Open Access

Production and immunogenicity of different prophylactic vaccines for hepatitis C virus (Review)

  • Authors:
    • Qianqian Zhao
    • Kun He
    • Xiuhua Zhang
    • Mingjie Xu
    • Xiuping Zhang
    • Huanjie Li
  • View Affiliations

  • Published online on: May 30, 2022     https://doi.org/10.3892/etm.2022.11401
  • Article Number: 474
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatitis C virus (HCV) infection is a global health challenge, and prophylactic vaccines are the most effective way to eliminate the infection. To date, numerous forms of preventive vaccines have entered the clinical trial stage, including the virus‑like particle (VLP) vaccine, recombinant subunit vaccine, peptide vaccine and nucleic acid vaccine. The rational design makes it easier to obtain specific vaccine structures with a broad spectrum and strong immunogenicity. Different vaccine antigens can evoke different immune responses, including humoral and T‑cell immune responses, and can be produced using different expression systems, such as bacteria, yeast, mammals, plants, insects or parasites. Intracellular and insoluble production and a narrow immune spectrum are two difficulties that limit the application of vaccines. The present study summarizes the immunogenicity of different preventive vaccines, evaluates the characteristics of different expression systems used for vaccine production, and analyzes the strategies to enhance the secretion and immune spectrum of vaccine proteins.

1. Introduction

Hepatitis C virus (HCV) infection is a global health problem that human beings have struggled with for ~30 years. Individuals infected with HCV develop chronic hepatitis, and a proportion might develop cirrhosis and liver carcinoma. The World Health Organization has set a goal to eliminate the public health burden of HCV by 2030(1). A platform in the use of medicines to treat chronic hepatitis C was constructed in Australia for the collection and management of treatment, virological outcome and other relevant clinical data of patients with HCV to better inform the limitations of HCV infection (2). Although the development of direct-acting antiviral agents (DAAs) for chronic HCV has resulted in a 95% cure rate for patients infected with HCV genotypes (3), there are still millions of new infections and tens of millions of re-infections worldwide every year. In such cases, an effective prophylactic vaccine is necessary to control HCV infection. Significant progress has been made in the development of vaccines against HCV in the past 10 years despite no commercial vaccine having appeared so far. The characteristics and properties of different forms of vaccines and the advantages and disadvantages of various vaccine expression systems are summarized in the present study, to provide new insights into the research and development of HCV preventive vaccines.

In the present review, a literature search was performed using the PubMed, Elsevier Science Direct and China National Knowledge Infrastructure databases with ‘Hepatitis C virus’ and ‘vaccine’ as the primary key words. On this basis, ‘immunogenicity’, ‘immune response’, ‘production’, ‘expression’ and ‘rational design’ were used as key words for the secondary search. Studies related to therapeutic vaccines were excluded, unless they described the immune protection on HCV genotypes different from the infected one. Studies not written in English or Chinese were excluded.

2. Different forms of preventive vaccines and their immunogenicity

HCV is a positive-sense, single-stranded RNA virus that belongs to the Flaviviridae family. The HCV genome contains two untranslated regions and an open reading frame encoding structural proteins [core, envelope protein 1 (E1) and envelope protein 2 (E2)] and non-structural proteins (p7, NS2, NS3, NS4A, NS4B, NS5A and NS5B) (Fig. 1) (4). The complete or partial genome has been used for vaccine preparation. Compared with the attenuated vaccine or inactivated vaccine prepared using live viruses, the preparation of vaccine using genetic engineering technology has further improved its safety and immunogenicity (5). Genetically engineered vaccines are widely used to prevent infection by various viruses (6). Different forms of genetically engineered vaccines have been developed, and their properties are summarized in Fig. 2.

VLP vaccine

The structural proteins of HCV can self-assemble into viral particles without infection in vitro. VLP vaccines can not only induce T-cell (CD4+ and CD8+) immune responses, but also stimulate the formation of protective neutralizing antibodies (7,8). Research has found that different assembly forms lead to different particle sizes (diameter ranging from 30-80 nm), in which only particles of a specific size (55-60 nm in diameter) are infectious and can cause specific antibody responses (9). VLPs with the same gene sequence can be assembled into different structures in different expression systems. For example, VLP vaccines containing the same structural protein sequences have distinct immunogenicity due to the different glycosylation mechanisms in insect and mammalian cells (10,11). This suggests that the composition of the sugar chain could affect the conformation of the whole virus particle. In addition, some chimeric VLPs have been widely used. To date, the hepatitis B virus (HBV) core protein (12), the small (S) envelope protein of HBV (13), and papaya mosaic virus coat protein (PapMVCP) (14) have been fused with partial or total sequences of HCV envelope protein to prepare VLP vaccines. However, the immunogenicity of chimeric VLPs is lower than that of vaccines prepared using HCV self-proteins.

VLPs have been proven to induce protective immune responses against viruses without adjuvants, and some studies have shown that the use of adjuvants can significantly enhance the immunogenicity of VLP vaccines (15-17). Use of an anionic self-adjuvanting lipopeptide containing the Toll-like receptor 2 agonist Pam2Cys (E8Pam2Cys) enhanced the immunogenicity of VLPs composed of HCV structural proteins (core, E1 and E2) (15). The improvement in VLP and E2-specific antibody responses in VLP + E8Pam2Cys vaccinated mice required up to three doses of non-adjuvanted VLPs to match the antibody titers obtained with a single dose of VLPs formulated with this lipopeptide. Further research found that co-formulation of this lipopeptide with VLPs could improve dendritic cell uptake and maturation, and could also induce better VLP-specific interferon (IFN)-γ-mediated responses.

Recombinant subunit vaccine

The recombinant subunit vaccine is mainly concentrated in the envelope proteins E1 and E2 of the HCV, as the epitopes on the envelope proteins are important for virus invasion and neutralizing antibody identification (18,19). Recombinant subunit vaccines based on envelope proteins can inhibit viral infections by stimulating protective human neutralizing antibodies (HMAbs). Topological analysis showed that E1 is a multi-pass transmembrane protein (crosses the membrane three times) with most domains inside, and E2 is a single-pass transmembrane protein with most domains outside (Fig. 3A). Antigenic epitopes are concentrated in the E2 ectodomain due to the existence of binding sites for cellular factors such as CD81, scavenger receptor class B type I and claudin-1 (20-22). Antigen epitopes on the surface of the envelope protein seriously affect a broad spectrum of vaccines. HMAbs generated by conservative antigen epitopes (divided into linear and conformational epitopes) have a wide range of neutralization characteristics and can cross-neutralize HCVs of different genotypes. HMAbs (AP33, 3/11, 95-2 and HCV-1) corresponding to the 412-423 amino acid (aa) linear epitope show extensive neutralizing activity against HCV pseudoparticles (HCVpp) of genotype 1-6 (Fig. 3B and C) (23); HMAbs (CBH2, HC11 and HC1) or antigen region 3 (AR3) corresponding to the conformational epitopes of two discontinuous sequences, 424-443/523-540 aa, or three discontinuous sequences, 394-424/437-447/523-540 aa, can also widely neutralize HCVpp of genotypes 1-6 (Fig. 3B and C) (24). Further experiements on virus-escaping mutants showed that no virus escape body was found in antibody HC1 at different experimental concentrations of 0.05-100 µg/ml (25). This indicated that the influence of the conformational epitope on the broad spectrum of the vaccine was greater than that of the linear epitope. The discovery that epitopes determine the broad spectrum of neutralizing antibodies will help in artificially designing specific vaccine structures to stimulate broadly neutralizing antibodies (26,27). Notably, not all epitopes could induce the production of protective neutralizing antibodies. The interfering antibody induced by the E2 region 434-446 aa does not have neutralizing activity; however, its binding to E2 can mask the binding of other neutralizing antibodies to the adjacent antigen epitope region 412-426 aa, thereby reducing the effective neutralizing activity corresponding to this epitope (28,29). Thus, elicitation of antibodies with interfering capacity should be avoided when producing an effective cross-neutralizing vaccine.

