• Open Access
    Review

    Self-replicating RNA viruses in vaccine development

    Kenneth Lundstrom *

    Explor Med. 2023;4:670–687 DOI: https://doi.org/10.37349/emed.2023.00168

    Received: March 16, 2023 Accepted: April 19, 2023 Published: October 10, 2023

    Academic Editor: Ragusa Marco, Università degli Studi di Catania, Italy

    This article belongs to the special issue RNA World in Health and Disease

    Abstract

    Self-replicating RNA viruses such as alphaviruses, flaviviruses, paramyxoviruses, and rhabdoviruses have been engineered as expression vectors for vaccine development. The prominent feature of self-replicating RNA viruses is their RNA-dependent RNA polymerase activity, which generates massive self-amplification of RNA in the cytoplasm of infected host cells leading to extreme levels of transgene expression. Infectious diseases have been targeted by overexpression of surface proteins of pathogens as antigens for vaccine development. Moreover, overexpression of tumor-associated antigens and immunostimulatory genes has been the basis for cancer vaccines. Proof-of-concept of robust antigen-specific immune responses and protection against challenges with lethal doses of infectious agents have been demonstrated. Likewise, vaccine development against various cancers has elicited strong immune responses and resulted in tumor regression and eradication, cure, and prolonged survival in animal tumor models. Good safety and immune responses have been achieved in clinical trials. The ERVEBO® vaccine, based on the vesicular stomatitis virus, has been approved for immunization against the Ebola virus disease.

    Keywords

    RNA viruses, self-replication, infectious diseases, cancer

    Introduction

    The classic approach for vaccine development against viral infections has relied on the application of killed or live-attenuated whole viruses [1]. The progress in recombinant protein expression in the 1980s and 1990s facilitated the development of protein subunit and peptide vaccines [2]. It also opened the possibility of utilization of viral and non-viral vectors for efficient delivery and overexpression of antigens suitable for eliciting robust antibody responses for vaccine development against infectious diseases and cancers [3]. The recent coronavirus disease 2019 (COVID-19) pandemic has taken vaccine development to unprecedented levels related to both viral vectors and nucleic acid-based vaccines [4]. Moreover, the application of viral vectors for the expression of tumor-associated antigens (TAAs) and immunostimulatory genes has been an attractive alternative for cancer vaccine development [57]. Additionally, therapeutic and prophylactic efficacy has been obtained by viral vector-based delivery of anti-tumor, and suicide genes [8].

    A variety of viral vectors have been utilized for vaccine development against infectious diseases and cancers as described before [9]. For this reason, some key findings are briefly presented below. For example, adenovirus (Ad)-based vaccines such as ChAdOx1 nCoV-19 [10], Ad26.COV2.S [11], and Ad5S-nb2 [12] have demonstrated robust immune responses and good vaccine efficacy against severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) as well as being granted emergency use authorization (EUA) for mass vaccinations in different countries [1315]. In the case of cancer therapy, Ad vectors demonstrated complete regression of pancreatic neuroendocrine tumors (PNETs) in mice [16]. The administration of the herpes simplex virus (HSV)-HF10 resulted in substantial breast tumor regression and prolonged survival in mice [17] and in partial response (PR) in three patients, stable disease (SD) in four patients and in progressive disease (PD) in two patients with pancreatic cancer in a phase I trial [18]. In the context of retroviruses, retroviral replicating vector (RRV) Toca 511 was administered to patients with recurrent or progressive high-grade glioma (HGG) in a phase I study, showing statistically prolonged survival [19]. However, in a phase II/III trial in HGG patients, no prolongation of overall survival was detected [20]. In a phase II/III trial, the administration of HSV talimogene laherparepvec (T-VEC) vector expressing the granular macrophage-colony stimulating factor (GM-CSF) demonstrated good tolerance and promising therapeutic efficacy in melanoma patients [21], which has contributed to the approval of HSV T-VEC for the treatment of advanced melanoma in the US, Europe, and Australia [22].

    In this review, the focus is solely on vaccine development by applying self-replicating RNA viruses. As detailed reviews have recently been published on both vaccine development against infectious diseases and cancers [23, 24], the goal here is to discuss the different types of viral vectors used and through the presentation of limited examples describe findings from both preclinical studies in animal models and evaluation in human volunteers and in cancer patients in clinical trials.

    Self-replicating RNA virus vectors

    Among self-replicating RNA viruses, alphaviruses, flaviviruses, paramyxoviruses, and rhabdoviruses have been engineered as expression vector systems. However, while the single-stranded RNA (ssRNA) genomes of alphaviruses [25] and flaviviruses [26] are of positive polarity, paramyxoviruses [27] and rhabdoviruses [28] possess negative sense ssRNA genomes. The polarity plays an essential role in the engineering and utilization of self-replicating RNA virus vectors as described below and illustrated in Figure 1. The positive sense ssRNA is directly translated in the cytoplasm, which is substantially enhanced by the formation of the replicase complex (RC) after processing of the non-structural polyprotein expressed from the non-structural protein 1–4 (nsP1–4) genes resulting in an estimated 106 copies of subgenomic RNA per cell [29].

    Lifecycle of positive- and negative-stranded self-replicating RNA viruses. In the case of positive-stranded RNA viruses (left), translation from the RNA plus strand can occur directly, whereas for negative-stranded RNA viruses (right) a plus strand copy needs to be synthesized first. In both cases, massive RNA replication takes place, RNA genomes are packaged into nucleocapsids, and viral particles are released from cells by budding. RdRp: RNA-dependent RNA polymerase

    Alphaviruses

    Alphavirus expression systems have been engineered to allow the utilization of recombinant viral particles, RNA replicons, and DNA replicons for the Semliki Forest virus (SFV) [30], Sindbis virus (SIN) [31], and Venezuelan equine encephalitis virus (VEE) (Figure 2) [32]. In the case of recombinant viral particles, both replication-deficient and replication-proficient systems have been developed. Briefly, the generation of replication-deficient particles takes place from co-electroporation or co-transfection of an alphavirus vector and a helper vector. The expression vector carries the nsP1–4 genes responsible for RC formation for massive viral RNA self-replication. The gene of interest (GoI) is introduced downstream of the highly efficient SFV subgenomic promoter (26S) located at the 3’ end of the nsP4 gene. As the alphavirus structural genes are deleted from the expression vector, they are provided in trans from the helper vector. RNA molecules from both expression and helper vectors are replicated in transfected host cells, but as the packaging signal is located in the nsP2 gene for SFV [33] or in the nsP1 gene for SIN and VEE [34], only RNA from the expression vector will be packaged into recombinant viral particles. These so-called suicide particles show the same broad host range as wild-type alphaviruses, but due to the absence of any viral structural genes, no further viral progeny will be produced. However, alphavirus suicide particles generate extreme levels of transgene expression. As no virus progeny is produced and the viral ssRNA is quickly degraded, active antigen production lasts for approximately 5–7 days in vivo, which is actually ideal for immunization with no risk of long-term extrachromosomal presence or chromosomal integration. In the case of replication-proficient alphavirus particles, transgene expression can be achieved by the introduction of the GoI either downstream of the non-structural genes or alternatively after the structural genes driven by a second 26S. Transfection of in vitro transcribed full-length alphavirus RNA and the GoI into mammalian host cells will generate fully replication-proficient particles. Importantly, as the generated alphavirus particles are both infectious and capable of generating viral progeny, the biosafety requirements are tighter compared to replication-deficient particles.

