Next Article in Journal / Special Issue
Vaccination against δ-Retroviruses: The Bovine Leukemia Virus Paradigm
Previous Article in Journal
Engineered RNase P Ribozymes Effectively Inhibit Human Cytomegalovirus Gene Expression and Replication
Previous Article in Special Issue
Infection with Non-Lethal West Nile Virus Eg101 Strain Induces Immunity that Protects Mice against the Lethal West Nile Virus NY99 Strain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alphavirus-Based Vaccines

by
Kenneth Lundstrom
PanTherapeutics, Rue des Remparts 4, CH1095 Lutry, Switzerland
Viruses 2014, 6(6), 2392-2415; https://doi.org/10.3390/v6062392
Submission received: 2 April 2014 / Revised: 3 June 2014 / Accepted: 4 June 2014 / Published: 16 June 2014
(This article belongs to the Special Issue Virus-based Vaccines)

Abstract

:
Alphavirus vectors have demonstrated high levels of transient heterologous gene expression both in vitro and in vivo and, therefore, possess attractive features for vaccine development. The most commonly used delivery vectors are based on three single-stranded encapsulated alphaviruses, namely Semliki Forest virus, Sindbis virus and Venezuelan equine encephalitis virus. Alphavirus vectors have been applied as replication-deficient recombinant viral particles and, more recently, as replication-proficient particles. Moreover, in vitro transcribed RNA, as well as layered DNA vectors have been applied for immunization. A large number of highly immunogenic viral structural proteins expressed from alphavirus vectors have elicited strong neutralizing antibody responses in multispecies animal models. Furthermore, immunization studies have demonstrated robust protection against challenges with lethal doses of virus in rodents and primates. Similarly, vaccination with alphavirus vectors expressing tumor antigens resulted in prophylactic protection against challenges with tumor-inducing cancerous cells. As certain alphaviruses, such as Chikungunya virus, have been associated with epidemics in animals and humans, attention has also been paid to the development of vaccines against alphaviruses themselves. Recent progress in alphavirus vector development and vaccine technology has allowed conducting clinical trials in humans.

1. Introduction

Alphaviruses are single-stranded RNA viruses with an envelope structure belonging to the family of Togaviridae [1]. Certain alphaviruses have been associated with pathogenicity, resulting in global fever epidemics, such as observed recently for Chikungunya virus [2]. Furthermore, Semliki Forest virus (SFV) [3] and Venezuelan equine encephalitis (VEE) virus [4] have been identified as the causes of an outbreak of febrile illness in Central Africa and an epidemic in horses and humans in South America, respectively. Despite this potential concern, several alphaviruses, including SFV [5], Sindbis virus (SIN) [6] and VEE [7] have been subjected to the engineering of vectors for heterologous gene expression. In these cases, attenuated strains have been employed.
Several types of vector systems have been engineered. There are three types of replication-deficient vectors consisting of naked RNA, recombinant particles and layered DNA vectors (Figure 1). The application of naked RNA vectors involves the use of in vitro transcribed RNA from an expression vector consisting of the viral nonstructural replicase genes and the foreign gene of interest downstream of the strong subgenomic promoter. The production of recombinant particles requires the co-transfection of in vitro transcribed RNA from an expression vector (as described above) and a helper vector supplying the viral structural genes into mammalian cell lines (for example, baby hamster kidney (BHK) cells). The generated particles are capable of one round of infection of a broad range of host cells, but due to the selective packaging of only expression vector RNA, no further virus production occurs. The layered DNA vector system consists of delivery of a DNA vector providing foreign gene expression from a CMV promoter. Furthermore, the engineering of vectors with an additional subgenomic promoter to the full-length genome allows for the generation of replication-proficient particles, which can provide improved delivery and extended gene expression. All alphavirus vectors described take advantage of the extremely efficient RNA replication, resulting in some 200,000 RNA copies from each RNA molecule. The essential question is: which vector system to use? Obviously, replication-proficient particles can provide efficient delivery, but suffer from potential insufficiency related to safety aspects. Although replication-deficient particles provide a higher level of safety, there is still a marginal risk of the generation of replication-proficient particles through non-homologous recombination. To minimize any unwanted recombination events, a split helper vector system with capsid and envelope genes expressed from separate helper vectors has been engineered [8].
So far, alphaviruses have been applied for the expression of a number of topologically different recombinant proteins [9]. Particularly, the use of SFV particles has resulted in high expression levels of integral membrane proteins in various mammalian host cell lines [10], in primary neurons [11] and in vivo [12]. For vaccine development, vectors based on SFV, SIN and VEE have been applied as naked RNA, recombinant virus particles and layered DNA vectors [13]. In this context, viral and tumor antigens have been administered in various animal models to elicit neutralizing antibodies and protection against challenges with tumor cells or lethal doses of viruses. Moreover, non-viral pathogens have been subjected to vaccine development. Replicon particles derived from VEE have furthermore demonstrated activity as safe and potent systemic, mucosal and cellular adjuvants when co-administered with antigen [14]. Finally, as alphaviruses have been identified as the cause of viral epidemics in animals and humans, a number of approaches have been initiated for immunization against alphavirus-based infections. In this review, the latest development on alphavirus vectors for vaccine production is summarized.
Figure 1. Alphavirus vector systems for vaccine delivery. (A) Naked RNA vector in vitro transcribed from plasmid DNA. (B) Replication-deficient alphavirus particles generated in baby hamster kidney (BHK) cells after co-transfection of in vitro transcribed RNA from the expression and helper vectors. (C) Layered DNA vector for plasmid immunization. SP6, polymerase promoter; 26S, subgenomic alphavirus promoter; CMV, cytomegalovirus promoter; polyA tail, polyadenylation signal.
Figure 1. Alphavirus vector systems for vaccine delivery. (A) Naked RNA vector in vitro transcribed from plasmid DNA. (B) Replication-deficient alphavirus particles generated in baby hamster kidney (BHK) cells after co-transfection of in vitro transcribed RNA from the expression and helper vectors. (C) Layered DNA vector for plasmid immunization. SP6, polymerase promoter; 26S, subgenomic alphavirus promoter; CMV, cytomegalovirus promoter; polyA tail, polyadenylation signal.
Viruses 06 02392 g001

