Next Article in Journal / Special Issue
Edwardsiella tarda OmpA Encapsulated in Chitosan Nanoparticles Shows Superior Protection over Inactivated Whole Cell Vaccine in Orally Vaccinated Fringed-Lipped Peninsula Carp (Labeo fimbriatus)
Previous Article in Journal
Targeting Immune Regulatory Networks to Counteract Immune Suppression in Cancer
Previous Article in Special Issue
Biodegradable Polymeric Nanoparticles-Based Vaccine Adjuvants for Lymph Nodes Targeting
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Replicon RNA Viral Vectors as Vaccines

PanTherapeutics, CH 1095 Lutry, Switzerland
Vaccines 2016, 4(4), 39; https://doi.org/10.3390/vaccines4040039
Submission received: 13 July 2016 / Revised: 14 October 2016 / Accepted: 28 October 2016 / Published: 7 November 2016
(This article belongs to the Special Issue Nanoparticles to Co-Deliver Immunopotentiators and Antigens)

Abstract

:
Single-stranded RNA viruses of both positive and negative polarity have been used as vectors for vaccine development. In this context, alphaviruses, flaviviruses, measles virus and rhabdoviruses have been engineered for expression of surface protein genes and antigens. Administration of replicon RNA vectors has resulted in strong immune responses and generation of neutralizing antibodies in various animal models. Immunization of mice, chicken, pigs and primates with virus-like particles, naked RNA or layered DNA/RNA plasmids has provided protection against challenges with lethal doses of infectious agents and administered tumor cells. Both prophylactic and therapeutic efficacy has been achieved in cancer immunotherapy. Moreover, recombinant particles and replicon RNAs have been encapsulated by liposomes to improve delivery and targeting. Replicon RNA vectors have also been subjected to clinical trials. Overall, immunization with self-replicating RNA viruses provides high transient expression levels of antigens resulting in generation of neutralizing antibody responses and protection against lethal challenges under safe conditions.

1. Introduction

Vaccine development against infectious diseases has classically been based on live attenuated or inactivated infectious agents [1]. Recently, the approach of vaccination with recombinantly expressed antigens and immunogens from viral and non-viral delivery systems has been introduced to the repertoire [2,3]. In this context, immunization with surface proteins and antigens has elicited strong humoral and cellular immune responses and vaccinated animals showed protection against challenges with lethal doses of infectious agents or tumor cells [4].
The types of non-viral vectors applied include liposomes [5], immunostimulatory complexes (ISCOMs) composed of adjuvant Quil A and peptides [6], and multiple antigen peptides (MAPs) also known as dendrimers [7]. A number of viral vectors based on adenoviruses, alphaviruses, avipoxiviruses, enteroviruses, flaviviruses, measles viruses (MV), rhabdoviruses, and vaccinia viruses have been engineered for vaccine development [3,8]. In this context, self-replicating RNA virus vectors have proven highly efficient for immunization studies in various animal models [9]. Among RNA viruses, rabies virus (RABV) and vesicular stomatitis virus (VSV) belonging to the rhabdovirus family carry a single-stranded RNA (ssRNA) genome of a negative polarity [10]. Likewise, MV possess a negative-sense ssRNA genome [11]. In contrast, flaviviruses and alphaviruses are of positive polarity. West Nile virus [12] and Kunjin virus [13] are the most common flaviviruses applied for immunization studies. Similarly, expression vectors have been engineered for alphaviruses such as Semliki Forest virus (SFV) [14], Sindbis virus (SIN) [15] and Venezuelan equine encephalitis virus (VEE) [16].
In this review, various self-replicating RNA virus vectors are described and their applications as recombinant virus particles, RNA replicons and layered DNA plasmids are compared. Moreover, examples are given of utilization of self-replicating RNA virus systems for immunization studies in various animal models to elicit humoral and cellular immune responses and to generate neutralizing antibodies, as well as protection against challenges with pathogens and tumor cells. Finally, a summary of clinical trials already conducted or in progress that apply self-replicating RNA viruses is presented. However, due to the large number of publications available, it is only possible to present key findings and examples of vaccine development for self-replicating viral vectors.

2. Self-Replicating RNA Expression Systems

Expression systems have been engineered for RNA viruses as described below. All ssRNA viruses share the feature of high level of RNA replication in the cytoplasm, which provides the basis for extreme transient expression of heterologous genes. However, the different polarities of the ssRNA genomes of self-replicating RNA viruses have required the design of vectors with specific features. Moreover, the viral vectors can be utilized in different forms as indicated for the individual types of viruses below.

2.1. Alphaviruses

Alphavirus-based expression systems have been developed in different formats for SFV [14], SIN [15] and VEE [16], here illustrated for SFV (Figure 1). In all cases, the basic component is represented by the alphavirus non-structural genes (nsP1-4), responsible for rapid and high quantity cytoplasmic RNA replication [17]. The replication-deficient system carries the gene of interest (GoI) downstream of the nsP1-4 genes in the alphavirus expression vector to be driven by the 26S subgenomic promoter. RNA replicon vectors can be generated by in vitro transcription for direct RNA immunization. In case of production of recombinant particles, in vitro transcribed RNA from an alphavirus helper vector is co-transfected or co-electroporated into baby hamster kidney (BHK) cells. The replication-proficient system utilizes a full-length vector, where the GoI can be introduced either downstream of the nsP1-4 genes or the structural genes. In vitro transcribed RNA can be applied for immunization, but due to the presence of full-length alphavirus genomic RNA, replication-proficient particles are generated. The DNA layered system applies plasmids carrying alternatively the nonstructural genes or the full-length genome and the GoI for direct immunization with DNA. All vector system approaches described above have proven efficient in immunization studies as presented below [4].

2.2. Flaviviruses

Among flaviviruses, Kunjin virus [18], West Nile virus [19,20], yellow fever virus [21,22], dengue virus [23,24] and tick-borne encephalitis virus [25,26] have been engineered for the development of vectors for DNA, RNA and recombinant particle delivery. In Kunjin virus vectors, the GoI is inserted between the first 20 codons of the core protein (C20) and the last 22 codons of the envelope gene (E22) in frame with the rest of the viral polyprotein (Figure 2) [27]. The GoI is expressed initially as a fusion with the Kunjin virus polyprotein, which is then processed into individual proteins. Introduction of flanking FMDV-2A protease sequences allows the cleavage of Kunjin virus sequences from the expressed recombinant protein [28]. To facilitate vector production, a system has been engineered for transfection of Kunjin virus replicon RNA into the tetKUNCprMEC packaging cell line.

2.3. Measles Viruses

Expression systems have been engineered, whereby replicating MV is rescued from cloned DNA expression constructs [29] (Figure 3). Reverse genetics has allowed the rescue of recombinant measles virus in an HEK293 helper cell line, where foreign genes were introduced between the phosphoprotein (P) and the matrix protein (M) or between the hemagglutinin (H) and the large protein (L), respectively, in the measles virus genome [30]. Transfection of the helper cell line with recombinant MV constructs and a plasmid expressing the MV polymerase L gene is followed by transfer of syncytia to Vero cell cultures after 3 days, and recombinant MV particles are harvested when reaching 80%–90% cytopathic effects.

2.4. Rhabdoviruses

Both RABV [10,31] and VSV [32,33] have been subjected to expression vector engineering (Figure 4). Similar to MV, reverse genetics has been applied for efficient recovery of VSV based on recombinant vaccinia virus, where the VSV N, P and L genes were inserted downstream of a T7 promoter and an internal ribosome entry site (IRES) [33]. The role of vaccinia virus has been to provide T7 RNA polymerase. However, vaccinia virus causes strong cytopathic effects in transfected cells, the vaccinia virus DNA polymerase contributes to homologous recombination between full-length genome and helper plasmids, and vaccinia virus may also contaminate recombinant virus stocks. For this reason, a BHK cell line stably expressing the T7 RNA polymerase was engineered as a vaccinia virus-free system for RABV [31]. In addition to rhabdovirus vectors, chimeric virus-like particles (VLPs) have been generated by expressing the VSV glycoprotein (VSV-G) in trans with the SFV replicon by introduction of a mutated SFV 26S promoter for packaging of infectious SFV pseudoparticles [34]. This system provides high biosafety standards as VSV-G shares no homology with the SFV genome.

3. Self-Replicating RNA Virus-Based Vaccines

Self-replicating RNA virus vectors have been frequently used for vaccine development against infectious diseases and various types of cancers [9]. Both vectors based on ssRNA viruses of positive (Kunjin virus, SFV, SIN, VEE) and negative (MV, RABV, VSV) polarity have been utilized for the expression of viral surface proteins and tumor antigens followed by immunization studies in animal models. Moreover, for vaccination, different approaches including recombinant particles, RNA replicons and layered DNA plasmids have been applied.