    Alphavirus expression systems illustrated by SFV vectors. (A) Recombinant viral particles. Co-electroporation/transfection of in vitro transcribed RNA from the expression vector carrying the nsP1–4 genes and the GoI and the helper vector providing the SFV E genes (C-p62-6K-E1) into mammalian host cells generates replication-deficient recombinant viral particles and expression of the GoI; (B) RNA replicons. In vitro transcribed RNA from the expression vector can be transfected to host cells or injected in vivo for expression of the GoI; (C) plasmid DNA replicons. Transfection of the plasmid DNA replicon vector, where the SP6 RNA polymerase promoter is replaced by a CMV promoter generated expression of the GoI. SP6: SP6 RNA polymerase promoter; pA: polyadenylation signal; C: capsid; p62: precursor of E2 and E2 envelope proteins; 6K: 6K protein; E1: E1 envelope protein; CMV: cytomegalovirus

    In the context of RNA-based delivery, in vitro transcribed RNA from the alphavirus expression vector containing the GoI can be directly administered to cell cultures or in vivo. However, the sensitivity of ssRNA to degradation [35] has triggered the encapsulation of RNA molecules in nanoparticles to improve their delivery, stability, and extended circulation [36]. Alternatively, DNA-based vectors can be used. In this context, the SP6 or T7 RNA polymerase promoter (T7) in alphavirus expression vectors has been replaced by a mammalian host cell compatible eukaryotic RNA polymerase type II promoter such as CMV [37]. The attractive features of DNA-based administration include their easy handling and rapid and inexpensive manufacturing. However, the application of these DNA replicons is dependent on efficient transfection methods, which are inferior to viral delivery. Another issue relates to the need for plasmid DNA to translocate to the nucleus for mRNA transcription followed by its transport to the cytoplasm for translation, which has substantially reduced transgene expression [38]. Different methods such as electroporation [39], gene gun [40], liposome [41], and polymer-based [42], nanoparticle formulation have been developed as well as the application of nuclear localization signals for optimized DNA [43].

    Flaviviruses

    In the case of flaviviruses, the Kunjin virus (KUN) has been subjected to the engineering of an expression system [44] (Figure 3). Generally, the GoI is introduced between the first 60 nucleotides of the C20 core protein and the last 22 codons of the E2 envelope protein [44]. As the GoI is expressed as part of a larger polyprotein, an FMDV2A protease sequence was engineered into the KUN vector for removing any foreign flanking regions [45]. Moreover, the generation of a packaging cell line has facilitated KUN particle production [46]. Other flaviviruses such as West Nile virus (WNV) [47], yellow fever virus (YFV) [48], Dengue virus (DENV) [49], tick-borne encephalitis virus (TBEV) [50], and Zika virus (ZIKV) [51] have been engineered. Additionally, expression vector systems have been constructed for the bovine viral diarrhea virus (BVDV) by insertion of the green fluorescent protein (GFP) reporter gene between the N(pro) and C genes in the non-cytopathic type-1 BVDV strain SD1 [52]. The classical swine fever virus (CSFV) has also been engineered as a bicistronic vector containing the GFP gene [53].

    Flavivirus expression vectors exemplified by KUN. (A) RNA replicon vector. The GoI is introduced between the first 60 nucleotides of the C20 core protein and the last 22 codons of the E2 envelope protein in the KUN genome; (B) DNA replicon vector. The SP6 RNA polymerase promoter has been replaced by a CMV promoter. UTR: untranslated region; U: ubiquitin; F: foot-and-mouth disease virus 2A protease; ns1–5: non-structural protein genes 1–5; HDVr: hepatitis D virus ribosome

    Rhabdoviruses

    In contrast to alphaviruses and flaviviruses, the negative-stranded ssRNA viruses such as rhabdoviruses the RNA self-replication is based on the large protein (L) gene coding for the RdRp and its cofactor in the phosphoprotein (P) gene [28]. Expression vectors for vesicular stomatitis virus (VSV) have been engineered by insertion of the GoI between the glycoprotein (G) and the L genes for the generation of recombinant VSV particles or alternatively by replacing the VSV G by the GoI for pseudovirus production (Figure 4A) [54, 55]. Pseudovirus VSV particles can be produced from plasmid DNA in transfected mammalian cells followed by infection with the VSV G-complemented VSVΔG, which can be used for infection of mammalian host cells leading to transgene expression but no generation of viral progeny [54]. Fully infectious VSV particles can be produced by the infection of mammalian producer cells with VSV G-complemented recombinant virus replacing the VSV G [55]. Reverse genetics has been applied for VSV based on vaccinia virus vectors [56]. Moreover, a vaccinia virus-free system has been engineered to allow the production of infectious VSV particles from DNA [57]. In the context of large-scale production, an inducible VSV L cell line has been engineered for the packaging of recombinant VSV particles [58]. In the context of rabies virus (RABV), expression vectors have been engineered, where the GoI is introduced between the nucleoprotein (N) and P genes, or alternatively, between the G and L genes (Figure 4B) [59]. A reverse genetics system has also been constructed for RABV based on vaccinia virus-free vectors [60].

    Rhabdovirus expression vectors. (A) VSV pseudovirus vector. The VSV-G protein gene is replaced by the GoI; (B) RABV vectors. The GoI can be inserted in the RABV vector either between the N and P genes or the G and L genes. M: matrix protein

    Paramyxoviruses

    Among the negative strand ssRNA paramyxoviruses, measles virus (MV) vectors have been engineered for the insertion of the GoI between the P and the M genes or alternatively between the hemagglutinin (HA) gene and the L genes (Figure 5) [27]. Packaging systems and helper cell lines have been engineered for MV, which will allow the rescue of replicating MV from plasmid DNA vectors [61, 62].

    Paramyxovirus expression vectors exemplified by MV. In MV vectors, the GoI is introduced either between the P and M genes or the HA and L genes. F: fusion protein; T7T: T7 terminator

    From bench to bedside

    Self-replicating RNA viruses have been used for a large number of studies in preclinical animal models and a limited number of clinical trials. This is also reflected in the very low number of approved vaccines, so far. In this section, examples of applications of self-replicating RNA viruses from preclinical studies to clinical trials are described and summarized in Table 1 and Table 2.

    Examples of preclinical studies and clinical trials on vaccines developed for infectious diseases

    DiseaseVectorFindings
    CHIKVMV-CHIKVProtection against CHIKV challenges in macaques [63]
    MV-CHIKVSeroconversion in 100% of volunteers in phase I [64]
    MV-CHIKVGood safety, and strong immune responses in phase II [65]
    EVDVSV-EBOV-GPProtection against two EBOV strains in macaques [66, 67]
    VSV-EBOV-GPGood safety and tolerability in phase I volunteers [68]
    VSV-ZEBOVHigh vaccine efficacy, and protection in phase III [69]
    VSV-ZEBOVHigh vaccine efficacy in phase III volunteers [70]
    VSV-ZEBOVApproval of ERVEBO® by the FDA and EMA [71]
    ZIKVMV-ZIKV-EZIKV-specific Abs, protection against ZIKV of mice fetus [72]
    MV-ZIKV-EDespite the completion of phase I, no results are available [73]
    MV-ZIKV-RSP-EPhase I study in progress [75]
    MV (TMV-083)Th1-biased Ab and T cell responses in mice [76]
    MV (TMV-083)Weak immune responses, phase I trial discontinued [77, 78]
    COVID-19VSV-SARS-CoV-2 SProtection against SARS-CoV-2 challenges in mice [79]
    VSV (V590)Weak immune responses, phase I trial discontinued [80, 81]
    VSVΔG-SARS-CoV-2 SProtection against SARS-CoV-2 in hamsters [82]
    VSVΔG-SARS-CoV-2 SPhase I/II study in progress [83]
    LNP-nCoVsaRNAStrong SARS-CoV-2 specific Ab responses in mice [86]
    LNP-nCoVsaRNASafe but not 100% seroconversion in phase I [87]
    Display full size

    Ab: antibody; CHIKV: chikungunya virus; EBOV-GP: Ebola virus-glycoprotein; EMA: European Medicines Agency; EVD: Ebola virus disease; FDA: US Food and Drug Administration; LNP: lipid nanoparticle; LNP-nCoVsaRNA: LNP-VEE-SARS-CoV-2 S RNA; SARS-CoV-2 S: SARS-CoV-2 spike; ZEBOV: EBOV-Zaire strain