2. Viral Vaccine Approaches

Due to their immunogenic properties, viral structural proteins have been popular targets for alphavirus-based vaccine development [13] (Table 1). In this context, immunization with SFV particles expressing influenza nucleoprotein (NP) elicited a strong immune response in mice [15]. Moreover, VEE-based expression of influenza hemagglutinin (HA) provided protection against challenges with H5N1 virus in chicken [16]. Similarly, SFV particles expressing the HIV envelope [17] and gp41 [18] and VEE particles expressing HIV MA/CA [19] showed humoral and CTL (cytotoxic T-lymphocyte) responses in mice. Furthermore, a VEE particle vaccine expressing the cluster IV H3N2 swine influenza HA gene demonstrated protection against challenges with homologous influenza virus [20]. Likewise, mice and guinea pigs vaccinated with VEE particles expressing Ebola NP [21] and GP [22], respectively, provided protection against challenges with lethal doses of Ebola virus.
Table 1. Alphavirus-based vaccine development for viral targets.
Table 1. Alphavirus-based vaccine development for viral targets.
VirusTargetVector/DeliveryImmunizationResponseReference
BVDVE2VEE/ParticlesCalfBVDV protection[23]
NS3 (p80)SFV/DNAMouseCTL, CMI[24]
CMVgB/pp65-1E1VEE/ParticlesHuman Phase INeutralizing Abs[25]
CSFVE2SFV/DNASwineCSFV protection[26]
DenguePrME, E85VEE/ParticlesMacaqueDengue protection[27]
EbolaNPVEE/ParticlesMouseEbola protection[21]
NP, GPVEE/ParticlesGuinea pigEbola protection[22]
VP24, 30, 35, 30VEE/ParticlesMouseEbola protection[28]
Hepatitis BcAgSIN/DNAMouseSpecific Abs[29]
sAgSIN/DNAMouseSpecific Abs[29]
Hepatitis CcAgSFV/Particles, DNAMouseCTL[30]
NS3SFV/ParticlesMouseCellular[31]
nsPsSFV/ParticlesMouseCD8+ T-cell response[32]
HeVGlycoproteinVEE/ParticlesMouseNeutralizing Abs[33]
HIV-1EnvSFV/ParticlesMouseHumoral[17]
gp41SFV/ParticlesMouseMonoclonal Abs[18]
MA/CAVEE/ParticlesMouseHumoral, CTL[19]
HPV16E7SFV/DNAMouseCTL[34]
16E7-VP22SIN/ParticlesMouseCD8+ T-cell response[35]
HSV-1gpBSIN/ParticlesMouseHSV protection[36]
gpBSIN/DNAMouseCTL, protection[37]
IBDVVP2SFV/Particles, DNAChickenSpecific Abs[38]
InfluenzaHASFV/ParticlesMouseSystemic response[15]
HAVEE/ParticlesChickenInfluenza protection[16]
HAVEE/ParticlesSwineInfluenza protection[20]
NPSFV/Particles, RNAMouseHumoral, CTL[39]
ISAVHESAV/ParticlesSalmonISAV protection[40]
JEVprM-E, NS1-2ASIN/ParticlesMouseJEV Abs[41]
LassaNVEE/ParticlesMouseImmune response[42]
LIVprMESFV/ParticlesMouseLIV protection[43]
prME, NS1SFV/ParticlesSheepLIV protection[44]
MBGVGP, NP, VP35VEE/ParticlesGuinea pigMBGV protection[45]
GP, NPVEE/ParticlesMacaqueMBGV protection[46]
MeaslesHA, FUdSIN/DNAMouseMeasles protection[47]
HA, FUdSIN-VEE/ParticlesMacaqueMeasles protection[48]
MVEprME, ESFV/ParticlesMouseNeutralizing Abs[49]
NiVGlycoproteinsVEE/ParticlesMouseNeutralizing Abs[33]
NLVVLPVEE/ParticlesMouseImmune response[50]
RabiesGSIN/DNAMouseRabies protection[51]
RSVF, GSFV/DNA, RNAMouseRSV protection[52]
F, GSFV/ParticlesMouseRSV protection[53]
RVFVGnVEE/ParticlesMouseRVFV protection[54]
SARS-CoVGlycoproteinVEE/ParticlesMouseSARS-CoV protection[55]
SEOVM, SSIN/Particles, DNAHamsterSEOV protection[56]
SHIVEnvSFV/ParticlesMacaqueT-cell proliferative response[57]
SUDVGPVEE/ParticlesPrimateSUDV protection[58]
VacciniaA33R, B5RVEE/ParticlesMouseVaccinia protection[59]
Abbreviations: Abs, antibodies; BVDV, bovine viral diarrhea virus; CMI, cell-mediated immune response; CMV, cytomegalovirus; CSFV, classical swine fever virus; CTL, cytotoxic T-lymphocyte activity; HBV, hepatitis B virus; HBC, hepatitis C virus; HE, hemagglutinin-esterase; HeV, Hendra virus; HIV, human immunodeficiency virus; HPV, human papillomavirus; HSV, herpes simplex virus; IBDV, infectious bursal disease virus; ISAV, infectious salmon anemia virus; JEV, Japanese encephalitis virus; LIV, Louping-ill virus; MBGV, Marburg virus; MVE, Murray Valley encephalitis virus; NiV, Nipah virus; NLV, Norwalk-like virus; RSV, respiratory syncytial virus; RVFV, Rift Valley fever virus; SARS‑CoV, severe acute respiratory syndrome corona virus; SAV, salmon anemia virus; SEOV, Seoul virus; SFV, Semliki Forest virus; SHIV, simian-human immunodeficiency virus; SIN, Sindbis virus; SUDV, Sudan virus; VEE, Venezuelan equine encephalitis virus.
Attempts to further improve the immunogenicity of vaccine candidates, the herpes simplex virus type I (HSV-1) VP22 protein was fused to the H5N1 subtype influenza HA [60]. The responses of both interleukin-4 (IL-4) of CD4+ T-cells and interferon-gamma (IFNɣ) of CD8+ T-cells were observed in vaccinated mice. VEE replicon particles expressing the severe acute respiratory syndrome coronavirus (SARS-CoV) glycoprotein managed to provide protection against challenges with lethal doses of SARS-CoV in vaccinated mice [55]. Furthermore, VEE particles were applied for the expression of glycoproteins from the zoonotic pathogenic Hendra virus (HeV) and Nipah virus (NiV), known to cause fatal infections in both animals and humans [33]. Immunization resulted in enhanced induction of cross-reactive neutralizing antibodies. In another study, mice were vaccinated with both DNA plasmids and alphavirus replicons expressing Rift Valley fever virus (RVFV) glycoprotein Gn fused to the C3d complement protein [54]. The immunization generated neutralizing antibodies and provided protection against challenges with RVFV, which suggested that plasmid DNA and alphavirus replicon approaches, as well as the combined DNA prime/replicon boost strategy show great promise for valid RVFV vaccine development. The combined approach included plasmid vaccinations at Weeks 0 and 3 followed by a replicon boost at Week 6.
In a combined vaccine approach, SFV DNA vectors and recombinant adenovirus expressing the classical swine fever virus (CSFV) E2 glycoprotein elicited higher titers of neutralizing antibodies in pigs [26]. After challenges with the virulent CSFV Shimen strain, no symptoms of viremia were observed, for the combined vaccine, whereas vaccination with adenovirus alone resulted in viremia in one pig of five. Furthermore, sequential immunization with SIN and VEE replicon particles expressing the type 1 HIV gp140 envelope (Env) and trimeric Env protein in MF59 adjuvant provided partial protection in macaques against intravenous challenges with high doses of simian-human immunodeficiency virus (SHIV) [61]. The administration could be further extended to intramuscular and mucosal delivery [62]. Different degrees of protection were observed against challenges with SHIV after mucosal administration. In contrast, intramuscular vaccination rendered macaques to be completely resistant to SHIV. In cotton rats, SIN DNA vectors carrying the hemagglutinin (pMSIN-H) and fusion proteins (pMSINH-FdU) elicited neutralizing antibodies, mucosal and systemic antibody-secreting cells, memory B-cells and IFNɤ secreting T-cells [47]. Priming with pMSIN-H provided 100% protection against challenges with pulmonary measles. However, pMSINH-FdU priming was observed only after a boost with live measles virus vaccine. In another study, chimeric VEE/SIN replicon particles were applied for the expression of measles virus hemagglutinin (H) and fusion (F) proteins, which elicited high-titer neutralizing antibody and IFNɤ-producing T-cells in macaques after intradermal vaccination [48]. Protection from rash and viremia was obtained after challenges with wild-type measles virus 12–17 months after vaccination. Alphaviruses have also been subjected to the development of smallpox vaccines by the introduction of A33R, B5R, A27L and L1R genes into VEE particles [59]. Vaccinated mice showed protective immunity. Furthermore, vaccination of macaques elicited strong antibody responses and was capable of neutralizing and inhibiting the spread of vaccinia and monkey pox viruses. SIN-based DNA vaccines have been developed against rabies [51]. In comparison to a conventional rabies DNA vaccine, the SIN DNA vaccine induced better humoral and cell-mediated immune responses in immunized mice and showed complete protection against challenge with the CVS rabies strain.
Recently, novel hepatitis C virus (HCV) vaccine candidates were developed by expressing all or a part of the HCV non-structural proteins (nsPs) from an SFV vector [32]. An insert as large as 6.1 kb allowed the expression of all nsPs leading to a strong and long-lasting NS3-specific CD8+ T-cell response. The level of T-cell response was similar to that observed for the expression of only NS3/4A. Immunization demonstrated significant growth delay of HCV-expressing EL4 tumors in a mouse model. In another study, glycoproteins for either Sudan virus (SUDV) or Ebola virus were expressed from VEE replicons and evaluated in vaccinated nonhuman primates [58]. A single intramuscular injection with VEE particles expressing SUDV GP provided complete protection against challenges with SUDV in cynomolgus macaques. However, VEE-SUDV GP vaccinated primates were not fully protected against back challenges with Ebola virus. On the other hand, co-injection of VEE particles expressing SUFV GP and Ebola virus GP showed protection against both virus types.
Recently, VEE replicon particles generated in Vero cells were used to express the E2 glycoprotein of bovine viral diarrhea virus (BVDV) [23]. Vaccination of BVDV free calves with 1 × 106 IU and 1 × 107 IU, respectively, resulted in neutralizing antibody titers, which were able to cross-neutralize both type 1 and type 2 BVDV genotypes after booster immunizations. Vaccination with the higher dose significantly reduced the viral-based leukopenia and showed some protection from clinical disease. Similarly, VEE replicon particles were engineered to express Dengue virus E antigen in two configurations as subviral particles (prME) and soluble dimers (E85) [27]. Immunization of macaques resulted in the rapid production of neutralizing antibodies and demonstrated protection against challenges with Dengue virus. Moreover, the tetravalent E85 VEE replicon particle vaccine induced a protective response to all four Dengue virus serotypes when two immunizations were administered six weeks apart.
A novel approach has been to combine alphavirus replicons with pseudotyped baculovirus [63]. In this context, pseudotyped baculovirus vectors containing the hybrid CM promoter and SFV replicon were used for the expression of GP5 and M proteins of porcine reproductive and respiratory syndrome virus (PRRSV) and compared to a pseudotyped baculovirus vector carrying only a CMV promoter. Immunization with the hybrid CMV/SFV showed the induction of strong GP5-specific antibodies, and in general, the Th1-dominant immune response was stronger than the one elicited by the CMV promoter alone.
Veterinary medicine has also gained from alphavirus vaccine development. In this context, salmonid alphavirus (SAV) replicons were applied for the expression of infectious salmon anemia virus (ISAV) hemagglutinin-esterase (HE) [40]. Intramuscular administration of SAV replicons provided protection against challenges with ISAV in Atlantic salmon. In contrast, intraperitoneal injection was not successful [64]. In another study, DNA vaccines based on the SAV E1 and E2 spike proteins were compared to whole virus vaccine in Atlantic salmon [65]. The whole virus vaccine showed superior immunogenicity to the DNA vaccine; the latter provided only marginal reduction in viral replication, and the protection against SAV challenges was no different from controls.

3. Non-Viral Targets

Alphavirus-based vaccine development has also been addressed for a number of other infectious pathogens (Table 2). For instance, SFV vectors were employed for the expression of the Plasmodium falciparum Pf332 antigen, which elicited immunological memory in vaccinated mice [66]. Moreover, vaccination of mice with SIN plasmid DNA vectors carrying the Mycobacterium tuberculosis 85A antigen (Ag85A) provided strong immunity and resulted in long-term protection against M. tuberculosis challenges [67]. In another approach, using SFV DNA replicons, the botulinum neurotoxin A Hc (BoNTA-Hc) gene provided both antibody and lymphoproliferative responses in vaccinated BALB/c mice [68]. The immunogenicity was enhanced when granulocyte-macrophage colony-stimulating factor (GM-CSF) was co-expressed as an adjuvant. Additionally, replication-deficient SFV particles were used for the expression of the Brucella abortus translation initiation factor 3 (IF3) [69]. Immunization of BALB/c mice demonstrated significant levels of protection against challenges with the virulent B. abortus strain 2308. Similarly, protective antigen (PA) for Bacillus anthracis were expressed from SIN vectors, resulting in specific and neutralizing antibodies in Swiss Webster mice and offered some protection against challenges with lethal doses of the pathogenic bacteria [70].
Table 2. Alphavirus-based vaccine development for non-viral infectious agents.
Table 2. Alphavirus-based vaccine development for non-viral infectious agents.
AgentTargetVector/DeliveryImmunizationResponseReference
B. anthracisPASIN/ParticlesMouseB. anthracis protection[70]
B. abortusIF3SFV/ParticlesMouseBrucella protection[69]
C. botulinumBoNTA-HcSFV/DNAMouseAbs, lymphoproliferative response[68]
MalariaCSSIN/ParticlesMouseMalaria protection[71]
M. tuberculosisAg85ASIN/DNAMouseProtection[67]
P. falciparumAg Pf332SFV/Particles-RNAMouseImmunological memory[66]
PrionNPSFV/ParticlesMouseMonoclonal Abs[72]
Staphylococcusenterotoxin BVEE/ParticlesMouseProtection[73]
Abbreviations: Abs, antibodies; SFV, Semliki Forest virus; SIN, Sindbis virus; VEE, Venezuelan equine encephalitis virus.

