1,930
Views
22
CrossRef citations to date
0
Altmetric
Reviews

Development of new hepatitis E vaccines

, , , &
Pages 2254-2262 | Received 14 Jan 2018, Accepted 21 Apr 2018, Published online: 18 Jun 2018

ABSTRACT

Hepatitis E virus (HEV) infection is an emerging zoonotic disease posing a severe threat to public health in the world, especially to pregnant women. Currently, no specific treatments are available for HEV infection. Therefore, it is crucial to develop vaccine to prevent this infection. Although several potential candidate vaccines against HEV have been studied for their immunogenicity and efficacy, only Hecolin® which is developed by Xiamen Innovax Biotech Co., Ltd. and approved by China Food and Drug Administration (CFDA) in 2012, is the licensed HEV vaccine in the world so far. Extensive studies on safety, immunogenicity and efficacy in phase III clinical trials have shown that Hecolin® is a promising vaccine for HEV prevention and control. In this article, the advances on HEV vaccine development and research are briefly reviewed.

Introduction

Hepatitis E virus (HEV) belongs to the genus of orthohepevirus within the family of Hepeviridae. As an emerging causative agent for Hepatitis E, this virus infection poses severe threats to the public health worldwide, especially in developing countries.Citation1-Citation3 HEV is the main cause of enterically transmitted hepatitis worldwide, and responsible for more than half of the cases with acute hepatitis in endemic countries such as India and China.Citation3-Citation7 In Europe, 5%-15% of the acute hepatitis of unknown origin are caused by HEV.Citation8,Citation9 In recent years, outbreaks of HEV on a large scale have been revealed among displaced people in South Sudan, Chad, Uganda and Nepal.Citation10-Citation12 Hepatitis E infection has a relatively high seropositive rate for HEV IgG ranging from 10.0% to 33.5% and a low fatality rate ranging from 0.5% to 4%, while infections in pregnant women induces acute liver failure, leading to approximately 10%-25% mortality.Citation13-Citation15 Due to the limitations of the detection methods, actual incidence of HEV exceeded the clinical positive carrier among general population. According to the estimates by World Health Organization, one third of the world population has been exposed to HEV, and 35 million people are infected by HEV annually, resulting in 70,000 deaths.Citation16-Citation19 The sporadic infection in industrialized countries and outbreaks in developing countries are becoming more and more serious. As the disease affects mainly young people and results in higher mortality among pregnant women, especially in the developing countries, much attention should be paid to people at high risk of severe disease and priority be given to the prevention and control of HEV infection in this crowd during outbreaks.Citation20,Citation21

HEV classification and global distribution

HEV is currently classified to four major genotypes I-IV and twenty-four subtypes, while all genotypes belong to one serotype.Citation22 HEV genotypes I and II are restricted to humans, whereas genotypes III and IV are zoonotic and mainly responsible for sporadic cases. In addition, genotype IV is restricted to Asia and genotype III is spread worldwide.Citation23,Citation24 Genotype I isolated from the Delhi epidemic from 1955 to 1956 is responsible for the epidemic and sporadic hepatitis E in the developing countries such as India and China (Xinjiang).Citation25-Citation27 Genotype II was initially identified from Mexico, and subsequently from Chad and Nigeria in Africa.Citation28 Genotype III was firstly isolated from the swine population in mid-western United States in 1997, and now is prevalent in the swine herd globally. The genotype III isolates were subsequently identified from human in the developed world including Italy, France, the United States and Japan.Citation29-Citation33 Genotype IV was first isolated from patients in Taiwan and subsequently found in China, Japan and India.Citation34-Citation37 The presence of acute viral hepatitis caused by HEV in developing countries has been documented for decades. Since the late 1990s, endemic hepatitis E caused by genotype III and genotype IV has been increasingly reported in developed countries.Citation38,Citation39

HEV genome organization

HEV is a non-enveloped virus with a diameter of 27-34 nm, which has a single-stranded positive-sense RNA genome. The genome is approximately 7.2 kb in length containing a relatively conserved 5′non-coding region (NCR), a variable 3′NCR, and three open reading frames (ORFs).Citation40-Citation42 The organization of the ORFs differs slightly among four HEV genotypes. However, the proteins they encoded have the same functions.

ORF1 encodes a polyprotein of approximately 1693 amino acids, which is post-translationally processed into multiple non-structural proteins including methyltransferase, putative papain-like cysteine protease, RNA helicase and RNA-dependent RNA polymerase that are required for virus replication.Citation43

HEV ORF2 encodes the capsid protein, a structural protein consisting of 660 amino acids with a predicted molecular mass of 72 kDa.Citation44 The N-terminal 112 residues of structural protein encoded by ORF2 are responsible for the packaging of the viral RNA genome.Citation45 The virus-like particles (aa 112-608, VLPs) have been made up of three definitive domains: S domain (aa 129-319), M domain (aa320-455), and P domain (aa 456-606). S domain forms the viral shell. M domain is involved in the formation of the 2, 3, and 5-fold icosahedral symmetries of the HEV capsid. P domain forms the protrusions that extend outward from the viral shell.Citation46-Citation48 The HEV capsid protein induces the antibodies that can neutralize the HEV in vitro, and protect primates against HEV infection, thus the complete and truncated capsid proteins are considered as vaccine candidate against HEV infection in humans.Citation49-Citation51

ORF3 overlaps with ORF2 and encodes a phosphoprotein of 123 amino acids that interacts with the cytoskeleton protein, suggesting it plays a role in the assembly of virus particles.Citation20

Hepatitis E vaccines

Since no specific treatment is available for hepatitis E at present, development of an effective vaccine against HEV infections is the only pragmatic approach to prevent and control this disease.Citation52,Citation53 The inability to reproducibly produce HEV in culture system makes it impossible to develop traditional live or inactivated vaccines. However, significant progress has been made in developing recombinant vaccines based on the ORF2 viral capsid protein VLPs (). Protein-based VLPs are self-assembling bionanoparticles that expose multiple epitopes on their surface and faithfully mimic the native virions.Citation54 Particle size of the nanometer scale particulate antigen and presence of key epitopes on the particle surface are two prerequisites for an effective VLP-based vaccine.Citation55 Progress and detailed information was summarized as follows.

Proteins expressed in prokaryotic system

Escherichia coli is the most economical and commonly used expression system for its high production of heterologous protein. Purdy et al. expressed a fusion protein TrpE-C2 containing aa 221- 660 of ORF2 protein from Burmese strain (genotype I) and tryptophan synthetase. The TrpE-C2 fusion protein can protect the cynomolgus macaques from wild-type homologous (genotype I) or heterologous (genotype II) HEV in preclinical challenge experiments.Citation56 Meng et al. mapped the neutralization epitopes for HEV at the minimal size fragment of 166 aa (p166) located in ORF2.Citation57 The HEV p179 (aa 439-617) vaccine with 13 aa extension at the N-terminus of p166, was demonstrated to self-assembly as VLPs and manufactured by Changchun Institute of Biological Products Co. Ltd.Citation58,Citation59 The immunogenicity and protective assessment showed that the HEV p179 vaccine could protect rhesus macaques from challenging with genotypes I and IV HEV. A randomized open-label controlled and a dose escalation scheduling of 20μg, 30μg and 40μg phase I clinical trial indicated that the HEV p179 vaccine was safe and well-tolerated for the 16-65 year adult population.Citation19 Another HEV p239 vaccine Hecolin® developed by Xiamen Innovax Biotech and expressed in Escherichia coli with truncated ORF2 (aa 368–606) was consisted of 23 nm VLPs and launched on market in China in 2012.Citation60-Citation62 The vaccine HEV p239 was tested in a randomized, controlled trial involving 112,604 participants in China. The vaccine showed an efficacy >99% in preventing clinical hepatitis E among population who were inoculated 3-doses of HEV p239 (n = 48,693) compared with a placebo (a commercial hepatitis B vaccine, n = 48,663).Citation63 Immunization with Hecolin® induced antibodies against HEV and provided protection against hepatitis E for up to 4.5 years.Citation64