    Examples of preclinical studies, clinical trials, and approved cancer vaccines

    DiseaseVectorFindings
    BreastVEE-HER-2Prevention of tumor growth and death in mice [89]
    VEE-HER-2Good tolerance, PR in 1 patient, SD in 2 patients in phase I [90]
    VEE-HER2Combination with pembrolizumab (ICB) in phase II [88]
    CervicalSFV-E6/E7Tumor regression or complete eradication in mice [91]
    SFV-E6/E7 (Vvax001)Good tolerability, HPV-specific responses in phase I [92]
    ColonVEE-CEAAnti-tumor activity in mice with MC38-CEA tumors [93]
    VEE-CEAImmune responses, prolonged survival in phase I patients [94]
    OvarianMVAnti-tumor activity in mice with ovarian tumor xenografts [95]
    MVSD in 67% of RROC patients in phase I [96]
    ProstateVEE-PSMARobust PSMA-specific immune responses in mice [97]
    VEE-PSMANo clinical changes in CRPC patients in phase I [98]
    PancreaticVEE-CEACEA-specific T-cell responses in transgenic mice [99]
    VEE-CEAAb and T-cell responses, prolonged survival in phase I [99]
    VariousChAd + VEE neoAgClinical benefits in cancer patients in phase I/II [100]
    Display full size

    CEA: carcinoembryonic antigen; ChAd: chimpanzee Ad; CRPC: castrate-resistant prostate cancer; E6/E7: human papillomavirus envelope proteins 6 and 7; HER-2: human epidermal growth factor receptor 2; HPV: human papillomavirus; ICB: immune checkpoint blockade; MC38: mouse colon cancer cell; neoAg: neo-antigen; PSMA: prostate-specific membrane antigen; RROC: refractory recurrent ovarian cancer

    Vaccines against infectious diseases

    In the case of infectious diseases, MV-based particles were applied for the expression of the CHIKV C and envelope (E) protein genes, which elicited strong immunogenicity and provided protection in immunized macaques [63]. Furthermore, the MV-CHIKV particles were subjected to a phase I trial in healthy volunteers [64]. After a single dose of MV-CHIKV, the seroconversion rate was 44–92%, and after a second dose, it reached 100% in all vaccinated volunteers. Moreover, in a phase II follow-up study, strong neutralizing antibodies were obtained without any serious adverse events registered [65].

    Related to EVD, VSV particles have been used for the expression of the EBOV-GP gene, which protected immunized macaques against challenges with the EBOV-Makona [66] and ZEBOV [67] strains. In the context of clinical evaluation, VSV-EBOV-GP showed good safety and tolerability in a phase I study in healthy volunteers [68]. Moreover, a good safety profile and excellent vaccine efficacy were demonstrated for the VSV-ZEBOV vaccine in 7,651 vaccinated individuals in a phase III trial in Guinea [69]. The VSV-ZEBOV vaccine efficacy was further confirmed in another phase III study in Guinea and Sierra Leone [70]. Approval of VSV-ZEBOV against EVD under the brand name ERVEBO® was granted in 2020 [71].

    In the case of ZIKV vaccine development, the ZIKV precursor membrane (prM) protein and the soluble form of the envelope protein (ZIKV-sE) were expressed from MV particles, which elicited E-protein specific and neutralizing antibodies in mice [72]. Moreover, challenges with ZIKV resulted in reduced virus load and prevented fetus infection in an allogenic mouse pregnancy model. Two clinical studies on MV-ZIKV have so far been conducted. Although a dose-finding phase I trial has been carried out in 48 volunteers with the MV-ZIKV (V186-001) vaccine candidate, no results have been published yet [73]. Additionally, the safety and immunogenicity of the MV-ZIKV-RSP (V187-001) vaccine candidate, similar to MV-ZIKV-E but with a different insert position [74], is under investigation in phase I study in volunteers in Austria [75].

    Due to the COVID-19 pandemic, massive efforts have been dedicated to the development of vaccines against SARS-CoV-2 resulting in EUA of vaccines based on Ad vectors and nucleic acids [13, 14]. Self-replicating RNA viruses have also been utilized. For example, MV-based expression of the SARS-CoV-2 S protein elicited strong Th1-biased antibody and T-cell responses in mice [76]. However, the MV-SARS-CoV-2 S (TMV-083) vaccine candidate induced disappointingly weak immune responses in volunteers in a phase I clinical study [77], which resulted in the termination of the trial [78]. The SARS-CoV-2 S gene has also been expressed from VSV vectors resulting in neutralizing antibody responses and protection against SARS-CoV-2 challenges in mice [79]. Moreover, the VSV-SARS-CoV-2 S (V590) vaccine candidate has been evaluated in 252 volunteers in a phase I trial [80]. Although good safety and tolerability profiles were achieved, the immune responses were weaker than those seen in COVID-19 patients, causing the discontinuation of the trial [81]. The replication-competent VSVΔG vector, where the VSV G protein was replaced by the SARS-CoV-2 S protein gene, was engineered [82]. Immunization of Syrian golden hamsters with VSVΔG-SARS-CoV-2 S induced strong neutralizing antibody responses and provided protection against challenges with SARS-CoV-2. Furthermore, the VSVΔG-SARS-CoV-2 S vaccine candidate has been subjected to a phase I/II trial where the safety and vaccine efficacy will be monitored for 12 months [83].

    Another application of self-replicating RNA viruses comprises RNA-based delivery. Due to the sensitivity of degradation of ssRNA, the VEE-SARS-CoV-2 S RNA has been formulated in LNPs. However, LNP formulations of self-replicating RNA molecules have proven more difficult than the smaller-size synthetic mRNA [84]. The application of the three FDA-approved ionizable lipids (6Z,9Z,28Z,31Z)-heptatriacont-6,9,28,31-tetrqene-19-yl 4-(dimethylamino) butanoate (MC3), 6-((2-hexyldecanoyl)oxy)hexyl-N-(4-hydroxybutyl)hexan-1-aminium (ALC-0315), and 9-heptadecanyl 8-{(2-hydroxyethyl)[6-oxo-6-(undecyloxy)hexyl]amino}octanoate (SM-102) for LNP formulations contributed to achieving maximal transgene expression. Another aspect to consider for LNP formulations is its effect on the acute systemic cytokine responses to self-replicating RNA [85]. For example, LNP formulations that induced stronger cytokine responses also elicited enhanced antibody responses showing an impact on the adaptive immune response after vaccinations with LNP-formulated self-replicating RNA. Intramuscular immunization of BALB/c mice with LNP-nCoVsaRNA induced strong dose-dependent SARS-CoV-2 specific antibody responses and neutralization of both pseudovirus and wild-type virus [86]. The LNP-nCoVsaRNA vaccine candidate has been subjected to the first-in-human dose-ranging phase I trial [87]. Compared to conventional mRNA-based vaccines, substantially lower doses of 0.1 to 10 μg of LNP-nCoVsaRNA were administered to 192 healthy volunteers. The immunization was safe with no serious adverse events detected. However, although seroconversion was obtained, it did not reach 100% and modifications are required to optimize humoral responses.