The epitopes of E2 are mostly concentrated in the ectodomain; therefore, the removal of the C-terminal transmembrane domain (TMD) has no significant effect on its immunogenicity (30). In addition, the truncated soluble E2, after removing hypervariable (HVR)1, HVR2 and intergenotypic variable region, can still be folded correctly, but the immunogenicity is reduced (31-33). Changing the glycosylation modification mode of the envelope protein also causes changes in its antigenicity and immunogenicity. A comparison of the two glycosylation modification modes in insect and mammalian cells showed that the sugar chain was essential for the immunogenicity of the E2 vaccine, but the complex sugar chain was not conducive to immunogenicity (34). Further research showed that insect E2 induced stronger neutralizing antibody responses against the homologous isolate used in the vaccine, but the two proteins elicited comparable neutralization titers against heterologous isolates (35). Adjuvants are required to exert the immunogenicity of recombinant subunit vaccines (36,37). Co-immunization with recombinant E2 vaccine and the saponin adjuvant QuilA, prepared by mammalian cells, can produce anti-E2 antibody titer much higher than that found using Freund's, monophosphoryl lipid A, cytosine phosphorothioate guanine oligodeoxynucleotide (CpG ODN) or α-galactosylceramide derivatives, and the effect of two adjuvant combinations (QuilA and CpG ODN) is better than that of a single adjuvant (36).

Peptide vaccine

Peptide vaccines with immunogenicity can be screened by constructing a phage expression peptide library or directly synthesized using cross-neutralization epitopes, specific major histocompatibility complex class I epitopes and T helper cell epitopes. Such vaccines contain only limited epitopes and cannot stimulate a wide range of immune responses. In patients, after standard antiviral treatment, peptide vaccines can induce HCV-specific T-cell responses to enhance the sustained virological response and reduce relapse rates (38). However, the immunogenicity of vaccines can be enhanced by modification. For example, in one study, the affinity and immunogenicity of peptide vaccines were improved after the leucine at position 8 of the cytotoxic T lymphocyte epitope of HCV core antigen (132-140 aa) was mutated to alanine (39). A nano-polypeptide vaccine based on an NS3 polypeptide mixture can stimulate stronger CD4+ T-cell responses and induce stronger CD8+ T-cell immune responses than the NS3 polypeptide vaccine (40). In addition, the latest research showed that an overlapping peptide nanoparticle vaccine prepared based on the p7 protein successfully stimulated CD4+ and CD8+ T-cell responses (41). This was the first study to demonstrate the immunogenicity of p7 as a vaccine target and provides a new idea for the preparation of peptide vaccines. Different application schedules and injection routes may also influence the immunogenicity of HCV peptide vaccines. In a phase I clinical trial for the dose and injection route of the HCV peptide vaccine IC41, when increasing the frequency of vaccinations from 4 or 6 to 8 or 16 times per cycle (16 weeks), and decreasing the time window from 4 weeks to 1 or 2 weeks, the T-cell response rates, in particular the rates measuring CD8+ T-cell function, were enhanced up to 2-fold compared with previous studies using the same formulation of the IC41 vaccine (42-44).

Nucleic acid vaccine

DNA vaccines are prepared based on the coding sequence of HCV structural proteins or non-structural proteins such as E1, E2, NS3, NS4A, NS4B, NS5A or inactivated NS5B (NSmut) gene. Viruses (adenovirus or vaccinia virus) and eukaryotic cytoplasmic particles are usually used as vaccine vectors. DNA vaccines made of structural protein-coding sequences usually stimulate humoral immune responses and produce protective neutralizing antibodies (45), whereas vaccines derived from non-structural protein sequences mainly sustain cellular immune responses. A prime-boost vaccine with chimpanzee-derived adenovirus-3 NSmut and modified vaccinia Ankara NSmut could successfully induce CD4+ and CD8+ T-cell responses to all candidate HCV antigen epitopes, but its phase I clinical trial results showed that the candidate vaccine did not provide better protection against chronic HCV infection than the placebo (clinicaltrials.gov identifier: NCT01436357) (46). It was found that the relative frequency of CD4+, CD25+ and Foxp3+ regulatory T cells (Tregs) was increased in the blood and liver in patients with chronic persistent HCV infection even after successful DAA treatment (47,48). Tregs were demonstrated to attenuate vaccine-induced protective CD4+ and CD8+ T-cell immune responses (49). Therefore, a novel HCV DNA vaccine, GLS-6150, consisting of the DNA plasmid encoding adjuvant IFNL3 with DNA plasmids encoding the HCV non-structural proteins (NS3/NS4A, NS4B and NS5A genes), reduced the frequency of Tregs and increased HCV-specific T-cell responses in a phase I clinical trial (NCT02027116) (50). Another study reported that the immunogenicity of a DNA vaccine mixture (vaccine cocktail) composed of conserved sequences of coding proteins of multiple HCV genotypes was much greater than that of DNA vaccines from a single genotype (51). The breadth and intensity of the T-cell response have been improved, although clinical experiments have not yet been performed. Experiments on safety showed that the injection of DNA vaccines does not impair the ability of the body to respond to non-HCV antigens (52).

Rationally designed vaccine

In the early stages of vaccine rational design research, neutralizing antibodies associated with specific virus clearance can be found by analyzing the antibody spectrum of the virus, and then specific immunogens can be rationally designed to facilitate the production of such antibodies. For example, reasonable design of a specific epitope in E2 can produce HC33.1 antibody with strong antiviral effect (53). A recent study successfully constructed an AP33 epitope structure simulant using an anti-idiotypic method. In a mouse model, an antibody with the same recognition site and residue as AP33 was induced as an immunogen, and the antibody had the ability to resist HCV infection (54). This research opens up a new method for the rational design of vaccines. Limited by technology, the rational design thus far is restricted to linear epitopes and the analog design of conformational epitopes cannot be realized.

