Abstract
Introduction:
Viral RNA replicons are self-amplifying RNA molecules generated by deleting genetic information of one or multiple structural proteins of wild-type viruses. Remaining viral RNA is used as such (naked replicon) or packaged into a viral replicon particle (VRP), whereby missing genes or proteins are supplied via production cells. Since replicons mostly originate from pathogenic wild-type viruses, careful risk consideration is crucial.
Methods:
A literature review was performed compiling information on potential biosafety risks of replicons originating from positive- and negative-sense single-stranded RNA viruses (except retroviruses).
Results:
For naked replicons, risk considerations included genome integration, persistence in host cells, generation of virus-like vesicles, and off-target effects. For VRP, the main risk consideration was formation of primary replication competent virus (RCV) as a result of recombination or complementation. To limit the risks, mostly measures aiming at reducing the likelihood of RCV formation have been described. Also, modifying viral proteins in such a way that they do not exhibit hazardous characteristics in the unlikely event of RCV formation has been reported.
Discussion and Conclusion:
Despite multiple approaches developed to reduce the likelihood of RCV formation, scientific uncertainty remains on the actual contribution of the measures and on limitations to test their effectiveness. In contrast, even though effectiveness of each individual measure is unclear, using multiple measures on different aspects of the system may create a solid barrier. Risk considerations identified in the current study can also be used to support risk group assignment of replicon constructs based on a purely synthetic design.
Keywords: viral replicon, biosafety, replication competent virus, RNA virus, self-amplifying
Introduction
Viral RNA replicons are defined as self-amplifying RNA molecules of viral origin and originate from single-stranded positive- or negative-sense RNA viruses. They are a form of viral vectors and are generated by deleting the genetic information of one or multiple structural proteins of a wild-type virus. The remaining viral RNA can be used as such and brought into the cells as a naked replicon by transfection or electroporation, or via synthetic nanoparticles. A schematic representation of a wild-type infection by an enveloped positive-sense RNA virus compared with the transfection of cells with a naked replicon is shown in Figure 1A and B, respectively.
Figure 1.
Schematic representation of a wild-type infection with an enveloped positive-sense RNA virus (A), compared with the transfection of cells with a viral replicon based on a positive-sense RNA virus (B). The figure does not aim at a complete representation of the gene expression strategies of all positive-sense RNA viruses and, for reader clarity, only shows a representative example. (A) Infection of a cell with a wild-type virus: (1) Attachment, entry, and uncoating of the virus. (2) Translation of vRNA (∼mRNA) into nonstructural proteins. (3) Genome replication and synthesis of additional viral RNAs coding for structural and nonstructural proteins. (4) Translation of RNA into structural proteins. (5) Assembly of viral proteins and RNA into a new viral particle. (6) Egress of the infectious virus particle. (B) Transfection of a cell with a naked replicon: (1) Transfection of RNA. (2) Translation of vRNA (∼mRNA) into nonstructural proteins. (3) Genome replication and synthesis of additional viral RNAs coding for nonstructural proteins. mRNA, messenger RNA; NS, nonstructural; S, structural; vRNA, viral RNA.
Alternatively, a viral replicon particle (VRP) can be generated by simultaneously providing (RNA coding for) the missing protein components to the cell, after which particles are formed which are able to infect a cell but are unable to subsequently form infectious virus particles due to the absence of the coding sequences for (part of) the structural proteins. Representative examples of the production of VRPs based on an enveloped positive-sense RNA virus using cells (stably) expressing structural proteins or using nonreplicating helper RNA coding for structural proteins are presented in Figure 2A and B, respectively. Finally, copy or complementary DNA (cDNA) can be delivered to the cells instead of viral RNA wherein it is converted into viral RNA. The latter approach is referred to as DNA-launched replicons (DREPs).
Figure 2.
Schematic example of the production of VRPs using cells (stably) expressing structural proteins (A), compared with the production of VRPs by means of transfecting cells with nonreplicating helper RNA coding for structural proteins (B). The figure does not aim at a complete representation of all VRP production systems and, for reader clarity, only shows two representative examples. VRP, viral replicon particle.
Due to their self-amplifying nature, RNA replicons have gained much attention in research and development as well as clinical applications. By removing genes coding for structural proteins and replacing them with transgenes of interest, they form ideal vectors to deliver genes to cells of interest or to help present specific proteins to the immune system. Since genome replication and transcription will occur in the replicon-containing cell, higher expression levels of the transgenes can be obtained in comparison with replication-defective vectors.
To work safely with viral RNA replicons, specific safety measures may be needed depending on local legislation, such as the legislation related to genetically modified organisms. To determine suitable safety measures, a thorough risk assessment is required. So far, limited information is available on the factors to consider for such risk assessment. While, on the one hand, the risk group of the parental virus can be considered, multiple genetic modifications, on the other hand, may warrant downscaling of the risk. The aims of the current review article were to provide an overview of replicon systems that are currently available or in development, to summarize their biosafety characteristics, and to recommend on an approach for future assessments and potential downscaling.
This review focuses on the application of, as well as the biosafety considerations for replicons originating from positive- and negative-sense single-stranded RNA viruses. Application of double-stranded RNA viruses, which so far has only been explored sporadically, as well as of single-stranded RNA viruses that use reverse transcriptase (i.e., retroviruses) remains outside the scope.
Overview of Single-Stranded RNA Viruses as Backbone for RNA Replicons
Single-stranded positive- and negative-sense RNA viruses have been associated with multiple diseases in vertebrates* and are classified in various risk groups. Table 1 provides a more detailed overview of the different viral families and species from which replicons have been generated. Risk classification mentioned in the table is based on the assessments of multiple international databases including the Netherlands Commission on Genetic Modification (COGEM), the Belgian Service of Biosafety and Biotechnology, the Central Committee on Biological Safety, Germany, and the Federal Office for the Environment, Switzerland (FOEN).
Table 1.
Overview of host range, disease, and risk group classification of viruses (wild type and, if relevant, vaccine strain) from which replicons have been derived (nonlimitative list)
| Classification | Family | Virus | Host range |
Disease
|
Risk group | |
|---|---|---|---|---|---|---|
| Human | Animal | |||||
| Positive-sense nonsegmented | Astroviridae | Human astrovirus | Humans |
|
— | 2a |
| Caliciviridae | Norovirus (Norwalk virus) | Humans; rodents; felines; canines; sea lions; pigs; sheep; cattle; bats |
|
— | 2 | |
| Coronaviridae | Middle East respiratory syndrome coronavirus | Humans; dromedary; camel; bats |
|
|
3 | |
| Kunjin virus | Humans; bats; civets |
|
|
3 | ||
| Severe acute respiratory syndrome coronavirus 2 | Humans; bats; minks; cats |
|
|
3 | ||
| Flaviviridae | Bovine viral diarrhea virus | Even-toed ungulates | — |
|
2 | |
| Classical swine fever | Pigs | — |
|
4 | ||
| Dengue virus | Humans; mosquitoes |
|
— | 3 | ||
| Hepatitis C virus | Humans; nonhuman primates; mosquitoes |
|
— | 2 | ||
| Japanese encephalitis virus | Humans; mosquitoes and vertebrate hosts, primarily pigs and wading birds |
|
|
3 | ||
| Kunjin virus | Humans; mosquitoes; birds; horses |
|
|
2b | ||
| Niénokoué virus | Mosquitoes | — | — | 1c | ||
| Tick-borne encephalitis virus | Humans; ruminants; birds; rodents; horses; carnivores; ticks |
|
— | 3 | ||
| WNV | Humans; nonhuman primates; equines; dogs; cats; sheep; llamas; alpacas; alligators; birds; squirrel; chipmunk; rabbits |
|
|
3 | ||
| Yellow fever virus | Humans |
|
|
3 | ||
| Yellow fever virus—vaccine strain 17D | 2 | |||||
| Zika virus | Humans; monkeys; sheep; goats; horses; cows; ducks; rodents; bats; orangutans; carabaos; mosquitoes |
|
— | 3 | ||
| Hepeviridae | Hepatitis E virus | Humans; fish; amphibians; moose; kestrels |
|
— | 2 | |
| Picornaviridae | Mengovirus | Humans; rodents; pigs; monkeys |
|
— | 2a | |
| Poliovirus | Humans |
|
— | 2 | ||
| Foot-and-mouth disease virus | Cloven-hoofed animals (including bovids) | — |
|
4 | ||
| Enterovirus 71 | Humans |
|
— | 2 | ||
| Coxsackie virus | Humans |
|
— | 2 | ||
| Hepatitis A virus | Humans; nonhuman primates |
|
— | 2 | ||
| Human rhinovirus | Humans; primates |
|
— | 2 | ||
| Togaviridae | Salmon pancreas disease virus | Atlantic salmon (farmed salmonid fish) | - |
|
2 | |
| Chikungunya virus | Humans; nonhuman primates; rodents; birds; mosquitoes |
|
|
3 | ||
| SFV | Humans; mosquitoes; wild birds; rodents; domestic animals and nonhuman primates |
|
|
2 | ||
| SINV | Humans; mammals; birds; insects; amphibians |
|
|
2 | ||
| VEEV | Humans; horses; burros; bats; and terrestrial mammals |
|
|
3 | ||
| VEEV—vaccine strain TC83 | 2 | |||||
| Western equine encephalitis virus | Humans; reptiles; bats; pheasants; wild birds; mosquitoes; horses; dogs; rodents |
|
|
3 | ||
| Eastern equine encephalitis virus | Humans; reptiles; bats; pheasants; wild birds; mosquitoes; horses; dogs; rodents |
|
|
3 | ||
| Negative-sense nonsegmented | Bornaviridae | Borna disease virus | Horses; ruminants; dogs; squirrels; foxes |
|
|
2 H; 3 Aa |
| Filoviridae | Ebola virus | Humans; chimpanzees; gorillas, baboons; duikers; rodents; shrew; bats |
|
|
4 | |
| Marburg virus | Humans; nonhuman primates |
|
|
4 | ||
| Paramyxoviridae | Measles virus | Humans |
|
— | 2 | |
| Parainfluenza virus | Humans; birds |
|
|
2 | ||
| Nipah virus | Humans; pigs; bats |
|
|
4 | ||
| Human metapneumovirus | Humans |
|
— | 2 | ||
| Respiratory syncytial virus | Humans; cattle | — |
|
2 | ||
| Newcastle disease virus | Birds | — |
|
3 | ||
| Newcastle disease virus—vaccine strain LaSota | 2 | |||||
| Sendai virus | Rodents (mice) | — |
|
2 | ||
| Rhabdoviridae | VSV | Humans; horses; cattle; pigs; mules; sand flies; grasshoppers; rodents |
|
|
3 | |
| Negative-sense segmented | Arenaviridae | Lassa virus | Humans; Mastomys natalensis (multimammate rat) |
|
|
4 |
| Junin virus | Humans; rodents |
|
|
4 | ||
| Tacaribe virus | Humans; bats |
a
|
|
2 | ||
| Machupo virus | Rodents; humans; ticks; mosquitoes |
|
|
4 | ||
| Hantaviridae | Hantaan virus | Humans; rodents |
|
— | 3a | |
| Nairoviridae | Crimean-Congo hemorrhagic fever virus | Humans; cattle; sheep; goats; birds; ticks |
|
|
4 | |
| Phenuiviridae | Huaiyangshan banyangvirus (also known as “severe fever with thrombocytopenia virus”) | Humans; goats; sheep; cattle; dogs; chickens, rodents, pigs; ticks |
|
|
4 | |
| Rift Valley fever virus | Humans; ruminants; camelids; mosquitoes |
|
|
3 | ||
| Uukuniemi uukuvirus | Humans; ticks; blackbirds; mosquitoes |
a
|
|
2 | ||
The table only refers to vaccine strains for which a risk group classification has been published by international databases.