Proteins expressed in insect cells

The HEV capsid antigen expressed in Baculovirus-infected insect cell system has been proposed as a candidate subunit vaccine for the prevention of hepatitis E. Most of the baculovirus-mediated ORF2 proteins have been expressed in Sf21, Sf9, Tn5 and Spodoptera Litura Larvae insect cell lines.Citation65 Expression of the HEV ORF2 capsid gene produced multiple immunoreactive proteins with molecular weight size ranging from 30 to 100 kDa, and the most abundant HEV proteins had molecular weights of 72, 63, 56, and 53 kDa.Citation66 The 56 kDa candidate vaccine showed highly immunogenic when administered to rhesus monkeys with two doses, and protected them from hepatitis E when challenged with a large intravenous dose of homologous or heterologous HEV. In this study, the immune procedure with two doses of HEV vaccine partially protected rhesus monkeys from hepatitis E following intravenous challenge at 6 or 12 months post-vaccination.Citation67 A randomized, double-blind, placebo-controlled phase II clinical trial was launched to evaluate the safety and efficacy of this candidate vaccine. A total of 1794 subjects received three vaccine doses, with 898 of them as vaccine group and 896 as placebo group. After three doses of immunization, 69 subjects developed in hepatitis E, of whom 66 were from the placebo group, with vaccine efficacy 95.5%.Citation68 Qi et al. constructed and expressed HEV ORF2 capsid protein using Baculovirus-infected insect cell system. In the study, two conformations of the HEV capsid proteins VLPs and non-VLPs were expressed in insect cells, and purified separately. The humoral immune response research showed that the immunogenicity of VLPs was better than HEV non-VLPs in mice.Citation69

Proteins expressed in yeast

The N-glycosylation plays an important role in the stability, immunogenicity and biological activity of protein. HEV ORF2 capsid protein contains several potential N-glycosylation sites, with N137 and N310 located in the S domain, and one site N562 in the P domain. The P domain forms a protruding spike from the shell with N562 in the apical center of the spike, which is also a cell-attachment region and neutralizing antigenic site.Citation70 Consideration of the influence of glycosylation, many research fellows attempted to express truncated HEV ORF2 protein in yeast.Citation71 Suet al. expressed truncated genotype IV HEV ORF2 (aa112–607) in a system of methylotrophic yeast Hansenula polymorpha. The 56 kDa truncated protein showed high immunoreactivity, providing foundation for the further development of recombinant vaccine against hepatitis E.Citation72 Xu et al. expressed the recombinant polypeptide p179 comprising of the region (aa 439-617) within the HEV ORF2 and a set of 4 mutant proteins where N at position 562 were substituted with Q, D, P and Y respectively, in Pichia pastoris. In vitro neutralization experiments showed the mice antibodies generated against p179N562P and p179N562Y were more efficient than the antibodies generated against the other 3 proteins.Citation70 Tong et al. studied the immunogenicity of the recombinant HEV ORF2 protein expressed in Pichia pastoris by immunizing BALB/c mice in different routes with different adjuvants. The study demonstrated high immunogenicity of this kind of ORF2 recombinant protein.Citation73

Vectored vaccine against HEV

A novel candidate vectored vaccine against HEV through adeno-associated virus (AAV) vector expressing the truncated capsid protein of HEV ORF2 (aa 112-660) in Sf9 cells using the Baculovirus Expression System was developed. The recombinant baculovirus expressing the truncated capsid protein was constructed and the capsid protein was confirmed by Western blot in transducted HEK 293 EBNA cells with rAAV.Citation74 Vaccinia virus was also used as vector to express recombinant HEV truncated△(aa1-111)-ORF2 protein in BHK-21 cells, facilitating the study of its glycosylation patterns and subcellular localization. The expressed proteins have good antigenicity for diagnosis and good immunogenicity to induce long-lasting specific antibodies for anti-HEV in mice.Citation75 In another study, a synthetic codon-optimized gene cassette encoding tPAsp-PADRE-truncated ORF2 (aa 112-660) protein was sub-cloned into the pVAX1 expression vector and expressed in eukaryotic cells with truncated ORF2 constructs as control. The expression of truncated ORF2 protein in eukaryotic cells was proven by Immunofluorescence assay (IFA) and RT-PCR, which can be used as a novel DNA candidate vaccine in future investigations.Citation76

Combined vaccine and recombinant chimeric vaccine

Hepatitis A virus (HAV) and HEV are causative agents of acute viral hepatitis transmitted via the fecal-oral route. To relieve burden on the public health in developing countries, approach of combined vaccine was adopted extensively. Dong et al. developed a combined vaccine against hepatitis A and E. Different groups of mice immunized with different formulations containing a commercial inactivated hepatitis A vaccine and a candidate recombinant hepatitis E vaccine. The experimental results showed that the combined vaccine can induce neutralizing antibodies against both HAV and HEV effectively in a mouse model.Citation77 The other chimeric recombinant vaccine HAV-HEp148 was created as a vector to express an HEV neutralization epitope HEp148 (aa 459-606 of HEV capsid protein). Immunization with the HAV-HEp148 virus induced a strong HAV- and HEV-specific immune response in mice.Citation78 Another novel combined protein vaccine by conjugating tuftsin to HEV-ORF2 (aa368-607) and HAV-VP1 (aa 1-198) used synthetic CpG oligodeoxynucleotides (ODNs) as the adjuvant. Mice in group intranasally immunized with tuftsin showed higher levels of IFN-γ-secreting splenocytes (Th1 response) and CD4+/CD8+ T cells than mice in group without tuftsin, demonstrating the tuftsin group generated stronger humoral and cellular immune responses than the no-tuftsin group. Moreover, intranasal immunization generated some enhanced immune responses compared with intramuscular injection to the combined protein vaccine.Citation79

Li et al. constructed the fusion gene fragment of HEV capsid protein (aa 551-607) and gene of HBsAg via synthetic glycine linker. The resulting fusion gene was then integrated into the genome of Pichia pastoris through transformation and expressed intracellularly. The chimeric VLPs showed high immunoreactive activity in ELISA assay, indicating that the chimeric VLPs may be a potential HBV/HEV bivalent vaccine candidate.Citation80

Norovirus (NoV) and HEV are both enteric-transmitted viruses causing gastroenteritis and hepatitis, respectively. Both NoV and HEV have surface protrusions formed by dimers of the protruding (P) domains of the viral capsids, which are responsible for virus-host interactions and inducing neutralizing antibody. A bivalent vaccine against the two viruses fusing the dimeric P domains of NoV and HEV together linked with the dimeric glutathione-S-transferase (GST). Both of the GST-NoV P(-)-HEV P and NoV P(-)-HEV P complexes vaccines can induce significantly higher antibody titers, indicating they are promising chimeric vaccine candidates against both NoV and HEV.Citation81

