Next Article in Journal
Angiotensin-(1-7) Improves Integrated Cardiometabolic Function in Aged Mice
Next Article in Special Issue
Adenovirus Receptor Expression in Cancer and Its Multifaceted Role in Oncolytic Adenovirus Therapy
Previous Article in Journal
Non-Thermal Plasma Application in Tumor-Bearing Mice Induces Increase of Serum HMGB1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Self-Amplifying RNA Viruses as RNA Vaccines

by
Kenneth Lundstrom
PanTherapeutics, CH1095 Lutry, Switzerland
Int. J. Mol. Sci. 2020, 21(14), 5130; https://doi.org/10.3390/ijms21145130
Submission received: 30 June 2020 / Revised: 15 July 2020 / Accepted: 16 July 2020 / Published: 20 July 2020
(This article belongs to the Special Issue Oncolytic Virotherapy)

Abstract

:
Single-stranded RNA viruses such as alphaviruses, flaviviruses, measles viruses and rhabdoviruses are characterized by their capacity of highly efficient self-amplification of RNA in host cells, which make them attractive vehicles for vaccine development. Particularly, alphaviruses and flaviviruses can be administered as recombinant particles, layered DNA/RNA plasmid vectors carrying the RNA replicon and even RNA replicon molecules. Self-amplifying RNA viral vectors have been used for high level expression of viral and tumor antigens, which in immunization studies have elicited strong cellular and humoral immune responses in animal models. Vaccination has provided protection against challenges with lethal doses of viral pathogens and tumor cells. Moreover, clinical trials have demonstrated safe application of RNA viral vectors and even promising results in rhabdovirus-based phase III trials on an Ebola virus vaccine. Preclinical and clinical applications of self-amplifying RNA viral vectors have proven efficient for vaccine development and due to the presence of RNA replicons, amplification of RNA in host cells will generate superior immune responses with significantly reduced amounts of RNA delivered. The need for novel and efficient vaccines has become even more evident due to the global COVID-19 pandemic, which has further highlighted the urgency in challenging emerging diseases.

1. Introduction

RNA-based vaccines have become potentially promising as alternative approaches to conventional vaccine development [1]. In this review, emphasis has been placed on tumor vaccines, but description of vaccines against pathogenic viruses has also been included as more progress has been made in this area, which can serve as encouragement for treatment and prevention of various types of cancer. Particularly, the utilization of mRNA due to its high potency, rapid production and development and safe administration has received plenty of attention. However, progress has been hampered by issues related to instability and low efficiency of in vivo delivery. Recent improvements in the field addressing optimization of mRNA translation and stability, modulation of immunogenicity and progress in mRNA vaccine delivery have contributed to vaccine platforms against infectious diseases and different types of cancers, showing encouraging results in both animal models and clinical trials. In this context, an mRNA vaccine targeting the rabies glycoprotein provided protection against lethal challenges with rabies virus in immunized mice and pigs [2]. In another study, mice and primates immunized with 30 μg and 50 μg of liposome-encapsulated nanoparticles containing mRNA for Zika virus (ZIKV) membrane and envelope glycoproteins, respectively, were protected against ZIKV challenges [3]. Moreover, lipid nanoparticle-encapsulated mRNA expressing influenza A virus hemagglutinin (HA) protected mice from lethal influenza virus challenges and reduced viral titers in ferret lungs [4]. Initial phase I clinical trial results demonstrated robust prophylactic immunity in humans with mild or moderate adverse events only. In another phase I clinical trial, mRNA encoding rabies glycoprotein was administered to healthy volunteers showing safety and a reasonable tolerability profile [5]. Functional antibodies against the viral antigen could be induced and boosted when administered with a needle-free device but not when a needle-syringe was used. In the context of mRNA-based vaccines, self-replicating RNA viruses present an interesting and attractive alternative as described below [6].
The common feature of self-amplifying RNA viruses is their single-stranded RNA (ssRNA) genome embedded in a capsid and envelope protein structure [6]. In the case of alphaviruses and flaviviruses, the ssRNA genome is of positive polarity in contrast to measles viruses (MVs) and rhabdoviruses, which carry a negative sense ssRNA genome. In any case, the RNA genome of self-amplifying RNA viruses can act directly in the cytoplasm without any need of nucleic acid delivery to the nucleus. In the case of positive polarity, the translation can be initiated directly from the incoming ssRNA genome [7], whereas negative sense RNA molecules require the generation of a positive strand RNA template [8]. All self-amplifying RNA viruses initially express their nonstructural genes resulting in the formation of the RNA replication complex (RNA replicon), responsible for extreme RNA replication in infected host cells [7]. It has been estimated that 200,000 copies of RNA are made from a single RNA molecule, providing together with strong subgenomic promoters the basis for extremely high expression levels of viral proteins. This feature has been taken advantage of in expression vectors engineered from self-amplifying RNA viruses, which have been applied for mammalian and non-mammalian cell lines, primary cells and in vivo [9]. Moreover, self-amplifying RNA virus vectors have been used for vaccine development, targeting both infectious diseases and different types of cancers. In this review, the basic function of self-replicating RNA virus vectors is described, and various preclinical and clinical applications are presented.

2. Self-Amplifying RNA Virus-Based Expression

Although efficient expression systems have been developed for alphaviruses, flaviviruses, MVs and rhabdoviruses, the differences in the polarity of the ssRNA genome have required alternative engineering. In the case of alphaviruses and flaviviruses, the procedure is straightforward, whereas MVs and rhabdoviruses require the utilization of packaging cell lines and reverse genetics. For simplicity, the focus is here on the illustration of the Semliki Forest virus (SFV)-based expression system, one of the most commonly applied alphavirus expression systems (Figure 1).
Alphaviruses belong to the family of Togaviruses [7]. Particularly, SFV [10], Sindbis virus (SIN) [11] and Venezuelan equine encephalitis virus (VEE) [12] have been used for the engineering of expression vector systems. In this context, engineering of an expression vector carrying the SFV nonstructural protein genes (nsP1-4), where the gene of interest can be inserted downstream of the strong 26S subgenomic promoter allows in vitro transcribed RNA to be directly translated in cell lines and in vivo (Figure 1). Alternatively, replacement of the SP6 or T7 RNA polymerase promoter upstream of the nsP1-4 region with a cytomegalovirus (CMV) promoter permits direct use of plasmid DNA in vitro or in vivo [13]. Another possibility is to co-transfect in vitro transcribed RNA from the expression vector and a helper vector expressing the SFV structural protein genes into mammalian host cells for production of replication deficient SFV particles. The reason for replication deficiency relates to the presence of the packaging signal in the nsP2 region of the expression vector, which prevents the packaging of the RNA from the helper vector [14]. Additionally, to reduce the formation of any replication proficient SFV particles, the second generation pSFV-Helper 2 vector was engineered [15]. Furthermore, for total elimination of RNA recombination and generation of replication proficient SFV particles a two-helper system with the capsid and spike protein genes on separated vectors was applied [16]. Finally, a full-length SFV genome with a second 26S subgenomic promoter and gene of interest introduced either downstream of the nsP genes or the structural genes can be applied, keeping in mind that replication proficient particles will be generated, obviously enhancing biosafety risks. In any case, the options and flexibility are excellent as alphavirus vectors can be used for vaccine development in the form of naked RNA replicons, DNA plasmid vectors and recombinant viral particles as described in more detail in the next sections.
Due to the positive polarity of the flavivirus RNA genome, similar expression vector systems to SFV have been engineered for Kunjin (KUN) virus-based delivery of RNA replicons, DNA plasmids and recombinant KUN particles [17]. However, foreign genes are introduced between the C20 core protein and the E22 envelope protein for expression as a large polyprotein, which will be processed into individual proteins. Introduction of an FMDV-2A protease sequence in the KUN vector will allow removal of remaining KUN flanking regions from the recombinant product [18]. Virus production from KUN vectors has been facilitated by the engineering of a packaging cell line [19]. In addition to KUN vectors, expression systems for West Nile virus [20,21], yellow fever virus [22,23], dengue virus [24,25] and tick-borne encephalitis [26,27] have been engineered.
In the context of MV vectors, the negative polarity of the ssRNA genome has demanded the application of adequate packaging systems [28] and reverse genetics [29]. The gene of interest can be introduced into the MV expression vector between the phosphoprotein (P) gene and the matrix protein (M) gene or between the HA gene and the large protein (L). Recombinant MV particles are generated by transfection of a helper cell line with plasmids carrying MV constructs and the MV polymerase gene, followed by harvesting of MV particles three days later.
In case of rhabdoviruses, expression vectors have been engineered for both rabies virus (RBAV) [30] and vesicular stomatitis virus (VSV) [31] using reverse genetics based on a recombinant vaccinia virus vector. Engineering of VSV vectors with the N, P and L genes inserted downstream of a T7 RNA polymerase promoter and an internal ribosome entry site (IRES) allowed recovery of DNA-transfected VSV particles in a vaccinia-free system [32]. Similar vectors have been constructed for RABV by insertion of the gene of interest between the RABV N and P genes [33]. A vaccinia-free reverse genetics system has also been engineered for RABV [34].