3.1. Vaccines against Infectious Diseases

The targets for vaccine development for infectious diseases comprise mainly surface antigens of pathogenic viruses (Table 1) and other infectious agents (Table 2). Obvious targets for vaccine development have been antigens of influenza virus and HIV. In this context, recombinant SFV particles expressing influenza nucleoprotein (NP) have demonstrated strong immune responses [35]. Similarly, VEE-based expression of hemagglutinin (HA) elicited strong immune responses and even provided protection against challenges with H5N1 virus in chicken [36]. Likewise, expression of the swine influenza virus HA H3N2 gene from VEE vectors protected swine from influenza virus challenges [37]. In another study, the swine influenza HA gene was expressed from replication-deficient alphavirus particles showing no spread of vaccine or reversion to virulence in the intended host (pig) or non-host (mouse) species [38]. Specific humoral and interferon-γ (IFN-γ) responses were observed in pigs, which were also protected against influenza virus challenges. Recombinant MV vectors carrying the HA gene have also been applied for vaccination studies [39]. Also VSV vectors have been utilized for vaccine development against influenza virus [40]. Instead of using full-length HA, expression of the stalk domain of HA generated chimeric HA (cHA) antigens. Both intramuscular and intranasal immunization of mice resulted in HA stalk-specific, cross-reactive antibodies. Prime-boost vaccination provided protection against lethal challenges with both homologous and heterologous influenza strains, which was significantly superior with intranasal administration.
For obvious reasons HIV has been a popular target for vaccine development. For instance, administration of Kunjin replicons expressing the HIV-1 gag antigen to BALB/c mice elicited gag-specific antibodies and protective gag-specific CD8+ T cell responses [41]. Interestingly, a single immunization with Kunjin virus particles induced 4.5-fold higher CD8+ T-cell responses and protection agains HIV challenges was obtained after two injections. Furthermore, RNA optimized Kunjin virus constructs for SIV Gag-Pol demonstrated improved effector memory and central memory responses as well as protection in primates [45]. Alphavirus vectors have also been employed for HIV vaccine development. Immunization with SFV particles expressing the Env [42] and gp41 [43] genes elicited humoral and cytotoxic T-lymphocyte (CTL) responses in mice. Interestingly, priming with a low dose (0.2 µg) DNA-based SFV replicon expressing the HIV Env and a Gag-Pol-Nef fusion prior to a heterologous boost with poxvirus (MVA) and/or HIV gp140 protein formulated in glycopyranosyl lipid A resulted in significantly enhanced immune responses [44]. Moreover, when macaques were immunized with a VSV vector carrying the SIV Env (smE660) gene neutralizing antibodies were obtained [46]. However, when challenged with SIVsmE660, all animals were infected. In contrast, vaccination with a combination of gag and Env resulted in immunity [47]. RABV vectors have also been employed for the expression of SIV Env and gag in macaques [48]. Although immune responses were detected for RABV glycoprotein G, no cellular responses were obtained against SIV antigens. However, replacing the RABV G with VSV G resulted in SIV-specific immune responses and immunized macaques were protected against SIV challenges.
A number of immunization studies have targeted such lethal viruses as Ebola and Lassa viruses. For instance, dose-dependent protection against Ebola virus was achieved in guinea pigs when immunized with Kunjin virus particles expressing the Ebola virus wild-type glycoprotein GP or a mutant GP (D637L) [49]. Similarly, African green monkeys were subcutaneously immunized with Kunjin particles carrying the Ebola GP D673L mutant [50]. Protection of three out of four primates was obtained against challenges with Zaire Ebola virus. Application of VSV vectors expressing the Ebola GP gene has also provided protection of macaques after challenges with the West African EBOV-Makuna strain [51]. Likewise, protection against three different Ebola strains was achieved by expression of Ebola GP from VSV vectors [52]. Alphavirus vectors have also been utilized for vaccine development against Ebola virus. In this context, RNA replicons derived from an attenuated VEE strain were applied for the expression of Ebola GP and nucleoprotein (NP) [53]. Immunization studies showed that VEE-GP alone or in combination with VEE-NP provided protection of both BALB/c mice and guinea pigs. In contrast, VEE-NP alone did not confer protection in guinea pigs, but did in mice. In another study, C57BL/6 mice were immunized with VEE particles expressing Ebola NP, which protected animals from Ebola virus challenges [54].
VSV vectors have been subjected to immunization studies for expression of the Lassa virus glycoprotein (strain Josiah, Sierra Leone), which generated protection in guinea pigs after a single prophylactic injection [55]. It was also shown that macaques were protected against challenges with the genetically distinct Liberian Lassa virus isolate. Importantly, previous VSV-based Lassa virus vaccination did not have an impact on immunization with VSV-Ebola GP particles [77]. Furthermore, alphaviruses have been used for vaccine development against Lassa virus [56]. Guinea pigs immunized with VEE particles expressing Lassa virus glycoprotein or nucleoprotein showed protection against lethal challenges with Lassa virus. Furthermore, a dual expression approach for Ebola and Lassa virus glycoproteins was engineered, which led to protection against both Ebola and Lassa virus challenges.
A number of other viral antigens have been subjected to vaccine development. Currently, relevant targets comprise dengue virus, severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome corona virus (MERS-CoV). For instance, mice immunized with VEE particles expressing the SARS-CoV glycoprotein provided protection against lethal SARS-CoV challenges [57]. Furthermore, mice were immunized with MV vectors expressing the MERS-CoV glycoprotein, which resulted in induction of T-cell and antibody responses and protection against lethal doses of MERS-CoV [58]. Respiratory syncytial virus (RSV) has also been targeted with recombinant MV vectors by expression of the RSV fusion protein (RSV-F) [39]. Immunization of cotton rats induced neutralizing antibodies against RSV and protected against RSV infection in the lungs. In another application, lipid nanoparticle (LNP) formulations were engineered for VEE RNA replicons, which demonstrated protection against RSV challenges in vaccinated mice [59]. Furthermore, immunization of African green monkeys with VEE particles expressing human RSV-F and metapneumovirus F (hMPV-F) proteins generated RSV-F and MPV-F-specific antibodies resulting in protection against RSV and MPV challenges [60]. In the context of dengue virus vaccines, a hybrid MV vector expressing the hepatitis B surface antigen (HBsAg) and the dengue virus 2 envelope protein (DV2) elicited neutralizing antibodies against MV, HBsAg and DV2 [61]. In another study, MV-DV2 vaccination of mice generated IFN-γ and DV2 antibody responses and protection against four DV serotypes [62]. Alphavirus vectors have also been evaluated for dengue vaccine development. Expression of two configurations of dengue virus E antigen (prME and E85) provided protection in macaques [63]. Moreover, a single immunization of BALB/c mice was sufficient to induce neutralizing antibodies and T-cell responses [64]. The neonatal immunization was durable, could be boosted later in life and provided protection against challenges with dengue virus. Additional viral targets evaluated for vaccine development are listed in Table 1.
Vaccine development has also been extended to other infectious diseases than caused by viral infections (Table 2). In this context, mice vaccinated with SFV vectors expressing the Plasmodium falciparum Pf332 antigen elicited immunological memory [68]. Similarly, strong immunity and long-term protection against Mycobacterium tuberculosis was obtained in mice immunized with SIN plasmid DNA vectors carrying the M. tuberculosis 85A antigen (Ag85A) [69]. Furthermore, expression of the botulinum neurotoxin A from layered SFV DNA plasmids elicited antibody and lymphoproliferative responses in immunized BALB/c mice [70]. Co-expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) enhanced the immune response. Replication-deficient SFV particles carrying the Brucella abortus translation initiation factor 3 (IF3) were subjected to immunization studies in BALB/c mice, which resulted in protection against challenges with the virulent B. abortus strain 2308 [71]. In another study, SIN vectors were utilized for the expression of the protective antigen (PA) for Bacillus antracis in Swiss Webster mice leading to the generation of specific and neutralizing antibodies and partial protection against challenges with pathogenic bacteria [72].
Recombinant SIN vectors were applied for the expression of a class I major histocompatibility complex-restricted 9-mer epitope of the Plasmodium yoelii circumsporozoite protein (CS), which induced a strong epitope-specific CD8+ T-cell response and a high degree of protection against malaria infection in mice [73]. Another approach to develop malaria vaccines involves the application of a live attenuated MV vaccine expressing recombinant antigens against malaria [78]. A modified replication-competent VSV vector pseudotyped with the glycoprotein of the lymphocytic choriomeningitis virus (VSV-GP) expressing ovalbumin (OVA) induced humoral and cellular immune responses after a single administration in mice [74]. Due to the generation of neutralizing antibodies against VSV, immunization boosters were only possible for VSV-GP-OVA. CTL responses of similar potency as obtained for state-of-the-art adenovirus administration were observed and complete protection against challenges with Listeria monocytogenes was obtained in mice. In the context of prion disease, SFV DNA, RNA and recombinant particles were employed for the expression of prion protein (PRNP), which allowed generation of monoclonal antibodies against PRNP in immunized mice [75]. Although not directly applied for vaccine development, the generated monoclonal antibodies will be useful for basic research and diagnostics for prions. Alphavirus vectors have also been applied for the development of vaccines against Staphylococcus enterotoxin B (SEB) [76]. Subcutaneous administration of VEE particles expressing SEB resulted in protection against challenge of wild-type SEB in mice.

