [CrossRef] [PubMed]
  165. Bukreyev, A.; Skiadopoulos, M.H.; Murphy, B.R.; Collins, P.L. Nonsegmented negative-strand viruses as vaccine vectors. J. Virol. 2006, 80, 10293–10306. [Google Scholar] [CrossRef] [Green Version]
  166. Li, A.; Yu, J.; Lu, M.; Ma, Y.; Attia, Z.; Shan, C.; Xue, M.; Liang, X.; Craig, K.; Makadiya, N.; et al. A Zika virus vaccine expressing premembrane-envelope-NS1 polyprotein. Nat. Commun. 2018, 9, 3067. [Google Scholar] [CrossRef] [Green Version]
  167. Roberts, A.; Buonocore, L.; Price, R.; Forman, J.; Rose, J.K. Attenuated vesicular stomatitis viruses as vaccine vectors. J. Virol. 1999, 73, 3723–3732. [Google Scholar] [CrossRef] [Green Version]
  168. Cox, F.; van der Fits, L.; Abbink, P.; Larocca, R.A.; van Huizen, E.; Saeland, E.; Verhagen, J.; Peterson, R.; Tolboom, J.; Kaufmann, B.; et al. Adenoviral vector type 26 encoding Zika virus (ZIKV) M-Env antigen induces humoral and cellular immune responses and protects mice and nonhuman primates against ZIKV challenge. PLoS ONE 2018, 13, e0202820. [Google Scholar] [CrossRef]
  169. Salisch, N.C.; Stephenson, K.E.; Williams, K.; Cox, F.; van der Fits, L.; Heerwegh, D.; Truyers, C.; Habets, M.N.; Kanjilal, D.G.; Larocca, R.A.; et al. A Double-Blind, Randomized, Placebo-Controlled Phase 1 Study of Ad26.ZIKV.001, an Ad26-Vectored Anti-Zika Virus Vaccine. Ann. Intern. Med. 2021, 174, 585–594. [Google Scholar] [CrossRef]
  170. López-Camacho, C.; Abbink, P.; Larocca, R.A.; Dejnirattisai, W.; Boyd, M.; Badamchi-Zadeh, A.; Wallace, Z.R.; Doig, J.; Velazquez, R.S.; Neto, R.D.L.; et al. Rational Zika vaccine design via the modulation of antigen membrane anchors in chimpanzee adenoviral vectors. Nat. Commun. 2018, 9, 2441. [Google Scholar] [CrossRef] [Green Version]
  171. Desprès, P.; Combredet, C.; Frenkiel, M.P.; Lorin, C.; Brahic, M.; Tangy, F. Live measles vaccine expressing the secreted form of the West Nile virus envelope glycoprotein protects against West Nile virus encephalitis. J. Infect. Dis. 2005, 191, 207–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Nürnberger, C.; Bodmer, B.S.; Fiedler, A.H.; Gabriel, G.; Mühlebach, M.D. A Measles Virus-Based Vaccine Candidate Mediates Protection against Zika Virus in an Allogeneic Mouse Pregnancy Model. J. Virol. 2019, 93, e01485-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Brault, A.C.; Domi, A.; McDonald, E.M.; Talmi-Frank, D.; McCurley, N.; Basu, R.; Robinson, H.L.; Hellerstein, M.; Duggal, N.K.; Bowen, R.A.; et al. A Zika Vaccine Targeting NS1 Protein Protects Immunocompetent Adult Mice in a Lethal Challenge Model. Sci. Rep. 2017, 7, 14769. [Google Scholar] [CrossRef] [Green Version]
  174. To, A.; Medina, L.O.; Mfuh, K.O.; Lieberman, M.M.; Wong, T.A.S.; Namekar, M.; Nakano, E.; Lai, C.Y.; Kumar, M.; Nerurkar, V.R.; et al. Recombinant Zika Virus Subunits Are Immunogenic and Efficacious in Mice. mSphere 2018, 3, e00576-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Tai, W.; He, L.; Wang, Y.; Sun, S.; Zhao, G.; Luo, C.; Li, P.; Zhao, H.; Fremont, D.H.; Li, F.; et al. Critical neutralizing fragment of Zika virus EDIII elicits cross-neutralization and protection against divergent Zika viruses. Emerg. Microbes Infect. 2018, 7, 7. [Google Scholar] [CrossRef] [PubMed]