The rational design of conformational epitope vaccines must be based on an in-depth study of protein structure and immune recognition information. Envelope glycoprotein E2 has a highly conserved neutralization surface that is not covered by the N-linked glycans and has three main overlapping neutralization sites: Antigen site 412, 434 and AR3. Although it is composed of highly conserved amino acid residues, its conformation is flexible (21). It is difficult to obtain ideal immune effects for peptides designed according to epitope sequences, possibly since they do not have the correct conformation. Analysis of the preferred conformation of the neutralization site provides a basis for the design of a conformational vaccine.

3. Different expression systems for vaccine preparation

To date, candidate vaccine proteins composed of partial or complete protein sequences of HCV have been successfully produced in a variety of expression systems, such as mammalian and insect cells, Pichia pastoris, Escherichia coli expression systems, plant leaf expression systems, and even the parasitic host Leishmania tarentolae. The hosts and expression vectors used in different expression systems are summarized in Table I.

Table I

Summary of different hepatitis C virus vaccine candidate expression systems.

Table I

Summary of different hepatitis C virus vaccine candidate expression systems.

Expression systemHost cell lineVectorVaccine typeVaccine antigenExpression positionMax. expression level(Refs.)
Mammalian cellsCOS-7PlasmidSubunit vaccineE2In vivo/solubleNot shown(57,58)
 CHOPlasmidSubunit vaccineE2In vivo/solubleNot shown(57)
 Hun7AdenovirusHomozygous VLPsCore-E1-E2In vivo/soluble4.75-8 mg/l(59,60)
 293TPlasmidChimeric VLPsHBV S-E1In vitro/soluble5.51 mg/l(13,61)
    HBV S-E2   
Insect cellsSpodoptera frugiperda Sf9AdenovirusVLPsCore-E1-E2In vivo/solubleNot shown(11)
 Drosophila S2 cellPlasmidSubunit vaccineE2In vitro/soluble100 mg/l(34,68)
    E1-E2 Not shown(69)
Yeast cellsPichia pastorisPlasmidVLPsCore-E1-E2In vivo/insolubleNot shown(70)
    CoreIn vitro/soluble1.61 mg/ml(71)
   Subunit vaccineE1-E2In vivo/insoluble35 mg/l(72)
    coreIn vivo/solubleNot shown(73)
    core-E1In vivo/solubleNot shown(73)
    core-E1-E2In vivo/solubleNot shown(73)
    E2 (384-650aa)In vitro/solubleNot shown(73)
    E2 (384-746aa)In vivo/insolubleNot shown(73)
    E2 (384-680aa)In vitro/solubleNot shown(62)
Bacterial cellsEscherichia coliPlasmidSubunit vaccineE2In vivo/inclusion body41.6-50%/ total somatic protein(58,74)
     PapMVCP27-215-E2In vivo/solubleNot shown(14)
   Homozygous VLPscore VLPsIn vivo/inclusion body10 mg/l(67)
   Chimeric VLPsPapMVCP-E2In vivo/solubleNot shown(14)
   Peptide vaccineNS3In vivo/inclusion bodyNot shown(40)
    p7In vivo/inclusion bodyNot shown(41)
Plant cellsNicotiana benthamiana plantsTobacco mosaic virusSubunit vaccineCholera toxin B subunit (CTB)- E2 HVR1In vivo/soluble6-80 µg/g of leaf tissue(65)
Parasite cellsLeishmania tarentolaePlasmidVLPsCore-E1-E2In vivo/insoluble4-6 mg/l(66)

[i] HBV S-E1, hepatitis B virus small envelope protein fused to hepatitis C virus envelope protein 1; PapMVCP-E2, papaya mosaic virus coat protein fused to hepatitis C virus envelope protein 2; NS3, non-structural proteins 3; p7, hepatitis C virus non-structural protein p7; E1, hepatitis C virus envelope protein 1; E2, hepatitis C virus envelope protein 2; HVR1, hypervariable 1; VLP, virus-like particle.

Eukaryotic expression system

A number of in-depth studies have focused on the preparation of HCV vaccines using eukaryotic expression system, as the recombinant vaccine protein is closest to the natural state of the vaccine and has post-translational protein modifications such as glycosylation. Mammalian cell-derived recombinant envelope proteins have been reported to have higher immunogenicity and human cell binding ability than those produced in yeast or insect cells (55,56). Different vaccine candidates have been successfully produced in mammalian cells, including Chinese hamster ovary (CHO) (57), COS-7 (57,58), Huh-7 (59,60) and human embryonic kidney 293T (13,61) cells. A comparison of envelope protein expression in COS-7 and CHO cells showed two different expression patterns: The former prepared full-length protein with transient expression form, while the latter produced truncated target protein in a stable manner (58). This suggests that transient expression is more suitable for maintaining protein integrity than transient expression. The Pichia pastoris expression system has the advantages of low cost and simple operation for recombinant protein preparation. The structural proteins of the core, E1 and E2, have been prepared as subunits or VLP vaccines in Pichia pastoris under methanol induction, and the target proteins have antigenicity. However, size-exclusion chromatography and SDS-PAGE experiments have suggested that E2 is mainly produced in a dimer or polymer form (62). The fragment from 612 to 620 aa has been reported to be a dimerization sequence (63). The tendency to aggregate is probably an intrinsic property of HCV glycoproteins, which leads to low protein synthesis when using a non-viral vector (64). In fact, the maximum yield of HCV glycoproteins prepared using yeast cells was 35 mg/l. The yield of vaccine protein prepared by different hosts ranged from 1-10 mg, and the highest yield was 100 mg/l from Drosophila S2 cells with an expression cycle of up to 9 days. In recent years, some new expression systems have been attempted for simpler genetic manipulation, higher production levels and lower-cost production. The HVR1/cholera toxin B subunit chimeric protein was expressed in Nicotiana benthamiana plants with a production of 6-80 µg/g of leaf tissue (65). Core-E1-E2 VLPs were successfully generated by the Leishmania expression system (66). These vaccines will not be suitable for use in clinics until their safety and efficacy is confirmed.

Prokaryotic expression system

Owing to the lack of protein modification by prokaryotic cells, the immunogenicity of the vaccine in a prokaryotic expression system is relatively lower than that developed using eukaryotic cells. Most HCV vaccine proteins prepared using the E. coli expression system are in the form of inclusion bodies, and a few truncated envelope proteins can be released into the periplasmic space of host cells using signal peptides. The expression of the target protein accounts for 40-50% of the total bacterial protein (58). It has been found that the expressed core protein can also be assembled into particles in vitro (with a diameter of 60 nm) (67). In addition, due to the influence of bacterial and toxin proteins, the purification cost is relatively high.