3. Vaccines Against Infectious Diseases

There are numerous examples of preclinical immunization studies against infectious diseases conducted with self-replicating RNA virus vectors, of which some selected examples are presented in Table 1. In the context of Dengue virus (DENV), a single immunization with a tetravalent vaccine based on VEE particles expressing the ectodomain of the DENV E protein (E85) elicited neutralizing antibodies and T cell responses for each of four serotypes tested [35]. Although the immune response was weaker in neonatal BALB/c mice than in adult animals, protective immunity against lethal challenges was obtained after a single vaccine administration. In another approach, the domain III of DENV envelope protein 2 (DV2) was expressed from an MV vector [36]. Immunization of MV-susceptible mice induced robust neutralizing antibodies. In another study on DENV, mice immunized with MV vectors expressing domain II of DV1-4, generated neutralizing antibodies and were protected against four serotypes of DENV [37]. Another flavivirus, ZIKV, was evaluated in a two-vial strategy of highly stable nanostructured lipid carriers (NLCs) and replicon RNA [38]. In this approach, the codon-optimized prM and E genes were introduced into a VEE replicon vector and NLC-RNA complexes, which provided protection against challenges with lethal ZIKV in mice subjected to a single immunization. In another study, a chimeric VSV vector expressing both the membrane-envelope (ME) glycoproteins of ZIKV and Chikungunya (CHIKV) envelope polyprotein (E3-E2-6K-E1) elicited neutralizing antibodies against both ZIKV and CHIKV in wild-type mice and in interferon-receptor-deficient A129 mice [39]. Protection against lethal challenges of both CHIKV and ZIKV was obtained after a single vaccination.
Filoviruses have for obvious reasons received a lot of attention due to such members as Ebola virus (EBOV), presenting urgent needs for the development of novel vaccines especially during the Ebola virus disease (EVD) outbreak in 2014–2016. In this context, KUN virus-like particles (VLPs) expressing the EBOV glycoprotein with the D637L mutation (GP/D637L), which by displaying a cleavage site improved cleavability and shedding of GP, were subcutaneously injected into nonhuman primates [40]. Complete protection was observed in three immunized primates, while one vaccinated and all control animals died. In another approach, it was demonstrated that a recombinant VSV vector expressing the EBOV glycoprotein provided complete protection of macaques challenged with the West African EBOV-Makona strain [41]. A single dose given only seven and three days before challenge resulted in complete and partial protection, respectively. Additionally, VSV-based immunization with EBOV GP provided protection against lethal challenges with three EBOV strains in immunized nonhuman primates [42]. Furthermore, a vaccination study against another filovirus, Marburg virus (MARV), showed protection in nonhuman primates after immunization with VSV-MARV-GP particles [42]. Vaccination against another filovirus, Sudan virus (SUDV), by a single intramuscular administration of VEE particles expressing the SUDV GP provided complete protection in cynomolgus macaques [43]. The immunization did not fully protect the macaques from intramuscular challenges with EBOV. However, intramuscular co-immunization with VEE-SUDV-GP and VEE-EBOV-GP led to complete protection against challenges with both SUDV and EBOV. The intramuscular immunization also resulted in complete protection against challenges with aerosolized SUDV, although two vaccinations were required to reach efficacy. In another study, it was demonstrated that C57BL/6 mice immunized with VEE particles expressing EBOV nucleoprotein (NP) survived challenges with EBOV [44]. Furthermore, immunizations with VEE-EBOV-GP and VEE-EBOV-NP VLPs were evaluated in BALB/c mice and guinea pigs [45]. Vaccination with VEE-EBOV-NP particles showed protection in mice, but not in guinea pigs. In contrast, co-immunization with VEE particles expressing both EBOV-GP and -NP rendered both mice and guinea pigs resistant to EBOV challenges. Recently, immunization with SFV DNA replicons co-expressing EBOV-GP and -VP40, generated both binding and neutralizing antibodies, which were superior to those elicited by a Modified Vaccinia virus Ankara (MVA) vaccine [46].
In the context of arenaviruses, development of Lassa virus (LASV) vaccines has been initiated by expressing LASV glycoproteins from VSV vectors [47]. Immunization studies in guinea pigs and macaques provided protection against LASV originating from Liberia, Mali and Nigeria. In another study, the wild-type LASV glycoprotein (GPCwt) and a non-cleavable C-terminally deleted modification (ΔGPfib) expressed from individual VEE 26S subgenomic promoters were immunogenic and protective in immunized mice [48]. The vaccination induced cross-reactive multifunctional T cell responses. Furthermore, a single-cycle replication system for LASV-based replicon particles devoid of the essential LASV GPC gene but supported by a Vero cell line providing the expressed GPC, enabled vaccine propagation [49]. Immunized guinea pigs were protected against challenges with lethal doses of LASV. VEE VLPs have also been applied for expression of Junin virus (JUNV) GPC and Machupo (MACV) GPC eliciting humoral immune responses, which correlated with complete protection against challenges with JUNV and MACV, respectively [50].
Lentiviruses have also been commonly chosen as vaccine targets, obviously because of human immunodeficiency virus (HIV) causing AIDS. For instance, recombinant SFV particles expressing HIV-1 envelope glycoprotein (Env) were applied for immunization studies in mice in comparison to a DNA vaccine and recombinant Env gp160 protein [51]. The highest antibody titers were observed in animals immunized with SFV particles. Furthermore, an SFV RNA replicon vector carrying the HIV-1 Env gene was applied for intramuscular injection into mice [52]. The immunization resulted in induction of Env-specific antibody responses in four out of five mice. Moreover, the monoclonal antibody 12H2 directed against gp41 was produced. In another study, mice were immunized with recombinant SFV particles expressing the Indian HIV-1C env/gag/polRT genes resulting in significant T cell immune responses [53]. The immune responses were stronger for SFV VLPs than RNA replicons. In another approach, BALB/c mice were immunized with plasmid DNA-based SFV vectors expressing Env and a Gag-Pol-Nef fusion protein [54] Initially, immunization studies revealed that lower HIV-specific T cell and IgG responses were obtained for 0.2 μg of SFV DNA compared to a dose of 10 μg, but no differences in immune responses between the doses were detected after boosting with MVA or HIV gp40 protein. It was also confirmed that immunization with the lower dose of SFV DNA elicited superior immune responses in comparison to MVA or HIV gp40 alone. In RNA-based approaches, RNA replicons have been delivered by nanoparticles [55,56]. In this context, the HIV-1 glycoprotein 140 (TV1 gp140) was introduced into the VEE replicon vector containing the 3′ end untranslated region and the packaging signal of SIN and formulated with a cationic nano-emulsion consisting of squalene, 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) and sorbitan trioleate [55]. The encapsulated RNA replicons intramuscularly administered to rhesus macaques elicited potent cellular immune responses, which were stronger than those observed for VEE VLPs. It was further demonstrated that doses of only 50 µg of encapsulated RNA replicons were safe and provided strong immune responses in primates. In another study, alphavirus RNA replicons expressing the HIV glycoprotein 120 (gp120) were encapsulated in DOTAP-based lipid nanoparticles [56]. Intramuscular administration generated high levels of recombinant protein expression for 30 days in mice compared to only brief and low levels expression of modified conventional mRNA. A single injection resulted in high titers of gp120-specific antibodies. Simian immunodeficiency virus (SIV) has also been subjected to vaccine development. For instance, KUN-based expression of four SIV gag constructs designed for the wild-type SIV gag gene (WT), an RNA-optimized nucleic acid sequence (DX), a human codon-optimized SIV gag gene (OPT) and wild-type matrix and capsid from gag-linked in-frame to reverse transcriptase from pol (Gag-pol) [57]. The Gag-pol vaccine was superior to WT, DX and OPT related to immune response induction and protection of immunized mice against SIV challenges. In another approach, macaques were immunized with SFV and MVA vectors expressing SIV env, gag-pol, nef, rev and tat, which resulted in only low or undetectable cytotoxic T cell responses [58]. In contrast, when macaques were first vaccinated with SFV and boosted with MVA, enhanced antibody responses and high T cell proliferation responses were observed. However, the immunizations did not provide protection against challenges with SIV three months after the last vaccination. In another study in rhesus macaques, immunization was conducted with two injections of VSV particles followed by a single administration of SFV particles expressing SIVsmE660 gag-env alone or in combination with rhesus granular macrophage-colony stimulating factor (GM-CSF) [59]. Two out six animals immunized with SIV gag-pol showed no protection, whereas the combination with GM-CSF resulted in four out of six macaques being infected, suggesting that GM-CSF abrogated protection. Not surprisingly, all animals in the control group receiving influenza virus hemagglutinin protein were infected by SIV.
Due to the recurrent annual threats of epidemics, influenza viruses have been sought after targets for vaccine development. In this context, intravascular administration of SFV particles expressing the influenza virus nucleoprotein (NP) resulted in systemic immune responses in mice [60]. Furthermore, intranasal delivery elicited mucosal immune responses. In another study, VEE particles expressing the hemagglutinin (HA) gene from the Hong Kong influenza A isolate (A/HK/156/97) provided partial protection against lethal challenges of influenza virus after immunization of chicken of one day of age and complete protection of two weeks old chicken [61]. Additionally, 10 µg of SFV RNA replicons expressing the influenza HA gene induced significant antibody titers after a single intramuscular administration [62]. Moreover, 90% of BALB/c mice immunized twice with SFV-HA RNA replicons were protected against influenza virus challenges, while 90% of control mice died. In comparison to synthetic mRNA, VEE RNA expressing the influenza HA gene required only 1.25 µg self-replicating RNA compared to 80 µg of synthetic mRNA to achieve protection against influenza strains H1N1, H3N2 and B in immunized BALB/c mice [63]. In another approach, the truncated derivative of VEE targeting dendritic cells was used as an adjuvant for inactivated influenza virus (iFlu) [64]. The truncated VEE lacks five nucleotides between the nsP4 stop codon and the beginning of the 3′ end untranslated region (UTR). Immunization of neonatal BALB/c mice elicited strong innate immune responses demonstrating significantly higher and sustained influenza virus-specific IgG antibodies compared to mice immunized with antigen only. Recently, liposome-encapsulation of RNA replicons of the flavivirus classical swine fever virus (CSFV) expressing influenza virus NP elicited immune responses both in vitro and in vivo [65].
The current timely coronavirus pandemic has cast a special light on the need of novel vaccines [75,76]. Naturally, vaccine development against SARS-CoV-2, the virus responsible for the COVID-19 pandemic, is currently at an early stage of development although as of May 27, 2020, 115 preclinical studies and 10 clinical trials are in progress according to the WHO [77]. These studies focus on live attenuated viruses, protein subunit vaccines, adenovirus-based delivery and mRNA and mRNA nanoparticles complexes. Two preclinical studies using MV vectors are included. However, previous studies have been conducted for other coronaviruses. In this context, VEE replicon particles were applied for the expression of SARS-CoV S and N proteins from the Urbani strain [66]. Immunization of mice with VEE-SARS-CoV S provided complete short- and long-term protection against challenges with homologues SARS-CoV strains in both young and senescent mice. In contrast, no protection was obtained after immunization with VEE-SARS-CoV N. Related to heterologous SARS-CoV strains, the chimeric icGDO3-S virus encoding a synthetic S gene of the most genetically divergent human GDO3 strain showed strong resistance to neutralization with antisera directed against the Urbani strain. Despite that, immunization with VEE-SARS-CoV S resulted in complete short-term protection against icGDO3-S challenges in young mice, but not in senescent animals. The age-related protection in mice was addressed in a study, where mice were immunized with VEE-SARS-CoV S particles packaged with either attenuated (3014) or wild-type (3000) VEE glycoproteins [67]. It was demonstrated that aged animals immunized with the VEE 3000-based vaccine were protected against SARS-CoV, while immunization with the VEE 3014-based vaccine did not result in survival after the challenge. Furthermore, superior protection was observed in a lethal influenza challenge model. In the context of Middle East respiratory syndrome coronavirus (MERS-CoV), the MERS-S full-length and a soluble variant (MERS-solS) were expressed from a replication-proficient MV vector, which both elicited robust MV- and MERS-CoV-neutralizing antibodies in mice [68]. Vaccination also provided protection against challenges with MERS-CoV.
Among hepatotropic viruses, hepatitis B virus surface antigen (HBsAg) has been expressed from three MV vectors generating humoral responses in MV-susceptible genetically modified mice and rhesus monkeys [69]. However, the differences in HBsAg levels from the MV vectors elicited significantly different HBsAg antibody levels. Moreover, only the MV vector with the highest HBsAg levels showed protection and even so in two out of four animals. In another approach, SFV RNA replicons were applied for the expression of the HBV middle surface envelope glycoprotein (MHB) and the core antigen (HBcAg) [70]. The SFV-G-MHB and SFV-G-HBcAg RNAs were packaged into an VSV G envelope and subjected to immunization of mice. The CD8+ T cell responses in mice were greater in magnitude and broader in specificity compared to recombinant protein- and DNA-based vaccines. A single injection with the SFV-G-MHB vaccine protected mice from HBV challenges. In contrast, immunization with SFV-G-HBcAg failed to provide protection. In another study, BALB/c mice were intravenously immunized with 107 and 108 SFV particles expressing the HBV small surface (S) protein, which elicited antibodies reacting with both yeast-derived S antigen and patient-derived S antigen [71]. Immunization with SFV particles expressing pre-S1 resulted in production of pre-S1 and S-specific IgG, and pre-S1 antibodies completely neutralized HBV infectivity.
Although commonly used as delivery vectors, alphavirus replicons have also shown potential as vaccine candidates, particularly for VEE, western equine encephalitis virus (WEE) and eastern equine encephalitis virus (EEE) [72]. In each replicon vector, the furin cleavage site between the E2 and E3 envelope proteins was deleted to prevent cleavage of the p62 precursor and generation of infectious particles, followed by RNA in vitro transcription and generation of VLPs with a two-helper system [78] by co-electroporation of replicon and capsid and glycoprotein (GP) helpers. Immunization in combination (VEE/WEE/EEE) or individually elicited strong neutralizing antibody responses in mice. Moreover, mice were protected against subcutaneous or aerosol challenges with VEE, WEE and EEE for 12 months. Combination immunization with VEE, WEE and EEE generated robust neutralizing antibody responses in macaques, and also provided good protection against aerosol challenges with an epizootic VEE virus and a North American variety of EEE, respectively. In contrast, both the WEE replicon and the VEE-WEE-EEE combination elicited poor neutralizing antibodies and weak protection against WEE challenges. In another approach, the genome of the attenuated VEE V4020 strain was rearranged to include two subgenomic promoters for the expression of the capsid and glycoprotein genes, respectively [79]. The full-length V4020 RNA was expressed from the plasmid pMG4020 from a CMV promoter, which elicited high titers of neutralizing antibodies in BALB/c mice. Challenges with wild-type VEE did not kill the vaccinated mice, while all control mice died. In another study the VEE V4020 vaccine was evaluated in intramuscularly immunized cynomolgus macaques [80]. The vaccination induced high levels of virus-neutralizing antibodies and primates were protected against aerosol challenges with wild-type VEE.