3.2. Vaccines against Cancer

A number of immunization studies have been carried out with self-replicating RNA virus vectors in the area of oncology (Table 3). For instance, attenuated oncolytic MV strains such as the Edmonston-B (MV-Edm) strain demonstrated anti-tumor activity [79]. The MV-Edm strain does not cause any significant cytopathic effect in normal tissue, but can selectively infect and replicate in tumor cells based on evaluations in cell lines, primary cancer cells and xenograft and syngeneic models for B-cell Non-Hodgkin lymphoma [80], ovarian cancer [81], glioblastoma multiforme [82], breast [83] and prostate [79] cancers. In this context, tumor regression was obtained in SCID mice with human lymphoma xenografts after intratumoral injection of MV-Edm [80]. Moreover, co-administration of MV vectors expressing carcinoembryonic antigen (CEA) and thyroidal sodium iodide symporter (NIS) in mice with SKOV3ip.1 ovarian xenografts showed superior tumor regression in comparison to treatment with either MV-CEA or MV-NIS alone [81]. To improve delivery and enhance efficacy, CD46 and signaling lymphocytic activation molecule (SLAM) ablating mutations in the hemagglutinin protein in combination with the display of a single-chain antibody against the epidermal growth factor receptor (EGFR) were incorporated into MV vectors for tumor targeting [82]. Tumor regression and significantly extended survival were observed after intratumoral administration of MV. Evaluation of MV-CEA delivery in an MDA-MB-231 mammary tumor model revealed a significant delay in tumor growth and prolonged survival [83]. Moreover, intratumoral administration of MV-CEA vectors showed tumor growth delay and improved survival in a subcutaneous PC-3 xenograft model [79].
Rhabdoviruses have also been applied in cancer therapy [84]. VSV vectors lack pre-existing immunity in humans and have demonstrated high susceptibility of cancer cells. Particularly, VSV vectors have been subjected to aggressive pancreatic ductal adenocarcinoma (PDAC) showing superiority to Sendai virus and RSV in 13 clinically relevant human pancreatic cell lines, although the response varied from one cell line to another [85]. Moreover, evaluation in ten PDAC cell lines of three VSV vectors expressing the wild-type matrix protein or ∆M51 showed activation of VSV-mediated apoptosis [86]. However, high constitutive expression of IFN-stimulated genes (ISGs) was discovered in three cell lines, which also contributed to resistance to apoptosis.
Kunjin virus replicons expressing the granulocyte colony-stimulating factor (GM-CSF) have been subjected to intratumoral administration, which resulted in cure in less than 50% of mice with established CT26 colon carcinoma and B16-OVA melanomas [87]. Subcutaneous injection led to regression in CT26 lung metastasis. Moreover, Kunjin vectors were engineered to express a CTL epitope of HPV16 E7 protein, which induced E7-directed T-cell responses and provided protection against challenges with an E7-expressing epithelial tumor in mice [88]. In this study, the Kunjin VLPs were more effective than RNA replicons or DNA vectors.
Alphavirus vectors have been applied in many studies on cancer vaccines [4,8]. In principle, tumor-associated antigens (TAAs), immunomodulating cytokines and combination therapies of TAAs and cytokines, TAAs and antibodies, cytokines and antibodies and even microRNAs (miRNAs) have been evaluated. In this context, intratumoral injection of SFV particles expressing enhanced green fluorescent protein (EGFP) showed apoptosis induction in mice implanted with human non-small cell lung carcinoma H353a cells in mice [109]. Furthermore, intratumoral administration of SFV particles into BALB/c mice with implanted sarcoma K-BALB sarcoma and CT26 colon tumors resulted in significant tumor growth inhibition [103]. Also, vaccination of mice with SFV RNA replicons expressing β-galactosidase showed protection against challenges with colon tumor cells [100]. Only a single intratumoral injection of 1 μg of SFV-LacZ RNA resulted in 10–20 days of survival extension in mice with existing tumors. Similarly, SIN-LacZ vectors demonstrated therapeutic efficacy in a mouse CT26 colon carcinoma model [101]. Despite not targeting specifically CT26 cells, SIN vectors showed susceptibility to mediastinal lymph nodes (MLNs), which induced effector and memory CD8+ T-cells displaying robust cytotoxicity. The well-characterized human carcinoembryonic antigen (CEA) elicited neutralizing antibodies after VEE VLP immunization [120]. Moreover, melanoma antigens such as tyrosine-related proteins TRP-1 and TRP-2, gp100 and melanoma antigen tyrosinase (Tyr) have been expressed from VEE vectors [94,112]. For instance, immunization of mice with VEE-TRP-2 particles resulted in growth inhibition of B16 transplantable melanoma and strong therapeutic potency [111]. Vaccination with VEE-TRP-2 VLPs was more efficient than the combination of VEE-gp100 and VEE-Tyr particles. Furthermore, VEE particles carrying the Tyr gene induced immune responses and tumor protection in mice after administration of VEE VLPs alone or an initial vaccination with plasmid DNA followed by boosting with VEE VLPs [112].
Breast cancer has been targeted in several therapeutic and prophylactic vaccine studies. SIN plasmid DNA carrying the neu gene were subjected to immunization of mice resulting in inhibition of growth of challenged A2L2 tumor cells [94]. Interestingly, vaccination two days after tumor challenge was inefficient. In contrast, immunization in a prime-boost protocol with SIN-neu DNA followed by adenovirus vectors carrying the neu gene prolonged the survival of mice. Due to their potency of stimulation of antigen-specific T-cells, dendritic cells (DCs) were transduced by VEE-neu particles, which resulted in high-level transgene expression, DC maturation and secretion of pro-inflammatory cytokines [95]. Robust neu-specific CD8+ T-cell and anti-neu IgG responses were observed after a single immunization. Moreover, regression of large established tumors was obtained. Another TAA attractive for immunotherapy is the six-transmembrane epithelial antigen of the prostate (STEAP), which has demonstrated up-regulation in multiple cancer cell lines [121]. Transgenic adenocarcinoma of mouse prostate (TRAMPC-2) tumor-bearing mice pre-immunized with VEE VLPs expressing STEAP demonstrated a strong immune response and a significantly prolonged overall survival [116]. The therapeutic affect was assessed for mice with 31-day-old tumors, which resulted in a modest but significant delay in tumor growth. Furthermore, VEE VLPs have been applied for expression of the prostate stem cell antigen (PSCA) in TRAMP mice, where the initial immunization with a PSCA DNA plasmid was followed by VEE-PSCA VLP delivery [117]. The outcome was a specific immune response and protection against tumor challenges in 90% of TRAMP mice. Also, the prostate-specific membrane antigen (PSMA) has been expressed from VEE vectors demonstrating strong humoral and cellular immune responses in subcutaneously immunized mice [115]. VEE VLPs expressing the prostate-specific antigen (PSA) were used for immunization of mice followed by a challenge with TRAMP cells [118]. The VEE VLPs were capable of infecting DCs in vitro and induced a robust PSA-specific response in vivo. Tumors in vaccinated animals showed low PSA expression levels and tumor growth was significantly delayed.
The P815A antigen is expressed in P815 mastocytoma tumors, which triggered an immunization study on the P1A gene coding for the PP815A antigen [119]. SFV particles expressing the P1A gene elicited strong CTL responses and protected immunized mice from challenges with P815 tumors. Other interesting TAA vaccine targets have been the E6 and E7 proteins of the human papilloma virus (HPV). Immunization with SFV particles expressing HPV type 16 E6,7 showed strong HPV-specific CTL activity and eradicated HPV-transformed tumors [97]. Similarly, immunization of mice with VEE particles carrying the HPV16 E7 gene prevented tumor development and eliminated established tumors in 67% of vaccinated animals [99]. In another study, tattoo injection [122] of SFV-HPV E6,7 particles resulted in antigen expression in both the skin and draining lymph nodes leading to ten-fold lower antigen levels in comparison to intramuscular administration [98]. However, tattoo injection provided higher or equal levels of immune responses.
Cytokines have played an important role in immunotherapy and vaccine development [8]. For instance, interleukin-12 (IL-12) has been expressed from both SFV and SIN vectors. In this context, SFV vectors expressing IL-12 induced tumor regression with long-term tumor-free survival in the MC38 colon carcinoma model [102]. Repeated intratumoral administration increased the anti-tumor response. In another study, immunization with SFV-luciferase and SFV-IL-12 particles was evaluated in a woodchuck model in which hepatocellular carcinoma (HCC) is induced by the infection by woodchuck hepatitis virus (WHV) [105]. High luciferase expression levels were observed in tumors and IL-12 secretion was measured in the serum after intratumoral injections. In tumor-bearing woodchucks, partial tumor remission was seen. Tumor volumes were reduced by 80%, but tumor growth was restored with time. The plasmid vector pTonL2(T)-mIL12, which provides liver-specific and inducible IL-12 expression, has been compared to SFV-IL-12 particle delivery in a L-PK/c-myc transgenic mouse model of HCC [106]. Overexpression of the c-myc gene in the liver of the transgenic animals induces spontaneous hepatic tumors with characteristics similar to human HCCs. Intratumoral administration of SFV-IL-12 resulted in tumor growth arrest and 100% survival rates. Mice treated with plasmid DNA showed a slightly lower survival rate despite higher IL-12 and IFN-γ levels in serum. The strong anti-tumor response in SFV-IL-12-treated mice was most likely due to the apoptosis and type 1 IFN response induced by SFV particles. Recombinant SIN particles were demonstrated to target tumor cells in SCID mice, which encouraged intraperitoneal injection of SIN-IL-12 particles in mice with established ovarian tumors [113]. The treatment resulted in systemic targeting and eradication of tumor cells without any adverse effects observed. Glioma-bearing mice were immunized with SFV-IL-12 particles, which induced apoptosis of glioma cells and facilitated the uptake of apoptotic cells by DCs and provided prolonged survival of vaccinated animals [91]. Moreover, DCs isolated from bone marrow were transduced with SFV vectors expressing IL-12 for the treatment of brain tumor-bearing mice [92]. The outcome was prolonged survival of immunized animals. In another study, SFV-IL-12 particles were tested in rat RG2 gliomas [93]. Low dose (5 × 107 VLPs) treatment resulted in a 70% reduction in tumor volume, whereas high-dose (5 × 108 VLPs) showed an 87% reduction in tumor volume. Moreover, intratumoral administration of 106 oncolytic SFV particles expressing EGFP generated significant tumor regression in melanoma-bearing SCID mice [123]. Other cytokines such as Il-18 have also been evaluated for alphavirus-based expression in ovarian and colon cancer models [104]. The enhanced SFV10E vector, which provides ten-fold higher levels of expression than the conventional SFV vector [124], was applied for immunization of BALB/c mice [114]. After in vitro verification of secretion of active IL-18, mice with subcutaneous K-BALB and CT26 tumors were injected with SFV-IL-18 particles, which led to tumor regression and disappearance of tumors in some treated animals. Moreover, GM-CSF, an immunostimulatory cytokine, has been expressed from SFV vectors [114]. Intraperitoneal administration of SFV-GM-CSF particles was evaluated in an ovarian mouse tumor model, which resulted in activation of macrophages to tumor cytotoxicity. Although no prolongation in survival of tumor-bearing mice was achieved, tumor growth was inhibited for two weeks.
Among the growth factors targeted for vaccine development, the vascular endothelial growth factor receptor 2 (VEGFR-2) was introduced into the SFV vector [96]. Immunization of mice with SFV-VEGFR-2 particles resulted in substantial inhibition of both tumor growth and spread of pulmonary metastases. Furthermore, vaccination led to tumor inhibition in mice with established CT26 colon tumors and metastatic 4T1 mammary tumors. In another approach, SFV particles carrying the endostatin gene were administered to mice bearing B16 brain tumors [89]. The treatment resulted in a substantial reduction in intratumoral vascularization in tumor sections and a significant inhibition of tumor growth. Endostatin serum levels were three-fold higher 7 days after intravenous administration of SFV-endostatin in comparison to administration of the retrovirus-based GCsap-Endostatin promoting inhibition of angiogenesis in established tumors. In another approach, SIN vectors have been employed for the expression of a fusion protein of HPV16 E7 protein and calreticulin (CRT), an ER Ca2+-binding transporter participating in antigen processing and presentation with major histocompatibility complex (MHC) class I [108]. Immunization of mice bearing E7-expressing tumors with SIN-E7-CRT particles significantly increased E7-specific CD8+ T-cell precursors and a strong anti-tumor response. Furthermore, a significant reduction in lung tumor nodules was observed in immuno-compromised BALB/c mice.
Combination therapy has been evaluated for alphavirus-based gene delivery. For instance, SFV layered DNA vectors were engineered to express one to four domains of VEGFR-2 and IL-12 [110]. Co-immunization with SFV replicon DNA expressing survivin and β-hCG antigens was verified in mice resulting in efficient humoral and cellular immune responses against survivin, β-hCG and VEGFR-2. Moreover, tumor growth was inhibited and the survival rate in a B16 melanoma mouse model was improved. Furthermore, immunization with SFV HPV E6/E7 was combined with sunitib and a single low-dose of irradiation, which enhanced the intratumoral ratio of anti-tumor effector cells to myeloid-derived suppressor cells [107]. Triple treatment of tumor-bearing mice demonstrated enhanced anti-tumor efficacy and provided 100% tumor-free survival.
An interesting approach comprises the introduction of micro RNA-124 (miR-124) into an SFV4 vector [90]. As IFN-1 tolerance has been associated with the SFV nsP3-nsP4 genes, conditionally replicating SFV4-miR-124 virus was able to replicate in neurons and allowed targeting of gliomas otherwise sensitive to IFN-1. Evaluation of CT-2A mouse astrocytoma cells and IFN-1 pretreated human glioblastoma cells showed increased oncolytic potency. Moreover, a single intraperitoneal injection of SFV4-miR-124 into mice with implanted CT-2A orthotopic gliomas showed significant inhibition of tumor growth and improved survival rates.

3.3. Clinical Trials

Self-replicating RNA virus vectors have been subjected to several clinical studies, albeit at an inferior level in comparison to adenovirus, AAV and lentivirus vectors. For instance, healthy volunteers were subjected to low-dose (3 × 105 pfu) immunization with the VSV-based Ebola vaccine (rVSV-ZEBOV) expressing the Zaire Ebola virus glycoprotein in a double-blinded study in comparison to a previous study with a high dose (5 × 107 pfu) [125]. No serious adverse events occurred and the overall safety was good. The low-dose immunization improved early tolerability, but generated inferior antibody responses and failed to prevent vaccine-induced arthritis, dermatitis or vasculitis. Furthermore, VSV particles expressing the HIV-1 gag gene were evaluated in a clinical trial on safety and immunogenicity [126]. In the randomized double-blinded placebo-controlled dose-escalation study, healthy HIV-negative volunteers received 4.6 × 103 to 3.4 × 107 pfu of rVSV HIV-1 gag vaccine intramuscularly at months 0 and 2. All vaccinated individuals showed antibody responses against VSV, and gag-specific T-cell responses were detected in 63%. Overall, the safety profile was good.
Alphaviruses have been subjected to some gene therapy and vaccine studies. In one approach, replication-deficient SFV particles were encapsulated in liposomes to promote passive targeting of tumors [127]. Initially, intraperitoneal administration of encapsulated SFV-LacZ particles showed enhanced accumulation of β-galactosidase in SCID mice implanted with LNCaP prostate tumors. Liposome-encapsulated SFV particles expressing the p40 and p35 subunits of IL-12 generated active secreted IL-12 in BHK-21 cells [128]. Next, encapsulated SFV-IL-12 particles were administered intravenously in terminally ill melanoma and kidney carcinoma patients in a phase I clinical trial. The patients showed a five to ten-fold increase in IL-12 plasma levels. The maximum tolerated dose was determined to 3 × 109 infectious particles and the safety profile was good. A phase I dose-escalation trial was conducted in prostate cancer patients with VEE particles expressing PSMA [129]. Patients with castration-resistant metastatic prostate cancer (CRPC) received up to five doses of either 0.9 × 107 IU or 0.36 × 108 IU of VEE-PSMA particles at weeks 1, 4, 7, 10 and 18. The study showed no toxicity and good toleration of the vaccination. However, only weak PSMA-specific immune responses were detected and no clinical benefits obtained. In another clinical trial, VEE particles expressing the CEA tumor antigen were demonstrated to efficiently infect DCs [120]. The VEE particles could be repeatedly administered and overcame high titers of neutralizing antibodies and elevated regulatory T cells (Tregs), which allowed induction of clinically relevant CEA-specific T cell and antibody responses. In another approach, VEE particles expressing the cytomegalovirus (CMV) gB and pp65/IE1 fusion protein were evaluated in a phase I randomized, double-blinded clinical trial [67]. Intramuscular or subcutaneous immunization at weeks 0, 8 and 24 of CMV seronegative adult volunteers showed good tolerance with only mild to moderate local reactions and no clinically important changes. Neutralizing and multifunctional T-cell responses against CMV antigens were detected in all vaccinated individuals.