Based on classification published by the Belgian Service of Biosafety and Biotechnology (SBB).130
Based on classification published by the Federal Office for the Environment, Switzerland (FOEN).131
Based on classification published by the Central Committee on Biological Safety, Germany (ZKBS).132
SFV, Semliki Forest virus; SINV, Sindbis virus; VEEV, Venezuelan equine encephalitis virus; VSV, vesicular stomatitis virus; WNV, West Nile virus.
Literature Survey to Identify Applications and Risk Considerations of Viral RNA Replicons
A literature review was undertaken in view of a project issued by the COGEM. The aim of the study was to compile scientific information related to the developments and use of replicons, as well as their potential risks. To formulate search strings, the following keywords were selected: Replicon/Vector, Virus/Replication competent/RCV, Biosafety/Risk/Adverse effect. After selecting keywords, a typical search string was composed using Boolean operators: (VIR* OR “REPLICATI* COMPET*”) AND (REPLICON OR VECTOR) AND (BIOSAFE* OR RISK OR “ADVERSE EFFECT” OR SAFE* OR ENVIRONMENT*) AND (RNA).
Relevant scientific publications from peer reviewed journals were retrieved using the databases Scopus and Web of Science. No language or publication restrictions were applied, and studies were not selected based on quality. The literature search was concluded on January 25, 2021, and resulted in ∼1400 publications. Additional publications were retrieved until April 30, 2021, and were based on reference lists of publications identified in the primary literature study, on reference lists in guidance documents and advices, and on internet searches using terms relevant for the current study.†
Most publications presented applications of a previously generated construct, referring to a common original article in which that construct was initially described. While some described features that are claimed to be beneficial for biosafety, none had the intention to study biosafety.
Further insight into risk assessments was obtained through an analysis of guidance documents, advices, and recommendations issued by (inter)national organizations and institutions related to (bio)safety. These included but were not limited to the COGEM, the European Biosafety Association, the American Biosafety Association, the National Institutes of Health, the World Health Organization, the Canadian Centre for Veterinary Biologics, and the Central Committee on Biological Safety (Zentrale Kommission für die Biologische Sicherheit, ZKBS). A search was performed using the term “replicon” on the websites of the respective organizations and institutions.
Application of Viral RNA Replicons
In total, the application of 54 replicon systems based on a variety of single-stranded RNA viruses was identified, including 3 chimeric approaches, that is, replicons based on 2 viral species. A summary of applications of the different replicon systems is provided in Table 2. A more detailed description of replicon application is provided below.
Table 2.
Application of viral RNA replicons (nonlimitative overview)
| Classification | Family | Virus (wild type or vaccine strain) | Study viral (genome) replication | Study of highly infectious viruses | Antiviral drug discovery | Cell modification in vitro |
Vaccination against infectious disease
|
Gene therapy/anticancer therapy
|
References (nonlimitative list of [review] articles) |
|---|---|---|---|---|---|---|---|---|---|
| (Pre-) clinical | (Pre-) clinical | ||||||||
| Positive-sense nonsegmented | Astroviridae | Human astrovirus |
|
92 | |||||
| Caliciviridae | Norovirus (Norwalk virus) |
|
|
7,12,93 | |||||
| Coronaviridae | Middle East respiratory syndrome coronavirus |
|
|
94 | |||||
| Severe acute respiratory syndrome coronavirus |
|
|
|
12,35,95 | |||||
| Severe acute respiratory syndrome coronavirus 2 |
|
|
11,96 | ||||||
| Flaviviridae | Bovine viral diarrhea virus |
|
|
17,97 | |||||
| Classical swine fever |
|
|
|
16–19 | |||||
| Dengue virus |
|
|
|
|
9,12,93 | ||||
| Hepatitis C virus |
|
|
|
|
12,98 | ||||
| Japanese encephalitis virus |
|
|
|
|
|
9,99,100 | |||
| Kunjin virus |
|
|
|
|
3,18,93 | ||||
| Niénokoué virus |
|
9 | |||||||
| Tick-borne encephalitis virus |
|
|
16 | ||||||
| WNV |
|
|
|
|
12,17,101 | ||||
| Yellow fever virus |
|
|
|
|
9,12,18 | ||||
| Zika virus |
|
|
|
9,12 | |||||
| Hepeviridae | Hepatitis E virus |
|
|
|
4,12,102 | ||||
| Picornaviridae | Mengovirus |
|
|
24 | |||||
| Poliovirus |
|
|
|
1,12,27 | |||||
| Foot-and-mouth disease virus |
|
103 | |||||||
| Enterovirus 71 |
|
12,104 | |||||||
| Coxsackie virus |
|
12 | |||||||
| Hepatitis A virus |
|
12 | |||||||
| Human rhinovirus |
|
12 | |||||||
| Togaviridae | Salmon pancreas disease virus |
|
12,17 | ||||||
| Chikungunya virus |
|
|
|
18,32 | |||||
| SFV |
|
|
|
16–18,31,32,105 | |||||
| SINV |
|
|
|
17,18,106 | |||||
| VEEV |
|
|
|
16–19,32 | |||||
| Western equine encephalitis virus |
|
|
|
18,93 | |||||
| Eastern equine encephalitis virus |
|
|
18 | ||||||
| Chimeric | SINV/VEEV |
|
16,19 | ||||||
| SINV/Western equine encephalitis virus (Eastern equine encephalitis) |
|
107 | |||||||
| Vesicular stomatitis virus/chikungunya virus |
|
18 | |||||||
| Negative-sense nonsegmented | Bornaviridae | Borna disease virus |
|
|
108,109 | ||||
| Filoviridae | Ebola virus |
|
|
8,110 | |||||
| Marburg virus |
|
|
8,111 | ||||||
| Paramyxoviridae | Measles virus |
|
|
|
|
13,18,32 | |||
| Parainfluenza virus |
|
112 | |||||||
| Nipah virus |
|
|
113 | ||||||
| Human metapneumovirus |
|
114 | |||||||
| Respiratory syncytial virus |
|
|
93,115 | ||||||
| Newcastle disease virus |
|
116 | |||||||
| Sendai virus |
|
|
|
23,36,117,118 | |||||
| Rhabdoviridae | VSV |
|
|
|
17,18,32,119 | ||||
| Negative-sense segmented | Arenaviridae | Lassa virus |
|
|
|
|
8,18,120 | ||
| Junin virus |
|
|
8 | ||||||
| Tacaribe virus |
|
|
8 | ||||||
| Machupo virus |
|
|
8 | ||||||
| Hantaviridae | Hantaan virus |
|
121 | ||||||
| Nairoviridae | Crimean-Congo hemorrhagic fever virus |
|
|
|
8 | ||||
| Phenuiviridae | Huaiyangshan banyangvirus (also known as “severe fever with thrombocytopenia virus”) |
|
|
8,122,123 | |||||
| Rift Valley fever virus |
|
|
|
|
17,124 | ||||
| Uukuniemi uukuvirus |
|
8 |
, Preclinical trials only; 
, clinical trials.
Replicons in Research
Study viral (genome) replication
Several viruses do not amplify efficiently in cell culture. To overcome the need to use wild-type virus particles to study genome replication, a self-replicating naked RNA replicon of the virus can be created and transfected in a suitable cell line. These systems have been developed for the following: poliovirus,1 Semliki Forest virus (SFV),2 Kunjin virus,3 hepatitis C virus,4 hepatitis E virus,5,6 and norovirus.7 The resulting cell line, stably expressing the replicon RNA, can subsequently be used to study the effect of viral gene expression on intracellular events, as well as to study antiviral drugs, in particular substances that affect viral genome replication. In these cellular systems, no structural proteins are provided in trans, as they do not aim to generate VRPs.
Study of highly infectious viruses at lower containment level
Due to their high pathogenicity and the limitations in the prevention or treatment of infection, viruses of risk group 4 (RG4) can only be handled in BSL-4 containment conditions. To allow research on these highly pathogenic agents at a lower containment level, various replicon systems have been generated (reviewed by Hoenen et al.8). This is the case for multiple human RG4 viruses belonging to, for example, Arenaviridae, Phenuiviridae, Hantaviridae, Paramyxoviridae, and Filoviridae. For RG4 viruses of veterinary importance, RNA replicons (sometimes also referred to as minigenomes) have been designed for foot and mouth disease virus and classical swine fever virus.
Also for risk group 3 agents, such as for Flaviviridae (reviewed by Kummerer9) and, more recently for the Middle East severe respiratory syndrome coronavirus10 and the acute respiratory syndrome coronavirus 2,11 RNA replicon systems have been developed to allow research at a lower containment level.
Antiviral drug discovery
A replicon-based assay allows for a high-throughput approach to test large libraries of compounds. In general, structural genes are replaced by reporter genes within the replicon RNA, after which cells are transfected with naked replicons or infected with a VRP to allow genome replication. In case structural proteins are provided in trans, also viral egress and a subsequent single cycle of infection will occur. A potential inhibitory effect of drug substances can be assessed by means of reduced expression of the reporter protein.
It remains outside the scope of this publication to describe each system in detail. For that, the reader is referred to scientific reviews, such as, but not limited to, Hoenen et al.8 and Fernandes et al.12
Cell modification in vitro
Viral RNA replicons have also been used for cell modification and differentiation in vitro. Replicons originating from Paramyxoviridae have been used to make induced pluripotent stem cells (iPSCs). In brief, a measles virus-based replicon has been generated to introduce reprogramming factors, such as OCT4, SOX2, KLF4, or cMYC in cells. Replicon genomes are subsequently eliminated from the derived iPSCs, taking their transient nature into account, leaving a virus-free cell system for research.13
Also, Sendai virus vectors have been shown to efficiently generate human iPSCs from human fibroblasts14 and human blood cells.15 Here, the issue was raised of the sustained cytoplasmic replication of the Sendai viral vectors after the iPSCs have been established. Several approaches have been explored to overcome this sustained replication, as discussed further.
Since replicons can modify cells but do not bear the risk of insertional mutagenesis (see the Genome Integration section), they are promising tools in the generation of stem and progenitor cells for future treatment of genetic and degenerative disorders.
Human and veterinary medicinal applications of replicons
Vaccination against infectious disease
Replicons can be exploited in several ways to prime the immune system. Their main advantage over conventional (i.e., nonreplicating) messenger RNA (mRNA) is that the antigen being expressed is proportional to the number of RNA transcripts, whereby conventional mRNA may require large doses or repeated administrations.
Administration of replicon RNA can be achieved as VRPs, liposome-encapsulated particles (e.g., liposomes, dendrimers), naked RNA replicons, or synthetic cDNA. Such vaccination has resulted in transient expression of the gene of interest, strong immune responses, and protection against viral challenge.
A detailed overview of all replicon systems used for vaccine development remains outside the scope of this report. For that, the reader is referred to scientific reviews, such as, but not limited to, Bloom et al.,16 Hikke and Pijlman,17 and Lundstrom.18 Several replicon systems intended to be used as vaccines against infectious disease are currently being explored in clinical trials (reviewed by Blakney et al.19).
Gene therapy and anticancer treatment
Replicons can also be used to deliver genes of interest in the context of gene therapy and anticancer treatment. Due to their cytoplasmic localization and self-replicating nature, significantly elevated and prolonged levels of transgene expression can be achieved without the risk of genome integration.
Most current applications are related to cancer treatment. For example, viral replicons can be used to deliver transgenes that, in turn, can affect tumor growth and/or attract cytolytic effector cells. This has been explored for an SFV-based DREP encoding human papillomavirus early proteins E6 and E7.20 Using intradermal delivery followed by electroporation, it was demonstrated that the replicon induced effective, therapeutic antitumor immunity.20 Also, injection of SFV-based replicons coding for anti-programmed death ligand 1 monoclonal antibodies21 or a Sendai or Kunjin virus-based replicon encoding mouse granulocyte–macrophage colony-stimulating factor22,23 resulted in a potent antitumor response in in vivo models.