4. Difficulties in vaccine preparation

How to prepare HCV vaccine protein in a soluble secretory form

The vaccine protein, especially the envelope glycoprotein, is mainly expressed intracellularly and is insoluble through the addition of the signal peptide sequence during recombinant expression (71). It is generally believed that the strong hydrophobicity of the C-terminus of the E2 protein is the main reason for this (75,76). Therefore, several strategies have been developed to enhance protein secretion: i) Fusion preparation with proteins having strong secretory ability: The wild-type HBV S subviral particles used in current HBV vaccines can be efficiently secreted into the cell supernatant and are easily purified. Replacing the N-terminal TMD of the HBV S protein with the TMD of HCV E1 or E2, the chimeric HBV-HCV envelope proteins (E1-S or/and E2-S) can be effectively secreted, co-expressed and assembled into VLPs [S+E1-S, S+E2-S and S+(E1-S+E2-S)] with the wild-type HBV S protein (13). Unlike HCV VLPs, the chimeric HBV-HCV VLPs could only induce a humoral immune response but not a T-cell immune response. ii) Optimization of the structure of the target protein: Following removal of the partial domain of the C-terminal to prepare a truncated envelope protein, the truncated E2 protein (384-521 or 605-680 aa) could be effectively secreted by Pichia pastoris cells, while intact E2 (384-746 aa) was mainly located in the insoluble part of ruptured cells (73). This is in concordance with another study that found that C-terminal truncated enveloped proteins could be efficiently secreted to the culture medium by mammalian cells (77). Further research demonstrated that the C-terminus of E2 that began with aa 718 contained an endoplasmic reticulum retention signal (75,76), and topological analysis also showed that aa 718 was the beginning of the TMD (Fig. 3A). iii) Optimization of the signal peptide structure: When the α-factor signal peptide of the Pichia pastoris expression system was changed to the leader sequence of sucrose invertase 2, extracellular expression of E2 could be realized (62,72). However, the underlying mechanism remains unknown. In addition, using the 374-383 aa of envelope protein E1 as a signal sequence, the E2 protein could be expressed and secreted by mammalian cells (78).

How to widen the immune spectrum of a vaccine

Due to a number of HCV genotypes, the vaccine that is usually effective for one genotype is ineffective for other genotypes. Therefore, broadening the spectrum of vaccines has become a research challenge. Based on the published literature, there are the following strategies to broaden the spectrum of vaccines: i) Selection of antigen epitopes with strong conservation. The appearance of broadly neutralizing antibodies (bNAbs) is important for the body to remove HCV infection (79). Analyzing the distribution characteristics of antigen epitopes in the tertiary structure of HCV E2 protein, it was found that the antigen epitopes near the receptor-binding amino acids were conserved and could easily produce extensive neutralizing antibodies. HMAbs represented by AP33, 3/11, 95-2 and HCV-1 bind to the 412-423 aa linear epitope (23), which can show extensive neutralizing activity against HCVpp of genotype 1-6. CBH2, HC11, HC1 and other monoclonal antibodies bind to the conformational epitope composed of 424-443 aa and 524-540 aa sequences, which can widely neutralize the HCV 1-6 genotype (24). The molecular mechanism of the exact cell entry process for HCV remains undefined, however, and only limited antigen epitopes have been identified to this day. ii) The antigen sequences of multiple genotypes are combined to prepare a multi-antigen vaccine cocktail: A multi-antigen cocktail regimen created by comparing a DNA vaccine cocktail encoding genotype (Gt)1b and Gt3a NS3, NS4 and NS5B proteins elicited significantly higher T-cell responses to Gt1b and Gt3a NS5B proteins than single-genotype NS3/4/5B DNA vaccine (51). Obviously, the multi-antigen vaccine cocktail method is not suitable for VLPs and subunit vaccines, for which the correct conformation of the protein is necessary. iii) Rational designing of a conservative structure suitable for multiple genotypes: Structure-based vaccine designs have been successfully used in influenza virus, human immunodeficiency virus and other variable viruses for the purpose of optimizing the presentation of key conserved epitopes, masking sites using N-glycans or stabilizing the conformations of the envelope glycoproteins (80,81). Similarly, the E2 antigen, in which a mutation H449P was designed to stabilize the conformation of a conservative immunogen domain D in the internal fluidity neutralization surface, successfully induced bNAbs with cross-neutralizing activities against HCVpp of the 1b, 2a, and 4a genotypes (27). Analyzing the sequence or structure of cross-neutralizing antibodies, the corresponding epitope structure simulant was designed using anti-idiotypic technology. The immunogenic effectiveness of AP33 linear epitope mimics has been confirmed in mouse models (54) and the design of structural epitope mimics is the development direction of this strategy.

5. Conclusions

An effective vaccine for HCV is, essentially, an antigen that elicits immune responses to key conserved epitopes. In the present review, the composition, immunogenicity, advantages and disadvantages of all different types of vaccines were summarized and compared, and it was found that the source of antigen determines the immunogenicity of vaccine. Structural proteins are involved in the invasion of the virus into host cells and are used in preference to stimulate the humoral immune response of the host. Non-structural proteins are mainly involved in virus replication and cause specific T-cell immune responses. HCV can be divided into a number of genotypes due to its genetic variability. The conserved key epitopes form flexible conformation. Vaccines using the wild-type and full-length antigen cannot stimulate an ideal immune effect. Future research will still focus on improving the immunogenicity and broadening the spectrum of vaccines. Analysis and optimization of the epitope structure corresponding to some extremely effective neutralizing antibodies has significant guidance value for the correct conformational expression of vaccines. Therefore, the rational design, with structure as the purpose and function as the starting point, is the most effective way to obtain broad-spectrum vaccines. In addition, the vaccine cocktail model can stimulate stronger and broader spectrum T-cell responses and has higher immunogenicity and a broader spectrum than vaccines from a single source.

Compared with other mature preparation technologies of viral vaccines, improving the preparation level of an HCV vaccine is also an important research direction in the future. By summarizing the preparation methods and levels using different expression hosts, it is indicated that deleting adverse protein domains, sampling the sugar chains, or constructing multivalent vaccines with proteins from other viruses, can effectively neutralize the insoluble characteristics of HCV self-proteins. Moreover, with the in-depth study of rational design, a new soluble protein structure may be obtained.

In conclusion, all these successful advances indicate that the high-level preparation of new vaccines with high immunogenicity and a broad spectrum is possible. Rational design will become the main focus in the future. At the same time, the present review indicates an urgent need for in-depth research on the structure and function of HCV proteins. A new vaccine will be expected to completely eliminate new cases of HCV infection.

Acknowledgements

Not applicable.

Funding

Funding: This present study was supported by the Shandong Key Research and Development Fund (grant no. 2018GSF121011), the Jinan Clinical Medical Science and Technology Innovation Fund (grant no. 201805066) and the High-Level Project Cultivation Fund of Jinan Central Hospital (grant no. 202105006).

Availability of data and materials

Not applicable.