HEV, rotavirus (RV) and astrovirus (AstV) are important pathogens that transmit via common fecal-oral route, causing hepatitis (HEV) and gastroenteritis (RV and AstV) in humans respectively. A recombinant fusion protein comprising the HEV truncated ORF2 gene (aa 112-607) and Rotavirus NSP4 was expressed in Baculovirus System as a potential candidate vaccine.Citation82 In another study, two formats of subunit vaccine candidates consisted of spike protein antigens from HEV, rotavirus (RV) and astrovirus (AstV) were designed. One was fused vaccine and the other was mixed vaccine. The fused vaccine is a fusion protein of the three antigens into one molecule and the mixed vaccine is a mixture of the three free antigens together. The fused vaccine induced significantly higher antibody responses against the three viral antigens than those induced by the mixed vaccine in mice immunization experiments. The similar results were observed in neutralizing titers against HEV and blocking titers (BT50) against RV VP8-HBGA receptor interactions.Citation83

Oral immunization HEV vaccine

HEV is an important causative agent of enterically-transmitted hepatitis, thus oral immunization may be an efficient approach for HEV vaccine. An oral administration experiment in mice was designed via detecting the response of serum IgM, IgG, and IgA and fecal IgA to evaluate the potential of recombinant HEVVLPs with its capsids protein N-terminal 111 amino acids truncated as an oral immunogen. The study indicated that serum IgG and fecal IgA were reactive to the native HEV antigen, as detected by Western blot and antigen-capture ELISA.Citation84 Serum IgM, IgG and IgA antibodies were also detected in cynomolgus monkeys orally inoculated with 10mg of this purified HEVVLPs. The monkeys were protected against infection or developing hepatitis after they were challenged with native HEV by intravenous injection, suggesting this recombinant HEV VLPs is a candidate for the oral hepatitis E vaccine.Citation85 In another study, a recombinant (inaQ-ORF2 gene fusion) Lactococcus lactis (L. lactis) strain NZ3900 expressing the HEV ORF2 antigen utilizing an ice nucleation protein-based anchor system was constructed. The BALB/c mice vaccinated orally displayed significantly higher levels of ORF2-specific mucosal IgA, serum IgG and cellular immunity, suggesting that L. lactis-based HEV antigen vaccines can be used for protection against HEV infection.Citation86 Tomato is used as an ideal plant carrier for transgenic oral vaccine for being consumed raw and easily transformed. The fragment of HEV ORF2 (aa 394-604) was cloned into plant binary expression vector p1301E2 containing a constitutive CaMV35s promoter and nos terminator, then directly introduced into agrobacterium tumefaciens EHA105. The production of truncated recombinant HEV ORF2 protein was 61.22 ng/g fresh weight in fruits and 6.37-47.9 ng/g fresh weight in leaves of the transformants. This study laid the foundation for further research on the development of a new type of plant-derived hepatitis E oral vaccine.Citation87

HEV vaccine neutralization assay

For assessing vaccine potency as a surrogate for clinical efficacy, a viral neutralization assay is needed. The host humoral immune responses for vaccination, especially the level of neutralization antibody, reflect the potency for an effective candidate vaccine. Rhesus macaque has been used as a classical animal model to evaluate the protective effect against the challenging HEV. Study revealed that 5µg of HEV p179 vaccine could induce protective IgG, while 20 µg of p179 vaccine could prevent 75% of rhesus macaques from shedding virus in feces.Citation19 Meng et al. reported a new PCR-based seroneutralization assay for the serological diagnosis of HEV infection based on neutralization of the virus in cell culture.Citation88 Takahashi et al. generated one mAb (H6225) against HEV capsid protein efficiently neutralizing HEV in vitro using a hepatocarcinoma cell culture system.Citation89 Yang et al. developed a high-throughput neutralization titer evaluation model for HEV. In the study, the quantitative assay for neutralizing capacity of different antibodies and antiserum samples from HEV vaccine recipients was established.Citation90 In another study, a broad neutralizing mAb 8G12 was used to develop a competitive ELSIA assay and quantified 8G12 competitive antibody (8G12-like antibody) in serum samples. The 8G12-like antibody was approved to be a promising surrogate for vaccine induced HEV neutralizing antibody.Citation91

Summary

HEV is one of the common cause of acute viral hepatitis globally. The HEV prevalence rates are more than 25% in many regions including Central America, large parts of Africa, and all of Asia, especially in population living in refugee camps or overcrowded temporary housing after natural disasters.Citation92 Hepatitis E infection results in significant burden in some parts of the world especially in developing countries. China has been listed as one of the countries suffering the severe economic burden with HEV infection.Citation93,Citation94 It is crucial to use vaccine to prevent HEV infection, thus knowledge for the structure of the HEV genome and the function of ORFs are essential for vaccine development.Citation95-Citation98 Although four genotypes of HEV have been reported globally, they belong to one serotype. The antibodies induced by different genotype HEV can neutralize homologous or heterologous HEV. The finding that serum antibodies to HEV were developed and detected in human naturally infected by HEV and in the primate animal models experimentally infected with HEV ratified the capacity of the humoral responses elicited by HEV.Citation99 Epidemiological survey during a hepatitis E outbreak suggests that people previously infected with HEV can be protected during epidemics of the disease, further validating the vaccine as effective tools in fighting HEV infection.Citation100

Recombinant HEV VLPs, consisting of different lengths of the ORF2 capsid protein, were prepared to mimic the protective and neutralizing epitopes on the virion surface. These candidate vaccines were designed to elicit protective immunity when inoculated non-human primates or human volunteers in preclinical tests and clinical trials.Citation101 In addition, linked recognition is considered as important mechanism for antibody production of TD antigens, so cellular immune responses may play an important role in providing protection against viruses. Thus, viral proteins containing T-cell epitopes may prove useful in stimulating T-cell immunity and providing help for antibody production. Furthermore, activation of T-cell immune responses may inhibit viral replication through a cytokine-mediated noncytolytic pathway. Aggarwal et al. present their results on T cell epitope mapping of HEV ORF2 and ORF3 proteins using overlapping peptides encompassing the entire lengths of these proteins. The immunodominant T-cell epitopes of HEV ORF2 protein were located in the regions encompassed by aa 73-156, aa 289-444 and aa 505-588.Citation102 These findings provided structure basis for T-cell immune response of HEV candidate vaccines consisting of ORF2 proteins.

Since ORF2 protein contains T-cell and B-cell epitopes and induces neutralizing antibodies, effector cell and cytokines offering help for antibody production against the HEV infection, it becomes the focus for developing the safe and effective vaccine.Citation103-Citation106 Various forms of HEV vaccines based on the ORF2 protein expressed using different heterologous systems are reported. The other new forms of vaccines such as vectored HEV vaccine, combined vaccine and recombinant chimeric vaccine were also studied. Traditional recombinant vaccines such as Hecolin®, the only licensed HEV vaccine in the world so far, can induce protective humoral immune responses well, and can be produced in economical E.coli expression system using simple technological process. Construction of DNA candidate vaccines using AAV and pVAX1 expression vectors are significant attempts to explore the novel HEV vaccines. Whereas, the unknown scruple for safety and intricate technological process are the bottleneck for these novel vaccines. Combined vaccine and recombinant chimeric vaccine have become important development direction for their good performance on safety, efficacy and economic cost. FDA approved combined hepatitis A and B vaccine for its safety and highly immunogenicity, which has facilitated the interest for scientists to develop combined vaccine and recombinant chimeric vaccine associated with hepatitis.Citation107 The development of combined vaccines to prevent multiple diseases was considered desirable, especially in developing countries where these diseases are highly endemic.