4. Vaccines Against Cancer

Cancer therapy and immunotherapy are areas of great potential for self-amplifying RNA virus vectors. Not surprisingly, most types of cancers have been targeted in preclinical settings (Table 2). In the case of brain tumors, a retargeted MV vector with CD46 and signaling lymphocyte activation molecule (SLAM) ablating mutations in the HA protein combined with a single-chain antibody against the epidermal growth factor receptor (EGFR) showed potent antitumor activity against EGFR- or EGFRvIII-overexpressing primary glioblastoma multiforme (GBM) cell lines [79]. Intratumoral administration in orthotopic GBM12 xenografts resulted in tumor regression and significantly prolonged survival. Alphavirus vectors have also been applied for cancer therapy. In this context, SFV vectors expressing endostatin in comparison to SFV-LacZ and retrovirus-based endostatin expression provided superior inhibition of tumor growth and reduction in intratumoral vascularization in mice with B16 brain tumor xenografts [80]. Moreover, dendritic cells (DCs) transduced with SFV particles expressing interleukin-18 (IL-18) were administered intratumorally into mice bearing B16 brain tumors in combination with systemic administration of IL-12 [81]. The SFV-based treatment resulted in enhanced induction of T helper type 1 responses from tumor specific CD4+ and CD8+ T cells and natural killers and antitumor immunity. In another study, neuron-targeting micro-RNA sequences (miRT124) introduced into the SFV4 strain generated increased oncolytic potency in human glioblastoma cell lines and resulted in virus replication in tumors, significant inhibition of tumor growth and prolonged survival after intraperitoneal administration in C57BL/6 mice implanted with CT-2A orthotopic gliomas [82]. A chimeric vector was engineered, where the VSV G protein was replaced by the CHIKV envelope proteins (E3-E2-6K-E1) [83]. The VSVΔG-CHIKV vector selectively infected and eliminated tumors and the survival of tumor-bearing mice was extended from 40 to 100 days. Moreover, the chimeric virus also targeted intracranial xenografts from melanoma patients and intravenous administration only infected tumor cells and selectively infected mouse melanoma cells within the brain.
In the context of brain cancer, an MV vector based on the Edmonston strain expressing the carcinoembryonic antigen (CEA) generated cytotoxic effects in breast cancer cell lines and provided significant delay in tumor growth and prolonged survival in BALB/c mice [84]. In another study, the rat HER2/neu gene expressed from SIN and Adenovirus (Ad) vectors inhibited A2L2 tumor cell growth in immunized mice [85]. On the other hand, if immunization took place two days after tumor challenge it was ineffective. However, a prime-boost protocol of SIN-neu immunization followed by Ad-neu administration significantly improved survival rates in mice, which were intravenously challenged with tumor cells. In another study on the A2L2 breast cancer cell line, SIN DNA replicons expressing HER2/neu elicited strong antibody responses and immunized mice showed reduced tumor incidence and reduced tumor mass [86]. Furthermore, intradermal immunization provided tumor protection and required 80% less plasmid DNA compared to conventional DNA plasmids. Additionally, the incidence of lung metastasis was also reduced in vaccinated mice. Related to DCs, VEE particles expressing a truncated neu gene elicited strong neu-specific CD8+ T cell and anti-neu IgG responses in mice [87]. Furthermore, a single immunization with DCs transduced with VEE-neuΔ generated regression of large neu tumors in mice. In another study, SFV particles and RNA replicons were administered to tumor-free mice and mice implanted with 4T1 mammary tumors for distribution studies of the luciferase reporter gene [88]. Intravenous RNA replicon administration showed primary brain targeting both in tumor-free and 4T1 tumor bearing animals, while intratumoral injection resulted in high expression levels in tumors. Interestingly, tumor targeting was observed for SFV particles when reduced intravenous or intraperitoneal viral doses were applied. The study also demonstrated that RNA replicons can be successfully re-administered to prolong luciferase reporter gene expression without the risk of immune responses associated with viral delivery. However, neither SFV particle nor RNA replicon delivery enhanced transgene expression from what was observed after the primary administration. In a combination therapy approach, SFV particles expressing IL-12 were administered together with an aroC (-) strain of Salmonella typhimurium (LVR01) into 4T1 tumor nodules orthotopically implanted in mice [89]. The treatment resulted in complete inhibition of lethal lung metastasis and long-term survival in 90% of mice after tumors were surgically resected. Although SFV-IL-12 alone showed anti-angiogenic effect, inhibited tumor growth and prolonged survival, the prevention of distant metastasis was related to the synergistic effect of SFV-IL-12 and LVR01. Despite the antitumor potential of LVR01 alone, the combination therapy was superior. Moreover, the order of administration was important as the therapeutic effect was only achieved when SFV-IL-12 was administered first, while pretreatment with LVR01 suppressed the anti-angiogenic effects of SFV-IL-12. In another study on SFV-IL-12, inhibition of tumor growth and lung metastases was demonstrated in a metastatic 4T1 mouse tumor model [90].
Cervical cancer has been frequently addressed in search for vaccines based on alphavirus vectors. For instance, immunization of mice with VEE particles expressing the human papilloma virus-16 (HPV-16) E7 protein generated CD8+ T cell responses and prevented tumor development [91]. In another approach, immunization of mice with an SFV vector engineered with the translation enhancer signal from the SFV capsid gene and an HPV E6-E7 fusion protein resulted in tumor regression and complete elimination of established tumors [92]. Therapeutic antitumor immunity was established in mice after the combination of intradermal administration of an SFV DNA replicon expressing HPV E6/7 [93] and electroporation. In comparison to conventional DNA plasmid vectors, a 200-fold lower equimolar dose of 0.05 µg resulted in 85% of immunized mice becoming tumor-free. Alphavirus-based immunization has also been combined with local low-dose irradiation, which resulted in 10-fold increase in CD8+ T cells in tumors [94]. It was demonstrated that irradiation upregulated chemokines and the combination enhanced antitumor activity. In a triple combination regimen, SFV-HPV E6,7 immunization was combined with administration of 40 mg/Kg sunitinib and low-dose tumor irradiation, which strongly enhanced immunotherapeutic antitumor activity resulting in tumor growth inhibition and 100% tumor-free survival of immunized mice [95].
The classic example of alphavirus-based colon cancer therapy comprises immunization of mice bearing CT26 colon tumors with SFV-LacZ replicon RNA [96]. Antigen-specific antibody and CD8+ T cell responses were observed after a single intramuscular injection of 0.1 µg SFV-LacZ RNA. Moreover, pre-immunization provided protection against tumor challenges and therapeutic efficacy and prolonged survival were obtained in mice with pre-existing tumors. In another study, mice implanted with CT29 colon tumors and 4T1 metastasizing breast tumors were immunized with SFV particles expressing the vascular endothelial growth factor receptor-2 (VEGFR-2) [97]. The outcome was inhibition of tumor growth, reduction in tumor angiogenesis and prevention of the spread of metastases. However, co-immunization with SFV-VEGFR-2 and SFV-IL-12 particles resulted in inferior immune responses and reduced inhibition of tumor growth. In contrast, immunization with the combination of SFV-VEGFR-2 and SF-IL-4 particles generated higher anti-VEGFR-2 antibody titers and resulted in prolonged survival. IL-12 expressed from an SFV vector containing the capsid translation enhancement signal showed high efficacy in the CT26 mouse tumor model [90]. It was demonstrated that SFV-based IL-12 expression induced immune cell stimulation and tumor necrosis. In another study, SIN particles expressing LacZ were administered to the mouse colon cancer CT26.CL25 model, showing potent therapeutic effect against existing tumors [98]. In addition to alphavirus vectors, noncytopathic KUN vectors expressing the granulocyte colony-stimulating factor (G-CSF) were subjected to intratumoral administration of mice implanted with CT26 colon tumors and B16-OVA melanomas [99]. The results from the study revealed that tumor regression was associated with the induction of anticancer CD8+ T cells and cure was achieved in more than 50% of immunized mice. Moreover, KUN-based immunization resulted in regression of CT26 lung metastases.
In the context of lung cancer, SFV-EGFP particles were demonstrated to induce cell death in human H358a non-small cell lung cancer (NSCLC) cells and also prevented growth of developing H358a spheroids [100]. Moreover, intratumoral administration of SFV-EGFP of nu/nu mice bearing H358a tumor xenografts induced apoptosis resulting in complete tumor regression in three out of seven mice. In another approach, nude mice with implanted A549 adenocarcinoma lung cells were locally injected with replication-competent SFV (VA7)-EGFP particles in comparison to a conditionally replicating Ad vector (Ad5-Delta24TK-GFP) [101]. SFV-based therapy resulted in superior survival rates in mice compared to Ad-based delivery. However, systemic administration was unable to elicit significant immune responses due to low efficacy of intratumoral replication. As previously described above for colon cancer, SIN-LacZ particles also enhanced therapeutic effects in a lung cancer model [98]. In this context, the efficacy was not dependent on tumor cell targeting, but related to the transient β-galactosidase expression in lymph nodes. The immunization produced long-lasting memory T cells and provided protection against challenges with both LacZ-positive and -negative tumor cells. In the case of VSV, NSCLC cell lines were transduced with VSV vectors expressing GFP and interferon-β (IFNβ), respectively, demonstrating oncolytic activity [102]. Moreover, intratumoral administration of VSV-GFP and VSV-IFNβ reduced tumor growth in nude mice with H2009 and A549 xenografts. Intratumoral injection of VSV-IFNβ into syngeneic LM2 lung tumors resulted in tumor regression, prolonged survival and cure of 30% of immunized mice. In the case of MV, the Edmonston strain was evaluated in NSCLC cell lines and immortalized lung epithelial Beas2B cells for viability, induction of apoptosis and viral transgene production by transduction with MV-GFP or MV-CEA [103]. MV transduction resulted in potent killing of most cell lines compared to Beas2B cells. Intratumoral administration of MV-CEA generated tumor regression in nude mice. In another application of MV, the modified rMV-SLAMblind vector unable to bind to its original SLAM receptor showed reduced cell viability in six out of nine human lung cancer cell lines [104]. Moreover, injection into subcutaneous NCI-H441 tumors in severe combined immune deficiency (SCID) mice suppressed tumor growth, and also targeted scattered tumor masses grown in the lungs of xenotransplanted mice.
A number of preclinical studies have been conducted for melanoma. In this context, mice were immunized with a yellow fever virus (YFV) vector expressing the cytotoxic T lymphocyte (CTL) epitope SIINFEKL derived from chicken ovalbumin, which elicited SIINFEKL-specific CD8+ lymphocytes and provided protection against challenges with malignant melanoma cells [105]. Furthermore, YFV-based immunization resulted in regression of solid tumors and lung metastases. In the context of alphaviruses, VEE-based expression of tyrosine-related protein-2 (TRP-2) was verified in a B16 mouse melanoma model, which demonstrated humoral immune responses, robust antitumor activity and prolonged survival [106]. In another study, VEE-TRP-2 immunization was combined with administration of immunomodulatory monoclonal antibodies (mAbs) for either antagonist anti-CTL antigen-4 (CTLA-4) or agonist anti-glucocorticoid-induced tumor necrosis factor receptor (GITR) [107]. The VEE-TRP-2 combination with anti-CTLA-4 or anti-GITR mAbs provided complete regression in 50% and 90% of mice, respectively. Studies on melanoma have also been conducted with DNA-based SFV vectors. In this context, VEGFR2 and IL-12 expressed from one SFV DNA vector and survivin and β-hCG antigens expressed from another SFV DNA vector were co-administered and evaluated in a B16 mouse melanoma model [108]. The combination therapy resulted in superior tumor growth inhibition and prolonged survival of mice compared to immunization with either SFV DNA vector alone. Live-attenuated MV strains have been evaluated for their oncolytic activity against a panel of human melanoma cell lines [109]. It was demonstrated that MV resistant melanoma cells possessed a fully functional type I IFN pathway, while sensitive cells showed defects in the pathway. Treatment with ruxolitinib rendered the resistant cells sensitive to MV. It was concluded that type I IFN responses determine whether melanoma cells are sensitive or resistant to MV. Furthermore, the MV Leningrad-16 (L-16) strain showed replication in a panel of human metastatic melanoma cell lines and mediated cell killing in tumor cells [110]. MV L-16 generated statistically significant inhibition of tumor growth in a melanoma xenograft model. Pseudotyping of VSV particles with the non-neurotropic lymphocytic choriomeningitis virus (LCMV) envelope glycoprotein generated VSV-GP vectors capable of efficient killing of mouse, canine and human melanoma cell lines [111]. Immunization of mice with VSV-GP provided prolonged survival in both A375 xenograft and B16-OVA syngeneic mouse models [112]. It has also been demonstrated that priming with intratumoral reovirus followed by boosting with VSV expressing a cDNA library of melanoma antigens (VSV-ASMEL) significantly improved survival of mice with subcutaneous B16 melanoma tumors [113]. When additional boosting with anti-PD1 immune checkpoint blockade was carried out, further enhanced survival and long-term cure was observed.
In the context of ovarian cancer, the above described VSV-GP vector, where the VSV-G protein was replaced by the lymphocytic choriomeningitis virus (LCMV) envelope glycoprotein, showed oncolytic activity both in ovarian cancer cell lines (A2780, HTB77, SKOV6, HOC7, OVCAR3, ID8 and HOSE) and in mice with A2780 tumor xenografts [114]. Combination therapy with ruxolitinib generated superior responses in both subcutaneous and orthotopic xenograft models. In the context of MV, a single-chain antibody (scFv) specific for alpha-folate receptor (FRalpha), overexpressed in 90% of ovarian cancers, was engineered on the MV attachment protein [115]. The tropism and the fusogenic activity of the MV-alphaFR vector was redirected to tumor cells showing no background infectivity of normal human cells. Moreover, tumor volume reduction and overall survival increase were observed in vivo. In the context of alphaviruses, combination therapy of SIN-IL-12 particles and the CPT-11 topoisomerase inhibitor irinotecan resulted in long-term survival in 35% of SCID mice with implanted aggressively growing human ovarian ES2 cancer [116]. In contrast, no long-term survival was observed for monotherapy with either SIN-IL-12 or CPT-11. Moreover, prime-boost immunization approaches with vaccinia virus (VV)-OVA followed by SFV-OVA particle administration or vice versa demonstrated enhanced OVA-specific CD8+ T cell immune responses in C57BL/6 mice [117]. In another study, it was demonstrated that SIN-GFP and SIN-Luc vectors induced cellular stress and apoptosis in the MOSEC tumor cell line derived from the ovarian epithelium [118]. In addition, the Maraba virus, belonging to rhabdoviruses, has been used in a prime-boost regimen with the MIS416 microparticle vaccine adjuvant derived from Propionibacterium acnes with immunostimulatory muramyl dipeptide and bacterial DNA [119]. Boosting with Maraba virus expressing the human melanoma antigen dopachrome tautomerase (DCT) generated robust tumor-specific CD8+ T cell responses resulting in improved tumor control and unique immunological changes in tumors.
Similar to the findings in ovarian tumor cells, SIN vectors expressing GFP and luciferase also induced apoptosis in the pancreatic adenocarcinoma cell line Pan02 [118]. In another approach, VSV vectors expressing GFP were compared to conditionally replicative adenovirus (CRAd), Sendai virus and Respiratory syncytial virus (RSV) for oncolytic activity in 13 pancreatic ductal adenocarcinoma (PDA) cell lines [120]. Generally, VSV demonstrated superior oncolytic ability compared to the other viral vectors tested, although the susceptibility was highly heterogeneous in different PDA cell lines. MIA PaCa2 and Panc-1 cells were highly permissive to both VSV and CRAd, SU86.86 highly permissive only to VSV and HPAF-II showed limited permissiveness to all viruses. A similar pattern was observed in vivo when tested in nude mice. The VSV resistance of HPAF-II cells could be reduced with a combination of polybrene, DEAE-dextran and ruxolitinib, which significantly improved VSV attachment and replication [121]. The triple combination should allow treatment of PDAC tumors highly resistant to oncolytic VSV. In the case of MV vectors, efficient killing of nectin-4 expressing pancreatic cell lines by MV-SLAMblind was observed [122]. Intratumoral injection of MV-SLAMblind showed significant tumor growth suppression in SCID mice implanted with KLM1 and Capan-2 xenografts.
Finally, prostate cancer has also been commonly targeted in preclinical tumor models. For instance, the prostate tumor cell lines PC-3, DU-145 and LnCaP were efficiently killed by MV vectors [123]. Intratumoral administration of MV-CEA vectors generated significant delay of tumor growth and prolonged survival of mice with PC-3 xenografts. In another study, mice and rabbits were immunized with VEE particles expressing the prostate-specific membrane antigen (PSMA) [124]. In comparison to purified PSMA protein, a single dose of 2 × 105 VSV-PSMA particles provided a superior immune response in mice. In another study, mice were immunized with VEE particles expressing the six-transmembrane epithelial antigen of the prostate (STEAP), which elicited specific CD8+ T cell responses and resulted in a prolonged survival rate [125]. Moreover, VEE particles expressing the prostate stem cell antigen (PSCA) provided long-term prostate cancer protection in a transgenic adenocarcinoma mouse prostate (TRAMP) model [126]. The survival rate was 90% at 12 months in immunized mice compared to control mice, who had either succumbed to prostate cancer or presented a heavy tumor load. In another study, the prostate-specific antigen (PSA) was expressed from a VEE vector showing efficient infection of mouse DCs in vitro and eliciting a robust PSA-specific response in vivo [127]. Furthermore, immunization with VEE-PSA strongly stimulated production of IgG2a/b anti-PSA antibodies and tumor growth was significantly delayed. The study showed that immunization can overcome immune tolerance to PSA and can mediate rapid clearance of PSA-expressing tumor cells. Related to VSV vectors, the VSV-GP-LCMV expressing luciferase showed great efficacy of infection of prostate cancer cell lines and long-term remission in Du145 and 22Rv1 prostate cancer models after intratumoral injections [127]. Moreover, remission was also observed in syngeneic TRAMP-C subcutaneous tumors and after intravenous vector administration in PC-3M-Luc bone metastases. In a combination therapy approach, oncolytic MV and mumps virus (MuV) vectors were evaluated in a human prostate cancer xenograft model [128]. Greater anti-tumor effect and prolonged survival were obtained from combination therapy compared to individual treatment either with MV or MuV vectors.