Authors' contributions

QZ conceived the idea of this study and was a major contributor in writing the manuscript. KH, XHZ and MX assisted in writing the manuscript. XPZ and HL reviewed the literature. All the authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Duncan JD, Urbanowicz RA, Tarr AW and Ball JK: Hepatitis C Virus vaccine: Challenges and prospects. Vaccines (Basel). 8(90)2020.PubMed/NCBI View Article : Google Scholar

2 

Ramsay J, Marsh J, Pedrana A, Andric N, Norman R, Cheng W, Webb S, Zeps N, Bellgard M, Graves T, et al: A platform in the use of medicines to treat chronic hepatitis C (PLATINUM C): Protocol for a prospective treatment registry of real-world outcomes for hepatitis C. BMC Infect Dis. 20(802)2020.PubMed/NCBI View Article : Google Scholar

3 

Falade-Nwulia O, Suarez-Cuervo C, Nelson DR, Fried MW, Segal JB and Sulkowski MS: Oral direct-acting agent therapy for hepatitis C virus infection: A systematic review. Ann Intern Med. 166:637–648. 2017.PubMed/NCBI View Article : Google Scholar

4 

Li D, Huang Z and Zhong J: Hepatitis C virus vaccine development: Old challenges and new opportunities. Natl Sci Rev. 2:285–295. 2015.

5 

Forns X, Bukh J and Purcell RH: The challenge of developing a vaccine against hepatitis C virus. J Hepatol. 37:684–695. 2002.PubMed/NCBI View Article : Google Scholar

6 

Buonaguro L, Tagliamonte M, Tornesello ML and Buonaguro FM: Developments in virus-like particle-based vaccines for infectious diseases and cancer. Expert Rev Vaccines. 10:1569–1583. 2011.PubMed/NCBI View Article : Google Scholar

7 

Murata K, Lechmann M, Qiao M, Gunji T, Alter HJ and Liang TJ: Immunization with hepatitis C virus-like particles protects mice from recombinant hepatitis C virus vaccinia infection. Proc Natl Acad Sci USA. 100:6753–6758. 2003.PubMed/NCBI View Article : Google Scholar

8 

Christiansen D, Earnest-Silveira L, Grubor-Bauk B, Wijesundara DK, Boo I, Ramsland PA, Vincan E, Drummer HE, Gowans EJ and Torresi J: Pre-clinical evaluation of a quadrivalent HCV VLP vaccine in pigs following microneedle delivery. Sci Rep. 9(9251)2019.PubMed/NCBI View Article : Google Scholar

9 

Gastaminza P, Dryden KA, Boyd B, Wood MR, Law M, Yeager M and Chisari FV: Ultrastructural and biophysical characterization of hepatitis C virus particles produced in cell culture. J Virol. 84:10999–11009. 2010.PubMed/NCBI View Article : Google Scholar

10 

Matsuura Y, Harada S, Suzuki R, Watanabe Y, Inoue Y, Saito I and Miyamura T: Expression of processed envelope protein of hepatitis C virus in mammalian and insect cells. J Virol. 66:1425–1431. 1992.PubMed/NCBI View Article : Google Scholar

11 

Baumert TF, Ito S, Wong DT and Liang TJ: Hepatitis C virus structural proteins assemble into virus like particles in insect cells. J Virol. 72:3827–3836. 1998.PubMed/NCBI View Article : Google Scholar

12 

Mihailova M, Boos M, Petrovskis I, Ose V, Skrastina D, Fiedler M, Sominskaya I, Ross S, Pumpens P, Roggendorf M and Viazov S: Recombinant virus-like particles as a carrier of B- and T-cell epitopes of hepatitis C virus (HCV). Vaccine. 24:4369–4377. 2006.PubMed/NCBI View Article : Google Scholar

13 

Beaumont E, Patient R, Hourioux C, Dimier-Poisson I and Roingeard P: Chimeric hepatitis B virus/hepatitis C virus envelope proteins elicit broadly neutralizing antibodies and constitute a potential bivalent prophylactic vaccine. Hepatology. 57:1303–1313. 2013.PubMed/NCBI View Article : Google Scholar

14 

Denis J, Majeau N, Acosta-Ramirez E, Savard C, Bedard MC, Simard S, Lecours K, Bolduc M, Pare C, Willems B, et al: Immunogenicity of papaya mosaic virus-like particles fused to a hepatitis C virus epitope: Evidence for the critical function of multimerization. Virology. 363:59–68. 2007.PubMed/NCBI View Article : Google Scholar

15 

Chua BY, Johnson D, Tan A, Earnest-Silveira L, Sekiya T, Chin R, Torresi J and Jackson DC: Hepatitis C VLPs delivered to dendritic cells by a TLR2 targeting lipopeptide results in enhanced antibody and cell-mediated responses. PLoS One. 7(e47492)2012.PubMed/NCBI View Article : Google Scholar

16 

Qiao M, Murata K, Davis AR, Jeong SH and Liang TJ: Hepatitis C virus-like particles combined with novel adjuvant systems enhance virus-specific immune responses. Hepatology. 37:52–59. 2003.PubMed/NCBI View Article : Google Scholar

17 

Acosta-Rivero N, Poutou J, Alvarez-Lajonchere L, Guerra I, Aguilera Y, Musacchio A, Rodríguez A, Aguilar JC, Falcon V, Alvarez-Obregon JC, et al: Recombinant in vitro assembled hepatitis C virus core particles induce strong specific immunity enhanced by formulation with an oil-based adjuvant. Biol Res. 42:41–56. 2009.PubMed/NCBI

18 

Logan M, Law J, Wong JAJ, Hockman D, Landi A, Chen C, Crawford K, Kundu J, Baldwin L, Johnson J, et al: Native folding of a recombinant gpE1/gpE2 heterodimer vaccine antigen from a precursor protein fused with Fc IgG. J Virol. 91:e01552–16. 2016.PubMed/NCBI View Article : Google Scholar

19 

Krapchev VB, Rychlowska M, Chmielewska A, Zimmer K, Patel AH and Bienkowska-Szewczyk K: Recombinant flag-tagged E1E2 glycoproteins from three hepatitis C virus genotypes are biologically functional and elicit cross-reactive neutralizing antibodies in mice. Virology. 519:33–41. 2018.PubMed/NCBI View Article : Google Scholar

20 

Owsianka AM, Timms JM, Tarr AW, Brown RJ, Hickling TP, Szwejk A, Bienkowska-Szewczyk K, Thomson BJ, Patel AH and Ball JK: Identification of conserved residues in the E2 envelope glycoprotein of the hepatitis C virus that are critical for CD81 binding. J Virol. 80:8695–8704. 2006.PubMed/NCBI View Article : Google Scholar

21 

Tzarum N, Wilson IA and Law M: The neutralizing face of hepatitis C Virus E2 envelope glycoprotein. Front Immunol. 9(1315)2018.PubMed/NCBI View Article : Google Scholar

22 

Kumar A, Hossain RA, Yost SA, Bu W, Wang Y, Dearborn AD, Grakoui A, Cohen JI and Marcotrigiano J: Structural insights into hepatitis C virus receptor binding and entry. Nature. 598:521–525. 2021.PubMed/NCBI View Article : Google Scholar

23 

Desombere I, Fafi-Kremer S, Van Houtte F, Pessaux P, Farhoudi A, Heydmann L, Verhoye L, Cole S, Mckeating JA, Leroux-Roels G, et al: Monoclonal anti-envelope antibody AP33 protects humanized mice against a patient-derived hepatitis C virus challenge. Hepatology. 63:1120–1134. 2016.PubMed/NCBI View Article : Google Scholar