    Cancer vaccines

    In the context of cancer vaccines, examples from studies in animal tumor models and clinical trials [88] are presented and summarized in Table 2. For example, VEE particles expressing the HER-2/neuroblastoma (neu) gene significantly inhibited or prevented the growth of HER-2/neu-expressing mouse breast tumor cells in mice after injection into mammary tissue or after intravenous administration [89]. Moreover, tumor formation and death in mouse mammary tumor virus (MMTV)-C-neu transgenic mice were completely prevented. VEE particles expressing the extracellular domain (ECD) and transmembrane (TM) domains of HER-2 have been subjected to a phase I study in patients with stage IV HER-2 overexpressing breast cancer, which showed good tolerance and resulted in PR in one patient and continued SD in two other patients [90]. A phase II trial in combination with ICB has been initiated with pembrolizumab in women with advanced HER-2+ breast cancer [88]. Three patients received combination therapy, which was well tolerated and did not cause any dose-related toxicity. Moreover, enhanced lymphocyte infiltration in tumors and increased infiltration of both CD4+ and CD8+ T-cells and B-cells were observed.

    In the case of cervix cancer caused by the HPV, vaccines have been developed against the HPV oncoproteins E6 and E7. For example, expression of the E6/E7 fusion protein from SFV vectors generated tumor regression and complete eradication of established tumors in immunized C57BL/6 mice [91]. Moreover, the Vvax001 vaccine candidate, based on an SFV vector containing the translation enhancement signal from the SFV C gene expressing the HPV E6/E7 fusion protein, has been subjected to a phase I study in 12 cervical intraepithelial neoplasia patients [92]. High safety and tolerability were seen, and HPV-specific immune responses were obtained in all 12 patients in the study.

    Related to colon cancer, VEE particles expressing the CEA elicited immune responses and generated anti-tumor activity in mice implanted with MC38-CEA-2 colon tumors, which could be further enhanced by co-immunization with VEE-CEA and VEE particles expressing interleukin-12 (VEE-IL-12) [93]. In a clinical setting, VEE-CEA particles were evaluated in stage IV colorectal cancer in a phase I trial [94]. Moreover, stage III colorectal cancer patients were included in the study. Immunization elicited antigen-specific immune responses and also resulted in prolonged overall survival.

    In the context of ovarian cancer, MV vectors were applied for the expression of CEA and the sodium iodide symporter (NIS) in mice with implanted SKOV3ip.1 xenografts [95]. The combination of MV-CEA and MV-NIS showed superior anti-tumor activity compared to treatment with either MV-CEA or MV-NIS alone. In a phase I trial, the MV-CEA vector was evaluated in patients with taxol and RROC [96]. Intraperitoneal MV-CEA administration was safe and well tolerated. The dose-dependent biological activity resulted in SD in 14 out of 21 RROC patients.

    VEE particles have been applied for the expression of the PSMA as a vaccine candidate for prostate cancer [97]. Immunization of BALB/c and C57BL/6 mice elicited strong PSMA-specific immune responses. Furthermore, patients with progressive CRPC have been subjected to VEE-PSMA immunizations in the first-in-human phase I trial [98]. The immunization was well tolerated, but no significant clinical changes such as those measured by PSA or circulating tumor cells were detected.

    In the context of pancreatic cancer, VEE particles expressing the CEA gene were administered to transgenic C57BL6-CEA mice, which elicited a high magnitude of CEA-specific T-cell responses [99]. A phase I clinical trial has been conducted with VEE-CEA particles in pancreatic cancer patients [99]. Clinically relevant antibody and T-cell responses were seen in patients, which were intramuscularly injected with VEE-CEA particles. The overall survival was also improved.

    Finally, self-replicating RNA has been used in a prime-boost regimen, where the primary immunization has taken place with ChAd vectors followed by self-replicating VEE RNA expressing neoepitopes from autologous tumors [88]. Different types of cancers such as colorectal cancer, gastroesophageal adenocarcinoma, and non-small cell lung cancer have been evaluated in a phase I/II clinical trial according to the ChAd-VEE RNA prime-boost immunization strategy [100]. No dose-related toxicity was detected in any of the 26 patients enrolled and only minor adverse events such as fever and reactions at the injection site were recorded. After prime immunization, neoantigen-specific T-cell responses were recorded, which further increased after the booster vaccination with self-replicating RNA. Clinically, a complete response (CR) was confirmed in one gastroesophageal cancer patient, SD in five patients, PD in eleven patients, and no measurable disease in two individuals.

    What has been learned?

    The common factors for all vaccine development for both infectious diseases and cancers are the application of self-replicating RNA viral vectors and the overexpression of recombinant proteins used for antigen production. Moreover, the route of administration is of importance, where intramuscular injection is the most common approach. Below are summarized the findings from preclinical studies and clinical trials.

    Preclinical studies have provided proof-of-concept for the vaccine establishment to demonstrate on a small scale as cost-effectively as possible that robust immune responses can be induced and in the case of infectious diseases protection against challenges with lethal doses of infectious agents can be achieved. In the case of cancer vaccines, demonstration of tumor growth inhibition, tumor eradication, and complete cure in animal models have been indications that expensive clinical trials can be initiated. Moreover, cancer vaccines are expected to provide protection against challenges with tumor cells in animal models. As described above and summarized in Table 1 and Table 2, these criteria have been reached both for infectious diseases and various cancers, which has allowed moving forward to clinical evaluation.

    In the case of clinical assessment, relatively few studies have been conducted, so far. In quite a few cases of infectious diseases, relatively modest immune responses have been obtained in phase I and II trials in healthy volunteers, which is a clear indication of the less straightforward transition from animal studies to humans. For example, clinical evaluation of both MV- and VSV-based vaccine candidates against SARS-CoV-2 was discontinued in phase I despite promising results in animal models [78, 81]. In contrast, VSV-based vaccine development against EDV was highly successful, showing excellent vaccine efficacy in phase III studies [69, 70], leading to vaccine approval by both the FDA and the EMA [71].

    The trend for cancer vaccines is similar. Several studies have demonstrated tumor inhibition [89], tumor eradication [93], and even long-term tumor-free survival [91] in different animal models. However, the transition to humans has resulted in less efficient tumor eradication, although PR, SD, and CR have been obtained in individual cases [90, 97].

    What can be done?

    Although plenty of engineering has been dedicated to the development of more efficient self-replicating RNA virus vectors, there is still room for improvement (Table 3). Related to vector development, several efforts have been made to specifically generate less cytotoxic alphavirus vectors by introducing point mutations in the non-structural protein genes [101, 102]. In the case of cancer vaccines and especially targeting existing tumors, this approach is not necessarily of advantage. Instead, the application of naturally occurring or engineered oncolytic viruses, which specifically replicate in tumor cells leading to their killing [103] might be a better approach. Recently, trans-amplifying SFV vectors, a split-vector derivative of self-replicating RNA vectors, have been engineered for the amplification of transreplicon RNA coding for antigens, which requires minimal amounts of RNA for vaccine development [104].

    Potential vaccine improvement strategies and expectations

    StrategyApproachExpectations
    Vector engineeringMutations

    Less cytotoxic vectors, prolonged transgene expression

    Tumor targeting

    Targeting APCs for improved immunogenicity

    AntigenstaRNAUse of minimal vectors, easier handling, and readiness
    Oncolytic virusesTumor targeting and killing
    Pseudoviruses

    Introduction of tumor targeting epitopes

    Introduction of immunogenic epitopes

    Protein and epitope optimization

    Improved immune responses

    Combination of epitopes for a larger spectrum of immunogenicity

    Epitope selection

    Identification of tumor-specific epitopes on cancer cells

    More potent personalized immune responses

    RNA deliveryStabilizationImproved resistant to degradation, longer RNA half-life
    EncapsulationImproved stability, delivery, prolonged circulation in vivo
    Combination therapyPrime-boostPrime-boost vaccinations with Ad and other viral vectors
    ICB, other drugsSuperior to self-replicating vector alone
    Process optimizationVaccine doseOptimization of applied dose
    RouteEvaluation of administration route
    Display full size

    APCs: antibody-producing cells; taRNA: trans-amplifying RNA

    In the context of antigen targets, the choice of regions or epitopes generating the best possible immune responses is important. In the case of infectious viruses, surface proteins easily recognized by the host cell immune system and/or structures involved in viral replication are desirable. For example, the replacement of the VSV-G protein with target surface proteins such as the SARS-CoV-2 S protein [82]. As epitopes have proven good as antigens, the combination of various epitopes can provide a larger spectrum against infectious viruses or their variants providing enhanced vaccine efficacy. Related to cancer vaccines, desirable antigen targets are those that are commonly expressed or uniquely overexpressed in cancer cells. More recently, personalized antigen approaches have become feasible, where neoepitopes derived from autologous tumors can potentially elicit more potent and personalized immune responses [100].