4. Tumor Vaccine Approaches

Alphaviruses have found frequent applications in the area of tumor vaccine development (Table 3). In this context, naked RNA, replication-deficient particles and DNA layered vectors have been employed as delivery vehicles. For instance, mice have been subjected to immunization with naked SFV RNA replicons carrying the LacZ gene [74]. Interestingly, a single injection of only 1 μg of SFV‑LacZ RNA presented complete tumor protection. Furthermore, when tumors were administered two days prior to the immunization, the survival was extended by 10–20 days. Among DNA-based tumor vaccine approaches, SIN vectors expressing mouse and human tyrosine-related protein-1 (TRP-1) were evaluated in a B16 mouse melanoma model [75]. Intramuscular injection was capable of breaking immune tolerance and provided protection against melanoma when mice were vaccinated five days prior to cancer challenge. In another study, alphavirus replicon-based expression of melanoma differentiation antigen (MDA) tyrosine demonstrated the inhibition of the growth of B16 transplantable melanoma [76]. In this context, the vaccine encoding tyrosine related protein 2 (TRP-2) relied on a novel immune mechanism, which required the activation of both IgG and CD8+ cell effector responses.
Table 3. Alphavirus-based vaccine development for cancer targets.
Table 3. Alphavirus-based vaccine development for cancer targets.
TargetGeneVector/DeliveryImmunizationResponseReference
Brain tumorIL-12SFV/ParticlesMouseImmunogenicity[77]
EndostatinSFV/ParticlesMouseInhibited tumor growth[78]
LacZSFV/ParticlesMouseTumor protection[79]
gp100, IL-18SIN/DNAMouseTumor protection[80]
HER2/neuSIN/DNAMouseTumor protection[81]
HER2/neuSIN/DNAMouseTumor protection[82]
HER2/neuSIN/DNA, paclitaxelMouseTumor regression[83]
NeuVEE/ParticlesRatAnti-tumor immunity[84]
NeuVEE /Particles, DCsMouseTumor regression[85]
Cervical cancerHPVE6-E7SFV/ParticlesMouseTumor protection[86]
HPVE6-E7SFV/ParticlesMouseTumor regression[87]
HPV-CRTSIN/ParticlesMouseTumor protection[88]
HPVE7VEE/ParticlesMouseTumor protection[89]
HPVE6E7+IL12SFV/ParticlesMouseAnti-tumor activity[90]
HPVE7-VP22SIN/ParticlesMouseCD8+ T-cell response[91]
Colon cancerSFV vectorSFV/ParticlesMouseAnti-tumor effect[92]
EndothelialVEGFR-2SFV/ParticlesMouseAntibody response[93]
GliomaB16, 203SFV/ParticlesMouseTumor protection[94]
Kidney cancerIL-12SFV/Encapsulated particlesHuman Phase I5-fold IL-12 expression[95]
MelanomaMDA/trp-2VEE/ParticlesMouseTherapeutic effect[76]
IL-12SFV/ParticlesMouseTumor eradication[96]
IL-12SFV/Encapsulated particlesHuman Phase I5-fold IL-12 expression[95]
MUC18/MCAMSIN/DNAMouseTumor protection[97]
MetastaticCEAVEE/ParticlesHuman Phase ICEA Abs, extended survival[98]
PSMAVEE/ParticlesHuman Phase IPSMA Abs[99]
Prostate cancerPSMAVEE/ParticlesMouseTumor response[100]
STEAPVEE/DNAMouseAnti-tumor response[101]
PSCAVEE/ParticlesMouseTumor protection[102]
Tumorβ-galactosidaseSFV/RNAMouseTumor protection[74]
IL-12SFV/ParticlesMouseTumor protection[103]
Tumor antigenMHC class IISFV/Particles-DNAMouseImmunogenicity[104]
P815SFV/ParticlesMouseCTL, tumor protection[105]
trp-1SIN/DNAMouseAntitumor activity[91]
Abbreviations: CEA, carcinoembryonic antigen; CRT, calreticulin; CTL, cytotoxic T-lymphocyte activity; DCs, dendritic cells; HPV, human papillomavirus; IL, interleukin; MCAM, melanoma cell adhesion molecule; MDA, melanoma differentiation antigen; MHC, major histocompatibility complex; PSCA, prostate stem cell antigen; PSMA, prostate-specific membrane antigen; SFV, Semliki Forest virus; SIN, Sindbis virus; STEAP, six-transmembrane epithelial antigen of the prostate; trp, tyrosine-related protein; VEE, Venezuelan equine encephalitis virus.
Furthermore, vaccination with recombinant particles expressing the P1A gene [105] and the human papilloma virus (HPV) E7 gene [89] from SFV and VEE vectors, respectively, provided protection against further tumor development in mice. Attempts have also been made to improve the efficacy of SFV-based HPV vaccines by supplying SFV-based IL-12 expression in mice [90]. At low doses, IL-12 stimulated antigen-specific CTL responses and enhanced anti-tumor responses after SFV-based HPV16-E6E7 immunization. Subsequent increases in dosage, however, neither improved the immune responses, nor tumor regression. SIN DNA vectors have been employed for the expression of the murine melanoma cell adhesion molecule (MCAM/MUC18) as a vaccine against murine melanoma, which resulted in the induction of humoral and CD8+ T-cell immune responses against melanoma [97].
In the context of breast cancer, a DNA-based SIN vector expressing the neu gene was applied for intramuscular vaccination of mice 14 days prior to the injection of cancer cells overexpressing neu [81]. The immunization provided strong protection against tumor development. The incidence of lung metastasis from mammary fat pad tumors was reduced. Moreover, the number of lung metastases from intravenous injection of neu overexpressing cells decreased. Additionally, intradermal vaccination provided tumor protection applying 80% less plasmid than required for conventional DNA vectors. Further confirmation of successful cancer vaccination was obtained from the administration of SIN vectors expressing neu (pSINCP/neu) in a murine breast tumor model [82]. However, in this case, the prophylactic vaccine only showed efficacy when administered prior to the tumor challenge. Another approach was comprised of combining alphavirus-based delivery with the chemical anticancer agent, doxorubicin [83]. When pSINCP/neu DNA and VEE/neu particles were administered after injection of 5 mg/kg of doxorubicin, the tumor progression was significantly delayed. This phenomenon did not occur for doxorubicin alone. Similarly, a combination therapy with paclitaxel (25 mg/kg) and pSINCP/neu was ineffective. Moreover, VEE-neu particles were subcutaneously administered in a rat mammary tumor model, which resulted in the elimination of 36% of pre-existing aggressive mammary tumors [84]. The combination of dendritic cell (DC)-based cancer immunotherapy with VEE-neu particle administration induced both cellular and humoral immunity against neu in transgenic human breast tumor-bearing mice [85]. Moreover, this treatment resulted in the significant inhibition of tumor growth. Similarly, both tumor growth and pulmonary metastatic spread were significantly inhibited when mice with pre‑existing tumors were subjected to five immunizations with SFV10-E VLP expressing the vascular endothelial growth factor receptor 2 (VEGFR-2) [93]. Furthermore, co-immunization with SFV particles encoding VEGFR-2 and IL-4 generated significant tumor regression in mice.
Lung cancer has also been targeted by combined therapy with SFV-IL-12 particles and anti-CD137 monoclonal antibodies [96]. Syngeneic TC-1 lung carcinoma was inhibited after intratumoral SFV-IL-12 administration and co-stimulation with anti-CD137 mAbs. In the context of colon cancer vaccines, a SIN-based DNA vector carrying the LacZ gene was compared to conventional plasmid DNA vectors in mice with CT26.CL25 tumors [106]. Intramuscular immunization elicited immune responses at doses 100- to 1,000-fold lower for the SIN DNA replicon vector compared to the conventional CMV‑promoter-based DNA-LacZ vector. Similarly, SFV particles providing VEGFR-2 expressing in vaccinated mice inhibited CT26 colon carcinoma growth [93]. Closer analysis of microvessel density demonstrated that a significant inhibition of tumor angiogenesis occurred. Additionally, when mice were co-immunized with SFV-VEGFR-2 and SFV-IL-4 particles, their survival rate was significantly enhanced. Furthermore, oncolytic SFV vectors have been used for immune stimulation in a CT26 colon tumor model [92]. Intratumoral injections led to an immediate and intense inflammatory reaction and a significant improvement in survival rates. SFV particles expressing HPV16 E6 and E7 have been subjected to prophylactic vaccine development in a murine TC-1 model for cervical cancer [107]. Pre‑immunization with a low dose (104 particles) resulted in an HPV-specific CTL response in 50% of mice, whereas a higher dose (106 particles) elicited CTL responses in all animals. Furthermore, at a dose of 5 × 106 particles, 40% of mice were protected from tumor challenges. In another study, the SFV-enhE6,7 particle vaccine showed its potential after intravenous and intramuscular delivery, where exponentially growing tumors completely resolved [87]. Similarly, SIN virus RNA replicons expressing HPV E7 were evaluated in a TC-1 mouse model [108]. The humoral and cellular immune responses were poor, and no tumor protection was obtained; but, when the HPV E7 gene was fused to the secretory Sig protein and lysosome-associated membrane protein 1 (LAMP-1), enhanced E7‑specific CD4+ helper T-cell and CD8+ cytotoxic T-cell activity was observed. Moreover, strong in vivo anti-tumor activity was induced. In another study, SIN virus particles expressing both HPV E7 and calreticulin (CRT), an endoplasmic reticulum Ca2+ binding transporter, were tested as prophylactic vaccines [88]. Vaccinations generated antigen-specific immune responses, an anti‑angiogenic effect and a strong anti-tumor activity. Furthermore, intramuscular immunization one week prior to challenge with TC-1 carcinoma cells provided protection to all treated mice. Also VEE particles expressing HPV E7 were subcutaneously injected in mice two weeks prior to cancer cell inoculation, which prevented tumor formation [89]. Furthermore, vaccination induced long-term memory, as protection was observed for challenges three months after the immunization. The therapeutic efficacy was only 67% of treated tumor-bearing mice. However, co-expression of HPV E6 and E7 from the same vector significantly enhanced the therapeutic effect [109,110].
In a prostate tumor model, VEE particles expressing human prostate-specific membrane antigen (PSMA) showed strong cellular and humoral immunity after subcutaneous administration [100]. Furthermore, VEE particles have been employed for the expression of the predominantly prostate tissue-specific six transmembrane epithelial antigen of the prostate (STEAP) [101]. Pre‑immunization with VEE-STEAP particles induced a specific immune response and significantly prolonged the overall survival of mice bearing TRAMPC-2 tumors. When TRAMP mice were prophylactically immunized with a prostate stem cell antigen (PSCA) DNA plasmid followed by VEE‑PSCA particle administration, a specific immune response and anti-tumor protection were observed in 90% of vaccinated animals [102]. Several vaccine studies have targeted brain tumors. For instance, SFV particles expressing endostatin showed a significant reduction of intratumoral vascularization after intratumoral delivery [78]. In another approach, bone-marrow isolated dendritic cells (DCs) were transduced with SFV vectors carrying cytokine genes of specific cDNAs from melanoma and glioma cells [79]. Pre-vaccination with DCs transduced with SFV-based B16 and 203 glioma cDNAs, respectively, resulted in tumor challenge protection and the prolonged survival of tumor-bearing mice. The combination of DCs transduced with SFV-IL-12 particles and systemically administered IL-18 also provided increased survival rates [111]. Moreover, the expression of human melanoma-associated antigen gp100 and IL-18 from a SIN virus DNA vector induced specific anti-tumor CTL responses and provided anti-tumor protection [80]. Vaccination prevented B16-hgp100 tumor formation and demonstrated significant prolongation of survival in mice with established B16-hgp100 tumors.