Although there are several HEV candidate vaccines synthesized, only three of them have access to clinical trial registration to date. The first one developed by GSK had completed phase II clinical trial, indicating that the vaccine was effective in the prevention of hepatitis E.Citation108,Citation109 The other one is the previously mentioned Hecolin® which have completed phase III clinical trial and been licensed in China. The long-term efficacy of Hecolin® had been evaluated, which is 86.8% after 4.5 years in the modified intention-to-treat analysis.Citation64,Citation110 Clinical trial data indicated that Hecolin® had provided good short-term and long-term protection against HEV infection in 16-65 years old healthy Chinese population.Citation3 Another HEV candidate vaccines developed by Changchun Institute of Biological Products Co., Ltd (China) have finished phase I clinical trial, showing good safety and tolerance.Citation19

China is the first country where the HEV vaccine was licensed. This vaccine is demonstrated as an effective HEV vaccine. The cases of hepatitis E infection since 2012 were reported to decline by 7.58% according to the data released by Chinese Center for Disease Control and Prevention (CDC).Citation111 However, the application of Hecolin® worldwide was hindered by various complicated factors. First of all, the only clinical data available are based on clinical trial conducted in China, where HEV is prevalent with low morbidity about 0.002%.Citation111 The safety and efficacy data regarding Hecolin® need to be further verified worldwide especially in regions such as Africa, Southeast Asia and South Asia, where HEV genotype I, II are the dominated endemic types.,Citation112,Citation113 Moreover, safety and efficacy data from children, elderly patients, patients with chronic liver disease, such as hepatitis B and hepatitis C, HIV and those with immune disorders, especially pregnant women are further needed for the global launch and promotion of Hecolin®.Citation114 Additionally, Hecolin® has received neither WHO prequalification (PQ) nor licenses in other countries, thus it is not available to HEV endemic countries especially to the refugees at HEV high-epidemic regions.Citation115 The first and only licensed vaccine was developed based on data from China where almost exclusively genotype 4 is the circulating strain. Thus more clinical data are needed to verify whether this vaccine or any vaccines being developed work effectively against other genotypes. How to meet the WHO prequalification (PQ) for Hecolin® and gather more clinical data are becoming more and more imperative.

Although steady progress has been made on research and application for HEV vaccine, the immunity mechanism has remained unarticulated. Compared with humoral immune responses, only limited research data are available on cellular immune responses for HEV vaccines. More information on cellular immune are required for illuminating the mechanism of pathogenetic injury and protective immunity response in HEV natural infection or in hepatitis E vaccination. In addition, establishment of steady and ingenious assay for neutralization antibody quantitation is indispensable for vaccines efficacy evaluation. Moreover, consideration of the huge global burden of epidemic and sporadic hepatitis E, and the public health threat caused by HEV infection in different population worldwide, available and effective approaches should be taken. With the accumulation of safety and efficacy data on pregnant women, chronic disease patients, and children under the age of 16, the application and promotion globally for HEV vaccines will be accelerated.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgments

The authors would like to thank the volunteers who participated in this clinical trial.

Table 1. HEV candidate vaccines and commercialized vaccines.

Additional information

Funding

This study was supported by the National Key Research and Development Program of China (2017YFD0500104, 2016YFD0500904), and the National Natural Science Foundation of China (31572531).