    The delivery of viral vectors has been an area of optimization too. The introduction of targeting sequences by generating pseudoviruses or using strains or mutants capable of selectively targeting antigen-presenting cells (APCs) can improve and target delivery. Related to delivery, the route of administration and dose optimization are key factors for the improvement and optimization of immune responses and vaccine efficacy.

    In the case of using RNA replicons instead of recombinant viral particles, prevention of ssRNA degradation has been addressed by stabilization of RNA by chemical modifications [35] and encapsulation of RNA replicons in nanoparticles to protect against degradation, enhance delivery, and prolong circulation of RNA in vivo [36].

    Combination therapy of self-replicating RNA viruses has also been attractive. In this context, prime-boost vaccinations with other viral-based vaccines [100], other antiviral and anti-cancer drugs [105], and ICB drugs [88] have been tested.

    Conclusions

    In summary, self-replicating RNA viruses have been evaluated in preclinical animal models and clinical trials for vaccine development against infectious diseases and various cancers. Both viruses with ssRNA genomes of positive and negative polarity have been used. Proof-of-concept for vaccine candidates against both infectious diseases and cancers has been established in animal models. However, although good safety and tolerability profiles have been seen in clinical trials, similar levels of vaccine efficacy as seen in animal models have been difficult to reproduce in humans. Despite many years of experience, recent developments such as the approval of the VSV-based vaccine against EDV have encouraged further vaccine development efforts. Moreover, the recent success achieved for SARS-CoV-2 vaccines, not the least mRNA-based vaccines, has presented excellent opportunities for the development of novel vaccines based on self-replicating RNA viruses.

    Abbreviations

    26S:

    Semliki Forest virus subgenomic promoter

    Ab:

    antibody

    Ad:

    adenovirus

    CEA:

    carcinoembryonic antigen

    ChAd:

    chimpanzee adenovirus

    CHIKV:

    chikungunya virus

    CMV:

    cytomegalovirus

    COVID-19:

    coronavirus disease 2019

    E6/E7:

    human papillomavirus envelope proteins 6 and 7

    EBOV-GP:

    Ebola virus-glycoprotein

    EVD:

    Ebola virus disease

    FDA:

    US Food and Drug Administration

    G:

    glycoprotein

    GoI:

    gene of interest

    HA:

    hemagglutinin

    HER-2:

    human epidermal growth factor receptor 2

    HPV:

    human papillomavirus

    HSV:

    herpes simplex virus

    ICB:

    immune checkpoint blockade

    KUN:

    Kunjin virus

    L:

    large protein

    LNP:

    lipid nanoparticle

    LNP-nCoVsaRNA:

    lipid nanoparticle-Venezuelan equine encephalitis virus-severe acute respiratory syndrome-coronavirus-2 spike RNA

    M:

    matrix protein

    MV:

    measles virus

    N:

    nucleoprotein

    neu:

    neuroblastoma

    NIS:

    sodium iodide symporter

    nsP1–4:

    non-structural protein 1–4

    P:

    phosphoprotein

    PR:

    partial response

    PSMA:

    prostate-specific membrane antigen

    RABV:

    rabies virus

    RC:

    replicase complex

    RROC:

    refractory recurrent ovarian cancer

    SARS-CoV-2 S:

    severe acute respiratory syndrome-coronavirus-2 spike

    SARS-CoV-2:

    severe acute respiratory syndrome-coronavirus-2

    SD:

    stable disease

    SFV:

    Semliki Forest virus

    SP6:

    SP6 RNA polymerase promoter

    ssRNA:

    single-stranded RNA

    T7:

    T7 RNA polymerase promoter

    VEE:

    Venezuelan equine encephalitis virus

    VSV:

    vesicular stomatitis virus

    ZEBOV:

    Ebola virus-Zaire strain

    ZIKV:

    Zika virus

    Declarations

    Author contributions

    KL is the sole author of the manuscript and contributed to: Conceptualization, Writing—original draft, Writing—review & editing.