5. Vaccines against Alphaviruses

As alphaviruses have been suggested to be responsible for epidemics in various parts of the world, it has also become important to develop vaccines against alphaviruses themselves (Table 4). In this context, protection against airborne virus was observed in BALB/c mice vaccinated with an attenuated VEE strain [112]. In another study, improved protection against VEE challenges was achieved by using a live attenuated V3526 VEE vaccine [113]. Similarly, when a chimeric Eastern equine encephalitis (EEE) and Western equine encephalitis (WEE) virus were applied for the vaccination of C57BL/6 mice, complete protection was observed against lethal challenges with a virulent Eastern equine encephalitis (EEE) virus strain [114].
Table 4. Alphavirus-based vaccine development against alphaviruses.
Table 4. Alphavirus-based vaccine development against alphaviruses.
VirusGeneVector DeliveryImmunizationResponseReference
CHIKTSI-GSD-218CHIK infectionHuman Phase IINeutralizing Abs[115]
CHIKGlycoproteinCHIK infectionMacaquesNeutralizing Abs[116]
CHIKIRESCHIK infectionVero cellsMosquito resistance[117]
CHIKC, E1 VLPsCHIK infectionPrimatesCHIK protection[118]
CHIKnsP3, E1 siRNACHIK infectionVero cellsReduced CHIK titer[119]
CHIKmiRNAsCHIK infectionMouseReduced CHIK replication[120]
EEEEEE/WEEEEE infectionMouseEEE protection[114]
VEEVEE attVEE infectionMouseVEE protection[112]
VEEVEE V3526VEE infectionMouseVEE protection[113]
VEEVEE TC-83VEE infectionMouseVEE protection[121]
VEE26SVEE infectionMacaquesVEE protection[122]
VEECHIK genesChimeric VEE-CHIKMosquitoReduced infectivity[123]
VEERdRp miRNAVEE infectionBHK cellsInhibition of VEE replication[124]
WNVWNV attWNV NanopatchMouseAbs[125]
Abbreviations: Abs, antibodies; att, attenuated; C, capsid; CHIK, Chikungunya virus; EEE, Eastern equine encephalitis virus; miRNA, micro-RNA; RdRp, RNA-dependent RNA polymerase; SFV, Semliki Forest virus; SIN, Sindbis virus; siRNA, short interfering RNA; VEE, Venezuelan equine encephalitis virus; VLPs, virus-like particles; WNV, West Nile virus.
In attempts to design attenuated alphaviruses for vaccine development, the mechanisms of replication and virus-host interaction have been investigated. In this context, mosquito transmission of Chikungunya (CHIK) virus has been prevented by making their replication dependent on internal ribosome entry sites (IRES) [117]. Although replication does not occur in mosquito cells, replication proceeds efficiently in Vero cells. In another approach, the non-structural genes of the attenuated VEE strain TC-83 and the naturally attenuated EEV strain, respectively, were engineered with the structural genes of CHIK [123]. The chimeric vaccine candidates presented a significantly reduced infectivity of the common urban Aedes aegypti and A. albopictus vectors for CHIK, thereby providing a low risk of virus transmission. A new CHIK virus isolated from an acutely infected human patient was used for the engineering of a synthetic DNA vaccine [116]. When this DNA vaccine was electroporated into mice, robust antigen-specific cellular and humoral responses were obtained, which provided protection against further challenges with CHIK. Furthermore, immunization of macaques demonstrated the induction of neutralizing antibodies similar to those elicited in sera from convalescent human patients. In another DNA vector approach, the VEE 26S structural genes were expressed and administered as an aerosol in nonhuman primates [122]. Vaccination resulted in no viremia in two macaques and low viremia in one animal, whereas it was high in all control animals.
In an approach to target epidermal and dermal antigen presenting cells (APCs), the Nanopatch (NP) technology has been applied for skin vaccination of West Nile virus and CHIK in mice [125]. NP, comprised of arrays of densely packed projections two orders of magnitude smaller than standard needles, provided efficient delivery of inactivated whole CHIK vaccine and a DNA-based West Nile virus vaccine. Moreover, as virus-like particles (VLPs) consisting of CHIK capsid and envelope proteins have been demonstrated to protect nonhuman primates against infection of multiple strains of CHIK, it was of benefit to screen and optimize the solution conditions for stable vaccine formulations [118]. In this context, sugar, sugar alcohols and polyanions were identified as potential stabilizers.
The relatively recent discovery of the commonly occurring phenomenon of gene silencing has also made its impact on vaccine development. In this context, small interfering RNAs (siRNAs) were engineered against two conserved regions of the nsP3 and E1 genes of CHIK [119]. A significant reduction of virus titer was observed in Vero cells after 24 h (99%) and 48 h (65%), suggesting a potential new therapeutic approach. In another approach, microRNA (miRNA)-specific target sequences for replicon particle production were introduced into alphavirus helper RNAs [120]. In the presence of miRNA-specific inhibitors, CHIK particles were efficiently generated, whereas in their absence, cellular miRNAs downregulated helper RNA replication. When mice were inoculated with replicon RNA carrying engineered miRNA sequences, cellular miRNAs prevented the replication of replicon RNA, which opens the feasibility of using miRNAs as a therapeutic approach for the inhibition of viral replication. Moreover, the VEE RNA-dependent RNA polymerase (RdRp) was targeted by artificial miRNAs in attempts to inhibit VEE replication [124]. Of five miRNAs that were evaluated in BHK cells, three showed significant inhibition of VEE replication.

6. Clinical Trials for Alphavirus Vaccines

Despite the large number of studies conducted in various animal models, relatively few evaluations have been subjected to humans. A clinical trial conducted with alphaviruses involved intravenous administration of liposome encapsulated SFV particles expressing IL-12 particles to melanoma and kidney carcinoma patients [95]. High transient expression of recombinant IL-12 with a five-fold increase lasted for 4–7 days. As higher doses induced a fever response in patients, the maximum tolerated dose (MTD) was restricted to 3 × 109 encapsulated particles per m2. No liposome- or SFV-related toxicity was observed. Furthermore, the liposome-encapsulation prevented SFV particles from being recognized by the host immune system and, therefore, allowed repeated systemic administration. The phase I study clearly demonstrated the safe administration of encapsulated SFV particles.
A human phase II, randomized, double-blind, placebo-controlled, safety and immunogenicity study was conducted in 73 healthy adult volunteers on a serially passaged, plaque-purified, live CHIK vaccine [115]. A single subcutaneous injection of the CHIK vaccine was administered to 59 volunteers, and 14 individuals received placebo. There were no adverse events, except for transient arthralgia in five individuals receiving the CHIK vaccine. Of the 58 evaluable vaccinated individuals, 57 (98%) developed CHIK neutralizing antibodies by Day 28 and 85% still remained seropositive one year later. None of the individuals who received placebo were seropositive. In another vaccine-related alphavirus phase I randomized, double-blind clinical trial for cytomegalovirus (CMV), a two-component vaccine expressing CMV gB or pp65/1E1 fusion protein was administered intramuscularly or subcutaneously in CMV seronegative adult volunteers [25]. The vaccination showed only mild to moderate local reactions and no clinically important changes. It induced neutralizing antibody and multifunctional T-cell responses against CMV antigens.
In another clinical trial, alphavirus particles expressing the carcinoembryonic antigen (CEA) were repeatedly administered to patients with metastatic cancer [98]. CEA-specific antibodies were able to mediate antibody-dependent cellular cytotoxicity against tumor cells from human colorectal cancer metastases. Furthermore, patients with CEA-specific antibodies encouragingly showed extended overall survival. Similarly, VEE replicons expressing the prostate-specific membrane antigen (PSMA) were applied for a human trial of patients with metastatic cancers at five doses of 0.9 × 107 IU or 3.6 × 107 IU [99]. At the lower dose, no PSMA-specific cellular response was obtained, but a weak PSMA-specific signal was detected by ELISA. Disappointingly, the higher dose showed no PMSA‑specific response. The trial demonstrated that while there was neither clinical benefit nor robust immune responses, there were no toxicities associated with the immunization, and the VEE-PMSA particles were well tolerated. However, as neutralizing antibodies were elicited, the dosing might be suboptimal, requiring some further optimization.

7. Conclusions

A number of vaccine development studies have been conducted using mainly the three most commonly applied alphavirus vectors, SFV, SIN and VEE. To demonstrate the variety of approaches available, replicon particles, naked RNA and layered DNA vectors have been employed. Each approach has commonly generated responses detected by cellular or humoral responses. In the case of vaccine development against lethal viruses (Table 1), non-viral infectious targets (Table 2) and tumors (Table 3) immunization has provided long-term protection against challenges with the disease-causing agents. Moreover, due to the pathogenicity of several alphaviruses, they are themselves credible targets for vaccine development. Therefore, a number of studies, particularly for VEE and CHIK (Table 4), have provided protection against challenges with virulent alphavirus strains.
The potential of alphaviruses causing global epidemics has placed additional concern on the needs of addressing biosafety issues. Generally, the strains used for vaccine development are attenuated. Furthermore, in the case using alphavirus particles, second generation helper vectors [126] or split helper systems [8] have been applied to prevent any production of wild-type-like replication-proficient particles through non-homologous recombination. Only recently alphavirus-based vaccines have been subjected to clinical trials. In this context, SFV vectors have been used for delivery of immunostimulatory genes. Furthermore, immunizations for the treatment of CMV and vaccinations against prostate and metastatic cancers have been conducted with alphavirus vectors and particles. The strength of applying alphaviruses is the generation of rapid transgene expression and the transient nature of expression. However, the full potential of alphavirus-based vaccines has not been explored yet. In the future, it is anticipated that additional, positive observations, particularly in the form of providing protection against challenges with lethal pathogens and tumors, will attract the enhanced application of alphaviruses for vaccine development.

Conflicts of Interest

The author declares no conflict of interest.