5. Clinical Trials

In the context of clinical trials, the number of conducted studies using self-amplifying RNA virus vectors are relatively modest compared to for instance adenovirus and retrovirus vectors (Table 3). In a randomized, double-blind phase I trial, 40 CMV seronegative healthy volunteers were intramuscularly or subcutaneously injected with VEE particles expressing the CMV glycoprotein B or the fusion between CMV pp65 and IE1 proteins [129]. The vaccinations with a lower dose of 1 × 107 infectious units (IU) and a higher dose of 1 × 108 IU were well tolerated with only mild to moderate local reactogenicity, minimal systemic reactogenicity and no clinically important changes in laboratory parameters. Direct IFN-γ responses to CMV antigens were obtained in all vaccinated subjects. The procedure was safe and neutralizing antibodies and multifunctional T cell responses were generated against all three CMV antigens important for protective immunity. In another randomized, double-blind, placebo-controlled phase I trial, HIV-negative volunteers in the US and South Africa were subjected to subcutaneous injection of escalating doses of VEE particles expressing the non-myristoylated form of the HIV-1 subtype C Gag protein [130]. The vaccine was well tolerated with only modest local reactogenicity. However, five serious adverse events were reported, albeit none of them were considered to be related to the vaccine. Disappointingly, the immune responses comprised only low levels of binding antibodies and T cell responses at the highest dose of 1 × 108 IU.
Not surprisingly, EBOV has been the target for numerous vaccine development programs including the application of self-amplifying RNA virus vectors in the light of the EBOV outbreak 2014–2016. For instance, VSV particles expressing the glycoprotein of a Zaire EBOV strain (VSV-ZEBOV) were used for immunization of 78 volunteers with one of three doses (3 × 106, 2 ×107 or 1 × 108 pfu) in a phase I trial to assess safety and immunogenicity [131]. The immunization caused some adverse events such as injection-site pain, fatigue, myalgia and headache, but less adverse events were observed after the second immunization. The antibody titers against ZEBOV glycoprotein were higher for the two highest doses than for the lowest dose. Moreover, the antibody titers were significantly higher after the second vaccination, but the effect diminished after six months. Overall, the immunization elicited anti-EBOV antibody responses and the highest dose should be further evaluated for pre-exposure prophylaxis. A second immunization may further boost the antibody response. In a dose-ranging, observer-blind, placebo-controlled phase I trial, 40 volunteers received doses of 1 × 105, 5 × 105 or 3 × 106 pfu of an attenuated VSV vector, where the VSV G protein was replaced by ZEBOV GP [132]. No serious adverse events occurred, the immunogenicity was comparable across all doses tested and a single immunization elicited sustainable high IgG titers throughout the whole study (180 days). In another phase I trial, 30 healthy volunteers were immunized with 3 × 105, 3 × 106 or 2 × 107 pfu of VSV-ZEBOV [133]. The immunization was well tolerated without any serious vaccine-related adverse events at all doses. EBOV-specific neutralizing antibodies were recorded in nearly all vaccinees and those receiving the highest dose generated EBOV GP-specific T cells. In a follow-up study within the phase I trial, analysis of adaptive immune responses revealed that although no pre-existing immunity was detected, more than one-third of the immunized volunteers developed VSV-specific CTL responses and antibodies [134]. In a dose-finding, placebo-controlled, double-blind phase I/II study, reduction of the VSV-ZEBOV dose from 1–5 × 107 pfu to 3 × 105 pfu improved tolerability, but decreased antibody responses and did not prevent vaccine-induced arthritis, dermatitis or vasculitis [135].
Several phase II/III trials have been conducted for EBOV. For instance, a randomized placebo-controlled phase II study comprising 1500 adults immunized with a chimpanzee Ad3 vector (ChAd3-EBO-Z) and VSV∆G-ZEBOV-GP in Liberia showed adverse events such as injection-site reactions, headache, fever and fatigue [136]. One month after immunization, ChaAd3-EBO-Z and VSV∆G-ZEBOV-GP induced antibody responses in 70.8% and 83.7%, respectively, of vaccinees compared to 2.8% in the control group receiving placebo. After 12 months, 63.5% and 79.5% of the individuals vaccinated with Ad and VSV, respectively, showed immune responses with 6.8% in control subjects. In an open-label, cluster-randomized ring vaccination phase III trial in suspected cases of EVD, 4123 individuals were assigned for immediate vaccination with VSV-ZEBOV and 3528 persons were to receive delayed vaccination [137]. Ten days after randomization, no new cases of EVD were diagnosed in the immediate vaccination group, while 16 EVD cases were registered in the delayed vaccination group confirming a vaccine efficacy of 100%. The interim analysis suggested that the vaccination was safe and efficacious in preventing EVD. In another phase III study in Guinea and Sierra Leone, 2119 individuals were immediately immunized with a single dose of 2 × 107 pfu of VSV-ZEBOV and 2041 subjects were immunized 21 days after randomization [138]. The participants were followed for 84 days showing substantial protection against EVD with no cases of EVD found from day 10 after vaccination. However, the efficacy of vaccination was questioned by claiming of a bias of the medical study team not staying in the communities of the delayed vaccination group, which could be considered as important for disease transmission, even suggesting that EVD was averted due to behavioral changes and the effect of the vaccine was 0% [139]. In response, the team behind the study confirmed that both the immediate and the delayed vaccination communities had similar exposure to health workers during the period after detection and no bias existed [140]. Moreover, at 10 days or more after randomization, eight unvaccinated contacts developed confirmed symptoms of EVD. Therefore, if vaccination had no consequence and the presence of medical staff accounted for the entire effect, the number of cases should be zero or close to zero. In the individually controlled phase II/III trial STRIVE (Sierra Leone Trial to Introduce a Vaccine against Ebola) health care and frontline workers in the five most EVD-affected districts of Sierra Leone received a single intramuscular dose of VSV-ZEBOV at enrollment or 18–24 weeks later [141]. Among the 8673 enrolled participants no EVD cases and no vaccine-related serious adverse events were reported. In a randomized, double-blind, multicenter phase III study in Canada, Spain and the US, individuals were immunized with a single dose or 2 × 107 pfu or 1 × 108 pfu of VSV∆G-ZEBOV-GP and assessed for safety and immunogenicity [142]. The results demonstrated safety of vaccination with no vaccine-related severe adverse events or deaths reported and should support the vaccination of persons at risk of EVD.
Chikungunya virus, itself a self-amplifying RNA alphavirus, has a history of epidemics in for instance the Republic of Congo [152] and Reunion [153]. A randomized, placebo-controlled, double-blind, phase II clinical trial was conducted in 393 healthy adults in Haiti, Dominican Republic, Martinique, Guadeloupe and Puerto Rico [143]. Two intramuscular injections 28 days apart were well tolerated although 16 serious adverse events unrelated to the vaccine occurred four times in the vaccine group and 12 times in the placebo group. Sixteen mild to moderate adverse events potentially related to the injection procedure occurred 12 times in the vaccine group and four times in the placebo group. Although good safety and tolerability profiles were obtained, clinical efficacy needs to be addressed in phase III trials. In the case of VEE, periodic epidemics have occurred in humans and equines in Latin America making it an important target for vaccine development [154]. A phase I trial was performed by intramuscular and intradermal electroporation of a VEE DNA replicon vector expressing the VEE E3-E2-6K-E1 genes [144]. Participants received 0.5 mg and 2.0 mg of plasmid DNA intramuscularly or 0.08 mg or 0.3 mg DNA intradermally and monitoring continued for 360 days. No serious adverse events related to the vaccine or the device were recorded and both delivery procedures were considered as acceptable although the acute tolerability of the intradermal delivery was judged to be better than the intramuscular procedure. Intramuscular administration elicited detectable VEE-neutralizing antibodies in all vaccinees in the high and low dose groups after two or three immunizations, respectively. Related to the intradermal route, seven out of eight individuals developed VEE-neutralizing antibodies, while five out of eight subjects generated antibody responses after three vaccine injections. Furthermore, a correlation between DNA dose and the magnitude of VEE-neutralizing antibody responses for both intramuscular and intradermal administration routes could be established.
In the context of cancer, self-amplifying RNA virus vectors have been subjected to several clinical trials (Table 3). A phase I clinical trial was conducted in patients with recurrent glioblastoma multiforme applying doses ranging from 1 × 105 to 2 × 107 TCID50 of MV-CEA [145]. So far, a limited number of patients have received 1 × 105 or 1 × 106 TCID50 in the resection cavity showing no dose-limiting toxicity. Additional results from the study are expected shortly. An interesting approach has been to utilize liposome-encapsulated SFV particles expressing IL-12 in a phase I trial in kidney carcinoma and melanoma patients [146]. The encapsulation procedure can provide passive tumor targeting, extend the circulation in the blood and protect recognition by host immune defense systems. The encapsulated LipoVIL12 particles were intravenously administered resulting in transient 10-fold increase in IL-12 plasma levels in patients. A good safety profile was established with no toxicity related to liposomes or SFV particles. The study demonstrated that repeated administration of encapsulated SFV particles did not induce any immune response against SFV. Moreover, the maximum tolerated dose (MTD) was determined as 3 × 109 per m2, the limitation being the fever response induced by IL-12 expression.
In the context of lymphomas, the oncolytic MV-Edm-Zagreb vaccine strain (MVEZ) was administered to five patients with stage IIb cutaneous T-cell lymphoma (CTLC) in an open-label dose-escalation phase I trial [147]. Administration of doses of 102 to 103 of TCID50 of MVEZ resulted in no dose-limiting toxicity and good tolerability. Complete regression of one CTCL tumor was obtained in one patient after one treatment cycle of four injections. After two treatment cycles, partial regressions were seen in four out of five patients. Related to myeloma, a phase I trial was conducted in patients with recurrent or refractory multiple myeloma with MV vectors expressing the sodium iodide symporter (NIS) in combination with cyclophosphamide or alone [148]. Preliminary findings from two heavily pre-treated patients with a single injection of 1011 TCID50 of MV-NIS resulted in complete remission, which lasted for nine months in one patient and complete resolution of bone marrow plasmacytosis in both patients. Furthermore, NIS-mediated imaging illustrated tumor-specific targeting of MV-NIS [155]. Based on the phase I results, a phase II combination therapy of MV-NIS and cyclophosphamide in recurrent or refractory myeloma patients has been initiated. MV vectors expressing CEA have also been subjected to phase I/II clinical trials in patients with recurrent ovarian cancer [149]. Patients with platinum- and paclitaxel-refractory ovarian cancer confined to the peritoneal cavity received intraperitoneally seven doses of MV-CEA, ranging from 103 to 109 TCID50, six times monthly. The maximum MV-CEA dose was limited to manufacturing capability of clinical grade oncolytic MV preparation [156]. The immunization was well tolerated with no dose-limiting toxicity and only mild treatment-related adverse events such as non-neutropenic fever and abdominal discomfort. Repeated administration did not induce increase in anti-measles humoral immunity or development of anti-CEA antibodies. The CEA expression was dose-dependent resulting in elevated serum CEA levels in patients receiving 109 TCID50. According to the Response Evaluation Criteria in Solid Tumors (RECIST) assessment of antitumor response [157], stable disease was achieved in 14 patients. In this context, nine out of nine patients were from the 107-109 TCID50 dose range, while only five out of 12 patients receiving lower dose levels showed stable disease. The median overall survival was 12.15 months, twice as long as expected in this patient population [158]. However, patients receiving high doses of 108–109 TCID50 had a median overall survival of 38.4 months.
In the context of pancreatic cancer, VEE-CEA was applied for intramuscular administration of 4 × 107 to 4 × 108 IU in pancreatic cancer patients in a phase I study [150]. Clinically relevant CEA-specific T cell antibody responses were induced after repeated administration of VEE-CEA. Furthermore, CEA-specific antibodies mediated the antibody-dependent toxicity against tumor cells from human colorectal cancer metastases. The study also demonstrated that immunized patients developing CEA-specific T cell responses showed prolonged survival. Finally, VEE-PSMA particles have been applied in a phase I clinical study in patients with castration resistant metastatic prostate cancer (CRPC) [151]. Immunization with either 0.9 × 107 or 3.6 × 107 IU of VEE-PSMA was well tolerated, but only weak PSMA-specific immune responses were obtained. Despite the failure to generate robust immune responses and clinical benefits, the success in eliciting neutralizing antibodies indicated that dose optimization will further enhance the efficacy of the vaccine.