    Conflicts of interest

    The author declares that he has no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Delrue I, Verzele D, Madder A, Nauwynck HJ. Inactivated virus vaccines from chemistry to prophylaxis: merits, risks and challenges. Expert Rev Vaccines. 2012;11:695719. [DOI] [PubMed]
    Chen WH, Du L, Chag SM, Ma C, Tricoche N, Tao X, et al. Yeast-expressed recombinant protein of the receptor-binding domain in SARS-CoV spike protein with deglycosylated forms as a SARS vaccine candidate. Hum Vaccin Immunother. 2014;10:64858. [DOI] [PubMed] [PMC]
    Schott JW, Galla M, Godinho T, Baum C, Schambach A. Viral and non-viral approaches for transient delivery of mRNA and proteins. Curr Gene Ther. 2011;11:38298. [DOI] [PubMed]
    Bok K, Sitar S, Graham BS, Mascola JR. Accelerated COVID-19 vaccine development: milestones, lessons, and prospects. Immunity. 2021;54:163651. [DOI] [PubMed] [PMC]
    Sasso E, D’Alise AM, Zambrano N, Scarselli E, Folgori A, Nicosia A. New viral vectors for infectious diseases and cancer. Semin Immunol. 2020;50:101430. [DOI] [PubMed]
    Liu CC, Yang H, Zhang R, Zhao JJ, Hao DJ. Tumour-associated antigens and their anti-cancer applications. Eur J Cancer Care (Engl). 2017;26:e12446. [DOI] [PubMed]
    Zajakina A, Spunde K, Lundstrom K. Application of alphaviral vectors for immunomodulation in cancer therapy. Curr Pharm Des. 2017;23:490632. [DOI] [PubMed]
    Zarogoulidis P, Darwiche K, Sakkas A, Yarmus L, Huang H, Li Q, et al. Suicide gene therapy for cancer – current strategies. J Genet Syndr Gene Ther. 2013;4:16849. [DOI] [PubMed] [PMC]
    Lundstrom K. Viral vectors in gene therapy: Where do we stand in 2023? Viruses. 2023;15:698. [DOI] [PubMed] [PMC]
    Ramasamy MN, Minassian AM, Ewer KJ, Flaxman AL, Folegatti PM, Owens DR, et al.; Oxford COVID Vaccine Trial Group. Safety and immunogenicity of ChAdOx1 nCoV-19 vaccine administered in a prime-boost regimen in young and old adults (COV002): a single-blind, randomised, controlled, phase 2/3 trial. Lancet. 2021;396:197993. [DOI] [PubMed] [PMC]
    Stephenson KE, Le Gars M, Sadoff J, de Groot AM, Heerwegh D, Truyers C, et al. Immunogenicity of the Ad26.COV2.S vaccine for COVID-19. JAMA. 2021;325:153544. [DOI] [PubMed] [PMC]
    Zhu FC, Guan XH, Li YH, Huang JY, Jiang T, Hou LH, et al. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2020;396:47988. [DOI] [PubMed] [PMC]
    Lamb YN. BNT162b2 mRNA COVID-19 vaccine: first approval. Drugs. 2021;81:495501. [DOI] [PubMed] [PMC]
    Vasireddy D, Atluri P, Malayala SV, Vanaparthy R, Mohan G. Review of COVID-19 vaccines approved in the United States of America for emergency use. J Clin Med Res. 2021;13:20413. [DOI] [PubMed] [PMC]
    Lundstrom K. Viral vectors for COVID-19 vaccine development. Viruses. 2021;13:317. [DOI] [PubMed] [PMC]
    Nagasato M, Rin Y, Yamamoto Y, Henmi M, Hiraoka N, Chiwaki F, et al. A tumor-targeting adenovirus with high gene-transduction efficiency for primary pancreatic cancer and ascites cells. Anticancer Res. 2017;37:3599605. [DOI] [PubMed]
    Eissa IR, Naoe Y, Bustos-Villalobos I, Ichinose T, Tanaka M, Zhiwen W, et al. Genomic signature of the natural oncolytic herpes simplex virus HF10 and its therapeutic role in preclinical and clinical trials. Front Oncol. 2017;7:149. [DOI] [PubMed] [PMC]
    Hirooka Y, Kasuya H, Ishikawa T, Kawashima H, Ohno E, Villalobos IB, et al. A phase I clinical trial of EUS-guided intratumoral injection of the oncolytic virus, HF10 for unresectable locally advanced pancreatic cancer. BMC Cancer. 2018;18:596. [DOI] [PubMed] [PMC]
    Cloughesy TF, Landolfi J, Hogan DJ, Bloomfield S, Carter B, Chen CC, et al. Phase 1 trial of vocimagene amiretrorepvec and 5-fluorocytosine for recurrent high-grade glioma. Sci Transl Med. 2016;8:341ra75. [DOI] [PubMed] [PMC]
    Tocagen reports results of Toca 5 phase 3 trial in recurrent brain cancer [Internet]. San Diego (CA): Tocagen; c2019 [cited 2023 Mar 1]. Available from: https://bit.ly/2lPm19v
    Johnson DB, Puzanov I, Kelley MC. Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma. Immunotherapy. 2015;7:6119. [DOI] [PubMed] [PMC]
    Rehman H, Silk AW, Kane MP, Kaufman HL. Into the clinic: talimogene laherparepvec (T-VEC), a first-in-class intratumoral oncolytic viral therapy. J Immunother Cancer. 2016;4:53. [DOI] [PubMed] [PMC]
    Lundstrom K. Self-replicating RNA viruses for vaccine development against infectious diseases and cancer. Vaccines (Basel). 2021;9:1187. [DOI] [PubMed] [PMC]
    Lundstrom K. Applications of self-replicating RNA. In: Aranda F, Berraondo P, Galluzzi L, editors. International review of cell and molecular biology. Cambridge: Academic Press; 2022. pp. 97–157. [DOI] [PubMed]
    Strauss JH, Strauss EG. The alphaviruses: gene expression, replication, and evolution. Microbiol Rev. 1994;58:491562. [DOI] [PubMed] [PMC]
    Pugachev KV, Guirakhoo F, Trent DW, Monath TP. Traditional and novel approaches to flavivirus vaccines. Int J Parasitol. 2003;33:56782. [DOI] [PubMed]
    Gotoh B, Komatsu T, Takeuchi K, Yokoo J. Paramyxovirus strategies for evading the interferon response. Rev Med Virol. 2002;12:33757. [DOI] [PubMed]
    Lyles DS, Rupprecht CE. Rhabdoviridae. In: Knipe DM, Howley PM, editors. Fields Virology, 5th ed. Philadelphia: Wolters Kluwer Health/Lippincott Williams & Wilkins; 2007.
    Frolov I, Hoffman TA, Pragal BM, Dryga SA, Huang HV, Schlesinger S, et al. Alphavirus-based expression vectors: strategies and applications. Proc Natl Acad Sci U S A. 1996;93:113717. [DOI] [PubMed] [PMC]
    Liljestöm P, Garoff H. A new generation of animal cell expression vectors based on the Semliki Forest virus replicon. Nat Biotechnol. 1991;9:135661. [DOI] [PubMed]
    Xiong C, Levis R, Shen P, Schlesinger S, Rice CM, Huang HV. Sindbis virus: an efficient, broad host range vector for gene expression in animal cells. Science. 1989;243:118891. [DOI] [PubMed]
    Davis NL, Willis LV, Smith JF, Johnston RF. In vitro synthesis of infectious venezuelan equine encephalitis virus RNA from a cDNA clone: analysis of a viable deletion mutant. Virology. 1989;171:189204. [DOI] [PubMed]
    White CL, Thomson M, Dimmock NJ. Deletion analysis of a defective interfering Semliki Forest virus RNA genome defines a region in the nsP2 sequence that is required for efficient packaging of the genome into virus particles. J Virol. 1998;72:43206. [DOI] [PubMed] [PMC]
    Kim DY, Firth AE, Atasheva S, Frolova EI, Frolov I. Conservation of a packaging signal and the viral genome RNA packaging mechanism in alphavirus evolution. J Virol. 2011;85:802236. [DOI] [PubMed] [PMC]
    Wadhwa A, Aljabbari A, Lokras A, Foged C, Thakur A. Opportunities and challenges in the delivery of mRNA-based vaccines. Pharmaceutics. 2020;12:102. [DOI] [PubMed] [PMC]
    Raouane M, Desmaële D, Urbinati G, Massaad-Massade L, Couvreur P. Lipid conjugated oligonucleotides: a useful strategy for delivery. Bioconjug Chem. 2012;23:1091104. [DOI] [PubMed]
    DiCiommo DP, Bremner R. Rapid, high level protein production using DNA-based Semliki Forest virus vectors. J Biol Chem. 1998;273:180606. [DOI] [PubMed]
    Bai H, Lester GMS, Petishnok LC, Dean DA. Cytoplasmic transport and nuclear import of plasmid DNA. Biosci Rep. 2017;37:BSR20160616. [DOI] [PubMed] [PMC]
    Muramatsu T, Nakamura A, Park HM. In vivo electroporation: a powerful and convenient means of nonviral gene transfer to tissues of living animals (review). Int J Mol Med. 1998;1:55117. [DOI] [PubMed]