References and Notes

  1. Strauss, J.H.; Strauss, E.G. The Alphaviruses: Gene Expression, Replication and Evolution. Micobiol. Rev. 1994, 58, 491–562. [Google Scholar]
  2. Kelvin, A.A. Outbreak of Chikungunya in the Republic of Congo and the global picture. J. Infect. Dev. Countries 2011, 5, 441–444. [Google Scholar]
  3. Mathiot, C.C.; Grimaud, G.; Garry, P. An outbreak of Semliki Forest virus infections in Central African Republic. Am. J. Trop. Med. Hyg. 1990, 42, 386–393. [Google Scholar]
  4. Weaver, S.C.; Salas, R.; Rico-Hesse, R. Remergence of epidemic Venezuelan equine encephalomyelitis in South America. VEE Study Group. Lancet 1996, 348, 436–440. [Google Scholar] [CrossRef]
  5. Liljeström, P.; Garoff, H. A new generation of animal cell expression vectors based on the Semliki Forest virus replicon. Biotechnology 1991, 9, 1356–1361. [Google Scholar] [CrossRef]
  6. Xiong, C.; Levis, R.; Shen, P.; Schlesinger, S.; Rice, C.M.; Huang, H.V. Sindbis virus: An efficient, broad host range vector for gene expression in animal cells. Science 1989, 243, 1188–1191. [Google Scholar]
  7. Davis, N.L.; Brown, K.W.; Johnston, R.E. In vitro synthesis of infectious Venezuelan equine encephalitis virus RNA from a cDNA clone: Analysis of a viable deletion mutant. Virology 1989, 171, 189–204. [Google Scholar] [CrossRef]
  8. Smerdou, C.; Liljeström, P. Two-helper RNA system for production of recombinant Semliki Forest virus particles. J. Virol. 1999, 73, 1092–1098. [Google Scholar]
  9. Lundstrom, K.; Schweizer, C.; Rotmann, D.; Hermann, D.; Schneider, E.M. Semliki Forest virus vectors: efficient vehicles for in vitro and in vivo gene delivery. FEBS Lett. 2001, 504, 99–103. [Google Scholar] [CrossRef]
  10. Lundstrom, K. Semliki Forest virus vectors for rapid and high-level expression of integral membrane proteins. Biochim. Biophys. Acta 2003, 1610, 90–96. [Google Scholar] [CrossRef]
  11. Ehrengruber, M.U.; Lundstrom, K.; Schweitzer, C.; Heuss, C.; Schlesinger, S.; Gähwiler, B.H. Recombinant Semliki Forest virus and Sindbis virus infect efficiently neurons in hippocampal slice cultures. Proc. Natl. Acad. Sci. USA 1999, 96, 7041–7046. [Google Scholar]
  12. Lundstrom, K.; Richards, J.G.; Pink, J.R.; Jenck, F. Efficient in vivo expression of a reporter gene in rat brain after injection of recombinant replication-deficient Semliki Forest virus. Gene Ther. Mol. Biol. 1999, 3, 15–23. [Google Scholar]
  13. Lundstrom, K. Alphavirus vectors for vaccine production and gene therapy. Exp. Rev. Vacc. 2003, 2, 447–459. [Google Scholar] [CrossRef]
  14. Tonkin, D.R.; Whitmore, R.; Johnston, R.E.; Barro, M. Infected dendritic cells are sufficient to mediate the adjuvant activity generated by Venezuelan equine encephalitis virus replication particles. Vaccine 2012, 30, 4532–4542. [Google Scholar] [CrossRef]
  15. Malone, J.G.; Berglund, P.J.; Liljestrom, P.; Rhodes, G.H.; Malone, R.W. Mucosal immune responses associated with polynucleotide vaccination. Behring Inst. Mitt. 1997, 98, 63–72. [Google Scholar]
  16. Schultz-Cherry, S.; Dybing, J.K.; Davis, N.L.; Williamson, C.; Suarez, D.L.; Johnston, R.; Perdue, M.L. Influenza virus (A/HK/156/97) hemagglutinin expressed by an alphavirus replicon system protects against lethal infection with Hong Kong-origin H5N1 viruses. Virology 2000, 278, 55–59. [Google Scholar] [CrossRef]
  17. Brand, D.; Lemiale, F.; Turbica, I.; Buzelay, L.; Brunet, S.; Branet, F. Comparative analysis of humoral immune responses to HIV type 1 envelope glycoproteins in mice immunized with a DNA vaccine, recombinant Semliki Forest virus RNA, or recombinant Semliki Forest virus particles. AIDS Res. Hum. Retroviruses 1998, 14, 1369–1377. [Google Scholar] [CrossRef]
  18. Giraud, A.; Ataman-Onal, Y.; Battail, N.; Piga, N.; Brand, D.; Mandrand, B.; Verrier, B. Generation of monoclonal antibodies to native human immunodeficiency virus type 1 envelope glycoprotein by immunization of mice with naked RNA. J. Virol. Methods 1999, 79, 75–84. [Google Scholar] [CrossRef]
  19. Caley, I.J.; Betts, M.R.; Irlbeck, D.M.; Davis, N.L.; Swanstrom, R.; Frelinger, J.A.; Johnston, R.E. Humoral, mucosal, and cellular immunity in response to a human immunodeficiency virus type 1 immunogen expressed by a Venezuelan equine encephalitis virus vaccine vector. J. Virol. 1997, 71, 3031–3038. [Google Scholar]
  20. Bosworth, B.; Erdman, M.M.; Stine, D.L.; Harris, I.; Irwin, C.; Jens, M.; Loynachan, A.; Kamrud, K.; Harris, D.L. Replicon particle vaccine protects swine against influenza. Comp. Immunol. Microbiol. Infect. Dis. 2010, 33, e99–e103. [Google Scholar]
  21. Wilson, J.A.; Hart, M.K. Protection from Ebola virus mediated by cytotoxic T-lymphocytes specific for the viral nucleoprotein. J. Virol. 2001, 75, 2660–2664. [Google Scholar] [CrossRef]
  22. Pushko, P.; Bray, M.; Ludwig, G.V.; Parker, M.; Schmaljohn, A.; Sanchez, A.; Jahrling, P.B.; Smith, J.F. Recombinant RNA replicons derived from attenuated Venezuelan equine encephalitis virus protect guinea pigs and mice from Ebola hemorrhagic fever virus. Vaccine 2000, 19, 142–153. [Google Scholar] [CrossRef]
  23. Loy, J.D.; Grander, J.; Mogler, M.; Vander Veen, R.; Ridpath, J; Harris, D.H.; Kamrud, K. Development and evaluation of a replicon particle vaccine expressing the E2 glycoprotein of bovine viral diarrhea virus (BVDV) in cattle. Virol. J. 2013, 10, e35. [Google Scholar] [CrossRef]
  24. Reddy, J.R.; Kwang, J.; Varthakavi, V.; Lechtenberg, K.F.; Minocha, H.C. Semiliki forest virus vector carrying the bovine viral diarrhea virus NS3 (p80) cDNA induced immune responses in mice and expressed BVDV protein in mammalian cells. Comp. Immunol. Microbiol. Infect. Dis. 1999, 22, 231–246. [Google Scholar]
  25. Bernstein, D.I.; Reap, E.A.; Katen, K.; Watson, A.; Smith, K.; Norberg, P.; Olmsted, R.A.; Hoeper, A.; Morris, J.; Negri, S.; et al. Randomized, double-blind, Phase 1 trial of an alphavirus replicon vaccine for cytomegalovirus in CMV seronegative adult volunteers. Vaccine 2009, 28, 484–493. [Google Scholar] [CrossRef]
  26. Sun, Y.; Li, N.; Li, H.Y.; Li, M.; Qiu, H.J. Enhanced immunity against classical swine fever in pigs induced by prime-boost immunization using an alphavirus replicon-vectored DNA vaccine and a recombinant adenovirus. Vet. Immunol. Immunopathol. 2010, 137, 20–27. [Google Scholar] [CrossRef]
  27. White, L.J.; Sariol, C.A.; Mattocks, M.D.; Wahala, M.P.B.W.; Yingsiwaphat, V.; Collier, M.L.; Whitley, J.; Mikkelsen, R.; Rodriguez, I.V.; Martinez, M.I.; et al. An alphavirus vector-based tetravalent dengue vaccine induces a rapid and protective immune response in macaques that differs qualitatively from immunity induced by live virus infection. J. Virol. 2013, 87, 3409–3424. [Google Scholar] [CrossRef]
  28. Wilson, J.A.; Bray, M.; Bakken, R.; Hart, M.K. Vaccine potential of Ebola virus VP24, VP30, VP35, and VP40 proteins. Virology 2001, 286, 384–390. [Google Scholar] [CrossRef]
  29. Driver, D.A.; Latham, E.M.; Polo, J.M.; Belli, B.A.; Banks, T.A.; Chada, S.; Brumm, D.; Chang, S.M.; Mento, S.J.; Dolly, D.J.; et al. Layered amplification of gene expression with a DNA gene delivery system. Ann. N. Y. Acad. Sci. 1995, 772, 261–264. [Google Scholar] [CrossRef]
  30. Vidalin, O.; Fournillier, A.; Renard, N.; Chen, M.; Depla, E.; Boucreux, D.; Brinster, C.; Baumert, T.; Nakano, I.; Fukuda, Y.; et al. Use of conventional or replicating nucleic acid-based vaccines and recombinant Semliki forest virus-derived particles for the induction of immune responses against hepatitis C virus core and E2 antigens. Virology 2000, 276, 259–270. [Google Scholar] [CrossRef]
  31. Brinster, C.; Chen, M.; Boucreux, D.; Paranhos-Baccala, G.; Liljeström, P.; Lemmonier, F.; Inchasupé, G. Hepatitis C virus non-structural protein 3-specific cellular immune responses following single or combined immunization with DNA or recombinant Semliki Forest virus particles. J. Gen. Virol. 2002, 83, 369–381. [Google Scholar]
  32. Ip, P.P.; Boerma, A.; Regts, J.; Meijerhof, T.; Wilschut, J.; Nijman, H.V.; Daemen, T. Alphavirus-based vaccines encoding non-structural proteins of Hepatitis C virus induce robust and protective T cell responses. Mol. Ther. 2013. [Google Scholar] [CrossRef]
  33. Defang, G.N.; Khetawat, D.; Broder, C.C.; Quinnan, G.V., Jr. Induction of neutralizing antibodies to Hendra and Nipah glycoproteins using a Venezuelan equine encephalitis virus in vivo expression system. Vaccine 2010, 29, 212–220. [Google Scholar] [CrossRef]
  34. Hsu, K.F.; Hung, C.F.; Cheng, W.F.; He, L.; Slater, L.A.; Ling, M.; Wu, T.C. Enhancement of suicidal DNA vaccine potency by linking Mycobacterium tuberculosis heat shock protein 70 to an antigen. Gene Ther. 2001, 8, 376–383. [Google Scholar]