  176. Zhu, X.; Li, C.; Afridi, S.K.; Zu, S.; Xu, J.W.; Quanquin, N.; Yang, H.; Cheng, G.; Xu, Z. E90 subunit vaccine protects mice from Zika virus infection and microcephaly. Acta Neuropathol. Commun. 2018, 6, 77. [Google Scholar] [CrossRef]
  177. Hu, Y.; Sun, L. Systematic Analysis of Structure Similarity between Zika Virus and Other Flaviviruses. ACS Infect. Dis. 2019, 5, 1070–1080. [Google Scholar] [CrossRef]
  178. Boigard, H.; Alimova, A.; Martin, G.R.; Katz, A.; Gottlieb, P.; Galarza, J.M. Zika virus-like particle (VLP) based vaccine. PLoS Negl. Trop. Dis. 2017, 11, e0005608. [Google Scholar] [CrossRef] [Green Version]
  179. Salvo, M.A.; Kingstad-Bakke, B.; Salas-Quinchucua, C.; Camacho, E.; Osorio, J.E. Zika virus like particles elicit protective antibodies in mice. PLoS Negl. Trop. Dis. 2018, 12, e0006210. [Google Scholar] [CrossRef]
  180. De Lorenzo, G.; Tandavanitj, R.; Doig, J.; Setthapramote, C.; Poggianella, M.; Sanchez-Velazquez, R.; Scales, H.E.; Edgar, J.M.; Kohl, A.; Brewer, J.; et al. Zika Virus-Like Particles Bearing a Covalent Dimer of Envelope Protein Protect Mice from Lethal Challenge. J. Virol. 2020, 95, e01415-20. [Google Scholar] [CrossRef]
  181. Barba-Spaeth, G.; Dejnirattisai, W.; Rouvinski, A.; Vaney, M.C.; Medits, I.; Sharma, A.; Simon-Lorière, E.; Sakuntabhai, A.; Cao-Lormeau, V.M.; Haouz, A.; et al. Structural basis of potent Zika-dengue virus antibody cross-neutralization. Nature 2016, 536, 48–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Fernandez, E.; Dejnirattisai, W.; Cao, B.; Scheaffer, S.M.; Supasa, P.; Wongwiwat, W.; Esakky, P.; Drury, A.; Mongkolsapaya, J.; Moley, K.H.; et al. Human antibodies to the dengue virus E-dimer epitope have therapeutic activity against Zika virus infection. Nat. Immunol. 2017, 18, 1261–1269. [Google Scholar] [CrossRef] [Green Version]
  183. Shanmugam, R.K.; Ramasamy, V.; Shukla, R.; Arora, U.; Swaminathan, S.; Khanna, N. Pichia pastoris-expressed Zika virus envelope domain III on a virus-like particle platform: Design, production and immunological evaluation. Pathog. Dis. 2019, 77, ftz026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Cabral-Miranda, G.; Lim, S.M.; Mohsen, M.O.; Pobelov, I.V.; Roesti, E.S.; Heath, M.D.; Skinner, M.A.; Kramer, M.F.; Martina, B.E.E.; Bachmann, M.F. Zika Virus-Derived E-DIII Protein Displayed on Immunologically Optimized VLPs Induces Neutralizing Antibodies without Causing Enhancement of Dengue Virus Infection. Vaccines 2019, 7, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Zhang, S.; Kostyuchenko, V.A.; Ng, T.S.; Lim, X.N.; Ooi, J.S.G.; Lambert, S.; Tan, T.Y.; Widman, D.G.; Shi, J.; Baric, R.S.; et al. Neutralization mechanism of a highly potent antibody against Zika virus. Nat. Commun. 2016, 7, 13679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Cimica, V.; Galarza, J.M. Adjuvant formulations for virus-like particle (VLP) based vaccines. Clin. Immunol. 2017, 183, 99–108. [Google Scholar] [CrossRef] [PubMed]