6. Conclusions

In summary, self-amplifying RNA virus vectors have been subjected to a large number of preclinical studies for both infectious diseases (Table 1) and different types of cancers (Table 2). Moreover, self-replicating RNA viral vectors have also been applied for immunizations against bacterial and parasite infections as previously described [159]. Briefly, immunization with SIN DNA-based 85A antigen (Ag85A) expression provided protection of mice challenged with Mycobacterium tuberculosis [160] and SFV VLPs expressing the Plasmodium yoelii circumsporozoite protein (CS) likewise against malaria infections [161]. In the context of viral infections, such as EBOV, LASV, HIV and influenza virus, robust immune responses have been induced providing in many cases protection against challenges with lethal doses of pathogenic viruses in animal models. Moreover, a clinical phase I trial in healthy volunteers elicited modest antibodies against HIV-1 [130]. Similarly, neutralizing antibodies were elicited in individuals immunized with VEE in a phase I study targeting VEE [144]. Encouraging results have been obtained from phase III trials on EBOV in Africa demonstrating substantial protection against EVD [137,138]. The current COVID-19 pandemic has naturally shed a new light on viral diseases and has accelerated the development of novel vaccines [162].
In the case of cancer vaccines, studies in numerous animal models for various cancer indication have demonstrated tumor regression and prolonged survival. As most of the clinical trials conducted so far have comprised phase I studies, the focus has mainly been on safety and tolerability. Despite that, prolonged survival was established in pancreatic cancer patients treated with VEE-CEA [150], stable disease in ovarian cancer patients immunized with MV-CEA [149] and complete remission in one patient with refractory multiple myeloma after treatment with MV-NIS [148]. However, in many of the studies the immune responses were relatively modest, suggesting that optimization of doses, prime-boosting, vector choice and delivery method/route are necessary.
In the context of self-amplifying RNA virus vectors, their main attractive features comprise the immediate efficient amplification of mRNA directly in the cytoplasm and the flexibility of applying delivery vectors based on RNA replicons, recombinant virus particles or VLPs and layered DNA/RNA vectors. The efficacy of self-amplifying RNA virus vectors has been confirmed for RNA replicons by comparison to immunization with synthetic mRNA, where protection of mice against influenza virus challenges was achieved with 64 times less self-amplifying VEE RNA (1.25 µg) compared to 80 µg of synthetic mRNA [63]. Similarly, 100- to 1000-fold lower doses of DNA replicon SIN-HSV-1-gB compared to conventional DNA plasmids were required to obtain antibody responses and protection against lethal challenges with virus in mice [163]. In the context of cervical cancer, where conventional DNA-based immunization failed to prevent tumor outgrowth, already a 200-fold lower equimolar dose of 0.05 µg of SFV-HPV E6/7 DNA provided complete tumor regression in 85% of immunized mice [93].
In attempts to enhance immune responses for development of better vaccines, attention has also been paid to vector engineering and delivery improvements. Oncolytic viruses have been applied for specific infection of tumor cells, leading to their killing without affecting normal tissues [160]. Another approach involves liposome- and polymer-encapsulation strategies with the goals of tumor targeting and protection against degradation and recognition by the host immune system. For instance, liposome encapsulated SFV particles demonstrated tumor targeting and allowed repeated administration in cancer patients [146]. In another approach, polymer-coating of MV expressing the N, P and L proteins (MV-NLP) of the wild-type MV strain resulted in superior oncolytic activity and improved antitumor activity in mice compared to naked MV-NLP [164]. One area of interest has been the targeting of DCs as described for SFV-IL-18 transduced DCs in mice with B16 xenografts [80] and VEE-neu transduced DCs in a mouse tumor model for breast cancer [86]. Furthermore, RNA replicons of CSFV expressing influenza virus NP complexed with liposomes for delivery to DCs, elicited immune responses both in vitro and in vivo [65]. Another approach relates to combination therapy of self-amplifying RNA virus vectors with drugs and irradiation (Table 2), including tumor eradication with SFV-HPV E6,7 particles, sunitinib and low-dose irradiation [95] or enhanced immune responses of VSV and ruxolitinib co-administration [120]. Much attention has also been paid to delivery safety including engineering of vectors providing the highest safety standards. For instance, point mutations introduced into the SFV p62 precursor sequence prevented the cleavage of p62 into E2 and E3 proteins, which resulted in conditionally infectious particles and reduction of production of replication competent SFV particles [15]. Furthermore, split helper systems have been engineered for SFV [16], SIN [165] and VEE [166] by placing the capsid and envelope genes on separate helper vectors, eliminating the production of replication-proficient particles.
In conclusion, self-amplifying RNA virus vectors provide the flexibility of utilizing RNA replicons, recombinant VLPs or DNA replicon plasmids for immunization studies leading to robust antibody responses and protection against challenges with lethal doses of pathogens or tumor cells. Although so far, clinical responses have been relatively modest, vector development, dosage optimization and delivery improvement will support the production of efficient vaccines in the near future. These needs are even more crucial today with the current COVID-19 pandemic.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines – a new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef] [Green Version]
  2. Schnee, M.; Vogel, A.B.; Voss, D.; Petsch, B.; Baumhof, P.; Kramps, T.; Stitz, L. An mRNA vaccine encoding rabies virus glycoprotein induces protection against lethal infection in mice and correlates of protection in adult and newborn pigs. PLoS Negl. Trop. Dis. 2016, 10, e0004746. [Google Scholar] [CrossRef] [PubMed]
  3. Pardi, N.; Hogan, M.J.; Pelc, R.S.; Muramatsu, H.; Andersen, H.; DeMaso, C.R.; Dowd, K.A.; Sutherland, L.L.; Scearce, R.M.; Parks, R.; et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature 2017, 543, 248–251. [Google Scholar] [CrossRef] [PubMed]
  4. Bahl, K.; Senn, J.J.; Yuzhakov, O.; Bulychev, A.; Brito, L.A.; Hassett, K.J.; Laska, M.E.; Smith, M.; Almarsson, Ö.; Thompson, J.; et al. Preclinical and clinical demonstration of immunogenicity by mRNA vaccines against H10N8 and H7N9 influenza viruses. Mol. Ther. 2017, 25, 1316–1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Alberer, M.; Gnad-Vogt, U.; Hong, H.S.; Mehr, K.T.; Bacckert, L.; Finak, G.; Gottardo, R.; Bica, M.A.; Garofano, A.; Koch, S.D.; et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: An open label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet 2017, 390, 1511–1520. [Google Scholar] [CrossRef]
  6. Lundstrom, K. Self-amplifying RNA virus vectors: Clinical applications in cancer drug delivery. Exp. Opin Drug Deliv. 2019, 16, 1027–1029. [Google Scholar] [CrossRef] [Green Version]
  7. Strauss, J.H.; Strauss, E.G. The Alphaviruses: Gene Expression, Replication and Evolution. Micobiol. Rev. 1994, 58, 491–562. [Google Scholar] [CrossRef]
  8. Banerjee, A.K. Transcription and Replication of Rhabdoviruses. Microbiol. Rev. 1987, 51, 66–87. [Google Scholar] [CrossRef] [Green Version]
  9. Lundstrom, K. RNA Viruses as Tools in Gene Therapy and Vaccine Development. Genes 2018, 10, E189. [Google Scholar] [CrossRef] [Green Version]
  10. Liljestrom, 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]
  11. 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] [CrossRef] [PubMed]
  12. Davis, N.L.; Willis, L.V.; Smith, J.F.; Johnston, R.F. 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]
  13. DiCiommo, D.P.; Bremner, R. Rapid, high level protein production using DNA-based Semliki Forest virus vectors. J. Biol. Chem. 1998, 273, 18060–18066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. White, C.L.; Thomson, M.; Dimmock, N.J. Deletion analysis of a defective interfering Semliki Forest virus RNA genome defines a region in the nsP2 sequence that is required for efficient packaging of the genome into viral particles. J. Virol. 1998, 72, 4320–4326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Berglund, P.; Sjöberg, M.; Garoff, H.; Atkins, G.J.; Sheahan, B.J.; Liljeström, P. Semliki Forest virus expression system: Production of conditionally infectious recombinant particles. Biotechnology 1993, 11, 916–920. [Google Scholar] [CrossRef] [PubMed]
  16. Smerdou, C.; Liljeström, P. Two-helper system for production of recombinant Semliki Forest virus particles. J. Virol. 1999, 73, 1092–1098. [Google Scholar] [CrossRef] [Green Version]
  17. Pijlman, G.P.; Suhrbier, A.; Khromykh, A.A. Kunjin virus replicons: An RNA-based, non-cytopathic viral vector system for protein production, vaccine and gene therapy applications. Exp. Opin Biol Ther. 2006, 6, 134–145. [Google Scholar] [CrossRef] [Green Version]
  18. De Felipe, F. Skipping the co-expression problem: The new 2A ‘CHYSEL’ technology. Genet. Vaccines Ther. 2004, 2. [Google Scholar] [CrossRef] [Green Version]
  19. Khromykh, A.A.; Varnavski, A.N.; Westaway, E.G. Encapsidation of the flavivirus Kunjin replicon RNA by using a complementation system providing Kunjin virus structural proteins in trans. J. Virol. 1998, 72, 5967–5977. [Google Scholar] [CrossRef] [Green Version]
  20. Shi, P.Y.; Tilgner, M.; Lo, M.K. Construction and characterization of subgenomic replicons of New York strain of West Nile virus. Virology 2002, 296, 219–233. [Google Scholar] [CrossRef]
  21. Scholle, I.; Girard, Y.A.; Zhao, Q.; Higgs, S.; Mason, P.W. Trans-packaged West Nile virus-like particles: Infectious properties in vitro and in infected mosquito vectors. J. Virol. 2004, 78, 11605–11614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Molenkamp, R.; Kooi, E.A.; Lucassen, M.A.; Greve, S.; Thijssen, J.C.; Spaan, W.J.; Bredenbeek, P.J. Yellow fever virus replicons as an expression system for hepatitis C virus structural proteins. J. Virol. 2003, 77, 1644–1648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Jones, C.T.; Patkar, C.G.; Kuhn, R.J. Construction and applications of yellow fever virus replicons. Virology 2005, 331, 247–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Jones, M.; Davidson, A.; Hibbert, L.; Gruenwald, P.; Schlaak, J.; Ball, S.; Foster, G.R.; Jacobs, M. Dengue virus inhibits alpha interferon signaling by reducing STAT2 expression. J. Virol. 2005, 79, 5414–5420. [Google Scholar] [CrossRef] [Green Version]
  25. Pang, X.; Zhang, M.; Dayton, A.I. Development of dengue virus type 2 replicons capable of prolonged expression in host cells. BMC Microbiol. 2001, 1, 18. [Google Scholar]
  26. Gherke, R.; Ecker, M.; Aberle, S.W.; Allison, S.L.; Heinz, F.X.; Mandi, C.W. Incorporation of tick-borne encephalitis virus replicons into virus-like particles by a packaging cell line. J. Virol. 2003, 77, 8924–8933. [Google Scholar] [CrossRef] [Green Version]
  27. Hayasaka, D.; Yoshii, K.; Ueki, T.; Goto, A.; Mizutani, T.; Kariwa, H.; Iwasaki, T.; Gould, E.A.; Takashima, I. Sub-genomic replicons of tick-borne encephalitis virus. Arch. Virol. 2004, 149, 1245–1256. [Google Scholar] [CrossRef] [PubMed]
  28. Radecke, F.; Spielhofer, P.; Schneider, H.; Kaelin, K.; Huber, M.; Dötsch, C.; Christiansen, G.; Billeter, M.A. Rescue of measles viruses from cloned DNA. EMBO J. 1995, 14, 5773–5784. [Google Scholar] [CrossRef]
  29. Singh, M.; Cattaneo, R.; Billeter, M.A. A recombinant measles virus expressing hepatitis B surface antigen induces humoral responses in genetically modified mice. J. Virol. 1999, 73, 4823–4828. [Google Scholar] [CrossRef] [Green Version]
  30. Osakada, F.; Callaway, E.M. Design and generation of recombinant rabies virus vectors. Nat. Protoc. 2013, 8, 1583–1601. [Google Scholar] [CrossRef]
  31. An, H.; Kim, G.N.; Kang, C.Y. Genetically modified VSV(NJ) vector is capable of accommodating a large foreign gene insert and allows high level gene expression. Virus Res. 2013, 171, 168–177. [Google Scholar] [CrossRef] [PubMed]
  32. Harty, R.N.; Brown, M.E.; Hayes, F.P.; Wright, N.T.; Schnell, M.J. Vaccinia virus-free recovery of vesicular stomatitis virus. J. Mol. Microbiol Biotechnol. 2001, 3, 513–517. [Google Scholar] [PubMed]
  33. Ohara, S.; Inoue, K.; Yamada, M.; Yamawaki, T.; Koganezawa, N.; Tsuttsui, K.; Witter, M.P.; Iijima, T. Dual transneural tracing in the rat entorhoinal-hippocampal circuit by intracerebral injection of recombinant rabies virus vectors. Front. Neuroanat. 2009, 3, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ito, N.; Takayama-Ito, M.; Yamada, K.; Hosokawa, J.; Sugiyama, M.; Minamoto, N. Improved recovery of rabies virus from cloned cDNA using a vaccinia virus-free reverse genetics system. Microbiol Immunol. 2003, 47, 613–617. [Google Scholar] [CrossRef]
  35. Khalil, S.M.; Tonkin, D.R.; Mattocks, M.D.; Snead, A.T.; Johnston, R.E.; White, L.J. A tetravalent alphavirus-vector based dengue vaccine provides effective immunity in an early life mouse model. Vaccine 2014, 32, 4068–4074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Harahap-Carrillo, I.S.; Ceballos-Olvera, I.; Valle, J.R. Immunogenic subviral particles displaying domain III of dengue 2 envelope protein vectored by measles virus. Vaccines 2015, 3, 503–518. [Google Scholar] [CrossRef] [Green Version]
  37. Hu, H.M.; Chen, H.W.; Hsiao, Y.; Wu, S.H.; Chung, H.H.; Hsieh, C.H.; Chong, P.; Leng, C.H.; Pan, C.H. The successful induction of T-cell and antibody responses by a recombinant measles virus-vectored tetravalent dengue vaccine provides partial protection against dengue-2 infection. Hum. Vaccin Immunother. 2016, 12, 1678–1689. [Google Scholar] [CrossRef]
  38. Erasmus, J.H.; Khandhar, A.P.; Guderian, J.; Granger, B.; Archer, J.; Archer, M.; Cage, E.; Fuerte-Stone, J.; Larson, E.; Lin, S.; et al. A nanostructured lipid carrier for delivery of a replicating viral RNA provides single, low-dose protection against Zika. Mol. Ther. 2018, 26, 2507–2522. [Google Scholar] [CrossRef] [Green Version]