Finally, a mengovirus-based replicon has been used to modify tumor environment in the liver by inducing local recruitment of innate immunity effectors to the tumor, which complemented the effect of peptide-based vaccines in tumor regression.24
In addition, replicons proved useful as delivery vehicles for small RNAs (sRNAs) that target specific promotor regions. They can induce long-term transcriptional gene regulation mechanisms, including transcriptional gene silencing or transcriptional gene activation, with subsequent long-term therapeutic effects for various diseases. Their nonintegrating nature and potential for inducing short-term expression is also of interest. Furthermore, genome editing could be achieved using RNA replicons with clustered regularly interspaced short palindromic repeat/CRISPR-associated protein 9 (CRISPR/Cas9). The application of replicons as delivery vehicle for sRNAs and CRISPR/Cas has been reviewed by Baltusnikas et al.25,26
In view of cancer treatment, some replicon systems have intrinsic oncolytic properties. For example, RNA replicons derived from poliovirus showed a direct oncolytic effect on tumor cells of various origin in vitro based on their cytopathic effects.27 The oncolytic effect was confirmed in vivo where it was additionally shown that the replicons not only exerted an effect at the site of inoculation but were also able to reach tumor cells that had metastasized from the initial site of implantation. Also, for vesicular stomatitis virus (VSV) replicons, an intrinsic oncolytic effect was demonstrated without vector-associated toxicities. The preferred genome replication in tumor cells, due to significantly attenuated antiviral responses in these cells, further supports the safety of the VSV replicon application.28,29 Also, for recombinant SFV particles and naked viral RNA replicons, promising results have been obtained in view of cancer therapy because they ensure a high level of transgene expression, a rapid and strong cytopathic effect,30 and an upregulation of interferon-stimulated genes.21
Several replicon systems intended to be used as anticancer therapy are currently being explored in clinical trials31 (reviewed by Blakney et al.19 and Lundstrom.32).
Risk Considerations for Replicon Systems
This section focuses on specific considerations that may affect the risk of replicon systems for the user and/or the environment. While reviewing the different considerations, it must be emphasized that for each consideration, the relevance for the risk assessment will depend on the type of replicon system and its application. Additionally, it is noted that this review focuses on the hazards of the replicon system itself. When transgenes are inserted in the replicon RNA, these transgenes and their expression products may additionally affect the hazards of the system (e.g., oncogenes, toxins). The assessment of the transgene, however, is not part of the current review.
Naked Replicons
Naked replicons comprise the genetic information for nonstructural proteins and, if relevant, for some of the structural proteins in case they contain important elements necessary for efficient RNA replication and/or other sequences required for nonstructural protein or transgene expression. Consequently, transfection of cells with naked RNA allows for genome replication but not for generation of replicon particles.
Genome integration
Upon cell infection, some viruses integrate their genome into the host chromosome, either as part of their life cycle (e.g., retroviruses) or accidentally. Genome integration may not only potentially promote long-term persistence of a virus in the cell, but it could also lead to drastic consequences for the host cell, such as gene disruption, insertional mutagenesis, and/or cell death. For DNA viruses, the genome forms a potential substrate for host genome integration, without the need for prior processing. However, most RNA viruses are not able to deliberately integrate their genome into the host chromosome, as their genetic information resides in RNA molecules and not in DNA.
The only exception is retroviruses because they possess the ability to transcribe their viral RNA genome into DNA, a mandatory step for productive retroviral infection. For all other RNA viruses, genome integration has not been reported or has been reported only as an incidental event.33 No data were found on the actual consequences of such rare integration events. Furthermore, no data were found on the likelihood for and the potential consequences of integration of replicons based on cDNA (DREP). Whether or not the reverse transcriptase encoded by the second open reading frame (ORF) of the human Long Interspersed Nuclear Element 1 in eukaryotic cells could allow for genome integration of replicons remains to be studied.34
Persistence in the host cell
Several RNA viruses have developed specific mechanisms, for example, at the level of viral transcription and replication, that could lead to the establishment of persistent infection. To follow-up viral genome replication and to screen drug compounds in vitro, this feature has been exploited in RNA replicon systems based on, among others, hepatitis C virus,4 severe acute respiratory syndrome coronavirus,35 norovirus,7 and hepatitis E virus.5 In brief, cells are stably transfected with (cDNA for) replicon RNA carrying a drug resistance gene, after which RNA genome replication is maintained in the cell.
For other applications, for example, when cells will be used for additional applications such as stem cell therapy, or when subsequent activities must be performed with less containment measures, prolonged presence of the RNA replicon is undesired. In these cases, conditional replication of the RNA replicon provides a means to control replicon expression. Ban et al.36 described modified Sendai virus replicon systems in which various temperature-sensitive mutations were introduced. When transduced cells were maintained at permissive temperature (maximum 37°C), the temperature-sensitive Sendai virus replicon was detected on 3 dpi as based on a green fluorescent protein (GFP) signal. However, switching to nonpermissive temperatures (38°C or 39°C) resulted in a disappearance of the GFP signal. Moreover, when placing the cells again at 37°C, GFP was not detected in the cells by which the authors concluded that transgene-free human iPSCs were obtained.
Also, the use of micro (mi)RNA or small interfering (si)RNA provides an interesting means to express conditionally replicon RNA. For example, a Sendai virus-based replicon was extended with a sequence encoding a target sequence for the so-called miRNA 302. This miRNA is specifically expressed in pluripotent stem cells and targets the RNA of the polymerase L gene. As a consequence, the replicon system is expected to disappear from the reprogrammed cells. However, to conclude on the effectiveness of the approach, it is advised to check the cells for the absence of syncytia by microscopy, the absence of Sendai virus nucleoprotein (NP) expression by immunofluorescence, and the absence of Sendai virus NP mRNA by quantitative polymerase chain reaction (PCR).37
To control replicon replication, also the use of riboswitches has been studied. In brief, a riboswitch is a regulatory segment of an mRNA molecule that binds a specific small molecule such as a therapeutic compound, resulting in a change in production of the proteins encoded by the RNA. For example, for alphaviruses, Bell et al.38 incorporated a riboswitch into the replicon 3′ untranslated region (UTR). By doing so, they could successfully regulate the expression of DNA-launched and VRP-packaged replicons. When integrating riboswitches into the 3′ and 5′ UTR of the subgenomic RNA region of the TC-83 strain of Venezuelan equine encephalitis virus (VEEV), replication could be regulated at an even greater fold.
The results clearly indicate that riboswitches hold promise for the development of novel vaccination strategies, whereby gene expression can be controlled in such a way that only one injection will allow for priming and boosting of the immune response. Also, for gene therapy or cancer treatment, regulated expression of replicons will have clear benefits.39 Further study will, however, be needed on the safety limits of each of the approaches, as mi- or siRNAs may not completely prevent the expression of all genes they act on.
Rather than including an element for conditional replication in the replicon system itself, external control factors can also be used. For example, cells can be treated with specific small molecules as was shown for cells transduced with a Borna disease virus-based replicon. Treatment with a small molecule, T-705, completely eliminated the replicon in persistently transduced cells.40
Generation of virus-like vesicles
As mentioned above, the risk of the transgene remains outside the scope of this study. However, using viral envelope genes as the transgene in a naked replicon is relevant for this report since it may allow for the formation of the so-called virus-like vesicles (VLVs).‡ In brief, upon their expression, the envelope proteins support the formation of vesicles in which the replicon RNA is packaged and spread from cell to cell. This phenomenon has been described when using a transgene coding for the G protein of VSV, a member of the Rhabdoviridae, in VEEV-based replicons or SFV-based replicons (Togaviridae).
VLVs with RNA replicon genomes were generated that could spread throughout a cell culture.41–43 The majority of the infectious particles were smaller and less dense than either VEEV or SFV particles. Characterization by electron microscopy showed membrane-enveloped vesicles that contained the VSV-G protein. Infectious particles were apparently generated by budding of vesicles containing VSV-G protein and the RNA replicon.41 Similarly, VLVs were generated when an envelope protein of murine leukemia virus was expressed by an SFV replicon.41 From these studies, it is concluded that the formation of VLVs cannot be excluded when the coding sequence for (homologous or heterologous) envelope proteins is used as transgene.44
Effects on nontargeted cells
Replicons could potentially exert negative effects on cells that are not targeted for the delivery of the gene of interest. To limit the replicon genome replication and transgene expression to targeted cells or tissues, different approaches have been described. One such approach employs the use of miRNAs, which are involved in silencing gene expression. The miRNAs bind to their cognate target RNA that possesses a miRNA-binding sequence generally referred to as the miRNA recognition element (MRE). Subsequently, the miRNA will silence the target RNA. Lee et al.45 used this process to engineer a dengue virus-based replicon that could not replicate in the liver by inserting a liver-specific MRE into the viral mRNA. Binding of liver-specific miRNA with the MRE incorporated in the replicon RNA conferred an inhibitory effect on the replicon RNA.
Viral Replicon Particles
The risk considerations as mentioned for naked replicons are also relevant for VRP. The sections below elaborate on additional risk considerations that must be taken into account when producing and/or applying VRP.
Generation of replication competent viruses during production: “primary” replication competent virus
In the creation or production of VRP, there is a stage in which different viral genetic elements come together to assemble the infectious particle. In that stage, there is a potential risk for recombination due to homologous sequences, with the formation of “primary” replication competent virus (RCV). For nonsegmented single-stranded RNA viruses, recombination may theoretically occur between replicon RNA and co-replicating helper RNA (recombination is even less likely in case of nonreplicating helper RNA as it will only be translated but not transcribed or replicated).
For segmented, single-stranded negative-sense RNA viruses, recombination may occur between the transcription plasmids and the expression plasmids when homologous sequences are present, although the event appears to be rare (reviewed by Han and Worobey46). Depending on the virus family, the likelihood for recombination may differ. Based on that, different strategies have been developed to reduce the likelihood of such recombination, as explained below. The value of these strategies has already been described for other RNA viral vector systems, the most comprehensive for lentiviral vectors (reviewed by Pauwels et al.47).
Using bipartite helper RNA system
For those replicon systems where structural genes are provided via helper RNA, such as, but not limited to, those generated from Togaviridae, the likelihood of recombination can be reduced by dividing the genetic elements coding for the structural proteins over multiple helper RNAs. The rationale is that increasing the number of helper RNAs increases the number of recombination events that are required to generate a full functional viral genome. This is clearly illustrated in a study by Pushko et al.48 where a monopartite and a bipartite helper RNA system were used for a VEEV replicon. In the monopartite system, where complementing genes were present on only one helper RNA, plaque formation indicative for recombination was found for different types of helper RNAs used, even when only very limited sequence homology was present with the replicon RNA. However, when using the bipartite system where the complementing genes were divided over two helper RNAs, no infectious virus was detected in the plaque assay.
Similar observations were made for Sindbis virus (SINV), where no recombinants were detected using one helper RNA coding for the capsid protein and a second helper RNA coding for viral membrane proteins.49 These results were further confirmed in later studies by Hyvärinen et al.50 It is highlighted that in a bipartite system, single recombination events may still occur, but these will not result in plaque-forming virus since only capsid or membrane proteins will be included.48
Beissert et al.51 developed a novel bipartite vector system using trans-amplifying RNA. In brief, an alphavirus replicon encoding a particular transgene is used, but rather than including all nonstructural genes of the alphavirus, the genetic sequence encoding replicase is deleted from the replicon. Replicase activity is subsequently provided in trans by an optimized nonreplicating mRNA (nrRNA). In case the replicon could be complemented with genes encoding structural proteins, it is highly unlikely that additional recombination with the nrRNA would occur as recombination with nrRNA is highly infrequent.52 In consequence, a productive infection is equally unlikely to occur.