  187. Karaca, K.; Bowen, R.; Austgen, L.E.; Teehee, M.; Siger, L.; Grosenbaugh, D.; Loosemore, L.; Audonnet, J.C.; Nordgren, R.; Minke, J.M. Recombinant canarypox vectored West Nile virus (WNV) vaccine protects dogs and cats against a mosquito WNV challenge. Vaccine 2005, 23, 3808–3813. [Google Scholar] [CrossRef] [PubMed]
  188. Paoletti, E.; Tartaglia, J.; Taylor, J. Safe and effective poxvirus vectors--NYVAC and ALVAC. Dev. Biol. Stand. 1994, 82, 65–69. [Google Scholar]
  189. Minke, J.M.; Siger, L.; Karaca, K.; Austgen, L.; Gordy, P.; Bowen, R.; Renshaw, R.W.; Loosmore, S.; Audonnet, J.C.; Nordgren, B. Recombinant canarypoxvirus vaccine carrying the prM/E genes of West Nile virus protects horses against a West Nile virus-mosquito challenge. Arch. Virol. Suppl. 2004, 18, 221–230. [Google Scholar]
  190. Brandler, S.; Marianneau, P.; Loth, P.; Lacôte, S.; Combredet, C.; Frenkiel, M.P.; Desprès, P.; Contamin, H.; Tangy, F. Measles vaccine expressing the secreted form of West Nile virus envelope glycoprotein induces protective immunity in squirrel monkeys, a new model of West Nile virus infection. J. Infect. Dis. 2012, 206, 212–219. [Google Scholar] [CrossRef]
  191. Iyer, A.V.; Boudreaux, M.J.; Wakamatsu, N.; Roy, A.F.; Baghian, A.; Chouljenko, V.N.; Kousoulas, K.G. Complete genome analysis and virulence characteristics of the Louisiana West Nile virus strain LSU-AR01. Virus Genes 2009, 38, 204–214. [Google Scholar] [CrossRef] [PubMed]
  192. Julander, J.G.; Testori, M.; Cheminay, C.; Volkmann, A. Immunogenicity and Protection After Vaccination With a Modified Vaccinia Virus Ankara-Vectored Yellow Fever Vaccine in the Hamster Model. Front. Immunol. 2018, 9, 1756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Monath, T.P. Dengue and yellow fever--challenges for the development and use of vaccines. N. Engl. J. Med. 2007, 357, 2222–2225. [Google Scholar] [CrossRef] [PubMed]
  194. Douam, F.; Ploss, A. Yellow Fever Virus: Knowledge Gaps Impeding the Fight Against an Old Foe. Trends Microbiol. 2018, 26, 913–928. [Google Scholar] [CrossRef]
  195. Monath, T.P. Suspected yellow fever vaccine-associated viscerotropic adverse events (1973 and 1978), United States. Am. J. Trop. Med. Hyg. 2010, 82, 919–921. [Google Scholar] [CrossRef] [Green Version]
  196. Domingo, C.; Charrel, R.N.; Schmidt-Chanasit, J.; Zeller, H.; Reusken, C. Yellow fever in the diagnostics laboratory. Emerg Microbes Infect. 2018, 7, 129. [Google Scholar] [CrossRef] [Green Version]
  197. Heinz, F.X.; Stiasny, K. Flaviviruses and their antigenic structure. J. Clin. Virol. 2012, 55, 289–295. [Google Scholar] [CrossRef]
  198. Pierson, T.C.; Kielian, M. Flaviviruses: Braking the entering. Curr. Opin. Virol. 2013, 3, 3–12. [Google Scholar] [CrossRef] [Green Version]
  199. Pletnev, A.G.; Bray, M.; Hanley, K.A.; Speicher, J.; Elkins, R. Tick-borne Langat/mosquito-borne dengue flavivirus chimera, a candidate live attenuated vaccine for protection against disease caused by members of the tick-borne encephalitis virus complex: Evaluation in rhesus monkeys and in mosquitoes. J. Virol. 2001, 75, 8259–8267. [Google Scholar] [CrossRef] [Green Version]
Table 1. The development and evaluation of mosquito-borne flavivirus vaccines.
Table 1. The development and evaluation of mosquito-borne flavivirus vaccines.