  39. Chattopadhyay, A.; Aquilar, P.V.; Bopp, N.E.; Yarovinsky, T.O.; Weaver, S.C.; Rose, J.K. A recombinant virus vaccine that protects both against Chikungunya and Zika virus infections. Vaccine 2018, 36, 3894–3900. [Google Scholar] [CrossRef]
  40. Pyankov, O.V.; Bodnev, S.A.; Pyankova, O.G.; Solodkyi, V.V.; Pyankov, S.A.; Setoh, Y.X.; Volchokova, V.A.; Suhrbier, A.; Volchikov, V.V.; Agafonov, A.A.; et al. A Kunjin replicon virus-like vaccine provides protection against Ebola virus infection in nonhuman primates. J. Infect. Dis. 2015, 212 (Suppl. S2), S368–S371. [Google Scholar] [CrossRef] [Green Version]
  41. Marzi, A.; Robertson, S.J.; Haddock, E.; Feldmann, F.; Hanley, P.W.; Scott, D.-P.; Strong, J.E.; Kobinger, G.; Best, S.M.; Feldmann, H. Ebola vaccine. VSV-EBOV rapidly protects macaques against infection with the 2014/2015 Ebola virus outbreak strain. Science 2015, 349, 739–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Geisbert, T.W.; Feldmann, H. Recombinant vesicular stomatitis virus-based vaccines against Ebola and Marburg infections. J. Infect. Dis. 2011, 204 (Suppl. S3), S1075–S1081. [Google Scholar] [CrossRef] [PubMed]
  43. 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, 4852–4964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. 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] [PubMed] [Green Version]
  45. 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]
  46. Öhlund, P.; Garcia-Arriaza, J.; Zusinaite, E.; Szurgot, I.; Mànnik, A.; Kraus, A.; Ustav, M.; Merits, A.; Esteban, M.; Liljestrom, P.; et al. DNA-launched RNA replicon vaccines induce potent anti-Ebola immune responses that can be further improved by a recombinant MVA boost. Sci. Rep. 2018, 8, 12459. [Google Scholar] [CrossRef]
  47. Safronetz, D.; Mire, C.; Rosenke, K.; Feldmann, F.; Haddock, E.; Geissbert, T.; Feldmann, H. A recombinant vesicular stomatitis virus-based Lassa fever vaccine protects guinea pigs and macaques against challenge with geographically and genetically distinct Lassa viruses. PLoS Negl. Trop. Dis. 2015, 9, e0003736. [Google Scholar] [CrossRef]
  48. Wang, M.; Jokinen, J.; Tretvakova, I.; Pushko, P.; Lukashevich, I.S. Alphavirus vector-based replicon particles expressing multivalent cross-protective Lassa virus glycoproteins. Vaccine 2018, 36, 683–690. [Google Scholar] [CrossRef]
  49. Kainulainen, M.H.; Spengler, J.R.; Welch, S.R.; Coleman-McCray, J.D.; Harmon, J.R.; Klena, J.D.; Nichol, S.T.; Albarino, C.G.; Spiropoulou, C.F. Use of a scalable replicon-particle vaccine to protect against lethal Lassa virus infection in the guinea pig model. J. Infect. Dis. 2018, 217, 1957–1966. [Google Scholar] [CrossRef]
  50. Johnson, D.M.; Jokinen, J.D.; Wang, M.; Pfeiffer, T.; Tretyakova, I.; Carrion, R., Jr.; Griffiths, A.; Pushko, P.; Lukashevich, I.S. Bivalent Junin and Machupo experimental vaccine based on alphavirus RNA replicon vector. Vaccine 2020, 38, 2949–2959. [Google Scholar] [CrossRef]
  51. Brand, D.; Lemiale, F.; Turbica, I.; Buzelay, L.; Brunet, S.; Barin, 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] [PubMed]
  52. Giraud, A.; Ataman-Onal, Y.; Battail, N. 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]
  53. Ajbani, S.P.; Velhal, S.M.; Kadam, R.B.; Patel, V.V.; Lundstrom, K.; Bandivdekar, A.H. Immunogenicity of virus-like Semliki Forest virus replicon particles expressing Indian HIV-1C gag, env and pol RT genes. Immunol Lett. 2017, 190, 221–232. [Google Scholar] [CrossRef]
  54. Knudsen, M.L.; Ljungberg, K.; Tatoud, R.; Weber, J.; Esteban, M.; Liljestrom, P. Alphavirus replicon DNA expressing HIV antigens is an excellent prime for boosting with recombinant modified vaccinia Ankara (MVA) or with HIV gp140 protein antigen. PLoS ONE 2015, 10, e0117042. [Google Scholar] [CrossRef] [PubMed]
  55. Bogers, W.M.; Oostermeijer, H.; Mooij, P.; Koopman, G.; Verschoor, E.J.; Davis, D.; Ulmer, J.B.; Brito, L.A.; Cu, Y.; Bannerjee, K.; et al. Potent immune responses in rhesus macaques induced by nonviral delivery of self-amplifying RNA vaccine expressing HIV type 1 envelope with a cationic emulsion. J. Infect. Dis. 2015, 211, 947–955. [Google Scholar] [CrossRef] [PubMed]
  56. Melo, M.; Porter, E.; Zhang, Y.; Silva, M.; Li, N.; Dobosh, B.; Liquori, A.; Skog, P.; Landais, E.; Menis, S. Immunogenicity of RNA Replicons Encoding HIV Env Immunogens Designed for Self-Assembly into Nanoparticles. Mol. Ther. 2019, 27, 2080–2090. [Google Scholar] [CrossRef]
  57. Anraku, I.; Mokhonov, V.V.; Rattanasena, P.; Mokhonova, E.I.; Leung, J.; Pijlman, G.; Cara, A.; Schroder, W.A.; Khromykh, A.A.; Suhrbier, A. Kunjin replicon-based simian immunodeficiency virus gag vaccines. Vaccine 2008, 26, 3268–3276. [Google Scholar] [CrossRef]
  58. Nilsson, C.; Mäkitalo, B.; Berglund, P.; Bex, F.; Liljeström, P.; Sutter, G. Enhanced simian immunodeficiency virus-specific immune responses in macaques induced by priming with recombinant Semliki Forest virus and boosting with modified vaccinia virus Ankara. Vaccine 2001, 19, 3526–3536. [Google Scholar] [CrossRef]
  59. Gambhira, R.; Keele, B.F.; Schell, J.B.; Hunter, M.J.; Dufour, J.P.; Montefiori, D.C.; Tang, H.; Rose, J.K.; Rose, N.; Marx, P.A. Transmitter/founder simian immunodeficiency virus envelope sequences in vesicular stomatitis and Semliki Forest virus vector immunized rhesus macaques. PLoS ONE 2014, 9, e109678. [Google Scholar] [CrossRef] [Green Version]
  60. 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]
  61. 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] [Green Version]
  62. 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] [PubMed] [Green Version]
  63. Vogel, A.B.; Lambert, L.; Kinnear, E.; Busse, D.; Erbar, S.; Reufer, K.C.; Wicke, L.; Perkovic, M.; Beissert, T.; Haas, H.; et al. Self-amplifying RNA Vaccines Give Equivalent Protection against Influenza to mRNA Vaccines but at Much Lower Doses. Mol. Ther. 2018, 26, 446–455. [Google Scholar] [CrossRef] [Green Version]
  64. Khalil, S.M.; Tonkin, D.R.; Snead, A.T.; Parks, G.D.; Johnston, R.E.; White, L.J. An alphavirus-based adjuvant enhances serum and mucosal antibodies, T cells and protective immunity to influenza virus in neonatal mice. J. Virol. 2014, 88, 9182–9196. [Google Scholar] [CrossRef] [Green Version]
  65. Englezou, P.C.; Sapet, C.; Démoulins, T.; Milona, P.; Ebensen, T.; Schulze, K. Self-amplifying replicon RNA delivery to dendritic cells by cationic lipids. Mol. Ther Nucl Acids 2018, 12, 118–134. [Google Scholar] [CrossRef]
  66. Deming, D.; Sheahan, T.; Heise, M.; Yount, B.; Davis, N.; Sims, A.; Suthar, M.; Harkema, J.; Whitmore, A.; Pickles, R.; et al. Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS Med. 2006, 3, e525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Sheahan, T.; Whitmore, A.; Long, K.; Ferris, M.; Rockx, B.; Funkhouser, B.; 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] [PubMed] [Green Version]
  68. Malczyk, A.H.; Kupke, A.; Prüfer, S.; Scheuplein, V.A.; Hutzler, S.; Kreuz, D. A highly immunogenic and protective Middle East respiratory syndrome coronavirus vaccine based on a recombinant measles virus vaccine platform. J. Virol. 2015, 89, 11654–11667. [Google Scholar] [CrossRef] [Green Version]
  69. Del Valle, J.R.; Devaux, P.; Hodge, G.; Wegner, N.J.; McChesney, M.B.; Cattaneo, R. A vectored measles virus induces hepatitis B surface antigen antibodies while protecting macaques against virus challenge. J. Virol. 2007, 81, 10597–10605. [Google Scholar] [CrossRef] [Green Version]
  70. Reynolds, T.D.; Buonocore, L.; Rose, N.F.; Rose, J.K.; Robek, M.D. Virus-like vesicle-based therapeutic vaccine vectors for chronic hepatis B virus infection. J. Virol. 2015, 89, 10407–10415. [Google Scholar] [CrossRef] [Green Version]
  71. Niedre-Otomere, B.; Bogdanova, A.; Skrastina, D.; Zajakina, A.; Bruvere, R.; Ose, V.; Gerlich, W.H.; Garoff, H.; Pumpens, P.; Glebe, D.; et al. Recombinant Semliki Forest virus vectors encoding hepatitis B small surface and pre-S1 antigens induced broadly reactive neutralizing antibodies. J. Viral Hepat. 2012, 19, 664–673. [Google Scholar] [CrossRef] [PubMed]
  72. Reed, D.S.; Glass, P.J.; Bakken, R.R.; Barth, J.F.; Lind, C.M.; da Silva, L.; Hart, M.K.; Rayner, J.; Alterson, K.; Custer, M.; et al. Combined alphavirus replicon particle vaccine induces durable and cross-protective immune responses against equine encephalitis virus. J. Virol. 2014, 88, 12077–12086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Tretyakova, I.; Tibbens, A.; Jokinen, J.D.; Johnson, D.M.; Lukashevich, J.S.; Pushko, P. Novel DNA-launched Venezuelan equine encephalitis virus vaccine with rearranged genome. Vaccine 2019, 37, 3317–3325. [Google Scholar] [CrossRef] [PubMed]
  74. Tretyakova, I.; Plante, K.S.; Rossi, S.L.; Lawrence, W.S.; Peel, J.E.; Gudjohnsen, S.; Wang, E.; Mirchandani, D.; Tibbens, A.; Lamichhane, T.N. Venezuelan equine encephalitis vaccine with rearranged genome resists reversion and protects non-human primates from viremia after aerosol challenge. Vaccine 2020, 38, 3378–3386. [Google Scholar] [CrossRef]
  75. Kaplan, E.H. Containing 2019-nCoV (Wuhan) coronavirus. Health Care Manag. Sci. 2020, 7. [Google Scholar] [CrossRef]
  76. Yang, Y.; Peng, F.; Wang, R.; Guan, K.; Jiang, T.; Xu, G.; Sun, J.; Chang, C. The deadly coronavirus; The 2003 SARS epidemic and the 2020 novel coronavirus epidemic in China. J. Autoimmun. 2020, 3, 102434. [Google Scholar] [CrossRef]
  77. The World Health Organization. Draft landscape of COVID-19 candidate vaccines. Available online: https://www.who.int/who-documents-detail/draft-landscape-of-covid-19-candidate-vaccines (accessed on 27 May 2020).
  78. Kamrud, K.I.; Custer, M.; Dudek, J.M.; Owens, G.; Alterson, K.D.; Lee, J.S.; Groebner, J.L.; Smith, J.F. Alphavirus replicon approach to promoterless analysis of IRES elements. Virology 2007, 360, 376–387. [Google Scholar] [CrossRef] [Green Version]
  79. Paraskevakou, G.; Allen, C.; Nakamura, T.; Zollman, P.; James, C.D.; Peng, K.W. Epidermal growth factor receptor (EGFR)retargeted measles virus strains effectively target EGFR- or EGFRvIII expressing gliomas. Mol. Ther. 2007, 15, 677–686. [Google Scholar] [CrossRef]
  80. Yamanaka, R.; Zullo, S.A.; Ramsey, J.; Onodera, M.; Tanaka, R.; Blaese, M. 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] [CrossRef] [Green Version]
  81. Yamanaka, R.; Tsuchiya, N.; Yajima, N.; Honma, J.; Hasegawa, H.; Tanaka, R.; Ramsey, J.; Blasé, R.M.; Xanthopoulos, K.G. Induction of ab 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]
  82. Martikainen, M.; Niittykoski, M.; von und zu Frauenberg, M.; Immonen, A.; Koponen, S. MicroRNA-attenuated clone of virulent Semliki Forest virus overcomes antiviral type I interferon in resistant mouse CT-2A glioma. J. Virol. 2015, 89, 10637–10647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zhang, X.; Mao, G.; van den Pol, A.N. Chikungunya-vesicular stomatitis chimeric virus targets and eliminates brain tumors. Virology 2018, 522, 244–259. [Google Scholar] [CrossRef] [PubMed]
  84. McDonald, C.J.; Erlichman, C.; Ingle, J.N.; Rosales, G.A.; Allen, C.; Greiner, S.M. A measles virus vaccine strain derivative as a novel oncolytic agent against breast cancer. Breast Cancer Res. Treat. 2006, 99, 177–184. [Google Scholar] [CrossRef] [PubMed]
  85. 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] [PubMed] [Green Version]
  86. Lachman, L.B.; Rao, X.M.; Kremer, R.H.; Ozpolat, B.; Kirjakova, 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] [CrossRef] [PubMed] [Green Version]
  87. Moran, T.P.; Burgents, J.E.; Long, B. 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] [Green Version]
  88. Vasilevska, J.; Skrastina, D.; Spunde, K.; Garoff, H.; Kozlovska, T.; Zajakina, A. Semliki Forest virus biodistribution in tumor-free and 4T1 mammary tumor-bearing mice: A comparison of transgene delivery by recombinant virus particles and naked RNA replicon. Cancer Gene Ther. 2012, 19, 579–587. [Google Scholar] [CrossRef] [Green Version]
  89. Kramer, M.G.; Masner, M.; Casales, E.; Moreno, M.; Smerdou, C.; Chabalgoity, J.A. Neoadjuvant administration of Semliki Forest virus expressing interleukin-12 combined with Salmonella eradicates breast cancer metastasis and achieves long-term survival in immunocompetent mice. BMC Cancer 2015, 15, 620. [Google Scholar] [CrossRef]
  90. Chikkanna-Gowda, C.P.; Sheahan, B.J.; Fleeton, M.N.; Atkins, G.J. Regression of mouse tumours and inhibition of metastases following administration of a Semliki Forest virus vector with enhanced expression of IL-12. Gene Ther. 2005, 12, 1253–1263. [Google Scholar] [CrossRef] [Green Version]
  91. Velders, M.P.; McElhiney, S.; Cassetti, M.C.; Eiben, G.L.; Higgins, T.; Kovacs, G.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]
  92. Daemen, T.; Riezebos-Brilman, A.; Bungener, L.; Regts, J.; Dontje, B.; Wilschut, J. Eradication of established HPV16-transformed tumours after immunisation with recombinant Semliki Forest virus expressing a fusion protein of E6 and E7. Vaccine 2000, 21, 1082–1088. [Google Scholar] [CrossRef]
  93. Van de Wall, S.; Ljungberg, K.; Ip, P.P.; Boerma, A.; Knudsen, M.L.; Nijman, H.W.; Liljeström, P.; Daemen, T. Potent therapeutic efficacy of an alphavirus replicon DNA vaccine expressing human papilloma virus E6 and E7 antigens. Oncoimmunology 2018, 7, e1487913. [Google Scholar] [CrossRef]
  94. Draghiciu, O.; Walczak, M.; Hoogeboom, B.N.; Franken, K.L.; Melief, K.J.; Nijman, H.W. Therapeutic immunization and local low-dose tumor irradiation, a reinforcing combination. Int. J. Cancer 2014, 134, 859–872. [Google Scholar] [CrossRef] [PubMed]
  95. Draghiciu, O.; Boerma, A.; Hoogeboom, B.N.; Nijman, H.W.; Daemen, T. A rationally designed combined treatment with an alphavirus-based cancer vaccine, sunitinib and low-dose tumor irradiation completely blocks tumor development. Oncoimmunology 2015, 4, e1029699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Ying, H.; Zaks, T.Z.; Wang, R.-F.; Irvine, K.R.; Kammula, U.S.; Marincola, F.M. Cancer therapy using a self-replicating RNA vaccine. Nat. Med. 1999, 5, 823–827. [Google Scholar] [CrossRef] [PubMed]
  97. Lyons, J.A.; Sheahan, B.J.; Galbraith, S.E. 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] [CrossRef]
  98. Granot, T.; Yamanashi, Y.; Meruelo, D. Sindbis viral vectors transiently deliver tumor-associated antigens to lymph nodes and elicit diversified antitumor CD8+ T-cell immunity. Mol. Ther. 2014, 22, 112–122. [Google Scholar] [CrossRef]
  99. Hoang-Le, D.; Smeenk, L.; Anraku, I.; Pijlman, G.P.; Wang, X.P.; de Vrij, J. A Kunjin replicon vector encoding granulocyte macrophage colony-stimulating factor for intra-tumoral gene therapy. Gene Ther. 2009, 16, 190–199. [Google Scholar] [CrossRef]