References

  • Song YJ, Park WJ, Park BJ, Lee JB, Park SY, Song CS, Lee NH, Seo KH, Kang YS, Choi IS. Hepatitis E virus infections in humans and animals. Clin Exp Vaccine Res. 2014;3:29−36. doi:10.7774/cevr.2014.3.1.29. PMID:24427760.
  • Reyes GR, Yarbough PO, Tam AW, Purdy MA, Huang CC, Kim JS, Bradley DW, Fry KE. Isolation of a cDNA from the virus responsible for enterically transmitted non-A, non-B hepatitis. Science. 1990;247:1335−9. doi:10.1126/science.2107574. PMID:2107574.
  • Wu X, Chen P, Lin H, Hao X, Liang Z. Hepatitis E virus: Current epidemiology and vaccine. Hum Vaccin Immunother. 2016;12:26032610. doi:10.1080/21645515.2016.1184806.
  • Kmush B, Wierzba T, Krain L, Nelson K, Labrique AB. Epidemiology of hepatitis E in low-and middle-income countries of Asia and Africa. Semin Liver Dis. 2013;33:15−29. doi:10.1055/s-0033-1338111. PMID:23564386.
  • Liu ZQ, Zuo JL, Yan Q, Fang QW, Zhang TJ. Epidemiologic and spatio-temporal characteristics of hepatitis E in China, 2004-2014. The Chinese Journal of Epidemiology. 2017;38:1380−5. PMID:29060984.
  • Khuroo MS, Rustgi VK, Dawson GJ, Mushahwar IK, Yattoo GN, Kamili S, Khan BA. Spectrum of hepatitis E virus infection in India. J Med Virol 1994;43:281−6. doi:10.1002/jmv.1890430316. PMID:7931190.
  • Pérez-Gracia MT, García M, Suay B. Mateos-Lindemann ML Current Knowledge on Hepatitis E. J Clin Transl Hepatol. 2015;28: 117−26.
  • Khuroo MS. Discovery of hepatitis E: the epidemic non-A, non-B hepatitis 30 years down the memory lane. Virus Res. 2011;161:3−14. doi:10.1016/j.virusres.2011.02.007. PMID:21320558.
  • Echevarría JM, Fogeda M, Avellón A. Diagnosis of acute hepatitis E by antibody and molecular testing: a study on 277 suspected cases. J Clin Virol. 2011;50:69−71. doi:10.1016/j.jcv.2010.09.016. PMID:21035388.
  • Herremans M, Vennema H, Bakker J, van der Veer B, Duizer E, Benne CA, Waar K, Hendrixks B, Schneeberger P, Blaauw G, et al. Swine-like hepatitis E viruses are a cause of unexplained hepatitis in the Netherlands. J Viral Hepat. 2007;14:140−6. doi:10.1111/j.1365-2893.2006.00786.x. PMID:17244254.
  • Shrestha A, Lama TK, Karki S, Sigdel DR, Rai U, Rauniyar SK, Al-Mahtab M, Takahashi K, Arai M, Akbar SM, et al. Hepatitis E epidemic, Biratnagar, Nepal, 2014. Emerg Infect Dis. 2015;21(4):711−3. doi:10.3201/eid2104.141512. PMID:25811975.
  • Teshale EH, Howard CM, Grytdal SP, Handzel TR, Barry V, Kamili S, Drobeniuc J, Okware S, Downing R, Tappero JW, et al. Hepatitis E epidemic, Uganda. Emerg Infect Dis. 2010;16:126−9. doi:10.3201/eid1601.090764.
  • Jin H, Zhao Y, Zhang X, Wang B, Liu P. Case-fatality risk of pregnant women with acute viral hepatitis type E: a systematic review and meta-analysis. Epidemiol Infect. 2016;144:2098−106. doi:10.1017/S0950268816000418. PMID:26939626.
  • Chaudhry SA, Verma N, Koren G. Hepatitis E infection during pregnancy. Can Fam Physician. 2015;61:607−8. PMID:26175368.
  • Rein DB, Stevens GA, Theaker J, Wittenborn JS, Wiersma ST. The global burden of hepatitis E virus genotypes 1 and 2 in 2005. Hepatology. 2012;55:988−97. doi:10.1002/hep.25505. PMID:22121109.
  • Pérez-Gracia MT, Mateos Lindemann ML, Caridad Montalvo Villalba M. Hepatitis E: current status. Rev Med Virol. 2013;23:384−98. doi:10.1002/rmv.1759. PMID:24038432.
  • Sridhar S, Lau SK, Woo PC. Hepatitis E: A disease of reemerging importance. J Formos Med Assoc. 2015;114:681−90. doi:10.1016/j.jfma.2015.02.003. PMID:25773541.
  • Aggarwal RGS. The global prevalence of hepatitis E virus infection and susceptibility: a systematic review Geneva, Switz, World Health Organization. 2010.
  • Cao YF, Tao H, Hu YM, Shi CB, Wu X, Liang Q, Chi CP, Li L, Liang ZL, Meng JH, et al. A phase 1 randomized open-label clinical study to evaluate the safety and tolerability of a novel recombinant hepatitis E vaccine. Vaccine. 2017;5:5073−80. doi:10.1016/j.vaccine.2017.05.072.
  • Teshale EH, Hu DJ. Hepatitis E: Epidemiology and prevention. World J Hepatol. 2011;27:285−91. doi:10.4254/wjh.v3.i12.285.
  • Lee GY, Poovorawan K, Intharasongkroh D, Sa-Nguanmoo P, Vongpunsawad S, Chirathaworn C, Poovorawan Y. Hepatitis E virus infection: Epidemiology and treatment implications. World J Virol. 2015;12:343−55. doi:10.5501/wjv.v4.i4.343.
  • Lu L, Li C, Hagedorn CH. Phylogenetic analysis of global hepatitis E virus sequences: genetic diversity, subtypes and zoonosis. Rev Med Virol. 2006;16: 5−36. doi:10.1002/rmv.482. PMID:16175650.
  • Cao Y, Zhu L, Liu D, Guo C, Yuan Y, Shi C, Liu Z, Wang X. A novel subgenotype 3a hepatitis E virus isolated from pigs in China. Virus Genes. 2017;53:483−6. doi:10.1007/s11262-017-1435-1. PMID:28205052.
  • Shrestha A, P Gupta B, K Lama T. Current Treatment of Acute and Chronic Hepatitis E Virus Infection: Role of Antivirals. Euroasian J Hepatogastroenterol. 2017;7:73−7. doi:10.5005/jp-journals-10018-1216. PMID:29201777.
  • Wong DC, Purcell RH, Sreenivasan MA, Prasad SR, Pavri KM. Epidemic and endemic hepatitis in India: evidence for a non-A, non-B hepatitis virus aetiology. Lancet 1980;25:876−9. doi:10.1016/S0140-6736(80)92045-0. PMID:6107544.
  • Khuroo MS. Study of an epidemic of non-A, non-B hepatitis. Possibility of another human hepatitis virus distinct from post-transfusion non-A, non-B type. Am J Med 1980;68:818−24. doi:10.1016/0002-9343(80)90200-4. PMID:6770682.
  • Chakraborty S, Datta M, Pasha ST, Kumar S. Non-A non-B viral hepatitis:a common-source outbreak traced to sewage contamination of drinking water. J Commun Dis 1982;14:41−6. PMID:6815259.
  • Velázquez O, Stetler HC, Avila C, Ornelas G, Alvarez C, Hadler SC, Bradley DW, Sepúlveda J. Epidemic transmission of enterically transmitted non-A, non-B hepatitis in Mexico, 1986-1987. JAMA 1990;263:3281−5. doi:10.1001/jama.1990.03440240071018. PMID:2112204.
  • Meng XJ, Purcell RH, Halbur PG, Lehman JR, Webb DM, Tsareva TS, Haynes JS, Thacker BJ, Emerson SU. A novel virus in swine is closely related to the human hepatitis E virus. Proc Natl Acad Sci USA 1997;94:9860−5. doi:10.1073/pnas.94.18.9860. PMID:9275216.
  • Kwo PY, Schlauder GG, Carpenter HA, Murphy PJ, Rosenblatt JE, Dawson GJ, Mast EE, Krawczynski K, Balan V. Acute hepatitis E by a new isolate acquired in the United States. Mayo Clin Proc 1997;72:1133−6. doi:10.4065/72.12.1133. PMID:9413292.
  • Legrand-Abravanel F, Mansuy JM, Dubois M, Kamar N, Peron JM, Rostaing L, Izopet J. Hepatitis E Virus Genotype 3 Diversity, France. Emerg Infect Dis. 2009;15:110−4. doi:10.3201/eid1501.080296. PMID:19116067.