Blakney et al.53 developed a system called splitzicon. Hereby, the genes encoding the nonstructural proteins and the gene of interest are located on separate RNA molecules but still exhibit the self-amplification properties of replicon RNA.
Limiting sequence homology between genetic elements
Whether or not recombination will occur, at least in part depends on the amount of sequence homology between the genetic elements. As a rule of thumb, the higher the homology between sequences, the higher the likelihood for recombination. Therefore, limiting sequence homology will reduce the likelihood of recombination. Depending on the replicon system, limitation of sequence homology can be achieved at different levels. Representative examples are described below.
For replicons based on VEEV, as an example of positive-sense single-stranded RNA viruses, it was shown that the rate of recombination in a monopartite helper RNA system was inversely related to the size of deletion in the nonstructural protein genes in the helper RNA.48 However, even when the helper RNA contained <1 kb of nonstructural gene sequence, recombinants were detected, as evidenced by the generation of plaques in a plaque assay. The authors suggested that the likelihood for recombination was not only determined by the amount of sequence homology but also that specific sequences in the helper RNA may have affected the rate of nonhomologous recombination independent of deletion size, although the sequences of the recombinants were not determined.
Sequence homology can also be diminished by introducing mutations in coding sequences for the structural genes. In this perspective, Widman et al.54 used a VEEV replicon to supplement the genetic information for the C protein in a West Nile virus (WNV) genome containing a 70-codon deletion in the C protein. With the aim of producing VRPs but avoiding recombination between the modified WNV genome and the VEEV replicon, 36 silent mutations were incorporated in the C gene, thereby generating a sequence that was no longer complementary to the WNV genome and thus preventing replication of a recombinant genome.
For negative-sense, single-stranded segmented RNA viruses such as the Phenuiviridae, recombination at the level of the UTR is a specific risk. Their UTRs direct replication and transcription of viral RNA are sufficient to allow encapsidation of viral RNA into ribonucleoprotein complexes.55–57 Their terminal panhandle sequences are conserved between the different genome segments.58 In the context of replicons, it is to be considered that, when segments previously and deliberately made devoid of their UTR recapture a homologous or heterologous UTR by recombination, an autonomously replicating virus particle may be the result.
For example, in a Rift Valley fever virus-based replicon system where an expression plasmid coding for the ORF of the M segment (no M-UTR) is co-transfected with transcription plasmids coding for the S or L segment, recombination may allow for the M ORF to obtain an UTR of the S or L segment and consequently allow replication and encapsidation of the M segment. The above-mentioned recombination events have never been described in nature, considering that the likelihood for a single crossover in two RNA segments is extremely low. Moreover, in the unlikely and mainly theoretical event that such recombination would occur, the resulting virus that harbors two genome segments with the same UTR is likely attenuated, as was demonstrated in experimental studies using reverse genetics for Bunyamwera virus.58
Avoiding packaging of genetic elements coding for structural proteins in VRPs
Depending on the virus family, different elements are involved to ensure that all genetic elements are packaged into new virus particles, such as, but likely not limited to, specific packaging signals (reviewed by Mendes and Kuhn59 and Twarock and Stockley60), specific RNA–protein interactions (reviewed by Hornak et al.61), or UTRs.57 During infection with a wild-type virus, these elements ensure that the various viral components are included in the virus particle, thereby releasing a fully infectious virus able to enter and replicate in new host cells. As a biological containment measure for replicons, it is reasoned that when the elements are removed or modified, the related RNAs will not be packaged into the VRP, and, consequently, the VRP itself will not harbor these RNAs and thus cannot express the encoded proteins. Depending on the replicon system, interference with the elements involved in packaging can be achieved at different levels. Representative examples are described below.
For those replicon systems where structural genes are provided via a co-replicating helper RNA, the likelihood of packaging of the helper RNA into the VRP can be reduced by not providing a packaging signal in the co-replicating helper RNA. That modifying packaging signals may result in hampered packaging has been demonstrated for RNA originating from SFV. Liljeström and Garoff2 described defective helper RNA, derived from SFV RNA, that lacked a large portion of the nonstructural region, including the region corresponding to the packaging signal identified for SINV.
This defective helper RNA was able to package SFV replicons but was not packaged itself. Similar results were obtained by White et al.62 However, for replicons generated from SINV, it was shown that even a co-replicating helper RNA lacking a packaging sequence is still co-packaged at a rate of about 1/300 of that of single RNA containing a packaging sequence.63,64 Of particular interest is the C protein. Packaging of the co-replicating helper RNA encoding the C protein without the packaging signal may, at least in part, be the result of the presence of other sequences in the C protein that support packaging.63 Even when using a C protein from a different alphavirus, formation of particles containing the co-replicating helper RNA could not be prevented.63
As explained above, for negative-sense, single-stranded segmented RNA viruses such as the Phenuiviridae, it is particularly relevant to remove the UTRs of the genome segments that are not to be packaged into the VRP, taking into account that the presence of the UTR is sufficient to allow encapsidation of that genome segment into ribonucleoprotein complexes.57
Reducing the hazard of potentially formed RCV
While the previous approaches address the likelihood of recombination, safety of the replicon can be enhanced by modifying the nonstructural and/or structural proteins in such a way that in the unlikely event an RCV is formed, it does not exhibit hazardous characteristics.
For example, for SFV-based replicons, mutations were introduced that result in removal of protease activity of the capsid protein, so that in case of recombination of helper and replicon RNAs, the newly formed viral particle remains replication defective.65 Also for SFV-based replicons, mutations were introduced in the cleavage site of the precursor protein p62.66 The rationale was to construct a cleavage-deficient variant of the precursor protein that would not be cleaved into envelope proteins without external addition of chymotrypsin and thus would render the particles noninfectious unless chymotrypsin is externally added. The resulting RCV indeed turned out to be largely noninfectious, due to reduced binding and uptake into endosomes as well as the impaired ability to induce membrane fusion in the endosome. Based on these results, the authors concluded that a p62 cleavage-deficient helper function could be used to enhance the biosafety of the SFV replicon system.
Apart from introducing mutations in the structural proteins, an alternative approach is the use of structural proteins of a less pathogenic virus homologue. Provided that the less pathogenic phenotype is associated with attenuating mutations in genes encoding structural proteins, the VRPs generated are likely to display the biological properties of that virus, rather than that of the more pathogenic virus. For a VEEV-based replicon, Pushko et al.48 described the use of helper glycoprotein genes derived from V3014, a recombinant VEE vaccine strain, which contain two strongly attenuating mutations. V3014 is avirulent upon subcutaneous inoculation of mice67 and horses (Smith et al., unpublished observations), and therefore, any recombinant generated during VRP packaging is very likely to be at least as attenuated as V3014.
Similarly, structural genes of less pathogenic heterologous viruses can be used, thereby generating chimeric viruses. An example is a VEEV-based replicon system in which structural proteins of SINV were provided in trans.68,69 In comparison to VEEV, SINV is less pathogenic to humans. Also, despite extensive use, no laboratory-acquired infections with SINV have been described that could be assigned to airborne transmission, whereas this has been described for VEEV.70,71
Finally, also the genetic sequence of nonstructural genes can be modified to ensure that RCV, if formed, are not hazardous. Such was described for a Rift Valley fever virus system in which the coding sequence for the nonstructural protein in the S genome segment (i.e. NSs) was replaced by the enhanced GFP gene.72 Since NSs is involved in suppressing the host innate immune responses,73–75 its absence could avoid those immunosuppressive effects.
Conditional generation of VRPs
As mentioned for the naked replicons, the production of VRPs can be made conditional, as illustrated below.
Harvey et al.76 generated a stable baby hamster kidney packaging cell line, carrying a Kunjin virus structural gene cassette and a Tet-Off system. In the presence of tetracycline, no structural proteins were produced. Withdrawal of tetracycline from the medium resulted in the production of Kunjin virus structural proteins that were capable of packaging transfected and self-amplified Kunjin virus replicon RNA into secreted virus-like particles. The same cells were also capable of packaging replicon RNA from closely (WNV) and distantly (dengue virus 2) related flaviviruses.
For alphavirus replicons, it was shown that by incorporating miRNA-specific target sequences (tissue-specific MRE) into the helper RNAs, VRPs were found to be efficiently produced if miRNA-specific inhibitors were introduced into cells. However, in the absence of such inhibitors, cellular miRNAs were capable of downregulating helper RNA replication and consequently inhibited the VRP production.77
Testing for the absence of “primary” RCV
Formation of RCV during production of VRP is a major risk consideration. Still, only limited data were identified on assays and/or limits of detection to provide evidence for successfully avoiding the presence of RCV (Table 3). In brief, data on RCV testing were identified for 5 of 34 identified replicon systems based on positive-sense single-stranded RNA viruses (including 1 chimeric system), 1 of 14 identified replicon systems based on nonsegmented, negative-sense single-stranded RNA viruses, and 1 of 10 identified replicon systems based on segmented, negative-sense single-stranded RNA viruses.
Table 3.
Examples of assays for the verification of replication competent virus presence
| Classification | Family | Virus (wild type or vaccine strain) | Assay for RCV | Results | Detection limit | References |
|---|---|---|---|---|---|---|
| Positive-sense nonsegmented | Flaviviridae | Kunjin virus | Microscopy (detection of E protein by immunofluorescence during 2 passages in Vero cells) | No fluorescence detected | Not described | 76 |
| Intracranial inoculation of suckling mice | No clinical signs upon inoculation with 4 × 106 IU | Not described | ||||
| Picornaviridae | Poliovirus | Intraspinal or intracranial inoculation of mice | No neurovirulence | Not described | 125,126 | |
| Togaviridae | SFV | Microscopy (infect BHK-21 cells and titrate supernatant in a PFU assay) | No PFU found in 4.6 × 109 particles from 13 independent SFV-lacZ recombinant stocks (frequency of recombination <4.6 × 109) | Not described | 127 | |
| VEEV | Microscopy (detection of syncytia upon 2 passages in Vero cells) | No RCV detected | 1 RCV/1 × 1010 gm-replicon particles | 128 | ||
| Microscopy (detection of syncytia upon 2 passages in Vero cells) | No RCV detected | <100 RCV/1 × 108 gm-replicon particles | 48 | |||
| Chimeric | SINV/VEEV | Microscopy (detection of syncytia upon 5 passages in BHK cells) | No RCV detected | Not described | 68 | |
| Reporter assay (test supernatant of 5 passages in BHK cells for β-galactosidase expression) | No RCV detected in 40 batches | Not described | ||||
| Negative-sense nonsegmented | Paramyxoviridae | Sendai virus | Microscopy (detection of syncytia) | Absence of syncytia | Not described | 37 |
| qPCR (detection of NP mRNA) | Negative | 2–3 Positive cells | ||||
| Fluorescence microscopy (detection of fluorescent-labeled antibodies to NP) | No fluorescence detected | Not described | ||||
| Negative-sense segmented | Phenuiviridae | Rift Valley fever virus | Microscopy (detection of fluorescence during 2 passages in BHK cells) | No fluorescence detected upon inoculation of cells at an m.o.i. of 1 | Not described | 72 |
| Inoculation of suckling mice | No clinical signs upon inoculation with 1.0 × 104 TCID50 | Not described | 129 |
BHK, baby hamster kidney; E, envelop; gm, genetically modified; IU, infectious units; m.o.i., multiplicity of infection; mRNA, messenger RNA; NP, nucleoprotein; PFU, plaque-forming units; qPCR, quantitative polymerase chain reaction; RCV, replication competent virus; TCID, tissue culture infectious dose.