  100. Murphy, A.M.; Morris-Downes, M.M.; Sheahan, B.J.; Atkins, G.J. Inhibition of human lung carcinoma cell growth by apoptosis induction using Semliki Forest virus recombinant particles. Gene Ther. 2000, 7, 1477–1482. [Google Scholar] [CrossRef] [Green Version]
  101. Määttä, A.M.; Mäkinen, K.; Ketola, A.; Liimatainen, T.; Yongabi, F.N.; Vähä-Koskela, M. Replication competent Semliki Forest virus prolongs survival in experimental lung cancer. Int. J. Cancer 2008, 123, 1704–1711. [Google Scholar] [CrossRef]
  102. Patel, M.R.; Jacobson, B.A.; Ji, Y.; Drees, J.; Tang, S.; Xiong, K. Vesicular stomatitis virus expressing interferon-β is oncolytic and promotes antitumor immune responses in a syngeneic murine model of non-small cell lung cancer. Oncotarget 2015, 6, 33165–33177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Patel, M.R.; Jacobson, B.A.; Belgum, H.; Raza, A.; Sadiq, A.; Drees, J.; Wang, H.; Jay-Dixon, J.; Etchison, R.; Federspiel, M.J.; et al. Measles vaccine strains for virotherapy of non-small cell lung carcinoma. J. Thorac. Oncol. 2014, 9, 1101–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Fujiyuki, T.; Yoneda, M.; Amagai, Y.; Obayashi, K.; Ikeda, F.; Shoji, K. A measles virus selectively blind to signalling lymphocytic activation molecule shows anti-tumor activity against lung cancer cells. Oncotarget 2015, 6, 24895–24903. [Google Scholar] [CrossRef] [Green Version]
  105. McAllister, A.; Arbetman, A.E.; Mandl, S.; Pena-Rossi, C.; Andino, R. Recombinant yellow fever viruses are effective therapeutic vaccines for treatment of murine solid tumors and pulmonary metastases. J. Virol. 2000, 74, 9197–9205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Avogadri, F.; Merghoub, T.; Maughan, M.F.; Hirschhorn-Cymerman, D.; Morris, J.; Ritter, E. 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] [CrossRef] [Green Version]
  107. Avogadri, F.; Zappasodi, R.; Yang, A.; Budhu, S.; Malandro, N.; Hisrchhorn-Cymerman, D. Combination of alphavirus replicon particle-based vaccination with immunomodulatory antibodies: Therapeutic activity in the B16 melanoma mouse model and immune correlates. Cancer Immunol. Res. 2014, 2, 448–458. [Google Scholar] [CrossRef] [Green Version]
  108. Yin, X.; Wang, W.; Zhu, X.; Wang, Y.; Wu, S.; Wang, Z. Synergistic antitumor efficacy of combined DNA vaccines targeting tumor cells and angiogenesis. Biochem. Biophys. Res. Comm. 2015, 465, 239–244. [Google Scholar] [CrossRef]
  109. Allaqui, F.; Achard, C.; Panterne, C.; Combredet, C.; Labarrière, N.; Dréno, B.; Elgaaied, A.B.; Pouliquen, D.; Tangy, F.; Fonteneau, J.F.; et al. Modulation of the Type I Interferon Response Defines the Sensitivity of Human Melanoma Cells to Oncolytic Measles Virus. Curr. Gene Ther. 2017, 16, 419–428. [Google Scholar] [CrossRef]
  110. Ammour, Y.; Ryabaya, O.; Shchetinina, Y.; Prokofeva, E.; Gavrilova, M.; Khochenkov, D.; Vorobyev, D.; Faizuloev, E.; Shohin, I.; Zverev, V.V.; et al. The Susceptibility of Human Melanoma Cells to Infection with the Leningrad-16 Vaccine Strain of Measles Virus. Viruses 2020, 12, E173. [Google Scholar] [CrossRef] [Green Version]
  111. Muik, A.; Kneiske, I.; Werbizki, M.; Wilflingseder, D.; Giroglou, T.; Ebert, O. Pseudotyping vesicular stomatitis virus with lymphocytic choriomeningitis virus glycoproteins enhances infectivity for glioma cells and minimizes neurotropism. J. Virol. 2011, 85, 5679–5684. [Google Scholar] [CrossRef] [Green Version]
  112. Kimpel, J.; Urbiola, C.; Koske, I.; Tober, R.; Banki, Z.; Wollmann, G. The Oncolytic virus VSV-GP is effective against malignant melanoma. Viruses 2018, 10, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ilett, E.; Kottke, T.; Thompson, J.; Rajani, K.; Zaidi, S.; Evgin, L.; Coffey, M.; Ralph, C.; Diaz, R.; Pandha, H.; et al. Prime-boost using separate oncolytic viruses in combination with checkpoint blockade improves anti-tumour therapy. Gene Ther. 2017, 24, 21–30. [Google Scholar] [CrossRef] [PubMed]
  114. Dold, C.; Rodriguez Urbiola, C.; Wollmann, G.; Egerer, L.; Muik, A.; Bellmann, L.; Fiegl, H.; Marth, C.; Kimpel, J.; von Laer, D. Application of interferon modulators to overcome partial resistance to ovarian cancers to VSV-GP oncolytic viral therapy. Mol. Ther Oncolytics 2016, 3, 16021. [Google Scholar] [CrossRef] [PubMed]
  115. Hasegawa, K.; Nakamura, T.; Harvey, M.; Ikeda, Y.; Oberg, A.; Figini, M.; Canevari, S.; Hartmann, L.C.; Peng, K.W. The use of a tropism-modified measles virus in folate receptor-targeted virotherapy of ovarian cancer. Clin Cancer Res. 2006, 12, 6170–6178. [Google Scholar] [CrossRef] [Green Version]
  116. Granot, T.; Meruelo, D. The role of natural killer cells in combinatorial anti-cancer therapy using Sindbis viral vector and irinotecan. Cancer Gene Ther. 2012, 19, 588–591. [Google Scholar] [CrossRef] [Green Version]
  117. Zhang, Y.Q.; Tsai, Y.C.; Monie, A.; Wu, T.C.; Hung, C.F. Enhancing the therapeutic effect against ovarian cancer through a combination of viral oncolysis and antigen-specific immunotherapy. Mol. Ther. 2010, 18, 692–699. [Google Scholar] [CrossRef]
  118. Venticinque, L.; Meruelo, D. Sindbis viral vector induced apoptosis requires translational inhibition and signaling through Mcl-1 and Bak. Mol. Cancer 2010, 9, 37. [Google Scholar] [CrossRef] [Green Version]
  119. McGray, A.J.R.; Huang, R.-Y.; Battaglia, S.; Eppolito, C.; Miliotto, A.; Stephenson, K.B.; Lugade, A.A.; Webster, G.; Lichty, B.D.; Seshadri, M.; et al. Oncolytic Maraba virus armed with tumor antigen boosts vaccine priming and reveals diverse therapeutic response patterns when combined with checkpoint blockade in ovarian cancer. J. Immunother Cancer 2019, 17, 189. [Google Scholar] [CrossRef] [Green Version]
  120. Murphy, A.M.; Besmer, D.M.; Moerdyk-Schauwecker, M.; Moestl, N.; Ornelles, D.A.; Mukherjee, P. Vesicular stomatitis virus as an oncolytic agent against pancreatic ductal adenocarcinoma. J. Virol. 2012, 86, 3073–3087. [Google Scholar] [CrossRef] [Green Version]
  121. Felt, S.A.; Droby, G.N.; Grdzelishvili, V.Z. Ruxolitinib and polycation combination treatment overcomes multiple mechanisms of resistance of pancreatic cancer cells to oncolytic vesicular stomatitis virus. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [Green Version]
  122. Msaouel, P.; Iankov, I.D.; Allen, C.; Morris, J.C.; von Messling, V.; Cattaneo, R. Engineered measles virus as a novel oncolytic therapy against prostate cancer. Prostate 2009, 69, 82–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Durso, R.J.; Andjelic, S.; Gardner, J.P.; Margitich, D.J.; Donovan, G.P.; Arrigale, R.R. 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] [PubMed] [Green Version]
  124. 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] [PubMed] [Green Version]
  125. Garcia-Hernandez, M.L.; Gray, A.; Hubby, B.; Klinger, O.J.; Kast, W.M. 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] [PubMed] [Green Version]
  126. Riabov, V.; Treykova, I.; Alexander, R.B.; Pushko, P.; Klyushnenkova, E.N. Anti-tumor effect of the alphavirus-based virus-like particle vector expressing prostate-specific antigen in a HLA-DR transgenic mouse model of prostate cancer. Vaccine 2015, 33, 5386–5395. [Google Scholar] [CrossRef] [Green Version]
  127. Urbiola, C.; Santer, F.R.; Petersson, M.; van der Pluijm, G.; Horninger, W.; Erlmann, P. Oncolytic activity of the rhabdovirus VSV-GP against prostate cancer. Int. J. Cancer 2018, 143, 1786–1796. [Google Scholar] [CrossRef]
  128. Son, H.A.; Zhang, L.; Cuong, B.K.; Van Tong, H.; Cuong, L.D.; Hang, N.T.; Nhung, H.T.M.; Yamamoto, N.; Toan, N.L. Combination of Vaccine-Strain Measles and Mumps Viruses Enhances Oncolytic Activity against Human Solid Malignancies. Cancer Invest. 2018, 7, 106–117. [Google Scholar] [CrossRef]
  129. Bernstein, D.I.; Reap, E.A.; Katen, K.; Watson, A.; Smith, K.; Norberg, P. Randomized, double-blind, phase I trial on an alphavirus replicon vaccine for cytomegalovirus in CMV negative volunteers. Vaccine 2010, 28, 484–493. [Google Scholar] [CrossRef]
  130. Wecker, M.; Gilbert, P.; Russell, N.; Hural, J.; Allen, M.; Pensiero, M. Phase I safety and immunogenicity evaluations of an alphavirus replicon HIV-1 subtype C gag vaccine in healthy HIV-1-uninfected adults. Clin. Vaccine Immunol. 2012, 19, 1651–1660. [Google Scholar] [CrossRef] [Green Version]
  131. Regules, J.A.; Beigel, J.H.; Paolino, K.M.; Voell, J.; Castellano, A.R.; Hu, Z.; Munoz, P.; Moon, J.E.; Ruck, R.C.; Bennett, J.W.; et al. A recombinant vesicular stomatitis virus ebola vaccine. N. Engl. J. Med. 2017, 376, 330–341. [Google Scholar] [CrossRef]
  132. ElSherif, M.S.; Brown, C.; MacKinnon-Cameron, D.; Li, L.; Racine, T.; Alimonti, J.; Rudge, T.L.; Sabourin, C.; Silvera, P.; Hooper, J.W.; et al. Assessing the safety and immunogenicity of recombinant vesicular stomatitis virus Ebola vaccine in healthy adults: A randomized clinical trial. CMAJ 2017, 189, E819–E827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Dahlke, C.; Kasonta, R.; Lunemann, S.; Krahling, V.; Zinser, M.E.; Biedenkopf, N.; Fehling, S.K.; Ly, M.L.; Rechtien, A.; Stubbe, H.C.; et al. Dose-dependent T-cell dynamics and cytokine cascade following rVSV-ZEBOV immunization. EBioMedicine 2017, 19, 107–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Poetsch, J.H.; Dahlke, C.; Zinser, M.E.; Kasonta, R.; Lunemann, S.; Rechtien, A.; Ly, M.L.; Stubbe, H.C.; Krähling, V.; Biedenkopf, N.; et al. Detectable Vesicular Stomatitis Virus (VSV)-Specific Humoral and Cellular Immune Responses Following VSV-Ebola Virus Vaccination in Humans. J. Infect. Dis. 2019, 219, 556–561. [Google Scholar] [CrossRef] [PubMed]
  135. Huttner, A.; Dayer, J.A.; Yerly, S.; Combescure, C.; Auderset, F.; Desmeules, J. The effect of dose on the safety and immunogenicity of the VSV Ebola candidate vaccine: A randomised double-blind, placebo-controlled phase 1/2 trial. Lancet Infect. Dis. 2015, 15, 1156–1166. [Google Scholar] [CrossRef]
  136. Kennedy, S.B.; Bolay, F.; Kieh, M.; Grandits, G.; Badio, M.; Ballou, R.; Eckes, R.; Feinberg, M.; Follmann, D.; Grund, B.; et al. Phase 2 placebo-controlled trial of two vaccines to prevent ebola in Liberia. N. Engl. J. Med. 2017, 377, 1438–1447. [Google Scholar] [CrossRef] [Green Version]
  137. Henao-Restrepo, A.M.; Longini, I.M.; Egger, M.; Dean, N.E.; Edmunds, W.J.; Camacho, A.; Carroll, M.W.; Doumbia, M.; Draguez, B.; Duraffour, S. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: Interim results from the Guinea ring vaccination cluster-randomised trial. Lancet 2015, 386, 857–866. [Google Scholar] [CrossRef]
  138. Henao-Restrepo, A.M.; Camacho, A.; Longini, I.M.; Watson, C.H.; Edmunds, W.J.; Egger, M.; Carroll, M.W.; Dean, N.E.; Diatta, I.; Doumbia, M.; et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: Final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ca Suffit!). Lancet 2017, 389, 505–518. [Google Scholar] [CrossRef] [Green Version]
  139. Metzger, W.G.; Vivas-Martinez, S. Questionable efficacy of the rVSV-ZEBOV Ebola vaccine. Lancet 2018, 391, 1021. [Google Scholar] [CrossRef] [Green Version]
  140. Longini, I.M.; Røttingen, J.A.; Kieny, M.P.; Edmunds, W.J.; Henao-Restrepo, A.M. Questionable efficacy of the rVSV-ZEBOV Ebola vaccine – Authors’ reply. Lancet 2018, 391, 1021–1022. [Google Scholar] [CrossRef] [Green Version]
  141. Widdowson, M.; Schrag, S.; Carter, R.; Carr, W.; Legardy-Williams, J.; Gibson, L.; Lisk, D.R.; Jalloh, M.I.; Bash-Taqi, D.A.; Kargbo, S.A.; et al. Implementing an Ebola vaccine study—Sierra Leone. MMWR Suppl. 2016, 65, 98–106. [Google Scholar] [CrossRef]
  142. Halperin, S.A.; Arribas, J.R.; Rupp, R.; Andrews, C.P.; Chu, L.; Das, R.; Simon, J.K.; Onorato, M.T.; Liu, K.; Martin, J.; et al. Six-month safety data of recombinant vesicular stomatitis virus-zaire ebola virus envelope glycoprotein vaccine in a phase 3 double-blind, placebo-controlled randomized study in healthy adults. J. Infect. Dis. 2017, 215, 1789–1798. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, G.L.; Coates, E.E.; Plummer, S.H.; Carter, C.A.; Berkowitz, N.; Conan-Cibotti, M.; Cox, J.H.; Beck, A.; O’Callahan, M.; Andrews, C.; et al. Effect of a Chikungunya Virus-Like Particle Vaccine on Safety and Tolerability Outcomes: A Randomized Clinical Trial. JAMA 2020, 323, 1369–1377. [Google Scholar] [CrossRef] [PubMed]
  144. Hannaman, D.; Dupuy, L.C.; Ellefsen, B.; Schmaljohn, C.S. A Phase 1 clinical trial of a DNA vaccine for Venezuelan equine encephalitis delivered by intramuscular or intradermal electroporation. Vaccine 2016, 34, 3607–3612. [Google Scholar] [CrossRef] [PubMed]
  145. Clinical Trials. Gov. Viral Therapy in Treating Patients with Recurrent Glioblastoma Multiforme. Available online: http://www.clinicaltrials.gov/ct2/show/NCT00390299 (accessed on 6 May 2020).
  146. Lundstrom, K. Biology and application of alphaviruses in gene therapy. Gene Ther. 2005, 12 (Suppl. S1), S92–S97. [Google Scholar] [CrossRef]
  147. Heinzerling, L.; Kunzi, V.; Oberholzer, P.A.; Kundig, T.; Naim, H.; Dummer, R. Oncolytic measles virus in cutaneous T-cell lymphomas mounts antitumor immune responses in vivo and targets interferon resistant tumor cells. Blood 2005, 106, 2287–2289. [Google Scholar] [CrossRef] [Green Version]
  148. Galanis, E.; Hartmann, L.C.; Cliby, W.A.; Long, H.J.; Peethambaram, P.P.; Barrette, B.A.; Kaur, J.S.; Haluska, P.J., Jr.; Aderca, I.; Zollman, P.J.; et al. Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer Res. 2010, 70, 875–882. [Google Scholar] [CrossRef] [Green Version]
  149. Russell, S.J.; Federspiel, M.J.; Peng, K.W.; Tong, C.; Dingli, D.; Morice, W.G.; Lowe, V.; O’Connor, M.K.; Kyle, R.A.; Leung, N.; et al. Remission of disseminated cancer after systemic oncolytic virotherapy. Mayo Clinic Proc. 2014, 89, 926–933. [Google Scholar] [CrossRef] [Green Version]
  150. Morse, M.A.; Hobelka, A.C.; Osada, T.; Berglund, P.; Hubby, B.; Negri, S. 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. Investig. 2010, 120, 3234–3241. [Google Scholar] [CrossRef]
  151. Slovin, S.F.; Kehoe, M.; Durso, R.; Fernandez, C.; Olson, W.; Gao, J.P. A phaseI 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]
  152. Kelvin, A.A. Outbreak of Chikungunya in the Republic of Congo and the global picture. J. Infect. Dev. Ctries. 2011, 5, 441–444. [Google Scholar] [CrossRef] [Green Version]
  153. Jansen, K.A. The 2005–2007 Chikungunya epidemic in Reunion: Ambiguous etiologies, memories, and meaning-making. Med. Anthropol. 2013, 32, 174–189. [Google Scholar] [CrossRef] [PubMed]