  35. Cheng, W.F.; Hung, C.H.; Chai, C.Y.; Hsu, K.F.; He, L.; Ling, M.; Slater, L.A.; Roden, R.B.; Wu, T.C. Enhancement of sindbis virus self-replicating RNA vaccine potency by linkage of herpes simplex virus type 1 VP22 protein to antigen. J. Virol. 2001, 75, 2368–2376. [Google Scholar] [CrossRef]
  36. Schlesinger, S.; Dubensky, T.W. Alphavirus vectors for gene expression and vaccines. Curr. Opin. Biotechnol. 1999, 10, 434–439. [Google Scholar] [CrossRef]
  37. Hariharan, M.J.; Driver, D.A.; Townsend, K.; Brumm, D.; Polo, J.M.; Belli, B.A.; Catton, D.J.; Hsu, D.; Mittelstaedt, D.; McCormack, J.E.; et al. DNA immunization against herpes simplex virus: enhanced efficacy using a Sindbis virus-based vector. J. Virol. 1998, 72, 950–958. [Google Scholar]
  38. Phenix, K.V.; Wark, K.; Luke, C.J.; Skinner, M.A.; Smyth, J.A.; Mawhinney, K.A.; Todd, D. Recombinant Semliki Forest virus vector exhibits potential for avian virus vaccine development. Vaccine 2001, 19, 3116–3123. [Google Scholar] [CrossRef]
  39. Zhou, X.; Berglund, P.; Rhodes, G.; Parker, S.E.; Jondal, M.; Liljestrom, P. Self-replicating Semliki Forest virus RNA as recombinant vaccine. Vaccine 1994, 12, 1510–1514. [Google Scholar] [CrossRef]
  40. Wolf, A.; Hodneland, K.; Frost, P.; Braaen, S.; Rimstad, E. A hemagglutinin-esterase-expressing salmonid alphavirus replicon protects Atlantic salmon (Salmo salar) against infectious salmon anemia (ISA). Vaccine 2013, 31, 661–669. [Google Scholar] [CrossRef]
  41. Pugachev, K.V.; Mason, P.W.; Shope, R.E.; Frey, T.K. Double-subgenomic Sindbis virus recombinants expressing immunogenic proteins of Japanese encephalitis virus induce significant protection in mice against lethal JEV infection. Virology 1995, 212, 587–594. [Google Scholar]
  42. Pushko, P.; Parker, M.; Ludwig, G.V.; Davis, N.L.; Johnston, R.E.; Smith, J.F. Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: Expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo. Virology 1997, 239, 389–401. [Google Scholar]
  43. Fleeton, M.N.; Liljeström, P.; Sheahan, B.J.; Atkins, G.J. Recombinant Semliki Forest virus particles expressing louping ill virus antigens induce a better protective response than plasmid-based DNA vaccines or an inactivated whole particle vaccine. J. Gen. Virol. 2000, 81, 749–758. [Google Scholar]
  44. Morris-Downes, M.M.; Phenix, K.V.; Smyth, J.; Sheahan, B.J.; Lileqvist, S.; Mooney, D.A.; Liljestrom, P.; Todd, D.; Atkins, G.J. Semliki Forest virus-based vaccines: Persistence, distribution and pathological analysis in two animal systems. Vaccine 2001, 19, 1978–1988. [Google Scholar] [CrossRef]
  45. Hevey, M.; Negley, D.; VanderZanden, L.; Tammariello, R.F.; Geisbert, J.; Smalljohn, C.; Smith, J.F.; Jahrling, P.B.; Smalljohn, A.L. Marburg virus vaccines: Comparing classical and new approaches. Vaccine 2001, 20, 586–593. [Google Scholar] [CrossRef]
  46. Hevey, M.; Negley, D.; Pushko, P.; Smith, J.; Schmaljohn, A. Marburg virus vaccines based upon alphavirus replicons protect guinea pigs and nonhuman primates. Virology 1998, 251, 28–37. [Google Scholar] [CrossRef]
  47. Pasetti, M.F.; Ramirez, K.; Resendiz-Albor, A.; Ulmer, J.; Barry, E.M.; Levine, M.M. Sindbis virus-based measles DNA vaccines protect cotton rats against respiratory measles: Relevance of antibodies, mucosal and systemic antibody-secreting cells, memory B cells, and Th1-type cytokines as correlates of immunity. J. Virol. 2009, 83, 2789–2794. [Google Scholar] [CrossRef]
  48. Pan, C.H.; Greer, C.E.; Hauer, D.; Legg, H.S.; Lee, E.Y.; Bergen, M.J.; Lau, B.; Adams, R.J.; Polo, J.M.; Griffin, D.E. A chimeric alphavirus replicon particle vaccine expressing the hemagglutinin and fusion proteins protects juvenile and infant rhesus macaques from measles. J. Virol. 2010, 84, 3798–3807. [Google Scholar] [CrossRef]
  49. Colombage, G.; Hall, R.; Pavy, M.; Lobigs, M. DNA-based and alphavirus-vectored immunisation with prM and E proteins elicits long-lived and protective immunity against the flavivirus, Murray Valley encephalitis virus. Virology 1998, 250, 151–163. [Google Scholar] [CrossRef]
  50. Harrington, P.R.; Yount, B.; Johnston, R.E.; Davis, N.; Moe, C.; Baris, R.S. Systemic, mucosal, and heterotypic immune induction in mice inoculated with Venezuelan equine encephalitis replicons expressing Norwalk virus-like particles. J. Virol. 2002, 76, 730–742. [Google Scholar] [CrossRef]
  51. Saxena, S.; Dahiya, S.S.; Sonwane, A.A.; Patel, C.L.; Saini, M.; Rai, A.; Gupta, P.K. A sindbis virus replicon-based DNA vaccine encoding the rabies virus glycoprotein elicits immune responses and complete protection in mice from lethal challenge. Vaccine 2008, 26, 6592–6601. [Google Scholar] [CrossRef]
  52. Fleeton, M.N.; Chen, M.; Berglund, P.; Rhodes, G.; Parker, S.E.; Murphy, M.; Atkins, G.J.; Liljestrom, P. Self-replicative RNA vaccines elicit protection against influenza A virus, respiratory syncytial virus, and a tickborne encephalitis virus. J. Infect. Dis. 2001, 183, 1395–1398. [Google Scholar] [CrossRef]
  53. Chen, M.; Hu, K.F.; Rozell, B.; Orvell, C.; Morein, B.; Liljestrom, P. Vaccination with recombinant alphavirus or immune-stimulating complex antigen against respiratory syncytial virus. J. Immunol. 2002, 169, 3208–3216. [Google Scholar] [CrossRef]
  54. Bhardwaj, N.; Heise, M.T.; Ross, T.M. Vaccination with DNA plasmids expressing Gn coupled to C3d or alphavirus replicons expressing gn protects mice against Rift Valley fever virus. PLoS Negl. Trop. Dis. 2010, 4, e725. [Google Scholar] [CrossRef] [Green Version]
  55. Sheahan, T.; Whitmore, A.; Long, K.; Ferris, M.; Rockx, B.; Funkhouser, W.; Donaldson, E.; Gralinski, L.; Collier, M.; Heise, M.; et al. Successful vaccination strategies that protect aged mice from lethal challenge from influenza virus and heterologous severe acute respiratory syndrome coronavirus. J. Virol. 2011, 85, 217–230. [Google Scholar] [CrossRef]
  56. Kamrud, K.I.; Hooper, J.W.; Elgh, F.; Schmaljohn, C.S. Comparison of the protective efficacy of naked DNA, DNA-based Sindbis replicon, and packaged Sindbis replicon vectors expressing Hantavirus structural genes in hamsters. Virology 1999, 263, 209–219. [Google Scholar] [CrossRef]
  57. Berglund, P.; Quesada-Rolander, M.; Putkonen, P.; Biberfeld, G.; Thorstensson, R.; Liljestrom, P. Outcome of immunization of cynomolgus monkeys with recombinant Semliki Forest virus encoding human immunodeficiency virus type 1 envelope protein and challenge with a high dose of SHIV-4 virus. AIDS Res. Hum. Retroviruses 1997, 13, 1487–1495. [Google Scholar] [CrossRef]
  58. Herbert, A.S.; Kuehne, A.I.; Barth, J.F.; Ortiz, R.A.; Nichols, D.K.; Zak, S.E.; Stonier, S.W.; Muhammad, M.A.; Bakken, R.R.; Prugar, L.I.; et al. Venezuelan equine encephalitis virus replicon particle vaccine protects nonhuman primates from intramuscular and aerosol challenge with ebolavirus. J. Virol. 2013, 87, 4952–4964. [Google Scholar] [CrossRef]
  59. Hooper, J.W.; Ferro, A.M.; Golden, J.W.; Silvera, P.; Dudek, J.; Alterson, K.; Custer, M.; Rivers, B.; Morris, J.; Owens, G.; et al. Molecular smallpox vaccine delivered by alphavirus replicons elicits protective immunity in mice and non-human primates. Vaccine 2009, 28, 494–511. [Google Scholar] [CrossRef]
  60. Yang, S.G.; Wo, J.E.; Li, M.W.; Mi, F.F.; Yu, C.B.; Lv, G.L.; Cao, H.C.; Lu, H.F.; Wang, B.H.; Zhu, H.; et al. Construction and cellular immune response induction of HA-based alphavirus replicon vaccines against human-avian influenza (H5N1). Vaccine 2009, 27, 7451–7458. [Google Scholar] [CrossRef]
  61. Xu, R.; Srivastava, I.K.; Greer, C.E.; Zarkikh, I.; Kraft, Z.; Kuller, L.; Polo, J.M.; Barnett, S.W.; Stamatatos, L. Characterization of immune responses elicited in macaques immunized sequentially with chimeric VEE/SIN alphavirus replicon particles expressing SIVGag and/or HIVEnv and with recombinant HIVgp140Env protein. AIDS Res. Hum. Retroviruses 2006, 22, 1022–1030. [Google Scholar]
  62. Barnett, S.W.; Burke, B.; Sun, Y.; Kan, E.; Legg, H.; Lian, Y.; Bost, K.; Zhou, F.; Goodsell, A.; Zur Megede, J.; et al. Antibody-mediated protection against mucosal simian-human immunodeficiency virus challenge of macaques immunized with alphavirus replicon particles and boosted with trimeric envelope glycoprotein in MF59 adjuvant. J. Virol. 2010, 84, 5975–5985. [Google Scholar] [CrossRef]
  63. Wu, Q.; Xu, F.; Fang, L.; Xu, J.; Li, B.; Jiang, Y.; Chen, H.; Xiao, S. Enhanced immunogenicity induced by an alphavirus replicon-based pseudotyped baculovirus vaccine against porcine reproductive and respiratory syndrome virus. J. Virol. Methods 2013, 187, 251–258. [Google Scholar] [CrossRef]
  64. Wolf, A.; Hodneland, K.; Frost, P.; Hoeijmakers, M.; Rimstad, E. Salmonid alphavirus-based replicon vaccine against infectious salmon anemia (ISA): Impact on infectious route and interactions of the replicon vector. Fish Shellfish Immunol. 2014, 36, 383–392. [Google Scholar] [CrossRef]