The amount of data is rather scarce compared with those for replication-deficient viruses, such as retro/lentiviruses and adeno-associated viruses, for which multiple guidelines have been developed. In brief, the FDA Guidance for Industry entitled “Testing of Retroviral Vector-Based Human Gene Therapy Products for Replication Competent Retrovirus During Product Manufacture and Patient Follow-up” provides recommendations regarding the testing for RCV during the manufacture of retroviral vector-based gene therapy products, and during follow-up monitoring of patients who have received retroviral vector-based gene therapy products. Recommendations include the identification and amount of material to be tested as well as general testing methods.
The requirement for testing adenovirus vectors for the presence of replication competent adenovirus (RCA) is described in Ph. Eur. 5.14 and the FDA guidance document entitled “Chemistry, Manufacturing, and Control (CMC) Information for Human Gene Therapy Investigational New Drug Applications (INDs)—Guidance for Industry” (2020). No acceptance criteria are specified in Ph. Eur. 5.14, but the FDA guidance document and the FDA BRMAC meeting number 30 (Adenovirus Titer Measurements and RCA levels, 2001) clearly state that the test is reported as “no RCA detected” if it complies with the acceptance criterion of <1 RCA/3 × 1010 virus particles.
Generation of VLVs
When using sequences of certain viral envelope proteins in helper RNA (or cDNA), generation of VLVs is a risk consideration during the production of VRPs, similar as described for the naked replicons (see section on Generation of virus-like vesicles).
Generation of RCVs During Application: “Secondary” RCV
Secondary events could lead to the generation of RCV, more specifically by recombination with or complementation by related viruses and/or unrelated viruses present in the host in vitro or in vivo during application of replicons.
In nature, recombination events do not appear to be a hallmark of negative-sense single-stranded RNA viruses.46 While sporadic authentic examples indicate that homologous recombination does occur, recombination seems to be generally rare or even absent in most negative-sense RNA viruses, and most of the homologous recombination events reported in the literature were likely generated artificially. Natural recombination has, however, been described for positive-sense single-stranded RNA viruses, where it is an important mechanism for promoting genetic variation. For example, for flaviviruses, recombination has been described under natural circumstances for dengue virus.78–80
Western equine encephalitis virus is believed to be a naturally occurring recombinant between SINV and Eastern equine encephalitis virus.81 Also, natural recombination among enteroviruses82,83 and among coronaviruses84 is frequently observed. Hereby, recombination of enteroviruses, in particular poliovirus, contributed to the emergence of pathogenic, circulating vaccine-derived polioviruses that have been complicating the World Health Organization's program for the global eradication of poliomyelitis,83 whereas recombination of coronaviruses may have contributed to the emergence of the pandemic severe acute respiratory syndrome coronavirus 2.85
Experimental conditions using cellular systems have confirmed and provided further insights in the recombination events. However, it is important to note that only one publication was identified in which recombination events were mimicked for viral RNA replicons.86 The authors described a system based on self-replicating subgenomic RNAs (replicons) derived from tick-borne encephalitis virus, WNV, and Japanese encephalitis virus for investigating the ability of the flavivirus genomes to recombine. They showed that, even though the replicon pairs shared ∼600 nucleotides of identical sequence where a precise homologous crossover event would have yielded a wild-type genome, recombination was not observed.
Only two aberrant recombination events were detected, both of which yielded unnatural genomes containing duplications. Moreover, infectious clones of these genomes yielded viruses with impaired growth properties. For tick-borne encephalitis virus and WNV, their systems did not yield any viable recombinant genomes at all. These data indicate that even in the case of high sequence homology, recombination was a rare event and resulted in impaired viruses only. The impaired character of chimeric viruses was also demonstrated for Togaviridae.87–89
Taking the above into account, recombination between replicon and (un)related viruses cannot be excluded. However, for recombination to happen, several conditions must be fulfilled. For example, replicon RNA and wild-type virus must be present in the same host cells. Therefore, if replicon and wild-type viruses enter different host cells, either because of differences in tropism or differences in route of application (e.g., injection in view of vaccination vs. natural infection), it is unlikely that recombination or complementation will occur. The likelihood of recombination or complementation is even further diminished if genome replication of the RNA replicon takes part in different cellular compartments compared with the wild-type virus replication.
Finally, even if replicon RNA and wild-type virus can infect the same host cell and replicate in the same cellular compartment, a phenomenon called superinfection resistance (also referred to as trans-inhibition or homologous interference) may prevent replication of the wild-type virus if the cell is already infected by the replicon. The phenomenon has been described for various single-stranded RNA viruses that are used for viral replicon design, including poliovirus,1 alphaviruses,90 measles virus, and hepatitis C virus. The mechanism of exclusion has been observed at various steps of the viral life cycle, including attachment, entry, viral genomic replication, transcription, and exocytosis.
Even though the data above indicate that the likelihood of recombination with or complementation by related and/or unrelated viruses is low, it cannot be excluded. Therefore, precautionary measures must be implemented mainly aiming at minimizing the risk of contact between replicon-containing materials or individuals on the one hand and the related and/or unrelated viruses on the other hand. In brief, measures can include, but are not necessarily limited to, selection of cells, experimental animals, and/or clinical trial subjects free of the related and/or unrelated viruses as tested by means of serological testing and/or PCR.
Conclusions
RNA replicons have gained much attention in research and development as well as clinical applications, such as gene therapy and vaccination. Despite these differences in origin and delivery mechanism, some common elements have been identified in the risk assessment for replicons as schematically represented in Figure 3.
Figure 3.
Summary of considerations justifying downscaling of the containment requirements of an activity with replicons. RCV, replication competent virus; RG, risk group.
In the case of VRP, the coding sequences for the structural proteins and those for the nonstructural proteins are separated from each other. The resulting VRP can enter the target cell as the wild-type virus would. However, while replication will occur, no new particles will be released from the cell. The risk classification of VRP can be downscaled substantially since they have turned into single round expression systems without pathogenic characteristics. Their classification is hardly related to that of the original virus. Nevertheless, uncertainty may still limit radical downscaling.
First, during the production of VRP when different viral genetic elements come together to assemble the infectious particle, there may be a theoretical or known possibility that recombination leads to the reconstitution of an RCV. Different approaches are discussed in this review for reducing the risk for primary RCV reconstitution. However, it should be noted that many considerations are of a theoretical nature. For example, different modifications in helper RNAs are expected to reduce the likelihood of recombination. Nonetheless, the exact impact of each modification (e.g., partial or complete deletion of structural genes, no sequence homology vs. limited sequence homology), as well as their (potential) cumulative nature (e.g., multiple plasmids and limited sequence homology) is not clear. Moreover, the (potential) impact is likely to differ between various virus families.
Second, although recombination leading to primary RCV is theoretically possible, there is little evidence that this recombination occurs at a significant frequency. Testing can provide further insights in the likelihood of recombination and the efficiency of the strategies for reducing the recombination potential. Still, each test method has its limitations. Robust data on which test to perform and what detection limits to consider are still restricted to only a few of the viral species for which replicons have been designed. One may question whether downscaling of the containment level for activities in which VRPs are produced can be allowed without confirmatory testing for the absence of RCV. Conversely, it can be questioned whether to perform repetitive testing once it has been established that RCV formation is unlikely.
Third, while replicons provide a safer approach for working with systems that are related to high-risk pathogens (in particular those belonging to RG4), the awareness of the high risk of the recipient may influence the willingness to downscale. Risk assessments are defined as an evaluation of likelihood and consequence. Even if the likelihood is low that recombination leads to a primary RCV, the possible consequences of handling a reconstituted RG4 virus at an inadequate containment level must be fully appreciated.
In the case of naked replicons, there is no phase during which the coding sequences for the structural genes are present. Primary recombination is therefore excluded. When solely using a coding sequence for nonstructural genes in the absence of any complementing sequences or sequences that allow for the formation of VLVs, the risk of the replicon seems negligible. Independent of the consideration of any inserted sequence, the risk group of the naked replicon can be downscaled as it lacks all pathogenic features. Although these elements provide a general argumentation, the specific nature of the replicon system (e.g., based on positive-sense vs. negative-sense RNA, use of co-replicating vs. nonreplicating helper RNA, segmented vs. nonsegmented RNA) may have to be used to refine the downscaling effort (e.g., different elements are involved in packaging for positive-sense compared with negative-sense RNA viruses, co-replicating helper RNAs are not used for negative-sense segmented RNA viruses).
While above we have highlighted the argumentation for downscaling the risk group of replicons, it must also be stressed that the situation differs depending on the type of activities performed with the replicon. For the worker handling the replicons, exposure is unintentional and unlikely even under low-level containment measures. In case of unintentional contact with the replicon, potential risks include persistence, VLV generation, effects of nontargeted cells, and for VRP production also secondary RCV formation. When replicons are intentionally administered in vivo (e.g., deliberate administration in view of vaccination of animals or delivery of genes for the treatment of human diseases), other risks may occur such as environmental risks.
For example, the participation of professional human research subjects in other studies besides that involving a replicon needs to be considered carefully, in particular when other studies involve vectors that may provide complementing sequences to the replicon. However, as replicons allow for a single infection only and do not have the capacity to cause a productive infection, the likelihood of recombination as a result of a consecutive study with a (potentially) complementing viral vector seems extremely low. Similarly, as the administration of the replicon will not result in viral replication and spread, shedding of replicons, if any, will be limited in time and space. Therefore, risk considerations are in a first instance relevant for and limited to the target cells. Cells or organisms treated with replicons can be handled in lower or no containment level once absence of replicon is demonstrated.
While secondary RCV formation based on recombination with a related or unrelated virus co-present in the host is a theoretical concern, its occurrence in reality remains unproven. The likelihood can be further reduced by avoiding simultaneous presence of the related or unrelated virus.
In conclusion, the most important concern is the primary production of RCV. In particular is the step where all genetic elements are present in the same cell. The recognition of this importance is illustrated by the fact that the majority of the publications address different strategies to reduce the risks for the formation of primary RCV. However, data on the actual need and/or efficiency of such measures remain scarcely documented. Even though it is not fully clear what the impact is of each of the measures, one might argue that the cumulative effect of different measures increases the safety profile.
In his “Swiss cheese model,” Reason91 visualized incidents as the result of the accumulation of multiple failures in defenses (represented as the holes in slices of cheese) that unfortunately align, creating an open “hazard trajectory” that results in harm. As long as enough layers of defense are in place, at least one with nonaligning holes, the probability of an accident can be minimized. For the risk measures of replicon systems, a similar reasoning could be proposed. Even though each individual measure is not yet proven leak tight, using multiple measures on different aspects of the system creates a solid barrier.
In the risk assessments summarized in this review, the starting point has been the recipient virus. The combination of safety measures allows a downscaling of the risk group as well as the containment measures. It is not clear if this approach, so embedded in the GMO legislation, is and will remain adequate. Design and application of replicons is a rapidly evolving field. Chimeric forms and even fully designed elements instead of those based on existing viruses are possible next steps. In such case, reference to a risk group determined for the recipient organism will not be possible, rather the risk classification will have to be developed de novo. The risk considerations identified in the current review can then be used to build up a “Swiss cheese model” that would support such risk group assignment, starting at the lowest level that would be applicable for self-amplifying RNA, that is, a naked replicon without a transgene.
Acknowledgments
The authors thank the following persons for critically reviewing the study and for sharing their technical insights and experience (in alphabetical order): Prof. Dr. Toos Daemen (University Medical Center Groningen/University of Groningen), Dr. E. Heylen (Rega Institute for Medical Research, KU Leuven), Dhr. Harmen Kloosterboer (National Institute for Public Health and the Environment), Prof. Dr. Jeroen Kortekaas (former Wageningen University and Research, currently Boehringer Ingelheim), Prof. Dr. J. Neyts (Rega Institute for Medical Research, KU Leuven), Dr. Ir. Gorben Pijlman (Wageningen University and Research), and Dr. Clara Posthuma (COGEM).