  • Montesano C, Giovanetti M, Ciotti M, Cella E, Lo Presti A, Grifoni A, Zehender G, Angeletti S, Ciccozzi M. Hepatitis E Virus Circulation in Italy: Phylogenetic and Evolutionary Analysis. HepatMon. 2016;16:e31951. doi:10.5812/hepatmon.31951. PMID:27226798.
  • Takahashi M, Nishizawa T, Sato H, Sato Y, Jirintai S, Okamoto H. Analysis of the full-length genome of a hepatitis E virus isolate obtained from a wild boar in Japan that is classifiable into a novel genotype. J Gen Virol. 2011;92:902−8. doi:10.1099/vir.0.029470-0. PMID:21228128.
  • Hsieh SY, Yang PY, Ho YP, Chu CM, Liaw YF. Identification of a novel strain of hepatitis E virus responsible for sporadic acute hepatitis in Taiwan. J Med Virol. 1998;55:300−4. doi:10.1002/(SICI)1096-9071(199808)55:4%3c300::AID-JMV8%3e3.0.CO;2-4. PMID:9661839.
  • Geng Y, Zhao C, Fan J, Harrison TJ, Zhang H, Lian H, Geng K, Wang Y. Genotype analysis of hepatitis E virus from sporadic hepatitis E cases in northern China. Infect Genet Evol. 2013;20:413−7. doi:10.1016/j.meegid.2013.10.003. PMID:24140474.
  • Nakano T, Takahashi K, Takahashi M, Nishigaki Y, Watanabe N, Ishida S, Fujimoto S, Kato H, Okano H, Takei Y. Investigating the origin and global dispersal history of hepatitis E virus genotype 4 using phylogeographical analysis. LiverInt. 2016;36:31−41. doi.110.1111/liv.12880. PMID:26037061.
  • Begum N, Polipalli SK, Husain SA, Kar P. Molecular analysis of swine hepatitis E virus from north India. Indian J Med Re. 2010;132:504−8. PMID:21149998.
  • Clemente-Casares P, Ramos-Romero C, Ramirez-Gonzalez E, Mas A. Hepatitis E Virus in Industrialized Countries: The Silent Threat. Biomed ResInt. 2016;2016:9838041. doi:10.1155/2016/9838041. PMID:28070522.
  • Wedemeyer H, PischkeS, Manns MP. Pathogenesis and treatment of hepatitis e virus infection. Gastroenterology. 2012;142:1388−97. doi:10.1053/j.gastro.2012.02.014. PMID:22537448.
  • Ahmad, I., HollaRP, JameelS. Molecular virology of hepatitis E virus. Virus Res. 2011;161:47−58. doi:10.1016/j.virusres.2011.02.011. PMID:21345356.
  • Panda SK, Thakral D, Rehman S. Hepatitis E virus. Rev Med Virol. 2007;17:151−80. doi:10.1002/rmv.522. PMID:17051624.
  • Chandra V, Taneja S, Kalia M, Jameel S. Molecular biology and pathogenesis of hepatitis E virus. J Biosci. 2008;33: 451−64. doi:10.1007/s12038-008-0064-1. PMID:19208971.
  • Nan Y, Zhang YJ. Molecular Biology and Infection of Hepatitis E Virus, Front Microbiol. 2016;7:1419. doi:10.3389/fmicb.2016.01419. PMID:27656178.
  • Wang H, Zhang W, Gu H, Chen W, Zeng M, Ji C, Song R, Zhang G. Identification and characterization of two linear epitope motifs in hepatitis E virus ORF2 protein. PLoS One. 2017;12: e0184947. doi:10.1371/journal.pone.0184947. PMID:28957334.
  • Reyes GR, Huang CC, Tam AW, Purdy MA. Molecular organization and replication of hepatitis E virus (HEV). Arch Virol Suppl 1993;7:15−25. doi:10.1007/978-3-7091-9300-6_2. PMID:8219799.
  • Li TC, Takeda N, Miyamura T, Matsuura Y, Wang JC, Engvall H, Hammar L, Xing L, Cheng RH. Essential elements of the capsid protein for self-assembly into empty virus-like particles of hepatitis E virus. J Virol. 2005;79:12999−3006. doi:10.1128/JVI.79.20.12999-13006.2005. PMID:16189002.
  • Xing L, Li TC, Mayazaki N, Simon MN, Wall JS, Moore M, Wang CY, Takeda N, Wakita T, Miyamura T. Structure of hepatitis E virion-sized particle reveals an RNA-dependent viral assembly pathway. J Biol Chem. 2010;285:33175−83. doi:10.1074/jbc.M110.106336. PMID:20720013.
  • Guu TS, Liu Z, Ye Q, Mata DA, Li K, Yin C, Zhang J, Tao YJ. Structure of the hepatitis E virus-like particle suggests mechanisms for virus assembly and receptor binding. Proc Natl Acad Sci USA. 2009;106:12992−7. doi:10.1073/pnas.0904848106. PMID:19622744.
  • Zhou YH, Purcell RH, Emerson SU. A truncated ORF2 protein contains the most immunogenic site on ORF2: Antibody responses to non-vaccine sequences following challenge of vaccinated and non-vaccinated macaques with hepatitis E virus. Vaccine. 2005;23:3157−65. doi:10.1016/j.vaccine.2004.12.020. PMID:15837215.
  • Emerson SU, Clemente-Casares P, Moiduddin N, Arankalle VA, Torian U, Purcell RH. Putative neutralization epitopes and broad cross-genotype neutralization of Hepatitis E virus confirmed by a quantitative cell-culture assay. J Gen Virol. 2006;87:697−704. doi:10.1099/vir.0.81545-0. PMID:16476993.
  • Taherkhani R, Makvandi M, Farshadpour F. Development of enzyme-linked immunosorbent assays using 2 truncated ORF2 proteins for detection of IgG antibodies against hepatitis E virus. Ann Lab Med. 2014;34:118−26. doi:10.3343/alm.2014.34.2.118. PMID:24624347.
  • Emerson SU, Purcell RH. Recombinant vaccines for hepatitis E. Trends Mol Med. 2001;7:462−6. doi:10.1016/S1471-4914(01)02106-2. PMID:11597521.
  • Kamar N, Izopet J, Pavio N, Aggarwal R, Labrique A, Wedemeyer H, Dalton HR. Hepatitis E virus infection. Nat Rev Dis Primers. 2017;3:17086. doi:10.1038/nrdp.2017.86. PMID:29154369.
  • Zhao Q, Li S, Yu H, Xia N, Modis Y. Virus-like particle-based human vaccines: quality assessment based on structural and functional properties. Trends Biotechnol. 2013;31(11):654−63. doi:10.1016/j.tibtech.2013.09.002. PMID:24125746.
  • Zhang X, Wei M, Pan H, Lin Z, Wang K, Weng Z, Zhu Y, Xin L, Zhang J, Li S, et al. Robust manufacturing and comprehensive characterization of recombinant hepatitis E virus-like particles in Hecolin®. Vaccine. 2014;32(32):4039−50. doi:10.1016/j.vaccine.2014.05.064. PMID:24892250.
  • Purdy MA, McCaustland KA, Krawczynski K, Spelbring J, ReyesGR, Bradley DW. Preliminary evidence that a trpE-HEV fusion protein protects cynomolgus macaques against challenge withwild-type hepatitis E virus (HEV). J Med Virol. 1993;41:90−4. doi:10.1002/jmv.1890410118. PMID:8228944.
  • Meng J, Dai X, Chang JC, Lopareva E, Pillot J, Fields HA, Khudyakov YE. Identification and characterization of the neutralization epitope(s) of the hepatitis E virus. Virol. 2001;288:203−11. doi:10.1006/viro.2001.1093.
  • Wen J, Behloul N, Dai X, Dong C, Liang J, Zhang M, Shi C, Meng J. Immunogenicity difference between two hepatitis E vaccines derived from genotype 1 and 4. Antiviral Res. 2016;128:36−42. doi:10.1016/j.antiviral.2016.02.002. PMID:26850829.
  • Dong C, Meng JH. Expression, purification and immunogenicity of a novel hepatitis E virus-like particle. Chinese J Cellular Mol Immunol. 2006;22:339−42.