  65. Xu, C.; Mutoloki, S.; Evensen, O. Superior protection against conferred by inactivated whole virus vaccine over subunit and DNA vaccines against salmonid alphavirus infection in Atlantic salmon (Salmon salar L). Vaccine 2012, 30, 3918–3928. [Google Scholar] [CrossRef]
  66. Andersson, C.; Vasconcelos, N.M.; Sievertzon, M.; Haddad, D.; Liljeqvist, S. Comparative immunization study using RNA and DNA constructs encoding a part of the Plasmodium falciparum antigen Pf332. Scand. J. Immunol. 2001, 54, 117–124. [Google Scholar] [CrossRef]
  67. Kirman, J.R.; Turon, T.; Su, H.; Li, A.; Kraus, C.; Polo, J.M.; Belisle, J.; Morris, S.; Seder, R.A. Enhanced immunogenicity to Mycobacterium tuberculosis by vaccination with an alphavirus plasmid replicon expressing antigen 85A. Infect. Immun. 2003, 71, 575–579. [Google Scholar] [CrossRef]
  68. Li, N.; Yu, Y.Z.; Yu, W.Y.; Sun, Z.W. Enhancement of the immunogenicity of DNA replicon vaccine of Clostridium botulinum neurotoxin serotype A by GM-CSF gene adjuvant. Immunopharmacol. Immunotoxicol. 2011, 33, 211–219. [Google Scholar] [CrossRef]
  69. Cabrera, A.; Sáez, D.; Céspedes, S.; Andrews, E.; Oñate, A. Vaccination with recombinant Semliki Forest virus particles expressing translation initiation factor 3 of Brucella abortus induces protective immunity in BALB/c mice. Immunobiology 2009, 214, 467–474. [Google Scholar] [CrossRef]
  70. Thomas, J.M.; Moen, S.T.; Gnade, B.T.; Vargas-Inchaustegui, D.A.; Foltz, S.M.; Suarez, G.; Heidner, H.W.; König, R.; Chopra, A.K.; Peterson, J.W. Recombinant Sindbis virus vectors designed to express protective antigen of Bacillus anthracis protect animals from anthrax and display synergy with ciprofloxacin. Clin. Vaccine Immunol. 2009, 16, 1696–1699. [Google Scholar] [CrossRef]
  71. Tsuji, M.; Bergmann, C.C.; Takita-Sonoda, Y.; Murata, K.; Rodrigues, E.G.; Nussenzweig, R.S.; Zavala, F. Recombinant Sindbis viruses expressing a cytotoxic T-lymphocyte epitope of a malaria parasite or of influenza virus elicit protection against the corresponding pathogen in mice. J. Virol. 1998, 72, 6907–6910. [Google Scholar]
  72. Krasemann, S.; Jürgens, T.; Bodemer, W. Generation of monoclonal antibodies against prion proteins with an unconventional nucleic acid-based immunization strategy. J. Biotechnol. 1999, 73, 119–129. [Google Scholar] [CrossRef]
  73. Lee, J.S.; Dyas, B.K.; Nystrom, S.S.; Lind, C.M.; Smith, J.F.; Ullrich, R.G. Immune protection against staphylococcal enterotoxin-induced toxic shock by vaccination with a Venezuelan equine encephalitis virus replicon. J. Infect. Dis. 2002, 185, 1192–1196. [Google Scholar] [CrossRef]
  74. Ying, H.; Zaks, T.Z.; Wang, R.F.; Irvine, K.R.; Kammula, U.S.; Marincola, F.M.; Leitner, W.W.; Restifo, N.P. Cancer therapy using a self-replicating RNA vaccine. Nat. Med. 1999, 5, 823–827. [Google Scholar] [CrossRef]
  75. Leitner, W.W.; Hwang, L.N.; deVeer, M.J.; Zhou, A.; Silverman, R.H.; Williams, B.R.; Dubensky, T.B.; Ying, H.; Restifo, N.P. Alphavirus-based DNA vaccine breaks immunological tolerance by activating innate antiviral pathways. Nat. Med. 2003, 9, 33–39. [Google Scholar]
  76. Avogadri, F.; Merghoub, T.; Maughan, M.F.; Hirschhorn-Cymerman, D.; Morris, J.; Ritter, E.; Olmsted, R.; Houghton, A.N.; Wolchok, J.D. Alphavirus replicon particles expressing TRP-2 provide potent therapeutic effect on melanoma through activation of humoral and cellular immunity. PLoS One 2010, 5, e12670. [Google Scholar]
  77. Yamanaka, R.; Zullo, S.A.; Ramsey, J.; Yajima, N.; Tsuchiya, N.; Tanaka, R.; Blaese, M.; Xanthopoulos, K.G. Marked enhancement of antitumor immune responses in mouse brain tumor models by genetically modified dendritic cells producing Semliki Forest virus-mediated interleukin-12. J. Neurosurg. 2002, 97, 611–618. [Google Scholar] [CrossRef]
  78. Yamanaka, R.; Zullo, S.A.; Ramsey, J.; Onodera, M.; Tanaka, R.; Blaese, M.; Xanthopoulos, K.G. Induction of therapeutic antitumor antiangiogenesis by intratumoral injection of genetically engineered endostatin-producing Semliki Forest virus. Cancer Gene Ther. 2001, 8, 796–802. [Google Scholar]
  79. Yamanaka, R.; Zullo, S.A.; Tanaka, R.; Blaese, M.; Xanthopoulos, K.G. Enhancement of antitumor immune response in glioma models in mice by genetically modified dendritic cells pulsed with Semliki forest virus-mediated complementary DNA. J. Neurosurg. 2001, 94, 474–481. [Google Scholar] [CrossRef]
  80. Yamanaka, R.; Xanthopoulos, K.G. Induction of antigen-specific immune responses against malignant brain tumors by intramuscular injection of sindbis DNA encoding gp100 and IL-18. DNA Cell Biol. 2005, 24, 317–324. [Google Scholar] [CrossRef]
  81. Lachman, L.B.; Rao, X.M.; Kremer, R.H.; Ozpolat, B.; Kiriakova, G.; Price, J.E. DNA vaccination against neu reduces breast cancer incidence and metastasis in mice. Cancer Gene Ther. 2001, 8, 259–268. [Google Scholar]
  82. Wang, X.; Wang, J.P.; Rao, X.M.; Price, J.E.; Zhou, H.S.; Lachman, L.B. Prime-boost vaccination with plasmid and adenovirus gene vaccines control HER2/neu+ metastatic breast cancer in mice. Breast Cancer Res. 2005, 7, R580–R588. [Google Scholar] [CrossRef]
  83. Eralp, Y.; Wang, X.; Wang, J.P.; Maughan, M.F.; Polo, J.M.; Lachman, L.B. Doxorubicin and paclitaxel enhance the antitumor efficacy of vaccines directed against HER 2/neu in a murine mammary carcinoma model. Breast Cancer Res. 2004, 6, R275–R283. [Google Scholar] [CrossRef] [Green Version]
  84. Laust, A.K.; Sur, B.W.; Wang, K.; Hubby, B.; Smith, J.F.; Nelson, E.L. VRP immunotherapy targeting neu: Treatment efficacy and evidence for immunoediting in a stringent rat mammary tumor model. Breast Cancer Res. Treat. 2007, 106, 371–382. [Google Scholar] [CrossRef]
  85. Moran, T.P.; Burgents, J.E.; Long, B.; Ferrer, I.; Jaffee, E.M.; Tisch, R.M.; Johnston, R.E.; Serody, J.S. Alphaviral vector-transduced dendritic cells are successful therapeutic vaccines against neu-overexpressing tumors in wild-type mice. Vaccine 2007, 25, 6604–6612. [Google Scholar] [CrossRef]
  86. Daemen, T.; Regts, J.; Holtrop, M.; Wilschut, J. Immunization strategy against cervical cancer involving an alphavirus vector expressing high levels of a stable fusion protein of human papillomavirus 16 E6 and E7. Gene Ther. 2002, 9, 85–94. [Google Scholar]
  87. Daemen, T.; Riezebos-Brilman, A.; Regts, J.; Dontje, B.; van der Zee, A.; Wilschut, J. Superior therapeutic efficacy of alphavirus-mediated immunization against human papilloma virus type 16 antigens in a murine tumour model: Effects of the route of immunization. Antivir. Ther. 2004, 9, 733–742. [Google Scholar]
  88. Cheng, W.F.; Lee, C.N.; Su, Y.N.; Chai, C.Y.; Chang, M.C.; Polo, J.M.; Hung, C.F.; Wu, T.C.; Hsieh, C.Y.; Chen, C.A. Sindbis virus replicon particles encoding calreticulin linked to a tumor antigen generate long-term tumor-specific immunity. Cancer Gene Ther. 2006, 13, 873–885. [Google Scholar]
  89. Velders, M.P.; McElhiney, S.; Cassetti, M.C.; Eiben, G.L.; Higgins, T.; Kovacs, G.R.; Elmishad, A.G.; Kast, W.M.; Smith, L.R. Eradication of established tumors by vaccination with Venezuelan equine encephalitis virus replicon particles delivering human papillomavirus 16 E7 RNA. Cancer Res. 2001, 61, 7861–7867. [Google Scholar]
  90. Riezebos-Brilman, A.; Regts, J.; Chen, M.; Wilschut, J.; Daemen, T. Augmentation of alphavirus vector-induced human papilloma virus-specific immune and anti-tumour responses by co-expression of interleukin-12. Vaccine 2009, 27, 701–707. [Google Scholar] [CrossRef]
  91. Cheng, W.F.; Hung, C.F.; Hsu, K.F.; Chai, C.Y.; He, L.; Polo, J.M.; Slater, L.A.; Ling, M.; Wu, T.C. Cancer immunotherapy using Sindbis virus replicon particles encoding a VP22-antigen fusion. Hum. Gene Ther. 2002, 13, 553–568. [Google Scholar]
  92. Smyth, J.W.; Fleeton, M.N.; Sheahan, B.J.; Atkins, G.J. Treatment of rapidly growing K-BALB and CT26 mouse tumours using Semliki Forest virus and its derived vector. Gene Ther. 2005, 12, 147–159. [Google Scholar]
  93. Lyons, J.A.; Sheahan, B.J.; Galbraith, S.E.; Mehra, R.; Atkins, G.J.; Fleeton, M.N. Inhibition of angiogenesis by a Semliki Forest virus vector expressing VEGFR-2 reduces tumour growth and metastasis in mice. Gene Ther. 2007, 14, 503–513. [Google Scholar]
  94. Yamanaka, R.; Zullo, S.A.; Tanaka, R.; Ramsey, J.; Blaese, M.; Xanthopoulos, K.G. Induction of a therapeutic antitumor immunological response by intratumoral injection of genetically engineered Semliki Forest virus to produce interleukin-12. Neurosurg. Focus 2000, 9, e7. [Google Scholar]
  95. Lundstrom, K. Biology and application of alphaviruses in gene therapy. Gene Ther. 2005, 12, S92–S97. [Google Scholar] [CrossRef]
  96. Quetglas, J.I.; Dubrot, J.; Bezunartea, J.; Sanmamed, M.F.; Hervas-Stubbs, S.; Smerdou, C.; Melero, I. Immunotherapeutic synergy between anti-CD137 mAb and intratumoral administration of a cytopathic Semliki Forest virus encoding IL-12. Mol. Ther. 2012, 20, 1664–1675. [Google Scholar] [CrossRef]