4. Conclusions

Self-replicating RNA viruses represented by ssRNA viruses of both negative and positive polarity have been subjected to engineering of efficient gene delivery vectors, which can be applied in the form of recombinant particles, RNA replicons and layered DNA plasmid vectors. In this context, measles (MV), rhabdoviruses, flaviviruses and alphaviruses expressing surface antigens from viruses and other infectious agents have been subjected to immunization studies in animal models. Moreover, similar studies have been conducted with tumor antigens. It seems that MV-, rabies virus (RABV)-, vesicular stomatitis virus (VSV)-, Kunjin virus-, Semliki Forest virus (SFV)-, Sindbis virus (SIN)- and Venezuelan equine encephalitis virus (VEE)-based delivery efficiently elicits humoral and cellular immune responses in immunized animals. Furthermore, numerous cases have demonstrated protection against challenges with lethal viruses/infectious agents or with tumor cells. Most of the studies have been conducted with replication-deficient recombinant particles. However, promising results have also been obtained with layered DNA plasmid vectors. A limited number of studies have applied administration of RNA replicons, but the results have been quite encouraging. The obvious advantage to using nucleic acid-based delivery is the elimination of any risk of virus progeny production through recombination events. On the other hand, superior delivery and prolonged duration of expression can be achieved with recombinant viral particles, especially applying replication-proficient oncolytic viruses. For this reason, it is difficult to make any recommendations related to which delivery format to use, and the choice of target will play an important role in decision making.
Similarly, it is practically impossible to favor one viral vector system over another. Reverse genetics systems engineered for MV and rhabdoviruses and packaging cell lines for flaviviruses surely facilitate recombinant particle production and ease of use. Although packaging cell lines have also been generated for alphaviruses, the straightforward in vitro RNA transcription has provided the means for sufficient preparation of replicon RNA and particles for immunization studies. Obviously, plasmid DNA can be directly applied for vaccinations. In comparison to other viral vectors and also non-viral delivery systems, self-replicating RNA viruses can surely be considered competitive (Figure 5 and Table 4). An extensive comparison to other delivery systems is not within the scope of this review, so only a few examples are addressed. Clearly, adenovirus-based vaccine development and gene therapy has a longer history, which has generated a multitude [130] of vector improvements and also resulted a number of clinical trials [131,132]. Similarly, herpes simplex virus (HSV) vectors have been frequently applied and HSV-GM-CSF have, for instance, been subjected to phase I−III human clinical trials in glioblastoma and melanoma patients [133]. HSV vectors were recently approved by the FDA for use in standard patient care [134]. Related to non-viral vectors, recently dendrimer-RNA nanoparticles have demonstrated protective immunity against lethal challenges with Ebola virus, influenza H1N1 virus and Toxoplasma gondii after a single injection in BALB/c mice [135].
Overall, self-replicating RNA viral vectors possess several attractive features. The presence of RNA replicons provides the efficient means for rapid generation of a large number of RNA copies for immediate protein translation in the cytoplasm of host cells. Moreover, the strong subgenomic promoter utilized by alphaviruses generates extreme levels of heterologous gene expression. The transient nature of expression is also an advantage for immunization studies. Furthermore, there is no risk of integration of viral genes in the host genome as the viral RNA is degraded within 3–5 days. In the case of immunization with layered alphavirus DNA vectors, approximately 100- to 1000-fold lower doses are required compared to immunizations with conventional plasmid DNA [136].
Although strong immune responses have been obtained and protection against challenges with lethal pathogens and tumor cells have been achieved and even tumor regression observed in animals with established tumors, some further technology development is necessary. Much development has been invested in vector design including mutant vectors, enhancement signals, targeting DCs and fusion constructs. Furthermore, quite an effort has been paid to the evaluation of different target antigens and immunogens. Several studies, particularly clinical trials, have indicated that although target-specific immune responses have been obtained, further investment is required in finding the right dose for the achievement of optimal response. One area which recently has received much attention is combination therapy. Tumor-associated antigens (TAAs) have been combined with cytokines and antibodies, as well as drugs and radiation co-administered with cytokines. Additionally, optimization of adjuvant composition and stability issues in case of RNA delivery needs to be addressed. Further research in these areas will certainly provide progress and should make immunotherapy an important approach in both prophylactic and therapeutic applications.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
MDPIMultidisciplinary Digital Publishing Institute
DOAJDirectory of open access journals
CEACarcinoembryonic antigen
CMVCytomegalovirus
CRPCCastration-resistant metastatic prostate cancer
CRTCalreticulin
CTLCytotoxic T-lymphocyte
DCDendritic cell
EGFPEnhanced green fluorescent protein
EGFREpidermal growth factor receptor
GFPGreen fluorescent protein
GM-CSFGranylocyte macrophage colony-stimulating factor
GoIGene of interest
HAHemagglutinin
HBVHepatitis B virus
HCCHepatocellular carcinoma
HPVzhuman papilloma virus
IF3Initiation factor 3
IFNInterferon
ILInterleukin
ISGInterferon-stimulated gene
MERS-CoVMiddle East respiratory syndrome coronavirus
MHCMajor histocompatibility complex
miRNAMicro RNA
MLNMediastinal lymph nodes
MVMeasles virus
NISSodium iodide symporter
NPNucleoprotein
PDACPancreatic ductal adenocarcinoma
PSMAProstate-specific membrane antigen
PSCAProstate stem cell antigen
RABVRabies virus;
RSVRespiratory syndrome virus
SARS-CoVSevere acute respiratory syndrome coronavirus
SFVSemliki Forest virus
SEBStaphylococcus enterotoxin B
SINSindbis virus
ssRNASingle-stranded RNA
STEAPSix-transmembrane epithelial antigen of the prostate
TAATumor-associated antigen
TRAMPTransgene adenocarcinoma of the mouse prostate
TRPTyrosine-related protein
TyrTyrosinase
VEEVenezuelan equine encephalitis virus
VEGFRVascular endothelial growth factor receptor
VLPsVirus-like particles
VSVVesicular stomatitis virus
WHVWoodchuck hepatitis virus