24 

Wang Y, Keck ZY and Foung SK: Neutralizing antibody response to hepatitis C virus. Viruses. 3:2127–2145. 2011.PubMed/NCBI View Article : Google Scholar

25 

Keck ZY, Saha A, Xia J, Wang Y, Lau P, Krey T, Rey FA and Foung SK: Mapping a region of HCV E2 that is responsible for escape from neutralizing antibodies and a core CD81-binding region that does not tolerate neutralization escape mutations. J Virol. 85:10451–10463. 2011.PubMed/NCBI View Article : Google Scholar

26 

Guest JD and Pierce BG: Structure-based and rational design of a hepatitis C Virus vaccine. Viruses. 13(837)2021.PubMed/NCBI View Article : Google Scholar

27 

Pierce BG, Keck ZY, Wang R, Lau P, Garagusi K, Elkholy K, Toth EA, Urbanowicz RA, Guest JD, Agnihotri P, et al: Structure-based design of hepatitis C Virus E2 glycoprotein improves serum binding and cross-neutralization. J Virol. 94:e00704–20. 2020.PubMed/NCBI View Article : Google Scholar

28 

Kachko A, Frey SE, Sirota L, Ray R, Wells F, Zubkova I, Zhang P and Major ME: Antibodies to an interfering epitope in hepatitis C virus E2 can mask vaccine-induced neutralizing activity. Hepatology. 62:1670–1682. 2015.PubMed/NCBI View Article : Google Scholar

29 

Zhang P, Zhong L, Struble EB, Watanabe H, Kachko A, Mihalik K, Virata ML, Alter HJ, Feinstone S and Major M: Depletion of interfering antibodies in chronic hepatitis C patients and vaccinated chimpanzees reveals broad cross-genotype neutralizing activity. Proc Natl Acad Sci USA. 106:7537–7541. 2009.PubMed/NCBI View Article : Google Scholar

30 

Ruwona TB, Giang E, Nieusma T and Law M: Fine mapping of murine antibody responses to immunization with a novel soluble form of hepatitis C virus envelope glycoprotein complex. J Virol. 88:10459–10471. 2014.PubMed/NCBI View Article : Google Scholar

31 

Vietheer PT, Boo I, Gu J, McCaffrey K, Edwards S, Owczarek C, Hardy MP, Fabri L, Center RJ, Poumbourios P and Drummer HE: The core domain of hepatitis C virus glycoprotein E2 generates potent cross-neutralizing antibodies in guinea pigs. Hepatology. 65:1117–1131. 2017.PubMed/NCBI View Article : Google Scholar

32 

McCaffrey K, Boo I, Owczarek CM, Hardy MP, Perugini MA, Fabri L, Scotney P, Poumbourios P and Drummer HE: An optimized hepatitis C virus E2 glycoprotein core adopts a functional homodimer that effciently blocks virus entry. J Virol. 91:e01668–16. 2017.PubMed/NCBI View Article : Google Scholar

33 

McCaffrey K, Boo I, Poumbourios P and Drummer HE: Expression and characterization of a minimal hepatitis C virus glycoprotein E2 core domain that retains CD81 binding. J Virol. 81:9584–9590. 2007.PubMed/NCBI View Article : Google Scholar

34 

Li D, von Schaewen M, Wang X, Tao W, Zhang Y, Li L, Heller B, Hrebikova G, Deng Q, Ploss A, et al: Altered glycosylation patterns increase immunogenicity of a subunit hepatitis C virus vaccine, inducing neutralizing antibodies which confer protection in mice. J Virol. 90:10486–10498. 2016.PubMed/NCBI View Article : Google Scholar

35 

Urbanowicz RA, Wang R, Schiel JE, Keck ZY, Kerzic MC, Lau P, Rangarajan S, Garagusi KJ, Tan L, Guest JD, et al: Antigenicity and immunogenicity of differentially glycosylated hepatitis C virus E2 envelope proteins expressed in mammalian and insect Cells. J Virol. 93:e01403–18. 2019.PubMed/NCBI View Article : Google Scholar

36 

Naarding MA, Falkowska E, Xiao H and Dragic T: Hepatitis C virus soluble E2 in combination with QuilA and CpG ODN induces neutralizing antibodies in mice. Vaccine. 29:2910–2917. 2011.PubMed/NCBI View Article : Google Scholar

37 

Frey SE, Houghton M, Coates S, Abrignani S, Chien D, Rosa D, Pileri P, Ray R, Di Bisceglie AM, Rinella P, et al: Safety and immunogenicity of HCV E1E2 vaccine adjuvanted with MF59 administered to healthy adults. Vaccine. 28:6367–6373. 2010.PubMed/NCBI View Article : Google Scholar

38 

Wedemeyer H, Schuller E, Schlaphoff V, Stauber RE, Wiegand J, Schiefke I, Firbas C, Jilma B, Thursz M, Zeuzem S, et al: Therapeutic vaccine IC41 as late add-on to standard treatment in patients with chronic hepatitis C. Vaccine. 27:5142–5151. 2009.PubMed/NCBI View Article : Google Scholar

39 

Sarobe P, Pendieton CD, Akatsuka T, Lau D, Engelhard VH, Feinstone SM and Berzofsky JA: Enhanced in vitro potency and in vivo immunogenicity of a CTL epitope from hepatitis C virus core protein following amino acid replacement at secondary HLA-A2.1 binding positions. J Clin Invest. 102:1239–1248. 1998.PubMed/NCBI View Article : Google Scholar

40 

Filskov J, Mikkelsen M, Hansen PR, Christensen JP, Thomsen AR, Andersen P, Bukh J and Agger EM: Broadening CD4+ and CD8+ T cell responses against hepatitis C virus by vaccination with NS3 overlapping peptide panels in cross-priming liposomes. J Virol. 91:e00130–17. 2017.PubMed/NCBI View Article : Google Scholar

41 

Filskov J, Andersen P, Agger EM and Bukh J: HCV p7 as a novel vaccine-target inducing multifunctional CD4+ and CD8+ T-cells targeting liver cells expressing the viral antigen. Sci Rep. 9(14085)2019.PubMed/NCBI View Article : Google Scholar

42 

Firbas C, Boehm T, Buerger V, Schuller E, Sabarth N, Jilma B and Klade CS: Immunogenicity and safety of different injection routes and schedules of IC41, a Hepatitis C virus (HCV) peptide vaccine. Vaccine. 28:2397–2407. 2010.PubMed/NCBI View Article : Google Scholar

43 

Firbas C, Jilma B, Tauber E, Buerger V, Jelovcan S, Lingnau K, Buschle M, Frisch J and Klade CS: Immunogenicity and safety of a novel therapeutic hepatitis C virus (HCV) peptide vaccine: A randomized, placebo controlled trial for dose optimization in 128 healthy subjects. Vaccine. 24:4343–4353. 2006.PubMed/NCBI View Article : Google Scholar

44 

Klade CS, Wedemeyer H, Berg T, Hinrichsen H, Cholewinska G, Zeuzem S, Blum H, Buschle M, Jelovcan S, Buerger V, et al: Therapeutic vaccination of chronic hepatitis C nonresponder patients with the peptide vaccine IC41. Gastroenterology. 134:1385–1395. 2008.PubMed/NCBI View Article : Google Scholar

45 

Yin D, Qi ZT, Yan S, Ding H, Hua X, Tong YM, Wang YZ, Liu Y, Xu FH and Zhao P: Study on the induction of mouse neutralization antibodies to HCVpp by HCV E2 DNA vaccines. Chin J Exp Clin Infect Dis (Electronic Version). 2:24–32. 2008.(In Chinese).