Acknowledge all sources of financial support (grants, fellowships, equipment, or remuneration of any kind) and any relationships that may be considered a conflict of interest (i.e., employment, stock holdings, retainers, paid or unpaid consultancies, patents or patent licensing arrangements, or honoraria) that may pertain to the manuscript (see Editorial and Ethical Policies).
Presentation details, if any: EBSA conference 2022, Ghent (Belgium), May 20, 2022.
Authors' Disclosure Statement
This article is based on research funded by the Netherlands Commission on Genetic Modification (COGEM).
Funding Information
No funding was received for this article.
Description of RNA viruses in plants (e.g., Nodaviridae) remains outside the scope of this study.
Between the end of the COGEM project and the time of submission of the review article, an additional literature search was performed but did not reveal significant new insights.
VLVs contain replicon RNA surrounded by an envelope composed of envelope proteins as encoded by the transgene, whereas VRPs contain replicon RNA surrounded by an envelope composed of envelope proteins as encoded by the helper plasmid or as expressed by the producer cells.
References
- 1. Kaplan G, Racaniello VR. Construction and characterization of poliovirus subgenomic replicons. J Virol 1988;62(5):1687–1696; doi: 10.1128/jvi.62.5.1687-1696.1988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Liljeström P, Garoff H. A new generation of animal cell expression vectors based on the Semliki Forest virus replicon. Biotechnology (N Y) 1991;9(12):1356–1361; doi: 10.1038/nbt1291-1356 [DOI] [PubMed] [Google Scholar]
- 3. Khromykh AA, Westaway EG. Subgenomic replicons of the flavivirus Kunjin: Construction and applications. J Virol 1997;71(2):1497–1505; doi: 10.1128/jvi.71.2.1497-1505.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Lohmann V, Körner F, Koch J, et al. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science 1999;285(5424):110–113; doi: 10.1126/science.285.5424.110 [DOI] [PubMed] [Google Scholar]
- 5. Cao DJ, Ni YY, Meng XJ. Substitution of amino acid residue V1213 in the helicase domain of the genotype 3 hepatitis E virus reduces virus replication. Virol J 2018;15(1):32; doi: 10.1186/s12985-018-0943-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Cao Y, Bing Z, Guan S, et al. Development of new hepatitis E vaccines. Hum Vacc Immunother 2018;14(9):2254–2262; doi: 10.1080/21645515.2018.1469591 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Chang KO, Sosnovtsev SV, Belliot G, et al. Stable expression of a Norwalk virus RNA replicon in a human hepatoma cell line. Virology 2006;353(2):463–473; doi: 10.1016/j.virol.2006.06.006 [DOI] [PubMed] [Google Scholar]
- 8. Hoenen T, Groseth A, de Kok-Mercado F, et al. Minigenomes, transcription and replication competent virus-like particles and beyond: Reverse genetics systems for filoviruses and other negative stranded hemorrhagic fever viruses. Antiviral Res 2011;91(2):195–208; doi: 10.1016/j.antiviral.2011.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Kummerer BM. Establishment and Application of Flavivirus Replicons. In: Dengue and Zika: Control and Antiviral Treatment Strategies. (Hilgenfeld R, Vasudevan SG. eds.) Springer: Singapore; 2018; pp. 165–173. [DOI] [PubMed] [Google Scholar]
- 10. Gutiérrez-Álvarez J, Honrubia JM, Sanz-Bravo A, et al. Middle East respiratory syndrome coronavirus vaccine based on a propagation-defective RNA replicon elicited sterilizing immunity in mice. Proc Natl Acad Sci U S A 2021;118(43):e2111075118; doi: 10.1073/pnas.2111075118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Zhang Y, Song WH, Chen SY, et al. A bacterial artificial chromosome (BAC)-vectored noninfectious replicon of SARS-CoV-2. Antiviral Res 2021;185:104974; doi: 10.1016/j.antiviral.2020.104974 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Fernandes RS, Freire M, Bueno RV, et al. Reporter replicons for antiviral drug discovery against positive single-stranded RNA viruses. Viruses 2020;12(6):598; doi: 10.3390/v12060598 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Wang Q, Vossen A, Ikeda Y, et al. Measles vector as a multigene delivery platform facilitating iPSC reprogramming. Gene Ther 2019;26(5):151–164; doi: 10.1038/s41434-019-0058-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Fusaki N, Ban H, Nishiyama A, et al. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proc Jpn Acad Ser B Phys Biol Sci 2009;85(8):348–362; doi: 10.2183/pjab.85.348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Seki T, Yuasa S, Fukuda K. Generation of induced pluripotent stem cells from a small amount of human peripheral blood using a combination of activated T cells and Sendai virus. Nat Protoc 2012;7(4):718–728; doi: 10.1038/nprot.2012.015 [DOI] [PubMed] [Google Scholar]
- 16. Bloom K, van den Berg F, Arbuthnot P. Self-amplifying RNA vaccines for infectious diseases. Gene Ther 2021;28(3–4):117–129; doi: 10.1038/s41434-020-00204-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Hikke MC, Pijlman GP. Veterinary replicon vaccines.Annu Rev Anim Biosci 2017;5:89–109. [DOI] [PubMed] [Google Scholar]
- 18. Lundstrom K. Self-Amplifying RNA viruses as RNA vaccines. Int J Mol Sci 2020;21(14):5130; doi: 10.3390/ijms21145130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Blakney AK, Ip S, Geall AJ. An update on self-amplifying mRNA vaccine development. Vaccines (Basel) 2021;9(2):97; doi: 10.3390/vaccines9020097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. van de Wall S, Ljungberg K, Ip PP, et al. Potent therapeutic efficacy of an alphavirus replicon DNA vaccine expressing human papilloma virus E6 and E7 antigens. Oncoimmunology 2018;7(10):487913; doi: 10.1080/2162402x.2018.1487913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Ballesteros-Briones MC, Martisova E, Casales E, et al. Short-term local expression of a PD-L1 blocking antibody from a self-replicating RNA vector induces potent antitumor responses. Mol Ther 2019;27(11):1892–1905; doi: 10.1016/j.ymthe.2019.09.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Hoang-Le D, Smeenk L, Anraku I, et al. A Kunjin replicon vector encoding granulocyte macrophage colony-stimulating factor for intra-tumoral gene therapy. Gene Ther 2009;16(2):190–199; doi: 10.1038/gt.2008.169 [DOI] [PubMed] [Google Scholar]
- 23. Inoue H, Iga M, Nabeta H, et al. Non-transmissible Sendai virus encoding granulocyte macrophage colony-stimulating factor is a novel and potent vector system for producing autologous tumor vaccines. Cancer Sci 2008;99(11):2315–2326; doi: 10.1111/j.1349-7006.2008.00964.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Couty JP, Crain AM, Gerbaud S, et al. Delivery of mengovirus-derived RNA replicons into tumoural liver enhances the anti-tumour efficacy of a peripheral peptide-based vaccine. Cancer Immunol Immunother 2008;57(8):1161–1171; doi: 10.1007/s00262-007-0448-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Baltusnikas J, Satkauskas S, Lundstrom K. Long-term transcriptional gene silencing by RNA viruses. Trends Biochem Sci 2018;43(6):397–401; doi: 10.1016/j.tibs.2018.03.003 [DOI] [PubMed] [Google Scholar]
- 26. Baltusnikas J, Satkauskas S, Lundstrom K. Constructing RNA viruses for long-term transcriptional gene silencing. Trends Biotechnol 2019;37(1):20–28; doi: 10.1016/j.tibtech.2018.07.015 [DOI] [PubMed] [Google Scholar]
- 27. Ansardi DC, Porter DC, Jackson CA, et al. RNA replicons derived from poliovirus are directly oncolytic for human tumor cells of diverse origins. Cancer Res 2001;61(23):8470–8479. [PubMed] [Google Scholar]
- 28. Shinozaki K, Ebert O, Kournioti C, et al. Oncolysis of multifocal hepatocellular carcinoma in the rat liver by hepatic artery infusion of vesicular stomatitis virus. Mol Ther 2004;9(3):368–376; doi: 10.1016/j.ymthe.2003.12.004 [DOI] [PubMed] [Google Scholar]
- 29. Shinozaki K, Ebert O, Woo SLC. Treatment of multi-focal colorectal carcinoma metastatic to the liver of immune-competent and syngeneic rats by hepatic artery infusion of oncolytic vesicular stomatitis virus. Int J Cancer 2005;114(4):659–664; doi: 10.1002/ijc.20772 [DOI] [PubMed] [Google Scholar]
- 30. Vasilevska J, Skrastina D, Spunde K, et al. Semliki Forest virus biodistribution in tumor-free and 4T1 mammary tumor-bearing mice: A comparison of transgene delivery by recombinant virus particles and naked RNA replicon. Cancer Gene Ther 2012;19(8):579–587; doi: 10.1038/cgt.2012.37 [DOI] [PubMed] [Google Scholar]
- 31. Komdeur FL, Singh A, van de Wall S, 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(2):611–625; doi: 10.1016/j.ymthe.2020.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Lundstrom K. Self-amplifying RNA virus vectors: Clinical applications in cancer drug delivery. Expert Opin Drug Deliv 2019;16(10):1027–1029; doi: 10.1080/17425247.2019.1653851 [DOI] [PubMed] [Google Scholar]
- 33. Desfarges S, Ciuffi A.. Viral Integration and Consequences on Host Gene Expression. In: Viruses: Essential Agents of Life. (Witzany G. ed.) Springer Netherlands: Dordrecht; 2012; pp. 147–175. [Google Scholar]
- 34. Feng Q, Moran JV, Kazazian HHJr., et al. Human L1 retrotransposon encodes a conserved endonuclease required for retrotransposition. Cell 1996;87(5):905–916; doi: 10.1016/s0092-8674(00)81997-2 [DOI] [PubMed] [Google Scholar]
- 35. Hertzig T, Scandella E, Schelle B, et al. Rapid identification of coronavirus replicase inhibitors using a selectable replicon RNA. J Gen Virol 2004;85:1717–1725; doi: 10.1099/vir.0.80044-0 [DOI] [PubMed] [Google Scholar]
- 36. Ban H, Nishishita N, Fusaki N, et al. Efficient generation of transgene-free human induced pluripotent stem cells (iPSCs) by temperature-sensitive Sendai virus vectors. Proc Natl Acad Sci U S A 2011;108(34):14234–14239; doi: 10.1073/pnas.1103509108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. CGM/130329-01—Advice by the Netherlands Commission on Genetic Modification (COGEM). Inschaling werkzaamheden gg-Sendai virus. Available from: https://cogem.net/publicatie/inschaling-werkzaamheden-met-gg-sendaivirus/ [Last accessed: March 28, 2023].