References

  1. Delrue, I.; Verzele, D.; Madder, A.; Nauwynck, H.J. Inactivated virus vaccines: From chemistry to prophylaxis: Merits, risks and challenges. Expert Rev. Vaccines 2012, 11, 695–719. [Google Scholar] [CrossRef]
  2. Deng, M.P.; Hu, Z.H.; Wan, H.L.; Deng, F. Developments of subunit and VLP vaccines against influenza A virus. Virol. Sin. 2012, 27, 145–153. [Google Scholar] [CrossRef]
  3. Apostolopoulos, V. Vaccine delivery methods into the future. Vaccines 2016. [Google Scholar] [CrossRef]
  4. Lundstrom, K. Alphavirus-based vaccines. Viruses 2014, 6, 2392–2415. [Google Scholar] [CrossRef]
  5. Schmidt, S.T.; Foged, C.; Korsholm, K.S.; Rades, T.; Christensen, D. Liposome-based adjuvants for subunit vaccines: Formulation strategies for subunit antigens and immunostimulators. Pharmaceutics 2016. [Google Scholar] [CrossRef]
  6. Cibulski, S.P.; Mourglia-Ettlin, G.; Teixeira, T.F.; Quirici, L.; Roehe, P.M.; Ferreira, F.; Silveira, F. Novel ISCOMs from Quillaja brasiliensis saponins induce mucosal and systemic antibody production, T-cell responses and improved antigen uptake. Vaccine 2016, 34, 1162–1171. [Google Scholar] [CrossRef]
  7. Sheng, K.C.; Kalkanidis, M.; Pouniotis, D.S.; Esparon, S.; Tang, C.K.; Apostolopoulos, V.; Pietersz, G.A. Delivery of antigen using a novel mannosylated dendrimer potentiates immunogenicity in vitro and in vivo. Eur. J. Immunol. 2008, 38, 424–436. [Google Scholar] [CrossRef]
  8. Lundstrom, K. Alphaviruses in gene therapy. Viruses 2015, 7, 2321–2333. [Google Scholar] [CrossRef]
  9. Lundstrom, K. Self-replicating RNA viral vectors in vaccine development and gene therapy. Future Virol. 2016, 11, 345–356. [Google Scholar] [CrossRef]
  10. Lyles, D.S.; Rupprecht, C.E. Rhabdoviridiae. In Fields’ Virology; Knipe, D.M., Howley, P.M., Eds.; Wolters Kluwer: Philadelphia, PA, USA, 2007; pp. 1364–1408. [Google Scholar]
  11. Rima, B.K.; Duprex, W.P. The measles virus replication cycle. Curr. Top. Microbiol. Immunol. 2009, 329, 77–102. [Google Scholar]
  12. Brinton, M.A. Replication cycle and molecular biology of West Nile virus. Viruses 2013, 6, 13–53. [Google Scholar] [CrossRef]
  13. Westaway, E.G.; Mckenzie, J.M.; Khromykh, A.A. Kunjin RNA replication and applications of Kunjin replicons. Adv. Virus Res. 2003, 59, 99–140. [Google Scholar]
  14. 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]
  15. 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]
  16. Davis, N.L.; Willis, L.V.; Smith, J.F.; Johnston, R.E. In vitro synthesis of infectious Venezuelan equine encephalitis virus RNA from a cDNA clone: Analysis of a viable deletion mutant. Virology 1989, 171, 189–204. [Google Scholar] [CrossRef]
  17. Strauss, J.H.; Strauss, E.G. The Alphaviruses: Gene Expression, Replication and Evolution. Micobiol. Rev. 1994, 58, 491–562. [Google Scholar]
  18. 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]
  19. 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]
  20. 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]
  21. 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]
  22. 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]
  23. 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]
  24. Pang, X.; Zhang, M.; Dayton, A.I. Development of Dengue virus Type 2 replicons capable of prolonged expression in host cells. BMC Microbiol. 2001. [Google Scholar] [CrossRef]
  25. 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]
  26. Hayasaka, D.; Yoshii, K.; Ueki, T.; Iwasaki, T.; Takashima, I. Sub-genomic replicons of Tick-borne encephalitis virus. Arch.Virol. 2004, 149, 1245–1256. [Google Scholar] [CrossRef]
  27. 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]
  28. De Felipe, F. Skipping the co-expression problem: The new 2A “CHYSEL” technology. Genet. Vaccines Ther. 2004. [Google Scholar] [CrossRef] [Green Version]
  29. 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]
  30. 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]
  31. 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]
  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]
  33. 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]
  34. Dorange, F.; Piver, E.; Bru, T.; Collin, C.; Roingeard, P. Vesicular stomatitis virus glycoprotein: A transducing coat for SFV-based RNA vectors. J. Gene Med. 2004, 6, 1014–1022. [Google Scholar] [CrossRef]
  35. Malone, J.G.; Berglund, P.J.; Liljestrom, P.; Rhodes, G.; Malone, R.W. Mucosal immune responses associated with polynucleotide vaccination. Behring Inst. Mitt. 1997, 98, 63–72. [Google Scholar]
  36. 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]
  37. Bosworth, B.; Erdman, M.M.; Stine, D.L.; Harris, I.; Irwin, C.; Jens, M.; Loynachan, A.; Kamrud, K.; Harris, D.L. Replicon particle vaccine protects swine against influenza. Comp. Immunol. Microbiol. Infect. Dis. 2010, 33, e99–e103. [Google Scholar] [CrossRef]
  38. Vander Veen, R.L.; Loynachan, A.T.; Mogler, M.A.; Russell, B.J.; Harris, D.L.; Kamrud, K. Safety, immunogenicity and efficacy of an alphavirus replicon-based swine influenza virus hemagglutinin vaccine. Vaccine 2012, 30, 1944–1950. [Google Scholar] [CrossRef]
  39. Sweft-Tapia, C.; Bogaert, L.; de Jong, P.; van Hoek, V.; Schouten, T.; Damen, I.; Spek, D.; Wanningen, P.; Radosevic, K.; Zahn, R.; et al. Recombinant measles virus incorporating heterlogous viral membrane proteins for use as vaccines. J. Gen. Virol. 2016, 97, 2117–2128. [Google Scholar] [CrossRef]
  40. Ryder, A.B.; Nachbaqauer, R.; Buonocore, L.; Palese, P.; Krammer, F.; Rose, J.K. Vaccination with Vesicular Stomatitis virus-vectored chimeric hemagglutinins protects mice against divergent influenza virus challenge strains. J. Virol. 2015, 90, 2544–2550. [Google Scholar] [CrossRef]
  41. Harvey, T.J.; Anraku, I.; Linedale, R.; Harrich, D.; Mackenzie, J.; Suhrbier, A.; Khromykh, A.A. Kunjin virus replicon vectors for human immunodefiency virus vaccine development. J. Virol. 2003, 77, 7796–7803. [Google Scholar] [CrossRef]
  42. 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. Retrovir. 1998, 14, 1369–1377. [Google Scholar] [CrossRef]
  43. Giraud, A.; Ataman-Onal, Y.; Battail, N.; Piga, N.; Brand, D.; Mandrand, B.; Vernier, B. Generation of monoclonal antibodies to native human immunodeficiency virus type 1 envelope glycoprotein by immunization of mice with naked RNA. J. Virol. Methods 1999, 79, 75–84. [Google Scholar] [CrossRef]
  44. 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]
  45. Anraku, I.; Mokhonov, V.V.; Rattanasena, P.; Mokhonova, E.I.; Leung, J.; Pijlman, G.; Cara, A.; Schroeder, W.A.; Khromykh, A.A.; Suhrbier, A. Kunjin replicon-based simian immunodeficiency virus gag vaccines. Vaccine 2008, 26, 3268–3276. [Google Scholar] [CrossRef]
  46. Schell, J.B.; Bahl, K.; Folta-Stogniew, E.; Rose, N.; Buonocore, L.; Marx, P.A.; Gambhira, R.A.; Rose, J.K. Antigenic requirement for Gag in a vaccine that protects against high-dose mucosal challenge with simian immunodeficiency virus. Virology 2015, 476, 405–412. [Google Scholar] [CrossRef]
  47. Van Rompay, K.K.; Abel, K.; Earl, P.; Kozlowski, P.A.; Easlick, J.; Moore, J.; Buonocore-Buzzelli, L.; Schmidt, K.A.; Wilson, R.L.; Simon, I.; et al. Immunogenicity of viral vector, prime-boost SIV vaccine regimens in infant rhesus macaques: Attenuated vesicular stomatitis virus (VSV) and modified vaccinia Ankara (MVA) recombinant SIV vaccines compared to live-attenuated SIV. Vaccine 2010, 28, 1481–1492. [Google Scholar] [CrossRef]
  48. McKenna, P.M.; Koser, M.L.; Carlson, K.R.; Montefiori, D.C.; Letvin, N.L.; Papaneri, A.B.; Pomerantz, R.J.; Dietzschold, B.; Silvera, P.; McGettigan, J.P.; et al. Highly attenuated rabies virus-based vaccine vectors expressing simian-human immunodeficiency virus89.6P Env and simian immunodeficiency virus mac239 Gag are safe in rhesus macaques and protect from an AIDS-like disease. J. Infect. Dis. 2007, 195, 980–988. [Google Scholar] [CrossRef]
  49. Reynard, O.; Mokhonov, V.; Mokhonova, E.; Leung, J.; Page, A.; Mateo, M.; Pyankova, O.; Georges-Courbot, M.C.; Raoul, H.; Khromykh, A.A.; et al. Kunjin virus replicon-based vaccines expressing Ebola virus glycoprotein GP protect the guinea pig against lethal Ebola virus infection. J. Infect. Dis. 2011, 204, S1060–S1065. [Google Scholar] [CrossRef]
  50. Pyankov, O.V.; Bodnev, S.A.; Pyankova, O.G.; Solodkyi, V.V.; Pyankov, S.A.; Setoh, Y.X.; Volchkova, V.A.; Suhrbier, A.; Volchkov, V.E.; 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, S368–S371. [Google Scholar] [CrossRef]
  51. 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]
  52. Geisbert, T.W.; Feldmann, H. Recombinant vesicular stomatitis virus-based vaccines against Ebola and Marburg infections. J. Infect. Dis. 2011, 204, S1075–S1081. [Google Scholar] [CrossRef]
  53. 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]
  54. 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]
  55. Safronetz, D.; Mire, C.; Rosenke, K.; Feldmann, F.; Haddock, E.; Geisbert, 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]
  56. Pushko, P.; Geisbert, J.; Parker, M.; Jahrling, P.; Smith, J. Individual and bivalent vaccines based on alphavirus replicons protect guinea pigs against infection with Lassa and Ebola viruses. J. Virol. 2001, 75, 11677–11685. [Google Scholar] [CrossRef]
  57. Sheahan, T.; Whitmore, A.; Long, K.; Ferris, M.; Rockx, B.; Funkhouser, D.; 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]
  58. Malczyk, A.H.; Kupke, A.; Prüfer, S.; Scheuplein, V.A.; Hutzler, S.; Kreuz, D.; Beissert, T.; Bauer, S.; Hubich-Rau, S.; Tondera, C.; et al. 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]
  59. Geall, A.J.; Verma, A.; Otten, G.R.; Shaw, C.A.; Hekele, A.; Banerjee, K.; Cu, Y.; Beard, C.W.; Brito, L.A.; Krucker, T.; et al. Nonviral delivery of self-amplifying RNA vaccines. Proc. Natl. Acad. Sci. USA 2012, 109, 14604–14609. [Google Scholar] [CrossRef]
  60. Bates, J.T.; Pickens, J.A.; Schuster, J.E.; Johnson, M.; Tollefson, S.J.; Williams, J.V.; Davis, N.L.; Johnston, R.E.; Schultz-Darken, N.; Slaughter, J.C.; et al. Immunogenicity and efficacy of alphavirus-derived replicon vaccines for respiratory syncytial virus and human metapneumovirus in nonhuman primates. Vaccine 2016, 34, 950–956. [Google Scholar] [CrossRef]
  61. 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]