46 

National Institute of Allergy and Infectious Diseases (NIH): Trial Evaluating Experimental Hepatitis C Vaccine Concludes. NIH, Bethesda, MD, 2019. https://www.niaid.nih.gov/news-events/trial-evaluating-experimental-hepatitis-c-vaccine-concludes. Accessed July 8, 2021.

47 

Shin EC, Sung PS and Park SH: Immune responses and immunopathology in acute and chronic viral hepatitis. Nat Rev Immunol. 16:509–523. 2016.PubMed/NCBI View Article : Google Scholar

48 

Lu L, Barbi J and Pan F: The regulation of immune tolerance by FOXP3. Nat Rev Immunol. 17:703–717. 2017.PubMed/NCBI View Article : Google Scholar

49 

Langhans B, Nischalke HD, Krämer B, Hausen A, Dold L, van Heteren P, Hüneburg R, Nattermann J, Strassburg CP and Spengler U: Increased peripheral CD4+ regulatory T cells persist after successful direct-acting antiviral treatment of chronic hepatitis C. J Hepatol. 66:888–896. 2017.PubMed/NCBI View Article : Google Scholar : (In Albanian, English).

50 

Han JW, Sung PS, Hong SH, Lee H, Koh JY, Lee H, White S, Maslow JN, Weiner DB, Park SH, et al: IFNL3-adjuvanted HCV DNA vaccine reduces regulatory T-cell frequency and increases virus-specific T-cell responses. J Hepatol. 73:72–83. 2020.PubMed/NCBI View Article : Google Scholar

51 

Wijesundara DK, Gummow J, Li Y, Yu W, Quah BJ, Ranasinghe C, Torresi J, Gowans EJ and Grubor-Bauk B: Induction of genotype cross-reactive, hepatitis C virus-specific, cell-mediated immunity in DNA-vaccinated mice. J Virol. 92:e02133–17. 2018.PubMed/NCBI View Article : Google Scholar

52 

Castellanos M, Cinza Z, Dorta Z, Veliz G, Vega H, Lorenzo I, Ojeda S, Dueñas-Carrera S, Alvarez-Lajonchere L, Martínez G, et al: Immunization with a DNA vaccine candidate in chronic hepatitis C patients is safe, well tolerated and does not impair immune response induction after anti-hepatitis B vaccination. J Gene Med. 12:107–116. 2010.PubMed/NCBI View Article : Google Scholar

53 

Pierce BG, Boucher EN, Piepenbrink KH, Ejemel M, Rapp CA, Thomas WD Jr, Sundberg EJ, Weng Z and Wang Y: Structure-based design of hepatitis C virus vaccines that elicit neutralizing antibody responses to a conserved epitope. J Virol. 91:e01032–17. 2017.PubMed/NCBI View Article : Google Scholar

54 

Cowton VM, Owsianka AM, Fadda V, Ortega-Prieto AM, Cole SJ, Potter JA, Skelton JK, Jeffrey N, Di Lorenzo C, Dorner M, et al: Development of a structural epitope mimic: An idiotypic approach to HCV vaccine design. NPJ Vaccines. 6(7)2021.PubMed/NCBI View Article : Google Scholar

55 

Choo QL, Kuo G, Ralston R, Weiner A, Chien D, Van Nest G, Han K, Berger K, Thudium K, Kuo C, et al: Vaccination of chimpanzees against infection by the hepatitis C virus. Proc Natl Acad Sci USA. 91:1294–1298. 1994.PubMed/NCBI View Article : Google Scholar

56 

Rosa D, Campagnoli S, Moretto C, Guenzi E, Cousens L, Chin M, Dong C, Weiner AJ, Lau JY, Choo QL, et al: A quantitative test to estimate neutralizing antibodies to the hepatitis C virus: Cytofluorimetric assessment of envelope glycoprotein 2 binding to target cells. Proc Natl Acad Sci USA. 93:1759–1763. 1996.PubMed/NCBI View Article : Google Scholar

57 

Shi X, Cao F, Du Y, Hou L, Ji Y and Wang H: Protein expression and antigenicity detection of Hepatitis C virus envelope protein E2. Medical Journal of Chinese People's Liberation Army. 27:17–19. 2002.

58 

Dai Z, Li G, Xu Q, Yao L, Han X, Yang L and Chen W: Expression and Identification of Hepatitis C Virus Envelop Protein E2. J Sun Yat-Sen Univ (Med Sci). 27:83–89. 2006.(In Chinese).

59 

Earnest-Silveira L, Chua B, Chin R, Christiansen D, Johnson D, Herrmann S, Ralph SA, Vercauteren K, Mesalam A, Meuleman P, et al: Characterization of a hepatitis C virus-like particle vaccine produced in a human hepatocyte-derived cell line. J Gen Virol. 97:1865–1876. 2016.PubMed/NCBI View Article : Google Scholar

60 

Earnest-Silveira L, Christiansen D, Herrmann S, Ralph SA, Das V, Gowans EJ and Torres J: Large scale production of a mammalian cell derived quadrivalent hepatitis C virus like particle vaccine. J Virol Methods. 236:87–92. 2016.PubMed/NCBI View Article : Google Scholar

61 

Shu F, Lei YF, Lin F, Wang X, Li B, Zhang LJ, Dong K, Zhang HZ and Wei SH: Construction of eukaryotic expression vector for chimeric gene of hepatitis C virus neutralizing epitopes with hepatitis B virus S antigen and its expression in 293T cells. Chinese Journal of Biologicals. 26:795–799. 2013.