    Mahvi DM, Sheehy MJ, Yang NS. DNA cancer vaccines: a gene gun approach. Immunol Cell Biol. 1997;75:45660. [DOI] [PubMed]
    Lundstrom K, Boulikas T. Viral and non-viral vectors in gene therapy: technology development and clinical trials. Technol Cancer Res Treat. 2003;2:47185. [DOI] [PubMed]
    Sunshine JC, Bishop CJ, Green JJ. Advances in polymeric and inorganic vectors for nonviral nucleic acid delivery. Ther Deliv. 2011;2:493521. [DOI] [PubMed] [PMC]
    Xu Y, Liang W, Qiu Y, Cespi M, Palmieri GF, Mason AJ, et al. Incorporation of a nuclear localization signal in pH responsive LAH4-L1 peptide enhances transfection and nuclear uptake of plasmid DNA. Mol Pharm. 2016;13:314152. [DOI] [PubMed]
    Pijlman GP, Suhrbier A, Khromykh AA. Kunjin virus replicons: an RNA-based, non-cytopathic viral vector system for protein production, vaccine and gene therapy applications. Expert Opin Biol Ther. 2006;6:13545. [DOI] [PubMed]
    de Felipe P. Skipping the co-expression problem: the new 2A “CHYSEL” technology. Genet Vaccines Ther. 2004;2:13. [DOI] [PubMed] [PMC]
    Khromykh AA, Varnavski AN, Westaway EG. Encapsidation of the flavivirus Kunjin replicon RNA by using a complementation system providing Kunjin virus structural proteins in trans. J Virol. 1998;72:596777. [DOI] [PubMed] [PMC]
    Shi PY, Tilgner M, Lo MK. Construction and characterization of subgenomic replicons of New York strain of West Nile virus. Virology. 2002;296:21933. [DOI] [PubMed]
    Molenkamp R, Kooi EA, Lucassen MA, Greve S, Thijssen JC, Spaan WJ, et al. Yellow fever virus replicons as an expression system for hepatitis C virus structural proteins. J Virol. 2003;77:16448. [DOI] [PubMed] [PMC]
    Jones M, Davidson A, Hibbert L, Gruenwald P, Schlaak J, Ball S, et al. Dengue virus inhibits alpha interferon signaling by reducing STAT2 expression. J Virol. 2005;79:541420. [DOI] [PubMed] [PMC]
    Gehrke R, Ecker M, Aberle SW, Allison SL, Heinz FX, Mandl CW. Incorporation of tick-borne encephalitis virus replicons into virus-like particles by a packaging cell line. J Virol. 2003;77:892433. [DOI] [PubMed] [PMC]
    Kuadkitkan A, Ramphan S, Worawichawong S, Sornjai W, Wikan N, Smith DR. Production of Zika virus virus-like particles. In: Pfeifer BA, Hill A, editors. Vaccine delivery technology. New York: Humana; 2021. pp. 183–203. [DOI] [PubMed]
    Fan ZC, Dennis JC, Bird RC. Bovine viral diarrhea virus is a suitable viral vector for stable expression of heterologous gene when inserted in between Npro and C genes. Virus Res. 2008;138:97104. [DOI] [PubMed]
    Stettler P, Devos R, Moser C, Tratschin JD, Hofmann MA. Establishment and application of bicistronic classical swine fever virus genomes for foreign gene expression and complementation of E2 deletion mutants. Virus Res. 2002;85:17385. [DOI] [PubMed]
    Schnell MJ, Buonocore L, Kretzschmar E, Johnson E, Rose JK. Foreign glycoproteins expressed from recombinant vesicular stomatitis viruses are incorporated efficiently into virus particles. Proc Natl Acad Sci U S A. 1996;93:1135965. [DOI] [PubMed] [PMC]
    Tani H, Morikawa S, Matsuura Y. Development and applications of VSV vectors based on cell tropism. Front Microbiol. 2012;2:272. [DOI] [PubMed] [PMC]
    Pfaller CK, Cattaneo R, Schnell MJ. Reverse genetics of Mononegavirales: How they work, new vaccines, and new cancer therapeutics. Virology. 2015;479–80:33144. [DOI] [PubMed] [PMC]
    Harty RN, Brown ME, Hayes FP, Wright NT, Schnell MJ. Vaccinia virus-free recovery of vesicular stomatitis virus. J Mol Microbiol Biotechnol. 2001;3:5137. [PubMed]
    Hong SK, Jung YT, Park SW, Paik SY. Inducible vesicular stomatitis virus (VSV) L cell line for packaging of recombinant VSV. Virus Genes. 2005;31:195201. [DOI] [PubMed]
    Osakada F, Callaway EM. Design and generation of recombinant rabies virus vectors. Nat Protoc. 2013;8:1583601. [DOI] [PubMed] [PMC]
    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;47:6137. [DOI] [PubMed]
    Singh M, Cattaneo R, Billeter MA. A recombinant measles virus expressing hepatitis B virus surface antigen induces humoral immune responses in genetically modified mice. J Virol. 1999;73:48238. [DOI] [PubMed] [PMC]
    Radecke F, Spielhofer P, Schneider H, Kaelin K, Huber M, Dötsch C, et al. Rescue of measles viruses from cloned DNA. EMBO J. 1995;14:577384. [DOI] [PubMed] [PMC]
    Rossi SL, Comer JE, Wang E, Azar SR, Lawrence WS, Plante JA, et al. Immunogenicity and efficacy of a measles virus-vectored chikungunya vaccine in nonhuman primates. J Infect Dis. 2019;220:73542. [DOI] [PubMed] [PMC]
    Ramsauer K, Schwameis M, Firbas C, Müllner M, Putnak RJ, Thomas SJ, et al. Immunogenicity, safety, and tolerability of a recombinant measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. Lancet Infect Dis. 2015;15:51927. [DOI] [PubMed]
    Reisinger EC, Tschismarov R, Beubler E, Wiedermann U, Firbas C, Loebermann M, et al. Immunogenicity, safety, and tolerability of the measles-vectored chikungunya virus vaccine MV-CHIK: a double-blind, randomised, placebo-controlled and active-controlled phase 2 trial. Lancet. 2018;392:271827. [DOI] [PubMed]
    Marzi A, Robertson SJ, Haddock E, Feldmann F, Hanley PW, Scott DP, et al. VSV-EBOV rapidly protects macaques against infection with the 2014/15 Ebola virus outbreak strain. Science. 2015;349:73942. [DOI] [PubMed]
    Geisbert TW, Feldmann H. Recombinant vesicular stomatitis virus-based vaccines against Ebola and Marburg virus infections. J Infect Dis. 2011;204:S107581. [DOI] [PubMed] [PMC]
    Clarke DK, Xu R, Matassov D, Latham TE, Ota-Setlik A, Gerardi CS, et al. Safety and immunogenicity of a highly attenuated rVSVN4CT1-EBOVGP1 Ebola virus vaccine: a randomised, double-blind, placebo-controlled, phase 1 clinical trial. Lancet Infect Dis. 2020;20:45566. [DOI] [PubMed]
    Henao-Restrepo AM, Longini IM, Egger M, Dean NE, Edmunds WJ, Camacho A, et al. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: interim results from the Guinea ring vaccination cluster-randomised trial. Lancet. 2015;386:85766. [DOI] [PubMed]
    Henao‐Restrepo AM, Camacho A, Longini IM, Watson CH, Edmunds WJ, Egger M, et al. Efficacy and effectiveness of an rVSV‐vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open‐label, cluster‐randomised trial (Ebola Ça Suffit!). Lancet. 2017;389:50518. [DOI] [PubMed] [PMC]
    Ollmann Saphire E. A vaccine against Ebola virus. Cell. 2020;181:6. [DOI] [PubMed]
    Nürnberger C, Bodmer BS, Fiedler AH, Gabriel G, Mühlebach MD. A measles virus-based vaccine candidate mediates protection against Zika virus in an allogeneic mouse pregnancy model. J Virol. 2019;93:e0148518. [DOI] [PubMed] [PMC]
    Zika-vaccine dose finding study regarding safety, immunogenicity and tolerability (V186-001) [Internet]. Bethesda (MD): U.S. National Library of Medicine; [cited 2023 Mar 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT02996890
    Kurup D, Wirblich C, Lambert R, Diba LZ, Leiby BE, Schnell MJ. Measles-based Zika vaccine induces long-term immunity and requires NS1 antibodies to protect the female reproductive tract. NPJ Vaccines. 2022;7:43. [DOI] [PubMed] [PMC]
    Safety and immunogenicity of a novel vaccine formulation MV-ZIKA-RSP (V187-001) (MV-ZIKA-RSP) [Internet]. Bethesda (MD): U.S. National Library of Medicine; [cited 2023 Mar 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT04033068