  62. Hu, H.M.; Chen, H.W.; Hsiao, Y.; Wu, S.H.; Chung, H.H.; Hsie, 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. Vaccines Immunother. 2016. [Google Scholar] [CrossRef]
  63. White, L.J.; Sariol, C.A.; Mattocks, M.D.; Wahala, M.P.B.W.; Yingsiwaphat, V.; Collier, M.L.; Whitley, J.; Mikkelsen, R.; Rodriguez, I.V.; Martinez, M.I.; et al. An alphavirus vector-based tetravalent dengue vaccine induces a rapid and protective immune response in macaques that differs qualitatively from immunity induced by live virus infection. J. Virol. 2013, 87, 3409–3424. [Google Scholar] [CrossRef]
  64. 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]
  65. 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]
  66. Del Valle, J.R.; Devaux, P.; Hodge, G.; Wegner, N.J.; McChesney, M.B.; Catteneo, 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]
  67. Bernstein, D.I.; Reap, E.A.; Katen, K.; Watson, A.; Smith, K.; Norberg, P.; Olmsted, R.A.; Hoeper, A.; Morris, J.; Negri, S.; et al. Randomized, double-blind, Phase 1 trial of an alphavirus replicon vaccine for cytomegalovirus in CMV seronegative adult volunteers. Vaccine 2009, 28, 484–493. [Google Scholar] [CrossRef]
  68. Andersson, C.; Vasconcelos, N.M.; Sievertzon, M.; Haddad, D.; Liljeqvist, S. Comparative immunization study using RNA and DNA constructs encoding a part of the Plasmodium falciparum antigen Pf332. Scand. J. Immunol. 2001, 54, 117–124. [Google Scholar] [CrossRef]
  69. Kirman, J.R.; Turon, T.; Su, H.; Li, A.; Kraus, C.; et al. Enhanced immunogenicity to Mycobacterium tuberculosis by vaccination with an alphavirus plasmid replicon expressing antigen 85A. Infect. Immun. 2003, 71, 575–579. [Google Scholar] [CrossRef]
  70. Li, N.; Yu, Y.Z.; Yu, W.Y.; Sun, Z.W. Enhancement of the immunogenicity of DNA replicon vaccine of Clostridium botulinum neurotoxin serotype A by GM-CSF gene adjuvant. Immunopharmacol. Immunotoxicol. 2011, 33, 211–219. [Google Scholar] [CrossRef]
  71. Cabrera, A.; Sáez, D.; Céspedes, S.; Andrews, E.; Oñate, A. Vaccination with recombinant Semliki Forest virus particles expressing translation initiation factor 3 of Brucella abortus induces protective immunity in BALB/c mice. Immunobiology 2009, 214, 467–474. [Google Scholar] [CrossRef]
  72. Thomas, J.M.; Moen, S.T.; Gnade, B.T.; Vargas-Inchaustegui, D.A.; Foltz, S.M.; Suarez, G.; Heidner, H.W.; König, R.; Chopra, A.K.; Peterson, J.W. Recombinant Sindbis virus vectors designed to express protective antigen of Bacillus anthracis protect animals from anthrax and display synergy with ciprofloxacin. Clin. Vaccine Immunol. 2009, 16, 1696–1699. [Google Scholar] [CrossRef]
  73. 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]
  74. Tober, R.; Banki, Z.; Egerer, L.; Muik, A.; Behmüller, S.; Kreppel, F.; Greczmiel, U.; Oxenius, A.; von Laer, D.; Kimpel, J. VSV-GP: A potent viral vaccine vector that boosts the immune response upon repeated applications. J. Virol. 2014, 88, 4897–907. [Google Scholar] [CrossRef]
  75. Krasemann, S.; Jürgens, T.; Bodemer, W. Generation of monoclonal antibodies against prion proteins with an unconventional nucleic acid-based immunization strategy. J. Biotechnol. 1999, 73, 119–129. [Google Scholar] [CrossRef]
  76. Lee, J.S.; Dyas, B.K.; Nystrom, S.S.; Lind, C.M.; Smith, J.F.; Ulrich, R.G. Immune protection against staphylococcal enterotoxin-induced toxic shock by vaccination with a Venezuelan equine encephalitis virus replicon. J. Infect. Dis. 2002, 185, 1192–1196. [Google Scholar] [CrossRef]
  77. Marzi, A.; Feldmann, F.; Geisbert, T.W.; Feldmann, H.; Safronetz, D. Vesicular stomatitis virus-based vaccines against Lassa and Ebola viruses. Emerg. Infect. Dis. 2015, 21, 305–307. [Google Scholar] [CrossRef]
  78. Tangy, F.; Naim, H.Y. Live attenuated measles vaccine as a potential multivalent pediatric vaccination vector. Viral Immunol. 2005, 18, 317–26. [Google Scholar] [CrossRef]
  79. Msaouel, P.; Iankov, I.D.; Dispenzieri, A.; Galanis, E. Attenuated oncolysis measles virus strains as cancer therapeutics. Curr. Pharm. Biotechnol. 2012, 13, 1732–1741. [Google Scholar] [CrossRef]
  80. Grote, D.; Russell, S.J.; Cornu, T.I.; Cattaneo, R.; Vile, R.; Poland, G.A.; Fielding, A.K. Live attenuated measles virus induces regression of human lymphoma xenografts in immunodeficient mice. Blood 2001, 97, 3746–3754. [Google Scholar] [CrossRef]
  81. Hasegawa, K.; Pham, L.; O’Connor, M.K.; Federspiel, M.J.; Russell, S.J.; Peng, K.W. Dual therapy of ovarian cancer using measles viruses expressing carcinoembryonic antigen and sodium iodide symporter. Clin. Cancer Res. 2006, 12, 1868–1875. [Google Scholar] [CrossRef]
  82. Paraskevakou, G.; Allen, C.; Nakamura, T.; Zollman, P.; James, C.D.; Peng, K.W.; Schroeder, M.; Russll, S.J.; Galanis, E. 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]
  83. McDonald, C.J.; Erlichman, C.; Ingle, J.N.; Rosales, G.A.; Allen, C.; Greiner, S.M.; Harvey, M.E.; Zollman, P.J.; Russell, S.J.; Galanis, E. 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]
  84. Hastie, E.; Grdzelishvili, V.Z. Vesicular stomatitis virus as a flexible platform for oncolytic virotherapy against cancer. J. Gen. Virol. 2012, 93, 2529–2545. [Google Scholar] [CrossRef]
  85. Murphy, A.M.; Besmer, D.M.; Moerdyk-Schauwecker, M.; Moesti, N.; Ornelles, D.A.; Mukherjee, P.; Grdzelishvili, V.Z. Vesicular stomatitis virus as an oncolytic agent against pancreatic ductal adenocarcinoma. J. Virol. 2012, 86, 3073–3087. [Google Scholar] [CrossRef]
  86. Felt, S.A.; Moerdyk-Schauwecker, M.J.; Grdzelishvili, V.Z. Induction of apoptosis in pancreatic cancer cells by vesicular stomatitis virus. Virology 2015, 474, 163–173. [Google Scholar] [CrossRef]
  87. Hoang-Le, D.; Smeenk, L.; Anraku, I.; Pijlman, G.P.; Wang, X.J.; de Vrij, J.; Liu, W.J.; Lee, T.T.; Schroeder, W.A.; Khromykh, A.A.; et al. A Kunjin replicon vector encoding granulocyte macrophage colony-stimulating factor for intra-tumoral gene therapy. Gene Ther. 2009, 16, 190–199. [Google Scholar] [CrossRef]
  88. Herd, K.A.; Harvey, T.; Khromykh, A.A.; Tindle, R.W. Recombinant Kunjin virus replicon vaccines induce protective T-cell immunity against human papillomavirus 16 E7-expressing tumour. Virology 2004, 319, 237–248. [Google Scholar] [CrossRef]
  89. Yamanaka, R.; Zullo, S.A.; Ramsey, J.; Onodera, M.; Tanaka, R.; Blaese, M.; Xanthopoulos, K.G. Induction of therapeutic antitumor antiangiogenesis by intratumoral injection of genetically engineered endostatin-producing Semliki Forest virus. Cancer Gene Ther. 2001, 8, 796–802. [Google Scholar] [CrossRef]
  90. Martikainen, M.; Niittykoski, M.; von und zu Frauenberg, M.; Immonen, A.; Koponen, S.; van Geenen, M.; Vähä-Koskela, M.; Ylösmäki, E.; Jääskeläinen, J.E.; Saksela, E.; et al. 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]
  91. Yamanaka, R.; Zullo, S.A.; Tanaka, R.; Ramsey, J.; Blaese, M.; Xanthopoulos, K.G. Induction of a therapeutic antitumor immunological response by intratumoral injection of genetically engineered Semliki Forest virus to produce interleukin-12. Neurosurg. Focus 2000, 9, e7. [Google Scholar] [CrossRef]
  92. Yamanaka, R.; Zullo, S.A.; Ramsey, J.; Yajima, N.; Tsuchiya, N.; Tanaka, R.; Blaese, M.; Xanthopoulos, K.G. Marked enhancement of antitumor immune responses in mouse brain tumor models by genetically modified dendritic cells producing Semliki Forest virus-mediated interleukin-12. J. Neurosurg. 2002, 97, 611–618. [Google Scholar] [CrossRef]
  93. Roche, F.P.; Sheahan, B.J.; O’Mara, S.M.; Atkins, G.J. Semliki Forest virus-mediated gene therapy of the RG2 rat glioma. Neuropathol. Appl. Neurobiol. 2010, 36, 648–660. [Google Scholar] [CrossRef]
  94. Wang, X.; Wang, J.P.; Rao, X.M.; Price, J.E.; Zhous, 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]
  95. Moran, T.P.; Burgents, J.E.; Long, B.; Ferrer, I.; Jaffee, E.M.; Tisch, R.M.; Johnston, R.E.; Serody, J.S. Alphaviral vector-transduced dendritic cells are successful therapeutic vaccines against neu-overexpressing tumors in wild-type mice. Vaccine 2007, 25, 6604–6612. [Google Scholar] [CrossRef]
  96. Lyons, J.A.; Sheahan, B.J.; Galbraith, S.E.; Mehra, R.; atkins, G.J.; Fleeton, M.N. Inhibition of angiogenesis by a Semliki Forest virus vector expressing VEGFR-2 reduces tumour growth and metastasis in mice. Gene Ther. 2007, 14, 503–513. [Google Scholar] [CrossRef]
  97. Daemen, T.; Riezebos-Brilman, A.; Bungener, L.; Regts, J.; Dontje, B.; Wiltschut, J. Eradication of established HPV16-transformed tumours after immunisation with recombinant Semliki Forest virus expressing a fusion protein of E6 and E7. Vaccine 2003, 21, 1082–1088. [Google Scholar] [CrossRef]
  98. Van de Wall, S.; Walczak, M.; van Rooij, N.; Hoogeboom, B.N.; Meijerhof, T.; Nijman, H.W.; Daemen, T. Tattoo delivery of a Semliki Forest virus based vaccine encoding Human Papillomavirus E6 and E7. Vaccines 2015, 3, 221–238. [Google Scholar] [CrossRef]
  99. Velders, M.P.; McElhiney, S.; Cassetti, M.C.; Eiben, G.L.; Higgins, T.; Kovacs, G.R.; Elmishad, A.G.; Kast, W.M.; Smith, L.R. Eradication of established tumors by vaccination with Venezuelan equine encephalitis virus replicon particles delivering human papillomavirus 16 E7 RNA. Cancer Res. 2001, 61, 7861–7867. [Google Scholar]
  100. Ying, H.; Zaks, T.Z.; Wang, R.F.; Irvine, K.R.; Kammula, U.S.; Marincola, F.M.; Leitner, W.W.; Restifo, N.P. Cancer therapy using a self-replicating RNA vaccine. Nat. Med. 1999, 5, 823–827. [Google Scholar]
  101. 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]
  102. Rodriguez-Madoz, J.R.; Prieto, J.; Smerdou, C. Semliki forest virus vectors engineered to express higher IL-12 levels induce efficient elimination of murine colon adenocarcinomas. Mol. Ther. 2005, 12, 153–163. [Google Scholar] [CrossRef]
  103. Smyth, J.W.; Fleeton, M.N.; Sheahan, B.J.; Atkins, G.J. Treatment of rapidly growing K-BALB and CT26 mouse tumors using Semliki Forest virus recombinant particles. Gene Ther. 2005, 12, 147–159. [Google Scholar] [CrossRef]
  104. Chikkanna-Gowda, C.P.; McNally, S.; Sheahan, B.J.; Fleeton, M.N.; Atkins, G.J. Inhibition of murine K-BALB and CT26 tumour growth using a Semliki Forest virus vector with enhanced expression of IL-18. Oncol. Rep. 2006, 16, 713–719. [Google Scholar] [CrossRef]