  154. Weaver, S.C.; Salas, R.; Rico-Hesse, R.; Ludwig, G.V.; Oberste, M.S.; Boshell, J.; Tesh, R.B. Re-emergence of epidemic Venezuelan equine encephalomyelitis in South America. VEE Study Group. Lancet 1996, 348, 436–440. [Google Scholar] [CrossRef]
  155. Stief, A.E.; McCart, J.A. Oncolytic virotherapy for multiple myeloma. Expert. Opin. Biol. Ther. 2008, 8, 463–473. [Google Scholar] [CrossRef] [PubMed]
  156. Russell, S.J.; Peng, K.W. Measles virus for cancer therapy. Curr. Top. Microbiol. Immunol. 2009, 330, 213–241. [Google Scholar] [PubMed] [Green Version]
  157. Therasse, P.; Arbuck, S.G.; Eisenhauer, E.A.; Wanders, J.; Kaplan, R.S.; Rubinstein, L.; Verweij, J.; Van Glabbeke, M.; van Oosterom, A.T.; Christian, M.C.; et al. New guidelines to evaluate the response to treatment in solid tumors European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J. Natl. Cancer Inst. 2000, 92, 205–216. [Google Scholar] [CrossRef] [Green Version]
  158. Markman, M.; Webster, K.; Zanotti, K.; Peterson, G.; Kulp, B.; Belinson, J. Survival following the documentation of platinum and taxane resistance in ovarian cancer: A single institution experience involving multiple phase 2 clinical trials. Gynecol. Oncol. 2004, 93, 699–701. [Google Scholar] [CrossRef]
  159. Lundstrom, K. Replicon RNA Viral Vectors as Vaccines. Vaccines 2016, 4, 39. [Google Scholar] [CrossRef]
  160. 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] [Green Version]
  161. 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] [CrossRef] [Green Version]
  162. Lundstrom, K. Coronaviruses Pandemic – Therapy and Vaccines. Biomedicines 2020, 8, 109. [Google Scholar] [CrossRef]
  163. 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. DNA immunization against herpes simplex virus: Enhanced efficacy using a Sindbis virus-based vector. J. Virol. 1998, 72, 950–958. [Google Scholar] [CrossRef] [Green Version]
  164. Nosaki, K.; Hamada, K.; Takashima, Y.; Sagara, M.; Matsumura, Y.; Miayamoto, S.; Hijikata, Y.; Okazaki, T.; Nakanishi, Y.; Tani, K. A novel, polymer-coated oncolytic measles virus overcomes immune suppression and induces robust antitumor activity. Mol. Ther. Oncolytics 2016, 3, 16022. [Google Scholar] [CrossRef] [PubMed]
  165. Ketola, A.; Schlesinger, S.; Wahlfors, J. Properties of Sindbis virus vectors produced with a chimeric split helper system. Int. J. Mol. Med. 2005, 15, 999–1003. [Google Scholar] [CrossRef] [PubMed]
  166. 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] [CrossRef] [PubMed] [Green Version]
Figure 1. Semliki Forest virus-based expression systems. A. Schematic illustration of expression and helper vectors. Dark triangle, SP6 RNA polymerase promoter; light triangle, Semliki Forest virus (SFV) 26S subgenomic promoter. B. RNA replicon (naked RNA or liposome-encapsulated RNA), recombinant virus particles or DNA replicon (DNA plasmid) can be used for transfection/infection of cell lines and immunization of animals and humans.
Figure 1. Semliki Forest virus-based expression systems. A. Schematic illustration of expression and helper vectors. Dark triangle, SP6 RNA polymerase promoter; light triangle, Semliki Forest virus (SFV) 26S subgenomic promoter. B. RNA replicon (naked RNA or liposome-encapsulated RNA), recombinant virus particles or DNA replicon (DNA plasmid) can be used for transfection/infection of cell lines and immunization of animals and humans.
Ijms 21 05130 g001
Table 1. Examples of preclinical immunizations against viral diseases.
Table 1. Examples of preclinical immunizations against viral diseases.
VirusTarget/AntigenVector TypeFindingRef
Flaviviruses
DENVE85 ectodomain VEE VLPs Dengue protection in mice [35]
DV2MVNeutralizing antibodies[36]
DV1-4MVDengue protection in mice [37]
Zika virusprM. ENLC-VEE RNAZIKV protection in mice[38]
M, EVSV VLPsZIKV protection in mice [39]
Filoviruses
EBOVGP/D637LKUN VLPsEBOV protection in 75% of primates[40]
EBOV-GPVSV VLPsEBOV protection in macaques[41]
EBOV-GPVSV VLPsEBOV protection in primates[42]
EBOV-GPVSV-VLPsEBOV protection in macaques[43]
EBOV-NPVEE VLPsEBOV protection in mice[44]
EBOV-GP, NPVEE VLPsEBOV protection in mice and guinea pigs[45]
EBOV-GP, VP40SFV DNANeutralizing antibodies[46]
MARVMARV-GPVSV VLPsMARV protection in primates[42]
SUDVSUDV-GPVEE VLPsSUDV protection in macaques[43]
Arenaviruses
LASVLASV-GPCVSV VLPsLASV protection in guinea pigs[47]
LASV-GPC/ΔGfibVEE VLPsLASV protection in mice[48]
LASV-GPCLASV VLPsLASV protection in guinea pigs[49]
JUNVJUNV-GPCVEE VLPsJUNV protection in guinea pigs[50]
MACVMACV-GPCVEE VLPsMACV protection in guinea pigs[50]
Lentiviruses
HIV-1HIV-1 Env gp100SFV VLPsHumoral immune responses[51]
HIV-1 EnvSFV RNAAntibody responses, mAbs[52]
Env/Gag/PolRTSFV VLPs/RNAAntigen-specific responses: VLPs > RNA[53]
Env/GagPolNefSFV DNASuperior to MVA, HIV gp40[54]
TV1 gp140VEE* RNA-NPsImmunogenicity in macaques[55]
Env gp120VEE RNA-NPsgp120-specific antibodies[56]
SIVGag-polKUN VLPsSIV protection in mice[57]
Env, Gag-pol, Nef, Rev, TatSFV + MVA VLPsHumoral and cellular responses[58]
Gag-polVSV + SFV VLPsPartial SIV protection in macaques[59]
Influenza
InfluenzaNPSFV VLPsMucosal immune response[60]
HAVEE-VLPsProtection in chicken[61]
HASFV RNAProtection in mice[62]
HAVEE RNAProtection in mice[63]
iFluVEE VLPsEnhanced immune response [64]
NPCSFV RNA-NPsImmune response in mice[65]
Coronaviruses
SARS-CoVSARS-CoV SVEE VLPsProtection in mice[66]
SARS-CoV SVEE VLPsProtection in mice[67]
MERS-CoVMERS-CoV SMVProtection in mice[68]
Hepatotropic
HBVHBsAgMVPartial protection[69]
MHB, HBcAgSFV-G VLPsProtection in mice by MHB[70]
HBV SSFV VLPsNeutralization of HBV infectivity[71]
Alphaviruses
CHIKVE1-E3, CVSVCHIKV protection in mice [39]
VEEVEE RepliconVEE VLPsProtection in mice, macaques[72]
EEEEEE RepliconEEE VLPsProtection in mice, macaques[72]
WEEWEE RepliconWEE VLPsWeak protection in mice, macaques[72]
VEEVEE V4020VEE DNAVEE protection in mice[73]
VEE V4020VEE DNAVEE protection in macaques[74]
CHIKV, Chikungunya virus; CSFV, Classical swine fever virus; DENV, Dengue virus; EBOV, Ebola virus; EEE, Eastern equine encephalitis virus; JUNV, Junin virus; HA, hemagglutinin; HBV, hepatitis B virus; HBcAg, hepatitis B core antigen; HBsAg, hepatitis B surface antigen; HIV, human immunodeficiency virus; iFlu, inactivated influenza virus; LASV, Lassa virus; MACV, Machupo virus; MARV, Marburg virus; mAbs, monoclonal antibodies; MERS-CoV, Middle East respiratory syndrome-coronavirus; MHB, Middle surface HBV glycoprotein; NLC, nanostructured lipid carrier; NP, nucleoprotein; SARS-CoV, severe acute respiratory syndrome-coronavirus; SFV, Semliki Forest virus; SFV-G, Semliki Forest virus with VSV G envelope; SUDV, Sudan virus; VEE, Venezuelan equine encephalitis virus; VEE*, VEE vector containing SIN 3′ untranslated and packaging signal sequences; VLPs, virus-like particles; VSV, vesicular stomatitis virus; WEE, Western equine encephalitis virus; ZIKV, Zika virus.
Table 2. Examples of preclinical immunizations targeting cancers.
Table 2. Examples of preclinical immunizations targeting cancers.
CancerTarget/AntigenVectorResponseRef
Brain
GBMSLAM, EGFRMVTumor regression, prolonged survival[79]
GBMEndostatinSFV VLPsTumor regression, prolonged survival[80]
GBMIL-18, IL-12DC-SFVEnhanced antitumor activity[81]
GBMmiR124SFV4Viral replication in tumors, tumor growth inhibition, prolonged survival[82]
GBMChimeric VLPsVSVΔG-CHIKVTumor cell infection, prolonged survival[83]
Breast
MDA-MBCEAMVProlonged survival in mice[84]
A2L2HER2/neuSIN VLPsProlonged survival in mice[85]
A2L2HER2/neuSIN DNAReduced tumor incidence and mass,
protection with 80% less DNA
[86]
A2L2neuΔVEE VLPs + DCsSpecific Abs, tumor regression in mice[87]
4T1luciferaseSFV-VLPs, -RNATumor targeting, re-administration[88]
4T1IL-12SFV-IL12, LVR01Long-term survival, lung metastasis prevention[89]
4T1IL-12SFV VLPsTumor regression, metastases[90]
Cervical
HPVHPV-16 E7VEE VLPsPrevention of tumor development[91]
HPVHPV E6-E7SFV VLPsComplete elimination of tumors[92]
HPVHPV E6/7SFV DNA85% of immunized mice tumor-free[93]
HPVHPV E6, 7SFV VLPs, RadEnhanced antitumor activity[94]
HPVHPV E6, 7SFV VLPs, Rad+
sunitinib
100% tumor-free survival[95]
Colon
CT26LacZSFV RNAAntibody response, protection in mice[96]
CT26VEGFR-2SFV VLPsInhibition of tumor growth[97]
CT26VEGFR-2 + IL-4SFV VLPsSuperior survival of mice[97]
CT26IL-12SFV VLPsTumor cell necrosis[90]
CT26LacZSIN VLPsTherapeutic effect in mouse model[98]
CT26G-CSFKUN VLPsCure in >50% of immunized mice[99]
Lung
H358aEGFPSFV VLPsComplete regression in 3 out of 7 mice[100]
A549EGFPSFV (VA7)Prolonged survival in mice[101]
CT26.CL25LacZSIN VLPsProtection against tumor challenges[98]
H2009, A549GFP, IFNβVSV VLPsReduced tumor growth in mice[102]
LM2IFNβVSV VLPsProlonged survival, cure of 30% of mice[102]
A549, LLCGFP, CEAMVTumor regression in mice[103]
NCI-H441EGFPMV-SLAMblindSuppression of tumor growth in mice[104]
Melanoma
B16-OVAG-CSFKUN VLPsCure in >50% of immunized mice[99]
B16-OVASIINFEKLYVF VLPsProtection against malignant melanoma[105]
B16TRP-2VEE VLPsProlonged survival in mice[106]
B16TRP-2VEE VLPs + anti-
CTLA-4/GITR
Tumor regression in mice[107]
B16VEGFR-2, IL-12+
Sur, β-hCG
SFV DNAProlonged survival in mice[108]
B16ASMEL, anti-PD1Oncolytic VSV + ReovirusExtended survival, long-term cure in mice [113]
Mel ZMV L-16MVInhibition of tumor growth[110]
A375, B16GFP, LucVSV-GP-LCMVProlonged survival in mice[112]
Ovarian
A2780LucVSV-GP-LCMVOncolytic activity in vitro and in vivo[114]
ARH77EGFPMV-alphaFRIncreased survival rate in vivo[115]
ES2IL-12 SIN VLPs, CPT-11Long-term survival in SCID mice[116]
MOSECOVASFV VLPs + VVEnhanced immune responses in mice[117]
MOSECGFP, LucSIN VLPsInduction of cellular stress, apoptosis[118]
ID9-mp1, ID8DCTOncolytic MarabaRobust immune response, tumor control[119]
Pancreatic
Pan02GFP, LucSIN VLPsInduction of cellular stress, apoptosis[118]
Panc-1, GFPVSV VLPsOncolytic activity in vitro and in vivo[120]
Su.86.86GFPVSV VLPsOncolytic activity in vitro and in vivo[120]
KLM1, SLAMMV-SLAMblindSuppression of tumor growth in mice[122]
Capan-2SLAMMV-SLAMblindSuppression of tumor growth in mice[122]
Prostate
PC-3 [122]
LnCaPCEAMVProlonged survival of mice[123]
TRAMP-CPSMAVEE VLPsRobust immune response in mice[124]
TRAMPSTEAPVEE VLPsProlonged survival in mice[125]
TRAMP-PSAPSCAVEE VLPs90% survival rate in mice[125]
Du145, 22Rv1PSAVEE VLPsTumor growth delay in mice[126]
TRAMP, PC-3LucVSV-GP-LCMVLong-term remission in mice[127]
LucVSV-GP-LCMVRemission in subcutaneous tumors and bone metastases [127]
PC-3MV, MuVMV + MuVProlonged survival in mice[128]
ASMEL, cDNA library of melanoma antigens; CEA, carcinoembryonic antigen; CHIKV, Chikungunya virus; CPT-11, topoisomerase inhibitor irinotecan; CTLA-4, cytotoxic T lymphocyte antigen-4; DC, dendritic cell; DCT, dopachrome tautomerase; EGFP, enhanced green fluorescent protein; EGFR, epidermal growth factor receptor; GBM, glioblastoma; GITR, glucocorticoid-induced tumor necrosis factor receptor; HPV, human papilloma virus; IL-4, interleukin-4; IL-12, interleukin-12; IL-18, interleukin-18; LLC, Lewis lung cancer; Luc, luciferase; MDA-MB, MDA-MB-231 tumor; MOSEC, ovarian surface epithelial carcinoma; MuV, mumps virus; MV, measles virus; MV-L16, Leningrad-16 strain; MV-SLAMblind, MV vector unable to bind to SLAM; Pan02, pancreatic tumor cell line; PSCA, prostate stem cell antigen; PSMA, prostate-specific membrane antigen; Rad, radiation; SFV, Semliki Forest virus; SFV (VA7), attenuated replication-proficient SFV; SIN, Sindbis virus; SLAM, signaling lymphocyte activating molecule; TRAMP-C, transgenic adenocarcinoma of the mouse prostate TRP-2, tyrosine related protein-2; VEE, Venezuelan equine encephalitis virus; VEGFR-2, vascular endothelial growth factor receptor-2; VLPs, virus-like particles; VSV, vesicular stomatitis virus; VSVΔG, VSV where the VSV-G protein replaced with CHIKV envelope; VSV-GP-LCMV, VSV where the VSV-G protein replaced by lymphocytic choriomeningitis virus envelope glycoprotein; YFV, yellow fever virus.
Table 3. Clinical trials conducted for self-amplifying RNA virus vectors.
Table 3. Clinical trials conducted for self-amplifying RNA virus vectors.
IndicationVector/AntigenPhaseResponseRef
Infections
CMVVEE-gB/p55-IE1Phase ICMV-specific Abs[129]
AIDSVEE-HIV-gagPhase IModest antibody responses[130]
EBOVVSV-ZEBOVPhase IAnti-ZEBOV Abs[131]
VSV∆G-ZEBOVPhase ISustainable IgG titers for 180 days[132]
VSV-ZEBOVPhase IEBOV-specific neutralizing Abs[133]
VSV-ZEBOVPhase I/IILower dose, improved tolerability[135]
VSV∆G-ZEBOVPhase IIAb-response in 80% of vaccines[136]
VSV-ZEBOVPhase III100% protection against EVD[137]
VSV-ZEBOVPhase IIISubstantial protection against EVD[138]
VSV-ZEBOVPhase II/IIINo EVD, no vaccine-related AEs[141]
VSV∆G-ZEBOVPhase IIISafe, no vaccine-related AEs[142]
CHIKCHIK VLPsPhase IISafe, well tolerated[143]
VEEVEE DNA Phase IVEE-specific neutralizing Abs[144]
Cancers
GlioblastomaMV-CEAPhase INo dose-limiting toxicity[145]
KidneyLipoVIL12Phase ISafe, tumor targeting[146]
LymphomaMVEZPhase ITumor regression[147]
MelanomaLipoVIL12Phase ISafe, tumor targeting[146]
MyelomaMV-NISPhase IComplete remission in one patient[148]
OvarianMV-CEAPhase I/IIStable disease (high dose treatment)[149]
PancreasVEE-CEAPhase IProlonged survival[150]
ProstateVEE-PSMAPhase INeutralizing Abs against PSMA[151]
Abs, antibodies; AEs, adverse events; CEA, carcinoembryonic antigen; CHIKV, Chikungunya virus; CMV, cytomegalovirus; EBOV, Ebola virus; EVD, Ebola virus disease; HIV, human immunodeficiency virus; LipoVIL12, liposome-encapsulate SFV expressing IL-12; MV, measles virus; MVEZ, MV Edmonston-Zagreb strain; NIS, sodium iodide symporter; PSMA, prostate-specific membrane antigen; SFV, Semliki Forest virus; VEE, Venezuelan equine encephalitis virus; VLPs, virus-like particles; VSV, vesicular stomatitis virus; ZEBOV, glycoprotein of Zaire EBOV.

Share and Cite

MDPI and ACS Style

Lundstrom, K. Self-Amplifying RNA Viruses as RNA Vaccines. Int. J. Mol. Sci. 2020, 21, 5130. https://doi.org/10.3390/ijms21145130

AMA Style

Lundstrom K. Self-Amplifying RNA Viruses as RNA Vaccines. International Journal of Molecular Sciences. 2020; 21(14):5130. https://doi.org/10.3390/ijms21145130

Chicago/Turabian Style

Lundstrom, Kenneth. 2020. "Self-Amplifying RNA Viruses as RNA Vaccines" International Journal of Molecular Sciences 21, no. 14: 5130. https://doi.org/10.3390/ijms21145130

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

Article Metrics

Back to TopTop