  • Zhang J, Liu CB, Li RC, Li YM, Zheng YJ, Li YP, Luo D, Pan BB, Nong Y, Ge SX. Randomized-controlled phase 2 clinical trial of a bacterially expressed recombinant hepatitis E vaccine. Vaccine. 2009;27:1869−74. doi:10.1016/j.vaccine.2008.12.061. PMID:19168109.
  • Li SW, Zhao Q, Wu T, Chen S, Zhang J, Xia NS. The development of a recombinant hepatitis E vaccine HEV 239. Hum Vaccin Immunother. 2015;11: 908−14. doi:10.1080/21645515.2015.1008870. PMID:25714510.
  • Wu T, Li SW, Zhang J, Ng MH, Xia NS, Zhao Q. Hepatitis E vaccine development: a 14 year odyssey. Hum Vaccin Immunother. 2012;8:823−7. doi:10.4161/hv.20042. PMID:22699438.
  • Zhu FC, Zhang J, Zhang XF, Zhou C, Wang ZZ, Huang SJ, Wang H, Yang CL, Jiang HM, Cai JP. Efficacy and safety of a recombinant hepatitis E vaccine in healthy adults: a large-scale, randomised, double-blind placebo-controlled, phase 3 trial. Lancet. 2010;376:895−902. doi:10.1016/S0140-6736(10)61030-6. PMID:20728932.
  • Zhang J, Zhang XF, Huang SJ, Wu T, Hu YM, Wang ZZ, Wang H, Jiang HM, Wang YJ, Yan Q. Long-term efficacy of a hepatitis E vaccine. N Engl J Med. 2015;372:914−22. doi:10.1056/NEJMoa1406011. PMID:25738667.
  • Sehgal D, Malik PS, Jameel S. Purification and diagnostic utilityof a recombinant hepatitis E virus capsid protein expressed in insect larvae. Protein Expr Purif. 2003;27:27−34. doi:10.1016/S1046-5928(02)00574-0. PMID:12509981.
  • Robinson RA, Burgess WH, Emerson SU, Leibowitz RS, Sosnovtseva SA, Tsarev S, Purcell RH. Structural characterization of recombinant hepatitis E virus ORF2 proteins in baculovirus-infected insect cells. Protein Expr Purif 1998;12:75−84. doi:10.1006/prep.1997.0817. PMID:9473460.
  • Zhang M, Emerson SU, Nguyen H, Engle R, Govindarajan S, Blackwelder WC, Gerin J, Purcell RH. Recombinant vaccine against hepatitis E: duration of protective immunity in rhesus macaques. Vaccine. 2002;20:3285−91. doi:10.1016/S0264-410X(02)00314-6. PMID:12213398.
  • Shrestha MP, Scott RM, Joshi DM, Mammen MP Jr, Thapa GB, Thapa N, Myint KS, Fourneau M, Kuschner RA, Shrestha SK. Safety and efficacy of a recombinant hepatitis E vaccine. N Engl J Med. 2007;356:895−903. doi:10.1056/NEJMoa061847. PMID:17329696.
  • Qi Y, Fan J, Huang W, Zhao C, Wang Y, Kong FT, Kong W, Jiang C. Expression and characterization of hepatitis E virus-like particles and non-virus-like particles from insect cells. Biotechnol Appl Biochem. 2016;63:362−70. doi:10.1002/bab.1379. PMID:25824972.
  • Xu M, Behloul N, Wen J, Zhang J, Meng J. Role of asparagine at position 562 in dimerization and immunogenicity of the hepatitis E virus capsid protein; Infect Genet Evol. 2016;37:99−107. doi:10.1016/j.meegid.2015.11.006. PMID:26584510.
  • Yang EC, Chi B R, Li X, Liu Y, Gao P, Jia P, Kan SF, Wen ZM, Wang W, Jin NY. Expression and Characterization of a Recombinant Truncated Capsid Protein of Hepatitis E Virus in Pichia pastoris. Chemical Research in Chinese Universities. 2010;26:235–9.
  • Su CX, Gu MR, Zhang P, Jin ZJ, Meng FH, Chen EJ, Yang Z, Liu Y, Wang YC. Expression of ORF2 protein of HEV genotype IV in Hansenula polymorpha. Chinese J Biotechnol. 2007;23:73−8; PMID:17366891.
  • Tong Y, Zhan M, Lu J, Bai Y, Bi S. Immunogenicity of recombinant HEV ORF2 protein expressed in pichia pastoris. Chinese J Exp Clin Virol. 2002;16:23−6; PMID:11986739.
  • Trabelsi K, Kamen A, Kallel H. Development of a vectored vaccine against hepatitis E virus. Vaccine. 2014;32:2808−11. doi:10.1016/j.vaccine.2014.02.041. PMID:24583005.
  • Jiménez de Oya N, Escribano-Romero E, Blázquez AB, Lorenzo M, Martín-Acebes MA, Blasco R, Saiz JC. Characterization of hepatitis E virus recombinant ORF2 proteins expressed by vaccinia viruses. J Virol. 2012;86:7880−6. doi:10.1128/JVI.00610-12. PMID:22593167.
  • Farshadpour F, Makvandi M, Taherkhani R. Design, Construction and Cloning of Truncated ORF2 and tPAsp-PADRE-Truncated ORF2 Gene Cassette From Hepatitis E Virus in the pVAX1 Expression Vector. Jundishapur J Microbiol. 2015;8: e26035. doi:10.5812/jjm.26035. PMID:26865938.
  • Dong C, Dai X, Meng JH. The first experimental study on a candidate combined vaccine against hepatitis A and hepatitis E. Vaccine. 2007;25:1662−8. doi:10.1016/j.vaccine.2006.11.001. PMID:17156900.
  • Xiang K, Kusov Y, Ying G, Yan W, Shan Y, Jinyuan W, Na Y, Yan Z, Hongjun L, Maosheng S. A Recombinant HAV Expressing a Neutralization Epitope of HEV Induces Immune Response against HAV and HEV in Mice. Viruses. 2017;9.pii: E260. doi:10.3390/v9090260.
  • Gao Y, Su Q, Yi Y, Jia Z, Wang H, Lu X, Qiu F, Bi S. Enhanced mucosal immune responses induced by a combined candidate mucosal vaccine based on Hepatitis A virus and Hepatitis E virus structural proteins linked to tuftsin. PLoS One. 2015;10: e0123400. doi:10.1371/journal.pone.0123400. PMID:25875115.
  • Li HZ, Gang HY, Sun QM, Liu X, Ma YB, Sun MS, Dai CB. Production in Pichia pastoris and characterization of genetic engineered chimeric HBV/HEV virus-like particles. Chin Med Sci J. 2004;19:78−83. PMID:15250239.
  • Wang L, Cao D, Wei C, Meng XJ, Jiang X, Tan M. A dual vaccine candidate against norovirus and hepatitis E virus. Vaccine. 2014;32:445−52. doi:10.1016/j.vaccine.2013.11.064. PMID:24291540.
  • Makvandi M, Teimoori A, Neisi N, Samarbafzadeh A. Designing, Construction and Expression of a Recombinant Fusion Protein Comprising the Hepatitis E Virus ORF2 and Rotavirus NSP4 in the Baculovirus Expression System. Jundishapur J Microbiol. 2016;9:e40303. doi:10.5812/jjm.40303. PMID:28138375.
  • Xia M, Wei C, Wang L, Cao D, Meng XJ, Jiang X, Tan M. Development and evaluation of two subunit vaccine candidates containing antigens of hepatitis E virus, rotavirus, and astrovirus. Sci Rep. 2016;6:25735. doi:10.1038/srep25735. PMID:27194006.
  • Li T, Takeda N, Miyamura T. Oral administration of hepatitis E virus-like particles induces a systemic and mucosal immune response in mice. Vaccine. 2001;19:3476−84. doi:10.1016/S0264-410X(01)00059-7. PMID:11348714.
  • Li TC, Suzaki Y, Ami Y, Dhole TN, Miyamura T, Takeda N. Protection of cynomolgus monkeys against HEV infection by oral administration of recombinant hepatitis E virus-like particles. Vaccine. 2004;22:370−7. doi:10.1016/j.vaccine.2003.08.004. PMID:14670318.
  • Gao S, Li D, Liu Y, Zha E, Zhou T, Yue X. Oral immunization with recombinant hepatitis E virus antigen displayed on the Lactococcus lactis surface enhances ORF2-specific mucosal and systemic immune responses in mice. Int Immunopharmacol. 2015;24:140−5. doi:10.1016/j.intimp.2014.10.032. PMID:25445956.