  105. Rodriguez-Madoz, J.R.; Liu, K.H.; Quetglas, J.I.; Ruiz-Guillen, M.; Otano, I.; Crettaz, J.; Butler, S.D.; Bellezza, C.A.; Dykes, N.L.; Tennant, B.C.; et al. Semliki forest virus expressing interleukin-12 induces antiviral and antitumoral responses in woodchucks with chronic viral hepatitis and hepatocellular carcinoma. J. Virol. 2009, 83, 12266–12278. [Google Scholar] [CrossRef]
  106. Rodriguez-Madoz, J.R.; Zabala, M.; Alfaro, M.; Prieto, J.; Kramer, M.G.; Smerdou, C. Short-term intratumoral interleukin-12 expressed from an alphaviral vector is sufficient to induce an efficient antitumoral response against spontaneous hepatocellular carcinomas. Hum. Gene Ther. 2014, 25, 132–143. [Google Scholar] [CrossRef]
  107. 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]
  108. Cheng, W.F.; Lee, C.N.; Su, Y.N.; Chai, C.Y.; Chang, M.C.; Polo, J.M.; Hung, C.F.; Wu, T.C.; Hsieh, C.Y.; Chen, C.A. Sindbis virus replicon particles encoding calreticulin linked to a tumor antigen generate long-term tumor-specific immunity. Cancer Gene. Ther. 2006, 13, 873–885. [Google Scholar] [CrossRef]
  109. 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]
  110. Yin, X.; Wang, W.; Zhu, X.; Wang, Y.; Wu, S.; Wang, Z.; Wang, L.; Du, Z.; Gao, J.; Yu, J. Synergistic antitumor efficacy of combined DNA vaccines targeting tumor cells and angiogenesis. Biochem. Biophys. Res. Commun. 2015, 465, 239–244. [Google Scholar] [CrossRef]
  111. Avogadri, F.; Merghoub, T.; Maughan, M.F.; Hirschhorn-Cymerman, D.; Morris, J.; Ritter, E.; Olmsted, R.; Houghton, A.N.; Wolchok, J.D. Alphavirus replicon particles expressing TRP-2 provide potent therapeutic effect on melanoma through activation of humoral and cellular immunity. PLoS ONE 2010, 5, e12670. [Google Scholar] [CrossRef]
  112. Goldberg, S.M.; Bartido, S.M.; Gardner, J.P.; Guevara-Patino, J.A.; Montgomery, S.C.; Perales, M.A.; Maughan, M.F.; Dempsey, J.; Donovan, G.P.; Olson, W.C.; et al. Comparison of two cancer vaccines targeting tyrosinase: Plasmid DNA and recombinant alphavirus replicon particles. Clin. Cancer Res. 2005, 11, 8114–8121. [Google Scholar] [CrossRef]
  113. Tseng, J.C.; Levin, B.; Hurtado, A.; Yee, H.; de Castro, I.P.; Jimenez, M.; Shamamian, P.; Jin, R.; Novick, R.P.; Pellicier, A.; et al. Systemic tumor targeting and killing by Sindbis viral vectors. Nat. Biotechnol. 2004, 22, 70–77. [Google Scholar] [CrossRef]
  114. Klimp, A.H.; van der Vaart, E.; Lansink, P.O.; Withoff, S.; de Vries, E.G.; Scherphof, G.L.; Wilschut, J.; Daemen, T. Activation of peritoneal cells upon in vivo transfection with a recombinant alphavirus expressing GM-CSF. Gene Ther. 2001, 8, 300–307. [Google Scholar] [CrossRef]
  115. Durso, R.J.; Andjelic, S.; Gardner, J.P.; Margitich, D.J.; Donovan, G.P.; Arrigale, R.R.; Wang, X.; Maughan, M.F.; Talarico, T.L.; Olmsted, R.A.; et al. 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]
  116. 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]
  117. 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]
  118. Riabov, V.; Tretyakova, 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]
  119. Colmenero, P.; Liljeström, P.; Jondal, M. Induction of P815 tumor immunity by recombinant Semliki Forest virus expressing the P1A gene. Gene Ther. 1999, 6, 1728–1733. [Google Scholar] [CrossRef]
  120. Morse, M.A.; Hobeika, A.C.; Osada, T.; Berglund, P.; Hubby, B.; Negri, S.; Niedzwiecki, D.; Devi, G.R.; Burnett, B.K.; Clay, T.M.; et al. 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]
  121. Gomes, I.M.; Maia, C.J.; Santos, C.R. STEAP proteins: From structure to applications in cancer therapy. Mol. Cancer Res. 2012, 10, 573–587. [Google Scholar] [CrossRef]
  122. Bins, A.D.; Jorritsma, A.; Wolkers, M.C.; Hung, C.F.; Wu, T.C.; Schumacher, T.N.; Haanen, J.B. A rapid and potent DNA vaccination strategy defined by in vivo monitoring of antigen expression. Nat. Med. 2005, 11, 899–904. [Google Scholar] [CrossRef]
  123. Vähä-Koskela, M.J.; Kallio, J.P.; Jansson, L.C.; Heikkilä, J.E.; Zakhartchenko, V.A.; Kallajoki, M.A.; Kähäri, V.M.; Hinkkanen, A.E. Oncolytic capacity of attenuated replicative Semliki Forest virus in human melanoma xenografts in severe combined immunodeficient mice. Cancer Res. 2006, 66, 7185–7194. [Google Scholar] [CrossRef]
  124. 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]
  125. Huttner, A.; Dayer, J.A.; Yerly, S.; Combescure, C.; Auderset, F.; Desmeules, J.; Eickmann, M.; Finckh, A.; Goncalves, A.R.; Hooper, J.W.; et al. 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]
  126. Fuchs, J.D.; Frank, I.; Elizaga, M.L.; Allen, M.; Frahm, N.; Kochar, N.; Li, S.; Edupuganti, S.; Kalams, S.A.; Tomaras, G.D.; et al. First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Vesicular Stomatitis Virus Human Immunodeficiency Virus-1 gag Vaccine (HVTN 090). Open Forum Infect. Dis. 2015. [Google Scholar] [CrossRef]
  127. Lundstrom, K. Biology and application of alphaviruses in gene therapy. Gene Ther. 2005, 12, S92–S597. [Google Scholar] [CrossRef]
  128. Ren, H.; Boulikas, T.; Lundstrom, K.; Söling, A.; Warnke, P.C.; Rainov, N.G. Immunogene therapy of recurrent glioblastoma multiforme with a liposomally encapsulated replication incompetent Semliki Forest virus vector carrying the human interleukin-12 gene—A phase I/II protocol. J. Neurooncol. 2003, 64, 147–154. [Google Scholar] [CrossRef]
  129. Slovin, S.F.; Kehoe, M.; Durso, R.; Fernandez, C.; Olson, W.; Gao, J.P.; Israel, R.; Scher, H.I.; Morris, S. A phase I dose escalation trial of vaccine replicon particles (VRP) expressing prostate-specific membrane antigen (PSMA) in subjects with prostate cancer. Vaccine 2013, 31, 943–949. [Google Scholar] [CrossRef]
  130. Alba, R.; Bosch, A.; Chillon, M. Gutless adenovirus: Last-generation adenovirus for gene therapy. Gene Ther. 2005, 12, S18–S27. [Google Scholar] [CrossRef]
  131. Schiedner, G.; Morral, N.; Parks, R.S.; Wu, Y.; Koopmans, S.C.; Langston, C.; Graham, F.L.; Beuadet, A.L.; Kochanek, S. Genomic DNA transfer with a high-capacity adenovirus vector results in improved in vivo gene expression and decreased toxicity. Nat. Genet. 1998, 18, 180–183. [Google Scholar] [CrossRef]
  132. Wang, F.; Wang, Z.; Tian, H.; Qi, M.; Zhai, Z.; Li, S.; Li, R.; Zhang, H.; Wang, W.; Fu, S.; et al. Biodistribution and safety assessment of bladder cancer specific oncolytic adenovirus in subcutaneous xenografts tumor model in nude mice. Curr. Gene Ther. 2012, 12, 67–76. [Google Scholar] [CrossRef]
  133. Andtbacka, R.H.; Agarwala, S.S.; Ollila, D.W.; Hallmeyer, S.; Milhem, M.; Amatruda, T.; Nemunaitis, J.J.; Harrington, K.J.; Chen, L.; Shilkrut, M.; et al. Cutaneous head and neck melanoma in OPTiM, a randomized phase 3 trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor for the treatment of unresected stage IIIB/IIIC/IV melanoma. Head Neck. 2016. [Google Scholar] [CrossRef]
  134. Zhang, L.; Tatsuya, T.; Nishiyama, Y. Oncotarget Strategies For Herpes Simplex Virus-1. Curr. Gene Ther. 2016, 16, 130–143. [Google Scholar] [CrossRef]
  135. Chahal, J.S.; Khan, O.F.; Cooper, C.L.; McPartlan, J.S.; Tsosie, J.K.; Tilley, L.D.; Sidik, S.M.; Lourido, S.; Langer, R.; Bavari, S.; et al. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges with a single dose. Proc. Natl. Acad. Sci. USA 2016, 113, E4133–E4142. [Google Scholar] [CrossRef]
  136. Leitner, W.W.; Ying, H.; Driver, D.A.; Dubensky, T.W.; Restifo, N.P. Enhancement of tumor-specific immune response with plasmid DNA replicon vectors. Cancer Res. 2000, 60, 51–55. [Google Scholar]
Figure 1. SFV-Based Expression Systems. (A) Replication-deficient system. In vitro transcribed RNA from expression and helper vectors are transfected into BHK-21 cells for generation of replication-deficient particles; (B) Replication-proficient system. In vitro transcribed RNA from full-length vector is transfected into BHK-21 cells for generation of replication-proficient particles; (C) DNA layered system. Plasmid DNA is transfected into host cells. 26S, SFV26S subgenomic promoter; CMV, cytomegalovirus promoter; pA, polyadenylation signal; SP6, SP6 phage RNA polymerase promoter.
Figure 1. SFV-Based Expression Systems. (A) Replication-deficient system. In vitro transcribed RNA from expression and helper vectors are transfected into BHK-21 cells for generation of replication-deficient particles; (B) Replication-proficient system. In vitro transcribed RNA from full-length vector is transfected into BHK-21 cells for generation of replication-proficient particles; (C) DNA layered system. Plasmid DNA is transfected into host cells. 26S, SFV26S subgenomic promoter; CMV, cytomegalovirus promoter; pA, polyadenylation signal; SP6, SP6 phage RNA polymerase promoter.
Vaccines 04 00039 g001
Figure 2. Kunjin Virus-Based Expression Systems. Expression vector based on Kunjin virus RNA and DNA expression systems. 3′ UTR, 3′ untranslated region; 5′ UTR, 5′ untranslated region; C20, first 20 amino acids of KUN C protein; CMV, cytomegalovirus promoter; E22, 22 last amino acids of KUN E protein; F, FMDV (foot-and-mouse disease virus) 2A autoprotease; HDVr, hepatitis delta virus ribozyme; ns1-5, nonstructural proteins; pA, polyadenylation signal; SP6, SP6 phage RNA polymerase promoter; U, mouse ubiquitin sequence.
Figure 2. Kunjin Virus-Based Expression Systems. Expression vector based on Kunjin virus RNA and DNA expression systems. 3′ UTR, 3′ untranslated region; 5′ UTR, 5′ untranslated region; C20, first 20 amino acids of KUN C protein; CMV, cytomegalovirus promoter; E22, 22 last amino acids of KUN E protein; F, FMDV (foot-and-mouse disease virus) 2A autoprotease; HDVr, hepatitis delta virus ribozyme; ns1-5, nonstructural proteins; pA, polyadenylation signal; SP6, SP6 phage RNA polymerase promoter; U, mouse ubiquitin sequence.
Vaccines 04 00039 g002
Figure 3. Measles Virus-Based Expression system. The measles virus structural proteins are flanked by T7 RNA polymerase promoter and the T7 RNA polymerase terminator. Foreign genes can be inserted between the P and M or H and L genes. H, MV hemagglutinin; L, MV L protein; M, MV matrix protein; N, MV nucleocapsid protein; T7, T7 RNA polymerase promoter; T7 term, T7 RNA polymerase terminator.
Figure 3. Measles Virus-Based Expression system. The measles virus structural proteins are flanked by T7 RNA polymerase promoter and the T7 RNA polymerase terminator. Foreign genes can be inserted between the P and M or H and L genes. H, MV hemagglutinin; L, MV L protein; M, MV matrix protein; N, MV nucleocapsid protein; T7, T7 RNA polymerase promoter; T7 term, T7 RNA polymerase terminator.