62 

Martínez-Donato G, Capdesuñer Y, Acosta-Rivero N, Rodríguez A, Morales-Grillo J, Martínez E, González M, Alvarez-Obregon JC and Dueñas-Carrera S: Multimeric HCV E2 protein obtained from Pichia pastoris cells induces a strong immune response in mice. Mol Biotechnol. 35:225–235. 2007.PubMed/NCBI View Article : Google Scholar

63 

Yagnik AT, Lahm A, Meola A, Roccasecca RM, Ercole BB, Nicosia A and Tramontano A: A model for the hepatitis C virus envelope glycoprotein E2. Proteins. 40:355–366. 2000.PubMed/NCBI View Article : Google Scholar

64 

Duvet S, Cocquerel L, Pillez A, Cacan R, Verbert A, Moradpour D, Eychoeski C and Dubuisson J: Hepatitis C virus glycoprotein complex localization in the endoplasmic reticulum involves a determinant for retention and not retrieval. J Biol Chem. 273:32088–32095. 1998.PubMed/NCBI View Article : Google Scholar

65 

Nemchinov LG, Liang TJ, Rifaat MM, Mazyad HM, Hadidi A and Keith JM: Development of a plant-derived subunit vaccine candidate against hepatitis C virus. Arch Virol. 145:2557–2573. 2000.PubMed/NCBI View Article : Google Scholar

66 

Bolhassani A, Davoudi N, Motevalli F and Agi E: Comparison of HCV Core and CoreE1E2 virus-like particles generated by stably transfected leishmania tarentolae for the Stimulation of Th1 immune responses in mice. Curr Drug Deliv. 14:1040–1049. 2017.PubMed/NCBI View Article : Google Scholar

67 

Kunkel M, Lorinczi M, Rijnbrand R, Lemon SM and Watowich SJ: Self-assembly of nucleocapsid-like particles from recombinant hepatitis C virus core protein. J Virol. 75:2119–2129. 2001.PubMed/NCBI View Article : Google Scholar

68 

Tarr AW, Backx M, Hamed MR, Urbanowicz RA, McClure CP, Brown RJP and Ball JK: Immunization with a synthetic consensus hepatitis C virus E2 glycoprotein ectodomain elicits virus-neutralizing antibodies. Antivir Res. 160:25–37. 2018.PubMed/NCBI View Article : Google Scholar

69 

Cao L, Yu B, Kong D, Cong Q, Yu T, Chen Z, Hu Z, Chang H, Zhong J, Baker D and He Y: Functional expression and characterization of the envelope glycoprotein E1E2 heterodimer of hepatitis C virus. PLoS Pathog. 15(e1007759)2019.PubMed/NCBI View Article : Google Scholar

70 

Fazlalipour M, Keyvani H, Monavari SH and Mollaie HR: Expression, purification and immunogenic description of a hepatitis C virus recombinant CoreE1E2 protein expressed by yeast Pichia pastoris. Jundishapur J Microbiol. 8(e17157)2015.PubMed/NCBI View Article : Google Scholar

71 

Pechelyulko A, Andreeva-Kovalevskaya Z, Dmitriev D, Lavrov V, Massino Y, Nagel A, Segal O, Sokolova OS, Solonin A, Tarakanova Y and Dmitriev A: A simple method to purify recombinant HCV core protein expressed in Pichia pastoris for obtaining virus-like particles and producing monoclonal antibodies. Protein Expr Purif. 183(105864)2021.PubMed/NCBI View Article : Google Scholar

72 

Cai W, Su L, Liao Q, Ye L, Wu Y, Wu Z and She Y: Expression, purification and immunogenic characterization of hepatitis C virus recombinant E1E2 protein expressed by Pichia pastoris yeast. Antivir Res. 88:80–85. 2010.PubMed/NCBI View Article : Google Scholar

73 

Martinez-Donato G, Acosta-Rivero N, Morales-Grillo J, Musacchio A, Vina A, Alvarez C, Figueroa N, Guerra I, Garcia J, Varas L, et al: Expression and processing of hepatitis C virus structural proteins in Pichia pastoris yeast. Biochem Biophys Res Commun. 342:625–631. 2006.PubMed/NCBI View Article : Google Scholar

74 

Yao M, Fang H, Yin W, Lei Y, Yang J, Sun M, Tian J and Lv X: Prokaryotic expression and purification of truncated Hepatitis C Virus envelope glycoprotein E2. Science Technology and Engineering,. 9:2296–2303. 2009.

75 

Cocquerel L, Meunier JC, Pillez A, Wychowski C and Dubuisson J: A retention signal necessary and sufficient for endoplasmic reticulum localization maps to the transmembrane domain of hepatitis C virus glycoprotein E2. J Virol. 72:2183–2191. 1998.PubMed/NCBI View Article : Google Scholar

76 

Cocquerel L, Wychowski C, Minner F, Penin F and Dubuisson J: Charged residues in the transmembrane domains of hepatitis C virus glycoproteins play a major role in the processing, subcellular localization, and assembly of these envelope proteins. J Virol. 74:3623–3633. 2000.PubMed/NCBI View Article : Google Scholar

77 

Matsuura Y, Suzuki T, Suzuki R, Sato M, Aizaki H, Saito I and Miyamura T: Processing of E1 and E2 glycoproteins of hepatitis C virus expressed in mammalian and insect cells. Virology. 205:141–150. 1994.PubMed/NCBI View Article : Google Scholar

78 

Mizushima H, Hijikata M, Asabe S, Hirota M, Kimura K and Shimotohno K: Two hepatitis C virus glycoprotein E2 products with different C termini. J Virol. 68:6215–6222. 1994.PubMed/NCBI View Article : Google Scholar

79 

Morin TJ, Broering TJ, Leav BA, Blair BM, Rowley KJ, Boucher EN, Wang Y, Cheslock PS, Knauber M, Olsen DB, et al: Human monoclonal antibody HCV1 effectively prevents and treats HCV infection in chimpanzees. PLoS Pathog. 8(e1002895)2012.PubMed/NCBI View Article : Google Scholar

80 

Impagliazzo A, Milder F, Kuipers H, Wagner MV, Zhu X, Hoffman RM, van Meersbergen R, Huizingh J, Wanningen P, Verspuij J, et al: A stable trimeric influenza hemagglutinin stem as a broadly protective immunogen. Science. 349:1301–1306. 2015.PubMed/NCBI View Article : Google Scholar

81 

Kulp DW, Steichen JM, Pauthner M, Hu X, Schiffner T, Liguori A, Cottrell CA, Havenar-Daughton C, Ozorowski G, Georgeson E, et al: Structure-based design of native-like HIV-1 envelope trimers to silence non-neutralizing epitopes and eliminate CD4 binding. Nat Commun. 8(1655)2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2022
Volume 24 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao Q, He K, Zhang X, Xu M, Zhang X and Li H: Production and immunogenicity of different prophylactic vaccines for hepatitis C virus (Review). Exp Ther Med 24: 474, 2022
APA
Zhao, Q., He, K., Zhang, X., Xu, M., Zhang, X., & Li, H. (2022). Production and immunogenicity of different prophylactic vaccines for hepatitis C virus (Review). Experimental and Therapeutic Medicine, 24, 474. https://doi.org/10.3892/etm.2022.11401
MLA
Zhao, Q., He, K., Zhang, X., Xu, M., Zhang, X., Li, H."Production and immunogenicity of different prophylactic vaccines for hepatitis C virus (Review)". Experimental and Therapeutic Medicine 24.1 (2022): 474.
Chicago
Zhao, Q., He, K., Zhang, X., Xu, M., Zhang, X., Li, H."Production and immunogenicity of different prophylactic vaccines for hepatitis C virus (Review)". Experimental and Therapeutic Medicine 24, no. 1 (2022): 474. https://doi.org/10.3892/etm.2022.11401