- 38. Bell CL, Yu D, Smolke CD, et al. Control of alphavirus-based gene expression using engineered riboswitches. Virology 2015;483:302–311; doi: 10.1016/j.virol.2015.04.023 [DOI] [PubMed] [Google Scholar]
- 39. Ketzer P, Kaufmann JK, Engelhardt S, et al. Artificial riboswitches for gene expression and replication control of DNA and RNA viruses. Proc Natl Acad Sci U S A 2014;111(5):E554–E562; doi: 10.1073/pnas.1318563111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Komatsu Y, Takeuchi D, Tokunaga T, et al. RNA virus-based episomal vector with a fail-safe switch facilitating efficient genetic modification and differentiation of iPSCs. Mol The Methods Clin Dev 2019;14:47–55; doi: 10.1016/j.omtm.2019.05.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Rolls MM, Webster P, Balba NH, et al. Novel infectious particles generated by expression of the vesicular stomatitis virus glycoprotein from a self-replicating RNA. Cell 1994;79(3):497–506; doi: 10.1016/0092-8674(94)90258-5 [DOI] [PubMed] [Google Scholar]
- 42. Rose NF, Buonocore L, Schell JB, et al. In vitro evolution of high-titer, virus-like vesicles containing a single structural protein. Proc Natl Acad Sci U S A 2014;111(47):16866–16871; doi: 10.1073/pnas.1414991111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. van den Pol AN, Mao G, Chattopadhyay A, et al. Chikungunya, Influenza, Nipah, and Semliki Forest Chimeric viruses with vesicular stomatitis virus: Actions in the brain. J Virol 2017;91(6):e02154-16; doi: 10.1128/jvi.02154-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. ZKBS. Allgemeine Stellungnahme der ZKBS zu häufig durchgeführten gentechnischen Arbeiten mit den zugrunde liegenden Kriterien der Vergleichbarkeit: Gentechnische Arbeiten mit dem Sindbis-Virusund dem Semliki-Forest-Virus-Expressionssystem. Az 6790-10-50 2017;13 pp. Available from: https://www.zkbs-online.de/ZKBS/SharedDocs/Downloads/01_Allgemeine%20Stellungnahmen/12_Stellungnahmen_Vergleichbarkeit/Sindbis-Virus_&_Semliki-Forest-Virus-Vektor_2017.html [Last accessed: March 28, 2023].
- 45. Lee TC, Lin YL, Liao JT, et al. Utilizing liver-specific microRNA-122 to modulate replication of dengue virus replicon. Biochem Biophys Res Commun 2010;396(3):596–601; doi: 10.1016/j.bbrc.2010.04.080 [DOI] [PubMed] [Google Scholar]
- 46. Han GZ, Worobey M. Homologous recombination in negative sense RNA viruses. Viruses 2011;3(8):1358–1373; doi: 10.3390/v3081358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Pauwels K, Gijsbers R, Toelen J, et al. State-of-the-art lentiviral vectors for research use: Risk assessment and biosafety recommendations. Curr Gene Ther 2009;9(6):459–474; doi: 10.2174/156652309790031120 [DOI] [PubMed] [Google Scholar]
- 48. Pushko P, Parker M, Ludwig GV, et al. Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: Expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo. Virology 1997;239(2):389–401; doi: 10.1006/viro.1997.8878 [DOI] [PubMed] [Google Scholar]
- 49. Frolov I, Frolova E, Schlesinger S. Sindbis virus replicons and Sindbis virus: Assembly of chimeras and of particles deficient in virus RNA. J Virol 1997;71(4):2819–2829; doi: 10.1128/JVI.71.4.2819-2829.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Hyvärinen A, Yongabi F, Mäkinen K, et al. Recombination of replicon and helper RNAs and the emergence of propagation-competent vectors upon Sindbis virus vector production. Int J Mol Med 2013;32(2):410–422; doi: 10.3892/ijmm.2013.1395 [DOI] [PubMed] [Google Scholar]
- 51. Beissert T, Perkovic M, Vogel A, et al. A Trans-amplifying RNA vaccine strategy for induction of potent protective immunity. Mol Ther 2020;28(1):119–128; doi: 10.1016/j.ymthe.2019.09.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Gmyl AP, Korshenko SA, Belousov EV, et al. Nonreplicative homologous RNA recombination: Promiscuous joining of RNA pieces? RNA 2003;9(10):1221–1231; doi: 10.1261/rna.5111803 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Blakney AK, McKay PF, Shattock RJ. Structural components for amplification of positive and negative strand VEEV splitzicons. Front Mol Biosci 2018;5:71; doi: 10.3389/fmolb.2018.00071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Widman DG, Ishikawa T, Fayzulin R, et al. Construction and characterization of a second-generation pseudoinfectious West Nile virus vaccine propagated using a new cultivation system. Vaccine 2008;26(22):2762–2771; doi: 10.1016/j.vaccine.2008.03.009 [DOI] [PubMed] [Google Scholar]
- 55. Barr JN, Elliott RM, Dunn EF, et al. Segment-specific terminal sequences of Bunyamwera bunyavirus regulate genome replication. Virology 2003;311(2):326–338; doi: 10.1016/s0042-6822(03)00130-2 [DOI] [PubMed] [Google Scholar]
- 56. Dunn EF, Pritlove DC, Jin H, et al. Transcription of a recombinant bunyavirus RNA template by transiently expressed bunyavirus proteins. Virology 1995;211(1):133–143; doi: 10.1006/viro.1995.1386 [DOI] [PubMed] [Google Scholar]
- 57. Osborne JC, Elliott RM. RNA binding properties of bunyamwera virus nucleocapsid protein and selective binding to an element in the 5’ terminus of the negative-sense S segment. J Virol 2000;74(21):9946–9952; doi: 10.1128/jvi.74.21.9946-9952.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Lowen AC, Boyd A, Fazakerley JK, et al. Attenuation of Bunyavirus replication by rearrangement of viral coding and noncoding sequences. J Virol 2005;79(11):6940–6946; doi: 10.1128/jvi.79.11.6940-6946.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Mendes A, Kuhn RJ. Alphavirus nucleocapsid packaging and assembly. Viruses 2018;10(3):138; doi: 10.3390/v10030138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Twarock R, Stockley PG. RNA-mediated virus assembly: Mechanisms and consequences for viral evolution and therapy. Annu Rev Biophys 2019;48(1):495–514; doi: 10.1146/annurev-biophys-052118-115611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Hornak KE, Lanchy JM, Lodmell JS. RNA encapsidation and packaging in the Phleboviruses. Viruses 2016;8(7):194; doi: 10.3390/v8070194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. 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(5):4320–4326; doi: 10.1128/jvi.72.5.4320-4326.1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Frolova E, Frolov I, Schlesinger S. Packaging signals in alphaviruses. J Virol 1997;71(1):248–258; doi: 10.1128/jvi.71.1.248-258.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Lu X, Silver J. Transmission of replication-defective Sindbis helper vectors encoding capsid and envelope proteins. J Virol Methods 2001;91(1):59–65; doi: 10.1016/s0166-0934(00)00247-0 [DOI] [PubMed] [Google Scholar]
- 65. CGM/111222-02—Advice by the Netherlands Commission on Genetic Modification (COGEM). Klinische studie met een Semliki forest virus vaccin tegen baarmoederhalskanker. Available from: https://cogem.net/publicatie/klinische-studie-met-een-semliki-forest-virus-vaccin-tegen-baarmoederhalskanker/ [Last accessed: March 28, 2023].
- 66. Berglund P, Sjöberg M, Garoff H, et al. Semliki Forest virus expression system: Production of conditionally infectious recombinant particles. Biotechnology (N Y) 1993;11(8):916–920; doi: 10.1038/nbt0893-916 [DOI] [PubMed] [Google Scholar]
- 67. Grieder FB, Davis NL, Aronson JF, et al. Specific restrictions in the progression of Venezuelan equine encephalitis virus-induced disease resulting from single amino acid changes in the glycoproteins. Virology 1995;206(2):994–1006; doi: 10.1006/viro.1995.1022 [DOI] [PubMed] [Google Scholar]
- 68. CGM/060314-01. Vaccinatie van apen met recombinante chimere alphavirusvectoren ten behoeve van vaccinonderzoek. 2006; p. 10. [Google Scholar]
- 69. Perri S, Greer CE, Thudium K, et al. An alphavirus replicon particle chimera derived from Venezuelan equine encephalitis and Sindbis viruses is a potent gene-based vaccine delivery vector. J Virol 2003;77(19):10394–10403; doi: 10.1128/jvi.77.19.10394-10403.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Pedrosa PBS, Cardoso TAO. Viral infections in workers in hospital and research laboratory settings: A comparative review of infection modes and respective biosafety aspects. Int J Infect Dis 2011;15(6):e366-e376; doi: 10.1016/j.ijid.2011.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Rusnak JM, Dupuy LC, Niemuth NA, et al. Comparison of aerosol- and percutaneous-acquired venezuelan equine encephalitis in humans and nonhuman primates for suitability in predicting clinical efficacy under the animal rule. Comp Med 2018;68(5):380–395; doi: 10.30802/AALAS-CM-18-000027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Kortekaas J, Oreshkova N, Cobos-Jimenez V, et al. Creation of a nonspreading Rift Valley Fever virus. J Virol 2011;85(23):12622–12630; doi: 10.1128/jvi.00841-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Billecocq A, Spiegel M, Vialat P, et al. NSs Protein of Rift Valley Fever virus blocks interferon production by inhibiting host gene transcription. J Virol 2004;78(18):9798–9806; doi: 10.1128/jvi.78.18.9798-9806.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Bouloy M, Janzen C, Vialat P, et al. Genetic evidence for an interferon-antagonistic function of Rift Valley Fever virus nonstructural protein NSs. J Virol 2001;75(3):1371–1377; doi: 10.1128/jvi.75.3.1371-1377.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Ikegami T, Won S, Peters CJ, et al. Rescue of infectious rift valley fever virus entirely from cDNA, analysis of virus lacking the NSs gene, and expression of a foreign gene. J Virol 2006;80(6):2933–2940; doi: 10.1128/jvi.80.6.2933-2940.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Harvey TJ, Liu WJ, Wang XJ, et al. Tetracycline-inducible packaging cell line for production of flavivirus replicon particles. J Virol 2004;78(1):531–538; doi: 10.1128/jvi.78.1.531-538.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Kamrud KI, Coffield VM, Owens G, et al. In vitro and in vivo characterization of microRNA-targeted alphavirus replicon and helper RNAs. J Virol 2010;84(15):7713–7725; doi: 10.1128/jvi.00310-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Chen SP, Yu M, Jiang T, et al. Identification of a recombinant dengue virus type 1 with 3 recombination regions in natural populations in Guangdong province, China. Arch Virol 2008;153(6):1175–1179; doi: 10.1007/s00705-008-0090-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Tolou HJG, Couissinier-Paris P, Durand JP, et al. Evidence for recombination in natural populations of dengue virus type 1 based on the analysis of complete genome sequences. J Gen Virol 2001;82(Pt 6):1283–1290; doi: 10.1099/0022-1317-82-6-1283 [DOI] [PubMed] [Google Scholar]
- 80. Worobey M, Rambaut A, Holmes EC. Widespread intra-serotype recombination in natural populations of dengue virus. Proc Natl Acad Sci U S A 1999;96(13):7352–7357; doi: 10.1073/pnas.96.13.7352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Hahn CS, Lustig S, Strauss EG, et al. Western equine encephalitis virus is a recombinant virus. Proc Natl Acad Sci U S A 1988;85(16):5997–6001; doi: 10.1073/pnas.85.16.5997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Combelas N, Holmblat B, Joffret ML, et al. Recombination between poliovirus and coxsackie A viruses of species C: A model of viral genetic plasticity and emergence. Viruses 2011;3(8):1460–1484; doi: 10.3390/v3081460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Muslin C, Mac Kain A, Bessaud M, et al. Recombination in Enteroviruses, a multi-step modular evolutionary process. Viruses 2019;11(9):859; doi: 10.3390/v11090859 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Su S, Wong G, Shi W, et al. Epidemiology, genetic recombination, and pathogenesis of coronaviruses. Trends Microbiol 2016;24(6):490–502; doi: 10.1016/j.tim.2016.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Zhu Z, Meng K, Meng G. Genomic recombination events may reveal the evolution of coronavirus and the origin of SARS-CoV-2. Sci Rep 2020;10(1):21617; doi: 10.1038/s41598-020-78703-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Taucher C, Berger A, Mandl CW. A trans-complementing recombination trap demonstrates a low propensity of Flaviviruses for intermolecular recombination. J Virol 2010;84(1):599–611; doi: 10.1128/jvi.01063-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Kuhn RJ, Griffin DE, Owen KE, et al. Chimeric Sindbis-Ross River viruses to study interactions between alphavirus nonstructural and structural regions. J Virol 1996;70(11):7900–7909; doi: 10.1128/JVI.70.11.7900-7909.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Paessler S, Fayzulin RZ, Anishchenko M, et al. Recombinant Sindbis/Venezuelan Equine encephalitis virus is highly attenuated and immunogenic. J Virol 2003;77(17):9278–9286; doi: 10.1128/jvi.77.17.9278-9286.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Schoepp RJ, Smith JF, Parker MD. Recombinant chimeric western and eastern equine encephalitis viruses as potential vaccine candidates. Virology 2002;302(2):299–309; doi: 10.1006/viro.2002.1677 [DOI] [PubMed] [Google Scholar]