    Hörner C, Schürmann C, Auste A, Ebenig A, Muraleedharan S, Dinnon KH III, et al. A highly immunogenic and effective measles virus-based Th1-biased COVID-19 vaccine. Proc Natl Acad Sci U S A. 2020;117:3265766. [DOI] [PubMed] [PMC]
    Clinical trial to evaluate the safety and immunogenicity of the COVID-19 vaccine (COVID-19-101) [Internet]. Bethesda (MD): U.S. National Library of Medicine; [cited 2023 Mar 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT04497298
    Merck discontinues development of SARS-CoV-2/COVID-19 vaccine candidates; continues development of two investigational therapeutic candidates [Internet]. Rahway (NJ): Merck & Co.; c2023 [cited 2023 Mar 8]. Available from: https://www.merck.com/news/merck-discontinues-development-of-sars-cov-2-covid-19-vaccine-candidates-continues-development-of-two-investigational-therapeutic-candidates/
    Case JB, Rothlauf PW, Chen RE, Kafai NM, Fox JM, Smith BK, et al. Replication-competent vesicular stomatitis virus vaccine vector protects against SARS-CoV-2-mediated pathogenesis in mice. Cell Host Microbe. 2020;28:465–74, e1–4. [DOI] [PubMed] [PMC]
    Dose ranging trial to assess safety and immunogenicity of V590 (COVID-19 vaccine) in healthy adults (V590-001) [Internet]. Bethesda (MD): U.S. National Library of Medicine; [cited 2023 Mar 8]. Available from: https://clinicaltrials.gov/ct2/show/NCT04569786
    Merck and IAVI discontinue development of COVID-19 vaccine candidate V590 [Internet]. New York (NY): International AIDS Vaccine Initiative; c2023 [cited 2023 Mar 8]. Available from: https://www.iavi.org/news-resources/press-releases/2021/merck-and-iavi-discontinue-development-of-covid-19-vaccine-candidate-v590
    Yahalom-Ronen Y, Tamir H, Melamed S, Politi B, Shifman O, Achdout H, et al. A single dose of recombinant VSV-∆G-spike vaccine provides protection against SARS-CoV-2 challenge. Nat Commun. 2020;11:6402. [DOI] [PubMed] [PMC]
    Evaluate the safety, immunogenicity and potential efficacy of an rVSV-SARS-CoV-2-S vaccine [Internet]. Bethesda (MD): U.S. National Library of Medicine; [cited 2023 Mar 9]. Available from: https://clinicaltrials.gov/ct2/show/NCT04608305
    Ly HH, Daniel S, Soriano SKV, Kis Z, Blakney AK. Optimization of lipid nanoparticles for saRNA expression and cellular activation using a design-of-experiment approach. Mol Pharm. 2022;19:1892905. [DOI] [PubMed] [PMC]
    Tregoning JS, Stirling DC, Wang Z, Flight KE, Brown JC, Blakney AK, et al. Formulation, inflammation, and RNA sensing impact the immunogenicity of self-amplifying RNA vaccines. Mol Ther Nucleic Acids. 2023;31:2942. [DOI] [PubMed] [PMC]
    McKay PF, Hu K, Blakney AK, Samnuan K, Brown JC, Penn R, et al. Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibodies in mice. Nat Commun. 2020;11:3523. [DOI] [PubMed] [PMC]
    Pollock KM, Cheeseman HM, Szubert AJ, Libri V, Boffito M, Owen D, et al.; COVAC1 study Group. Safety and immunogenicity of a self-amplifying RNA vaccine against COVID-19: COVAC1, a phase I, dose-ranging trial. EClinicalMedicine. 2022;44:101262. [DOI] [PubMed] [PMC]
    Morse MA, Crosby EJ, Force J, Osada T, Hobeika AC, Hartman ZC, et al. Clinical trials of self-replicating RNA-based cancer vaccines. Cancer Geen Ther. 2023;30:80311. [DOI] [PubMed] [PMC]
    Wang X, Wang JP, Maughan MF, Lachman LB. Alphavirus replicon particles containing the gene for HER2/neu inhibit breast cancer growth and tumorigenesis. Breast Cancer Res. 2004;7:R145. [DOI] [PubMed] [PMC]
    Crosby EJ, Gwin W, Blackwell K, Marcom PK, Chang S, Maecker HT, et al. Vaccine-induced memory CD8+ T cells provide clinical benefit in HER2 expressing breast cancer: a mouse to human translational study. Clin Cancer Res. 2019;25:272536. [DOI] [PubMed] [PMC]
    Daemen T, Riezebos-Brilman A, Bungener L, Regts J, Dontje B, Wilschut J. Eradication of established HPV16-transformed tumours after immunisation with recombinant Semliki Forest virus expressing a fusion protein of E6 and E7. Vaccine. 2003;21:10828. [DOI] [PubMed]
    Komdeur FL, Singh A, van de Wall S, Meulenberg JJM, Boerma A, Hoogeboom BN, et al. First-in-human phase I clinical trial of an SFV-based RNA replicon cancer vaccine against HPV-induced cancers. Mol Ther. 2021;29:61125. [DOI] [PubMed] [PMC]
    Osada T, Berglund P, Morse MA, Hubby B, Lewis W, Niedzwiecki D, et al. Co-delivery of antigen and IL-12 by Venezuelan equine encephalitis virus replicon particles enhances antigen-specific immune responses and antitumor effects. Cancer Immunol Immunother. 2012;61:194151. [DOI] [PubMed] [PMC]
    Crosby EJ, Hobeika AC, Niedzwiecki D, Rushing C, Hsu D, Berglund P, et al. Long-term survival of patients with stage III colon cancer treated with VRP-CEA(6D), an alphavirus vector that increases the CD8+ effector memory T cell to Treg ratio. J Immunother Cancer. 2020;8:e001662. [DOI] [PubMed] [PMC]
    Hasegawa K, Pham L, O’Connor MK, Federspiel MJ, Russell SJ, Peng KW. Dual therapy of ovarian cancer using measles viruses expressing carcinoembryonic antigen and sodium iodide symporter. Clin Cancer Res. 2006;12:186875. [DOI] [PubMed]
    Galanis E, Hartmann LC, Cliby WA, Long HJ, Peethambaram PP, Barrette BA, et al. Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer Res. 2010;70:87582. [DOI] [PubMed] [PMC]
    Durso RJ, Andjelic S, Gardner JP, Margitich DJ, Donovan GP, Arrigale RR, et al. A novel alphavirus vaccine encoding prostate-specific membrane antigen elicits potent cellular and humoral immune responses. Clin Cancer Res. 2007;13:39994008. [DOI] [PubMed]
    Slovin SF, Kehoe M, Durso R, Fernandez C, Olson W, Gao JP, et al. A phase I dose escalation trial of vaccine replicon particles (VRP) expressing prostate-specific membrane antigen (PSMA) in subjects with prostate cancer. Vaccine. 2013;31:9439. [DOI] [PubMed]
    Morse MA, Hobeika AC, Osada T, Berglund P, Hubby B, Negri S, 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 Invest. 2010;120:323441. [DOI] [PubMed] [PMC]
    Catenacci DV, Liao C, Maron S, Solomon BJ, Mahipal A, Johnson ML, et al. 960MO clinical outcomes and immune responses in a phase I/II study of personalized, neoantigen-directed immunotherapy in patients with advanced MSS-CRC, GEA and NSCLC. Ann Oncol. 2021;32:S83031. [DOI]
    Agapov EV, Frolov I, Lindenbach BD, Prágai BM, Schlesinger S, Rice CM. Noncytopathic Sindbis virus RNA vectors for heterologous gene expression. Proc Natl Acad Sci U S A. 1998;95:1298994. [DOI] [PubMed] [PMC]
    Lundstrom K, Abenavoli A, Malgaroli A, Ehrengruber MU. Novel Semliki Forest virus vectors with reduced cytotoxicity and temperature sensitivity for long-term enhancement of transgene expression. Mol Ther. 2003;7:2029. [DOI] [PubMed]
    Lundstrom K. Therapeutic applications for oncolytic self-replicating RNA viruses. Int J Mol Sci. 2022;23:15622. [DOI] [PubMed] [PMC]
    Perkovic M, Gawletta S, Hempel T, Brill S, Nett E, Sahin U, et al. A trans-amplifying RNA simplified to essential elements is highly replicative and robustly immunogenic in mice. Mol Ther. 2023;31:163646. [DOI] [PubMed] [PMC]
    Dold C, Rodriguez Urbiola C, Wollmann G, Egerer L, Muik A, Bellmann L, et al. Application of interferon modulators to overcome partial resistance of human ovarian cancers to VSV-GP oncolytic viral therapy. Mol Ther Oncolytics. 2016;3:16021. [DOI] [PubMed] [PMC]