  97. Leslie, M.C.; Zhao, Y.J.; Lachman, L.B.; Hwu, P.; Wu, G.J.; Bar-Eli, M. Immunization against MUC18/MCAM, a novel antigen that drives melanoma invasion and metastasis. Gene Ther. 2007, 14, 316–323. [Google Scholar]
  98. Morse, M.A.; Hobeika, A.C.; Osada, T.; Berglund, P.; Hubby, B.; Negri, S.; Niedzwiecki, G.; Devi, G.R.; Burnett, G.K.; Clay, T.M. An alphavirus vector overcomes the presence of neutralizing antibodies and elevated numbers of Tregs to induce immune responses in humans with advanced cancer. J. Clin. Invest. 2010, 120, 3234–3241. [Google Scholar] [CrossRef]
  99. Slovin, S.F.; Kehoe, M.; Durso, R.; Fernandez, C.; Olson, W.; Gao, J.P.; Israel, R.; Sher, H.I.; Morris, S. A phase I dose escalation trial of vaccine replicon particles (VRP) expressing prostate-specific membrane antigen (PSMA) in subjects with prostate cancer. Vaccine 2013, 31, 943–949. [Google Scholar] [CrossRef]
  100. Durso, R.J.; Andjelic, S.; Gardner, J.P.; Margitich, D.J.; Donovan, G.P.; Arrigale, R.R.; Wang, X.; Maughan, M.F.; Talarico, T.L.; Olmsted, R.A. A novel alphavirus vaccine encoding prostate-specific membrane antigen elicits potent cellular and humoral immune responses. Clin. Cancer Res. 2007, 13, 3999–4008. [Google Scholar] [CrossRef]
  101. Garcia-Hernandez, M.L.; Gray, A.; Hubby, B.; Kast, W.M. In vivo effects of vaccination with six-transmembrane epithelial antigen of the prostate: A candidate antigen for treating prostate cancer. Cancer Res. 2007, 67, 1344–1351. [Google Scholar] [CrossRef]
  102. Garcia-Hernandez, M.L.; Gray, A.; Hubby, B.; Klinger, O.J.; Kast, WM. Prostate stem cell antigen vaccination induces a long-term protective immune response against prostate cancer in the absence of autoimmunity. Cancer Res. 2008, 68, 861–869. [Google Scholar] [CrossRef]
  103. Colmenero, P.; Chen, M.; Castaños-Velez, E.; Liljeström, P.; Jondal, M. Immunotherapy with recombinant SFV-replicons expressing the P815A tumor antigen or IL-12 induces tumor regression. Int. J. Cancer 2002, 98, 554–560. [Google Scholar] [CrossRef]
  104. Ying, H.; Zeng, G.; Black, K.L. Innovative cancer vaccine strategies based on the identification of tumour-associated antigens. BioDrugs 2001, 15, 819–831. [Google Scholar] [CrossRef]
  105. Colmenero, P.; Liljeström, P.; Jondal, M. Induction of P815 tumor immunity by recombinant Semliki Forest virus expressing the P1A gene. Gene Ther. 1999, 6, 1728–1733. [Google Scholar] [CrossRef]
  106. Leitner, W.W.; Ying, H.; Driver, D.A.; Dubensky, T.W.; Restifo, N.P. Enhancement of tumor-specific immune response with plasmid DNA replicon vectors. Cancer Res. 2000, 60, 51–55. [Google Scholar]
  107. Daemen, T.; Pries, F.; Bungener, L.; Kraak, M.; Regts, J.; Wilschut, J. Genetic immunization against cervical carcinoma: induction of cytotoxic T lymphocyte activity with a recombinant alphavirus vector expressing human papillomavirus type 16 E6 and E7. Gene Ther. 2000, 7, 1859–1866. [Google Scholar]
  108. Cheng, W.F.; Hung, C.F.; Hsu, K.F.; Chai, C.Y.; He, L.; Ling, M.; Slater, L.A.; Roden, R.B.; Wu, T.C. Enhancement of sindbis virus self-replicating RNA vaccine potency by targeting antigen to endosomal/lysosomal compartments. Hum. Gene Ther. 2001, 12, 235–252. [Google Scholar]
  109. Eiben, G.L.; Velders, M.P.; Schreiber, H.; Cassetti, M.C.; Pullen, J.K.; Smith, L.R.; Kast, W.M. Establishment of an HLA-A*0201 human papillomavirus type 16 tumor model to determine the efficacy of vaccination strategies in HLA-A*0201 transgenic mice. Cancer Res. 2002, 62, 5792–5799. [Google Scholar]
  110. Cassetti, M.C.; McElhiney, S.P.; Shahabi, V.; Pullen, J.K.; Le, Poole, I.; Eiben, G.L.; Smith, L.R.; Kast, W.M. Antitumor efficacy of Venezuelan equine encephalitis virus replicon particles encoding mutated HPV16 E6 and E7 genes. Vaccine 2004, 22, 520–527. [Google Scholar] [CrossRef]
  111. Yamanaka, R.; Tsuchiya, N.; Yajima, N.; Honma, J.; Hasegawa, H.; Tanaka, R.; Ramsey, J.; Blaese, R.M.; Xanthopoulos, K.G. Induction of an antitumor immunological response by an intratumoral injection of dendritic cells pulsed with genetically engineered Semliki Forest virus to produce interleukin-18 combined with the systemic administration of interleukin-12. J. Neurosurg. 2003, 99, 746–753. [Google Scholar] [CrossRef]
  112. Bennett, A.M.; Elvin, S.J.; Wright, A.J.; Jones, S.M.; Phillpotts, R.J. An immunological profile of Balb/c mice protected from airborne challenge following vaccination with a live attenuated Venezuelan equine encephalitis virus vaccine. Vaccine 2000, 19, 337–347. [Google Scholar] [CrossRef]
  113. Hart, M.K.; Caswell-Stephan, K.; Bakken, R.; Tammariello, R.; Pratt, W.; Davis, N.; Johnston, R.E.; Smith, J.; Steele, K. Improved mucosal protection against Venezuelan equine encephalitis virus is induced by the molecularly defined, live-attenuated V3526 vaccine candidate. Vaccine 2000, 18, 3067–3075. [Google Scholar] [CrossRef]
  114. Schoepp, R.J.; Smith, J.F.; Parker, M.D. Recombinant chimeric western and eastern equine encephalitis viruses as potential vaccine candidates. Virology 2002, 302, 299–309. [Google Scholar] [CrossRef]
  115. Edelman, R.; Tacket, C.O.; Wasserman, S.S.; Bodison, S.A.; Perry, J.G.; Mangiafico, J.A. Phase II safety and immunogenicity study of live chikungunya virus vaccine TSI-GSD-218. Am. J. Trop. Med. Hyg. 2000, 62, 681–685. [Google Scholar]
  116. Mallilankaraman, K.; Shedlock, D.J.; Bao, H.; Kawalekar, O.U.; Fagone, P.; Ramanathan, A.A.; Ferraro, B. A DNA vaccine against chikungunya virus is protective in mice and induces neutralizing antibodies in mice and nonhuman primates. PLoS Negl. Trop. Dis. 2011, 5, e928. [Google Scholar] [CrossRef]
  117. Kim, D.Y.; Atasheva, S.; Foy, N.J.; Wang, E.; Frolova, E.I.; Weaver, S.; Frolov, I. Design of chimeric alphaviruses with a programmed, attenuated, cell type-restricted phenotype. J. Virol. 2011, 85, 4363–4376. [Google Scholar]
  118. Kramer, R.M.; Zeng, Y.; Sahni, N.; Kueltzo, L.A.; Schwartz, R.M.; Srivastava, I.K.; Crane, L.; Joshi, S.B.; Volkin, D.B.; Middaugh, C.R. Development of a stable virus-like particle vaccine formulation against Chikungunya virus and investigation of the effects of polyanions. J. Pharm. Sci. 2013, 102, 4305–4314. [Google Scholar] [CrossRef]
  119. Dash, P.K.; Tiwari, M.; Santhosh, S.R.; Parida, M.; Lakshmana Rao, P.V. RNA interference mediated inhibition of Chikungunya virus replication in mammalian cells. Biochem. Biophys. Res. Commun. 2008, 376, 718–722. [Google Scholar] [CrossRef]
  120. Kamrud, K.I.; Coffield, V.M.; Owens, G.; Goodman, C.; Alterson, K.; Custer, M.; Murphy, M.A.; Lewis, W.; Timberlake, S.; Wansley, E.K. In vitro and in vivo characterization of microRNA-targeted alphavirus replicon and helper RNAs. J. Virol. 2010, 84, 7713–7725. [Google Scholar] [CrossRef]
  121. Elvin, S.J.; Bennett, A.M.; Phillpotts, R.J. Role for mucosal immune responses and cell-mediated immune functions in protection from airborne challenge with Venezuelan equine encephalitis virus. J. Med. Virol. 2002, 67, 384–393. [Google Scholar] [CrossRef]
  122. Dupuy, L.C.; Richards, M.J.; Reed, D.S.; Schmaljohn, C.S. Immunogenicity and protective efficacy of a DNA vaccine against Venezuelan equine encephalitis virus aerosol challenge in nonhuman primates. Vaccine 2010, 28, 7345–7350. [Google Scholar] [CrossRef]
  123. Darwin, J.R.; Kenney, J.L.; Weaver, S.C. Transmission potential of two chimeric Chikungunya vaccine candidates in the urban mosquito vectors, Aedes aegypti and Ae. albopictus. Am. J. Trop. Med. Hyg. 2011, 84, 1012–1015. [Google Scholar] [CrossRef]
  124. Bhomia, M.; Sharma, A.; Gayen, M.; Gupta, P.; Maheshwari, R.K. Artificial microRNAs can effectively inhibit replication of Venezuelan equine encephalitis virus. Antivir. Res. 2013, 100, 429–434. [Google Scholar] [CrossRef]
  125. Prow, T.W.; Chen, X.; Prow, N.A.; Fernando, G.J.; Tan, C.S.; Raphael, A.P.; Chang, D.; Ruutu, M.D.; Jenkins, D.W.; Pyke, A. Nanopatch-targeted skin vaccination against West Nile Virus and Chikungunya virus in mice. Small 2010, 6, 1776–1784. [Google Scholar] [CrossRef]
  126. Berglund, P.; Sjöberg, M.; Garoff, H.; Atkins, G.J.; Sheahan, B.J.; Liljestrom, P. Semliki Forest virus expression system: Production of conditionally infectious recombinant particles. Biotechnology 1993, 11, 916–920. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Lundstrom, K. Alphavirus-Based Vaccines. Viruses 2014, 6, 2392-2415. https://doi.org/10.3390/v6062392

AMA Style

Lundstrom K. Alphavirus-Based Vaccines. Viruses. 2014; 6(6):2392-2415. https://doi.org/10.3390/v6062392

Chicago/Turabian Style

Lundstrom, Kenneth. 2014. "Alphavirus-Based Vaccines" Viruses 6, no. 6: 2392-2415. https://doi.org/10.3390/v6062392

Article Metrics

Back to TopTop