- 90. Karpf AR, Lenches E, Strauss EG, et al. Superinfection exclusion of alphaviruses in three mosquito cell lines persistently infected with Sindbis virus. J Virol 1997;71(9):7119–7123; doi: 10.1128/jvi.71.9.7119-7123.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Reason J. Managing the Risks of Organizational Accidents. Ashgate Publishing: Aldershot; 1997. [Google Scholar]
- 92. Lulla V, Firth AE. A hidden gene in astroviruses encodes a viroporin. Nat Commun 2020;11(1):4070; doi: 10.1038/s41467-020-17906-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Hannemann H. Viral replicons as valuable tools for drug discovery. Drug Discov Today 2020;25(6):1026–1033; doi: 10.1016/j.drudis.2020.03.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Chen J, Hu B-J, Zhao K, et al. Development of a MERS-CoV replicon cell line for antiviral screening. Virol Sin 2021;36(4):730–735; doi: 10.1007/s12250-020-00341-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Wang JM, Wang LF, Shi ZL. Construction of a non-infectious SARS coronavirus replicon for application in drug screening and analysis of viral protein function. Biochem Biophys Res Commun 2008;374(1):138–142; doi: 10.1016/j.bbrc.2008.06.129 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Kotaki T, Xie X, Shi P-Y, et al. A PCR amplicon–based SARS-CoV-2 replicon for antiviral screening. bioRxiv 2020;2020.08.28.267567; doi: 10.1101/2020.08.28.267567 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Behrens S-E, Grassmann CW, Thiel H-J, et al. Characterization of an autonomous subgenomic pestivirus RNA replicon. J Virol 1998;72(3):2364–2372; doi: 10.1128/jvi.72.3.2364-2372.1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Ward JC, Bowyer S, Chen SC, et al. Insights into the unique characteristics of hepatitis C virus genotype 3 revealed by development of a robust sub-genomic DBN3a replicon. J Gen Virol 2020;101(11):1182–1190; doi: 10.1099/jgv.0.001486 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Li S-H, Li X-F, Zhao H, et al. Development and characterization of the replicon system of Japanese encephalitis live vaccine virus SA14-14-2. Virol J 2013;10:64–64; doi: 10.1186/1743-422X-10-64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Yun S-I, Song B-H, Lee Y-M. SA14-14-2Rep: A replicon vector derived from the live-attenuated Japanese encephalitis vaccine SA14-14-2 virus for human and veterinary medicine. Pediatr Infect Dis Open Access 2016;1:4. [Google Scholar]
- 101. Scholle F, Girard YA, Zhao QZ, et al. Trans-packaged West Nile virus-like particles: Infectious properties in vitro and in infected mosquito vectors. J Virol 2004;78(21):11605–11614; doi: 10.1128/jvi.78.21.11605-11614.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Emerson SU, Nguyen H, Graff J, et al. In vitro replication of hepatitis E virus (HEV) genomes and of an HEV replicon expressing green fluorescent protein. J Virol 2004;78(9):4838–4846; doi: 10.1128/jvi.78.9.4838-4846.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Tulloch F, Pathania U, Luke GA, et al. FMDV replicons encoding green fluorescent protein are replication competent. J Virol Methods 2014;209:35–40; doi: 10.1016/j.jviromet.2014.08.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Xiong Q, Wang Y, Xie B, et al. Single-step construction of a picornavirus replicon RNA with precise ends. J Virol Methods 2017;248:87–91; doi: 10.1016/j.jviromet.2017.06.009 [DOI] [PubMed] [Google Scholar]
- 105. Zhang WL, Hagedorn C, Schulz E, et al. Viral hybrid-vectors for delivery of autonomous replicons. Curr Gene Ther 2014;14(1):10–23; doi: 10.2174/1566523213666131223130024 [DOI] [PubMed] [Google Scholar]
- 106. Pan CH, Valsamakis A, Colella T, et al. Modulation of disease, T cell responses, and measles virus clearance in monkeys vaccinated with H-encoding alphavirus replicon particles. Proc Natl Acad Sci U S A 2005;102(33):11581–11588; doi: 10.1073/pnas.0504592102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Atasheva S, Wang EY, Adams AP, et al. Chimeric alphavirus vaccine candidates protect mice from intranasal challenge with western equine encephalitis virus. Vaccine 2009;27(32):4309–4319; doi: 10.1016/j.vaccine.2009.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Ikeda Y, Makino A, Matchett WE, et al. A novel intranuclear RNA vector system for long-term stem cell modification. Gene Ther 2016;23(3):256–262; doi: 10.1038/gt.2015.108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Daito T, Fujino K, Honda T, et al. A novel Borna disease virus vector system that stably expresses foreign proteins from an intercistronic noncoding region. J Virol 2011;85(23):12170–12178; doi: 10.1128/jvi.05554-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Tao WY, Gan TY, Guo MZ, et al. Novel stable Ebola virus minigenome replicon reveals remarkable stability of the viral genome. J Virol 2017;91(22):e01316-17; doi: 10.1128/jvi.01316-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Mühlberger E, Lötfering B, Klenk H-D, et al. Three of the four nucleocapsid proteins of Marburg Virus, NP, VP35, and L, are sufficient to mediate replication and transcription of Marburg virus-specific monocistronic minigenomes. J Virol 1998;72(11):8756–8764; doi: 10.1128/jvi.72.11.8756-8764.1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Hara K, Fukumura M, Ohtsuka J, et al. Human Parainfluenza virus type 2 vector induces dendritic cell maturation without viral RNA replication/transcription. Hum Gene Ther 2013;24(7):683–691; doi: 10.1089/hum.2013.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Freiberg A, Dolores LK, Enterlein S, et al. Establishment and characterization of plasmid-driven minigenome rescue systems for Nipah virus: RNA polymerase I- and T7-catalyzed generation of functional paramyxoviral RNA. Virology 2008;370(1):33–44; doi: 10.1016/j.virol.2007.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. de Graaf M, Herfst S, Schrauwen EJA, et al. Specificity and functional interaction of the polymerase complex proteins of human and avian metapneumoviruses. J Gen Virol 2008;89(4):975–983; doi: 10.1099/vir.0.83537-0 [DOI] [PubMed] [Google Scholar]
- 115. Duvall JR, VerPlank L, Ludeke B, et al. Novel diversity-oriented synthesis-derived respiratory syncytial virus inhibitors identified via a high throughput replicon-based screen. Antiviral Res 2016;131:19–25; doi: 10.1016/j.antiviral.2016.03.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Viktorova EG, Khattar SK, Kouiavskaia D, et al. Newcastle disease virus-based vectored vaccine against poliomyelitis. J Virol 2018;92(17):e00976-18; doi: 10.1128/jvi.00976-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Park A, Hong P, Won ST, et al. Sendai virus, an RNA virus with no risk of genomic integration, delivers CRISPR/Cas9 for efficient gene editing. Mol Ther Methods Clin Dev 2016;3:16057; doi: 10.1038/mtm.2016.57 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Bitzer M, Ungerechts G, Bossow S, et al. Negative-strand RNA viral vectors: Intravenous application of Sendai virus vectors for the systemic delivery of therapeutic genes. Mol Ther 2003;7(2):210–217; doi: 10.1016/s1525-0016(02)00052-7 [DOI] [PubMed] [Google Scholar]
- 119. Pattnaik AK, Andrew Ball L, LeGrone AW, et al. Infectious defective interfering particles of VSV from transcripts of a cDNA clone. Cell 1992;69(6):1011–1020; doi: 10.1016/0092-8674(92)90619-N [DOI] [PubMed] [Google Scholar]
- 120. Hass M, Golnitz U, Muller S, et al. Replicon system for Lassa virus. J Virol 2004;78(24):13793–13803; doi: 10.1128/jvi.78.24.13793-13803.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Flick K, Hooper JW, Schmaljohn CS, et al. Rescue of hantaan virus minigenomes. Virology 2003;306(2):219–224; doi: 10.1016/S0042-6822(02)00070-3 [DOI] [PubMed] [Google Scholar]
- 122. Brennan B, Li P, Zhang S, et al. Reverse genetics system for severe fever with thrombocytopenia syndrome virus. J Virol 2015;89(6):3026–3037; doi: 10.1128/jvi.03432-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. CGM/130502-01—Advice by the Netherlands Commission on Genetic Modification (COGEM). Inschaling werkzaamheden met gg-Huaiyangshan virus. Available from: https://cogem.net/publicatie/inschaling-werkzaamheden-met-gg-huaiyangshan-virus/ [Last accessed: March 28, 2023].
- 124. Oreshkova N, Spel L, Vloet RPM, et al. Preliminary evaluation of a Bunyavirus vector for cancer immunotherapy. J Virol 2015;89(17):9124–9127; doi: 10.1128/jvi.01105-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Bledsoe AW, Jackson CA, McPherson S, et al. Cytokine production in motor neurons by poliovirus replicon vector gene delivery. Nat Biotechnol 2000;18(9):964–969; doi: 10.1038/79455 [DOI] [PubMed] [Google Scholar]
- 126. Bledsoe AW, Gillespie GY, Morrow CD. Targeted foreign gene expression in spinal cord neurons using poliovirus replicons. J Neurovirol 2000;6(2):95–105; doi: 10.3109/13550280009013153 [DOI] [PubMed] [Google Scholar]
- 127. Smerdou C, Liljestrom P. Two-helper RNA system for production of recombinant Semliki Forest virus particles. J Virol 1999;73(2):1092–1098; doi: 10.1128/jvi.73.2.1092-1098.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. CGM/170224-01—Advice by the Netherlands Commission on Genetic Modification (COGEM). Omlaagschaling van in vivo en in vitro werkzaamheden met Venezuelan equine encephalitis virus (VEEV) replicons. Available from: https://cogem.net/publicatie/omlaagschaling-van-in-vivo-en-in-vitro-werkzaamheden-met-venezuelan-equine-encephalitis-virus-veev-replicons/ [Last accessed: March 28, 2023].
- 129. Dodd KA, Bird BH, Metcalfe MG, et al. Single-dose immunization with virus replicon particles confers rapid robust protection against Rift Valley fever virus challenge. J Virol 2012;86(8):4204–4212; doi: 10.1128/jvi.07104-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Belgian Biosafety Server. Tools—Belgian classification for micro-organisms based on their biological risks. Revised lists of pathogens and their corresponding class of biological risk. Available from: https://www.biosafety.be/content/tools-belgian-classification-micro-organisms-based-their-biological-risks [Last accessed: March 28, 2023].
- 131. Federal Office for the Environment. Classification of organisms Part 2: Viruses. Available from: https://www.bafu.admin.ch/bafu/en/home/topics/biotechnology/publications-studies/publications/classification-of-organisms.html [Last accessed: March 28, 2023].
- 132. The Central Committee on Biological Safety. Database of safety-assessed microorganisms. Available from: https://zag.bvl.bund.de/organismen/index.jsf?dswid=1047&dsrid=267 [Last accessed: March 28, 2023].