Vaccines 04 00039 g003
Figure 4. Rabies Virus-Based Expression Systems. The structural genes are from the HEP-Flury strain except the G protein from the CVS strain. Foreign genes can be inserted between the N and P or G and L genes, respectively. CMV, cytomegalovirus promoter; G, rabies G protein; L, rabies L protein; M, rabies matrix protein; N, rabies nucleocapsid protein.
Figure 4. Rabies Virus-Based Expression Systems. The structural genes are from the HEP-Flury strain except the G protein from the CVS strain. Foreign genes can be inserted between the N and P or G and L genes, respectively. CMV, cytomegalovirus promoter; G, rabies G protein; L, rabies L protein; M, rabies matrix protein; N, rabies nucleocapsid protein.
Vaccines 04 00039 g004
Figure 5. Schematic Presentation of the Life-Cycle of Self-Replicating RNA Viruses and Their Advantages. Several cell receptors are recognized providing a broad range of susceptible host cells. RNA released in the cytoplasm is immediately subjected to RNA replication and translation. Extreme RNA replication is the basis for highly efficient transgene expression.
Figure 5. Schematic Presentation of the Life-Cycle of Self-Replicating RNA Viruses and Their Advantages. Several cell receptors are recognized providing a broad range of susceptible host cells. RNA released in the cytoplasm is immediately subjected to RNA replication and translation. Extreme RNA replication is the basis for highly efficient transgene expression.
Vaccines 04 00039 g005
Table 1. Self-replicating RNA viral vector-based immunizations against viral diseases.
Table 1. Self-replicating RNA viral vector-based immunizations against viral diseases.
VirusTargetVectorImmunizationResponseReference
InfluenzaNPSFV VLPsmousesystemic NP immune response[35]
HAVEE VLPschickenprotection against influenza virus[36]
HAVEE VLPsswineprotection against influenza virus[37]
HAVEE VLPsswineprotection against influenza virus[38]
HArMVmouseneutralizing Abs[39]
cHAVSVmouseprotection against influenza virus[40]
HIVGagKunjin VLPsmouseprotection against HIV[41]
EnvSFV VLPsmouseneutralizing Abs, humoral response[42]
gp41SFV-VLPsmousegeneration of mAbs[43]
EnvSFV DNAmouseT cell and IgG immune responses[44]
SIVGag-PolKunjin VLPsmacaquesprotection against SIV[45]
EnvVSV VLPsmacaquesneutralizing Abs [46]
Gag-EnvVSV VLPsmacaquesprotection against SIV[47]
Gag-EnvRABV VLPsmacaquesprotection against SIV[48]
EbolaGPKunjin VLPsguinea pigprotection against Ebola[49]
GPKunjin VLPsprimateprotection against Ebola[50]
GPVSV VLPsmacaquesprotection against Ebola[51,52]
GP, NPVEE VLPsmouseprotection against Ebola[53]
NPVEE VLPsmouseprotection against Ebola[54]
LassaGVSV VLPsguinea pigprotection against Lassa[55]
GVEE VLPsguinea pigprotection against Lassa[56]
SARS-CoVGVEE VLPsmouseprotection against SARS-CoV[57]
MERS-CoVGMVmouseprotection against SARS-CoV[58]
RSVFMVratprotection against RSV[39]
FVEE LNPsmouseprotection against RSV[59]
FVEE VLPsprimateprotection against RSV [60]
MPVFVEE VLPsprimateprotection against MPV[60]
DengueDV2-HBsAgMVmouseneutralizing Abs[61]
DV2MVmouseprotection against dengue virus[62]
prME-E85VEE VLPsmacaquesprotection against dengue virus[63]
prME-E85VEE VLPsmouseprotection against dengue virus[64]
HBVMHBSFV-VSV Gmouseprotection against HBV[65]
DV2-HBsAgMVmouseprotection against HBV[62]
HBsAgMVmacaquesprotection against HBV[66]
CMVgB-pp65/IE1VEE VLPshumanneutralizing Abs[67]
Abs, antibodies; cHA, chimeric hemagglutinin; CMV, cytomegalovirus; DV2, dengue virus 2; G, glycoprotein; HA, hemagglutinin; HBV, hepatitis B virus; HBsAg, HBV surface antigen; LNPs, lipid nanoparticles; mAbs, monoclonal antibodies; MERS-CoV, Middle East respiratory syndrome coronavirus; MV, measles virus; MPV, metapneumonia virus; NP, nucleoprotein; RABV, rabies virus; RSV, respiratory syncytial virus; SARS-CoV, severe acute respiratory syndrome coronavirus; SFV, Semliki Forest virus; VEE, Venezuelan equine encephalitis virus; VLPs, virus-like particles.
Table 2. Self-Replicating RNA Viral Vector-Based Immunizations against Infectious Diseases.
Table 2. Self-Replicating RNA Viral Vector-Based Immunizations against Infectious Diseases.
AgentTargetVectorImmunizationResponseReference
P. falciparumAg Pf332SFV VLPs/RNAmouseimmunological memory[68]
M. tuberculosisAg 85ASIN DNAmouseprotection against M. tuberculosis[69]
C. botulinumBoNTA-HcSFV DNAmouseAb and lymphoproliferative response[70]
B. abortusIF3SFV VLPsmouseprotection against Brucella[71]
B. antracisPASIN VLPsmouseprotection against B. antracis[72]
MalariaCSSIN VLPsmouseprotection against malaria[73]
L. monocytogenesOVAVSV-GPmouseprotection against Listeria[74]
PrionPRNPSFV VLPsmousemonoclonal Abs[75]
StaphylococcusSEBVEE VLPsmouseprotection against enterotoxin[76]
Abs, antibodies; CS, circumsporozoite protein; IF3, translation initiation factor 3; MV, measles virus; MPV, metapneumonia virus; OVA, ovalbumin; PRNP, prion protein; SEB, staphylococcus enterotoxin B; SFV, Semliki Forest virus; SIN, Sindbis virus; VEE, Venezuelan equine encephalitis virus; VLPs, virus-like particles; VSV-GP, vesicular stomatitis virus pseudotyped with lymphocytic choriomeningitis glycoprotein.
Table 3. Self-Replicating RNA Viral Vector-Based Immunizations against Cancers.
Table 3. Self-Replicating RNA Viral Vector-Based Immunizations against Cancers.
CancerTargetVectorResponseReference
BrainGFP, SLAM, EGFRMVreplication in/lysis of cancer cells[79]
EndostatinSFV VLPstumor inhibition[89]
miR-124SFV-miR-124prolonged survival[90]
IL-12SFV-IL-12prolonged survival[91,92,93]
BreastCEAMVtumor growth delay, better survival[83]
NeuSIN DNAimmune responses, tumor protection[94]
NeuVEE VLPs + DCstumor regression by transduced DCs[95]
VEGFR-2SFV VLPstumor inhibition[96]
CervicalHPV E6, 7SFV VLPstumor eradication[97,98]
HPV E7VEE VLPseradication of existing tumors[99]
HPV E7 EpitopeKunjin VLPs/RNA/DNAtumor protection in mice[88]
ColonGM-CSFKunjin VLPsregression of tumors and metastasis[87]
VEGFR-2SFV VLPsreduced tumor and metastasis growth[96]
LacZSFV RNAtumor protection in mice[100]
Lac ZSIN VLPsanti-tumor CD8+ T-cell immunity[101]
IL-12SFV VLPstumor elimination[102]
SFVSFV VLPstumor growth inhibition[103]
IL-18SFV VLPstumor regression in mice[104]
LiverIL-12SFV VLPsanti-tumor responses in woodchucks[105,106]
LungHPV E6/E7SFV + Sun + Radtumor-free survival[107]
HPV E7-CRTSIN VLPslong-term anti-tumor effect[108]
EGFPSFV VLPsapoptosis, tumor regression in mice[109]
MelanomaGM-CSFKunjin VLPstumor regression[87]
VEGF-2-IL-12 + Sur + β-hCGSFV VLPstumor inhibition[110]
TRP-2VEE VLPshumoral and cellular immunity[111]
TyrVEE VLPsT-cell responses, tumor protection in mice[112]
OvarianCEA, NISMVsuperior dual therapy[81]
IL-12SIN VLPstumor targeting, eradication[113]
IL-18SFV VLPstherapeutic anti-tumor response[91]
GM-CSFSFV VLPstumor growth inhibition[114]
PancreaticMatrix proteinVSV VLPskilling of tumor cells in vitro and in vivo[85]
ProstateCEAMVreplication in/lysis of cancer cells[79]
PSMAVEE VLPscellular and humoral immunity in mice[115]
STEAPVEE VLPsCD8+ T-cell response, tumor growth delay[116]
PSCADNA + VEE VLPslong-term protective immune response[117]
SarcomaPSAVEE VLPsPSA-cell clearance, tumor growth delay[118]
SkinSFVSFV VLPstumor growth inhibition[103]
P1ASFV VLPsstrong CTL-response, tumor protection[119]
CEA, carcinoembryonic antigen; CRT, calreticulin; CTL, cytotoxic T lymphocyte; EGFP, enhanced green fluorescent protein; EGFR, epidermal growth factor receptor; GFP, green fluorescent protein; GM-CSF, granulocyte macrophage colony-stimulating factor; HPV, human papilloma virus; MV, measles virus; NIS, sodium iodide symporter; PSMA, prostate-specific membrane antigen; PSCA, prostate stem cell antigen; RABV, rabies virus; SFV, Semliki Forest virus; SIN, Sindbis virus; SLAM, signaling lymphocytic activation molecule; STEAP, six-transmembrane epithelial antigen of the prostate; Sun, sunitab; Sur, survivin; TRP, tyrosine-related protein; Tyr, melanoma antigen tyrosinase; VEE, Venezuelan equine encephalitis virus; VEGFR, vascular endothelial growth factor receptor; VLPs, virus-like particles; VSV, vesicular stomatitis virus.
Table 4. Comparison of Self-replicating RNA Viral Vectors with other Viral Vectors.
Table 4. Comparison of Self-replicating RNA Viral Vectors with other Viral Vectors.
Viral VectorGenomeCapacitySpecial Features
AlphavirusssRNA6–8 kbbroad host range, high titer, cytoplasmic RNA, extreme transient expression, no chromosomal integration, choice of DNA, RNA replicon and particle delivery
FlavivirusssRNA5 kbbroad host range, packaging system, choice of DNA, RNA replicon and particle delivery
Measles virusssRNA5 kbpackaging cell line, measles virus strains for immunization, cytoplasmic RNA
RhabdovirusssRNA5 kbreverse genetics systems, broad host range cytoplasmic RNA
AdenovirusdsDNA>8 kbbroad host range, packaging cell line, nuclear translocation necessary, transient expression, potential integration
AAVssDNA<4 kbmultiple AAV serotypes for avoiding immune responses, nuclear translocation necessary, chromosomal integration
Herpes simplex virusdsDNA30–40 kblarge packaging capacity, nuclear translocation necessary, latent long-term transgene expression after integration
LentivirusdsRNA8 kbtransduction of dividing and non-dividing cells, nuclear translocation necessary, chromosomal integration
RetrovirusdsRNA4 kbtransduction of only dividing cells, nuclear translocation necessary, chromosomal integration
VacciniadsDNA25 kblarge packaging capacity, nuclear translocation necessary
AAV, adeno-associated virus; dsDNA, double-stranded DNA; ssDNA, single-stranded DNA; ssRNA, single-stranded RNA.

Share and Cite

MDPI and ACS Style

Lundstrom, K. Replicon RNA Viral Vectors as Vaccines. Vaccines 2016, 4, 39. https://doi.org/10.3390/vaccines4040039

AMA Style

Lundstrom K. Replicon RNA Viral Vectors as Vaccines. Vaccines. 2016; 4(4):39. https://doi.org/10.3390/vaccines4040039

Chicago/Turabian Style

Lundstrom, Kenneth. 2016. "Replicon RNA Viral Vectors as Vaccines" Vaccines 4, no. 4: 39. https://doi.org/10.3390/vaccines4040039

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