  • Ma Y, Lin SQ, Gao Y, Li M, Luo WX, Zhang J, Xia NS. Expression of ORF2 partial gene of hepatitis E virus in tomatoes and immunoactivity of expression products. World J Gastroenterol. 2003;9:2211−5. doi:10.3748/wjg.v9.i10.2211. PMID:14562380.
  • Meng J, Dubreuil P, Pillot J. A new PCR-based seroneutralization assay in cell culture for diagnosis of hepatitis E. J Clin Microbiol 1997;35(6):1373−7. PMID:9163446.
  • Takahashi M, Hoshino Y, Tanaka T, Takahashi H, Nishizawa T, Okamoto H. Production of monoclonal antibodies against hepatitis E virus capsid protein and evaluation of their neutralizing activity in a cell culture system. Arch Virol. 2008;153(4):657−66. doi:10.1007/s00705-008-0045-6. PMID:18266052.
  • Yang F, Tang Z, Wang S, Cai W, Wen G, Ji W, Yu J, Zhang K, Xia N, Zheng Z. The establishment of high-through put neutralization titer evaluation model for hepatitis E virus (HEV). Bing Du Xue Bao. 2015;31(1):1−6. PMID:25997322.
  • Wu X, Chen P, Lin H, Su Y, Hao X, Cao Y, Li L, Zhu F, Liang Z. Dynamics of 8G12 competitive antibody in "prime-boost" vaccination of Hepatitis E vaccine. Hum Vaccin Immunother. 2017;13(6):1−6. doi:10.1080/21645515.2017.1291105.
  • Hepatitis E FAQs for Health Professionals. 2017. Available online: http://www.cdc.gov/hepatitis/hev/hevfaq.htm.
  • Ogholikhan S, Schwarz KB. Hepatitis Vaccines. Vaccines (Basel). 2016;4. pii:E6. doi:10.3390/vaccines4010006. PMID:26978406.
  • Haffar S, Bazerbachi F, Lake JR. Making the case for the development of a vaccination against hepatitis E. Virus Liver Int. 2015;35:311−6. doi:10.1111/liv.12590. PMID:24836400.
  • Kamar N, Mansuy J.M, Cointault O, Selves J, Abravanel F, Danjoux M, Otal P, Esposito L, Durand D, Izopet J. Hepatitis E virus-related cirrhosis in kidney-and kidney-pancreas-transplant recipients. Am J Transplant. 2008;8:1744−8. doi:10.1111/j.1600-6143.2008.02286.x. PMID:18557740.
  • Larralde O, Petrik J. Phage-displayed peptides that mimic epitopes of hepatitis E virus capsid. Med Microbiol Immunol. 2017;206:301−9. doi:10.1007/s00430-017-0507-0. PMID:28434129.
  • Wang H, Ji F, Liang H, Gu H, Ning Z, Liu R, Zhang G. A Proline-Rich Domain in the Genotype 4Hepatitis E Virus ORF3 C-Terminus is Crucial for Downstream V105DLP108 Immunoactivity. PLoS ONE. 2015;10:e0133282. doi:10.1371/journal.pone.0133282. PMID:26177202.
  • Wang Y, Zhao C, Qi Y, Geng Y. Hepatitis E Virus. Adv Exp Med Biol. 2016;948:1−16. doi:10.1007/978-94-024-0942-0_1. PMID:27738976.
  • Tsarev SA, Tsareva TS, Emerson SU, Govindarajan S, Shapiro M, Gerin JL. Successful passive and active immunization of cynomolgus monkeys against hepatitis E. Proc Natl Acad Sci 1994;91:10198−202. doi:10.1073/pnas.91.21.10198. PMID:7937861.
  • Bryan JP, Tsarev SA, Iqbal M, Ticehurst J, Emerson S, Ahmed A, Duncan J, Rafiqui AR, Malik IA, Purcell RH, et al. Epidemic hepatitis E in Pakistan: patterns of serologic response and evidence that antibody to hepatitis E virus protects against disease. J Infect Dis 1994;170:517−21. doi:10.1093/infdis/170.3.517. PMID:8077708.
  • Zhao Q, Zhang J, Wu T, Li SW, Ng MH, Xia NS, Shih JW. Antigenic determinants of hepatitis E virus and vaccine-induced immunogenicity and efficacy. J Gastroenterol. 2013;48(2):159−68. doi:10.1007/s00535-012-0701-1. PMID:23149436.
  • Aggarwal R, Shukla R, Jameel S, Agrawal S, Puri P, Gupta VK, Patil AP, Naik S. T-cell epitope mapping of ORF2 and ORF3 proteins of human hepatitis E virus. J Viral Hepat. 2007;14(4):283−92. doi:10.1111/j.1365-2893.2006.00796.x. PMID:17381721.
  • Behloul N, Wei W, Baha S, Liu Z, Wen J, Meng J. Effects of mRNA secondary structure on the expression of HEV ORF2 proteins in Escherichia coli. Microb Cell Fact. 2017;16:200. doi:10.1186/s12934-017-0812-8. PMID:29137642.
  • Behloul N, Wen J, Dai X, Dong C, Meng J. Antigenic composition and immunoreactivity differences between; HEV recombinant capsid proteins generated from different genotypes. Infect Genet Evol. 2015;34:211−20. doi:10.1016/j.meegid.2015.06.026. PMID:26122075.
  • Zhao M, Li XJ, Tang ZM, Yang F, Wang SL, Cai W, Zhang K, Xia NS, Zheng ZZ. A Comprehensive Study of Neutralizing Antigenic Sites on the Hepatitis E Virus (HEV) Capsid by Constructing, Clustering, and Characterizing a Tool Box. J Biol Chem. 2015;290:19910−22. doi:10.1074/jbc.M115.649764. PMID:26085097.
  • Shima R, Li TC, Sendai Y, Kataoka C, Mori Y, Abe T, Takeda N, Okamoto T, Matsuura Y. Production of hepatitis E virus-like particles presenting multiple foreign epitopes by co-infection of recombinant baculoviruses. Sci Rep. 2016;6:21638. doi:10.1038/srep21638. PMID:26905478.
  • Beran J, Van Der Meeren O, Leyssen M, D'silva P. Immunity to hepatitis A and B persists for at least 15 years after immunisation of adolescents with a combined hepatitis A and B vaccine. Vaccine. 2016;34(24):2686−91. doi:10.1016/j.vaccine.2016.04.033. PMID:27105563.
  • Labrique AB, Sikder SS, Krain LJ, West KP Jr, Christian P, Rashid M, Nelson KE. Hepatitis E, a vaccine-preventable cause of maternal deaths. Emerg Infect Dis. 2012;18:1401−4. doi:10.3201/eid1809.120241. PMID:22931753.
  • Andrews J. Military sponsored vaccine trials and la resistance in Nepal. Am J Bioeth. 2005;5: W1−3. doi:10.1080/15265160591002962. PMID:16006352.
  • Zhang J, Shih JW, Wu T, Li SW, Xia NS. Development of the hepatitis E vaccine: from bench to field. Semin Liver Dis. 2013;33:79−88. doi:10.1055/s-0033-1338116. PMID:23564392.
  • Reports on Notifiable Infectious Disease in China. Chinese Centerfor Disease Control and Prevention. Available online at: http://www.chinacdc.cn/tjsj/fdcrbbg/.
  • Utsumi T, Hayashi Y, Lusida MI, Amin M, Soetjipto, Hendra A, Soetjiningsih, Yano Y, Hotta H. Prevalence of hepatitis E virus among swine and humans in two different ethnic communities in Indonesia. Arch Virol. 2011;156:689−93. doi:10.1007/s00705-010-0883-x. PMID:21191625.
  • Kim JH, Nelson KE, Panzner U, Kasture Y, Labrique AB, Wierzba TF. A systematic review of the epidemiology of hepatitis E virus in Africa. BMC Infect Dis. 2014;14:308. doi:10.1186/1471-2334-14-308. PMID:24902967.
  • Khuroo MS, Khuroo MS. Hepatitis E: an emerging global disease from discovery towards control and cure. J Viral Hepat. 2016;23: 68−79. doi:10.1111/jvh.12445. PMID:26344932.
  • Lam E, McCarthy A, Brennan M. Vaccine-preventable diseases in humanitarian emergencies among refugee and internally displaced populations. Hum Vaccin Immunother. 2015;11:2627−36. doi:10.1080/21645515.2015.1096457. PMID:26406333.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.