Vaccine TypesVirusVaccine CandidateDeveloper/
Manufacturer
EvaluationReferences
LAVDENVDengvaxia®Sanofi PasteurLicensed [17]
TDVTakeda Pharmaceutical CompanyPhase III (NCT02747927) [71]
LATVNIAIDPhase III (NCT02406729) [69]
TDENWRAIR and GSKPhase II (NCT01843621) [69]
ZIKVChinZIKVBeijing Institute of Microbiology and EpidemiologyIn vivo (animal) [95]
ChimeriVax-ZikaSanofi PasteurIn vivo (animal) [27]
rZIKV/D4Δ30–713NIAIDPhase I (NCT03611946) [27]
WNVChinWNVBeijing Institute of Microbiology and EpidemiologyIn vivo (animal) [97]
WN/DEN4Δ30NIH in USPhase I (NCT00094718 and NCT00537147) [98]
ChimeriVax-WN02Acambis in UKPhase II (NCT00442169) [102]
RepliVAX WNDepartment of Microbiology and Immunology in USIn vivo (animal) [66]
JEVSA14-14-2Chengdu Institute of Biological Products in ChinaLicensed [13]
ChimeriVax-JEAcambis in UKPhase III (NCT01092507) [110]
RepliVAX-JE.2Department of Pathology, University of Texas Medical Branch et al.In vivo (animal) [112]
YFVYFV-17DTheiler et al.Licensed [13]
INVDENVDPIVWRAIR and GSKPhase I (NCT01666652) [121]
ZIKVZPIVNIAD, WRAIR and BIDMCPhase I (NCT03008122, NCT02963909, NCT02952833 NCT02937233) [96]
PIV vaccineR&D Department Bharat Biotech International Ltd.In vivo (animal) [124]
WNVWN-VAXMuraki et al.In vivo (animal) [125,126]
HydroVax-001 WNVNajít Technologies, Inc, Beaverton, OR in USIn vivo (animal) [117]
HydroVax-IINajít Technologies, Inc, Beaverton, OR in USIn vivo (animal) [127]
JEVJE-VAXResearch Foundation for Microbial Diseases of Osaka UniversityLicensed [130]
IC51Intercell Biomedical Livingston in UKLicensed [106]
CVI-JELiaoning Cheng Da Biotechnology Co., Ltd.Licensed [48,106]
Nucleic acid vaccinesDENVTVDVAMRDCk, WRAIR, NMRC and Vical Inc. in USPhase I (NCT01502358) [148]
ZIKVGLS-5700GeneOne Life Science/Inovio PharmaceuticalsPhase I (NCT02887482, NCT02809443) [150]
VRC5283NIAIDPhase II (NCT03110770) [149]
VRC5288NIAIDPhase I (NCT02840487) [149]
mRNA-1325ModernaPhase I (NCT03014089) [151]
mRNA-1839ModernaPhase I (NCT04064905) [152]
SAMGSKIn vivo (animal) [153,154]
WNVpCBWNDavis et al.In vivo (animal) [158]
pKUN1 plasmid DNAHall et al.In vivo (animal) [159]
Viral vector vaccinesDENVcAdVaxD12/
cAdVaxD34
Raviprakash et al.In vivo (animal) [143]
ZIKVAd26.ZIKV.001Janssen VaccinesPhase I (NCT03356561) [168]
ChAdOx1 ZikaUniversity of OxfordPhase I (NCT04015648) [170]
MV-ZikaThemis BiosciencePhase I (NCT02996890) [172]
MV-Zika RSPThemis BiosciencePhase I (NCT04033068) [27,172]
WNVALVAC®-WNVMinke et al.In vivo (animal) [59,188]
YFVMVA-BN-YFJu lander et al.In vivo (animal) [192]
subunit protein vaccinesDENVV180Merck & Co., Inc.Phase I (NCT00936429, NCT01477580) [69]
WNVMVSchwsEWNVUnite’ des Interactions Mole´culaires Flavivirus-Hoˆtes and Unite´ des Virus Lents in FranceIn vivo (animal) [190]
VLP vaccinesZIKVZIKVLPsUniversity of WisconsinIn vivo (animal) [179]
VLP-cvDMRC—University of Glasgow Centre for Virus ResearchIn vivo (animal) [180]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wu, B.; Qi, Z.; Qian, X. Recent Advancements in Mosquito-Borne Flavivirus Vaccine Development. Viruses 2023, 15, 813. https://doi.org/10.3390/v15040813

AMA Style

Wu B, Qi Z, Qian X. Recent Advancements in Mosquito-Borne Flavivirus Vaccine Development. Viruses. 2023; 15(4):813. https://doi.org/10.3390/v15040813

Chicago/Turabian Style

Wu, Bingan, Zhongtian Qi, and Xijing Qian. 2023. "Recent Advancements in Mosquito-Borne Flavivirus Vaccine Development" Viruses 15, no. 4: 813. https://doi.org/10.3390/v15040813

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop