1
|
Nazarenko AS, Vorovitch MF, Biryukova YK, Pestov NB, Orlova EA, Barlev NA, Kolyasnikova NM, Ishmukhametov AA. Flaviviruses in AntiTumor Therapy. Viruses 2023; 15:1973. [PMID: 37896752 PMCID: PMC10611215 DOI: 10.3390/v15101973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 10/29/2023] Open
Abstract
Oncolytic viruses offer a promising approach to tumor treatment. These viruses not only have a direct lytic effect on tumor cells but can also modify the tumor microenvironment and activate antitumor immunity. Due to their high pathogenicity, flaviviruses have often been overlooked as potential antitumor agents. However, with recent advancements in genetic engineering techniques, an extensive history with vaccine strains, and the development of new attenuated vaccine strains, there has been a renewed interest in the Flavivirus genus. Flaviviruses can be genetically modified to express transgenes at acceptable levels, and the stability of such constructs has been greatly improving over the years. The key advantages of flaviviruses include their reproduction cycle occurring entirely within the cytoplasm (avoiding genome integration) and their ability to cross the blood-brain barrier, facilitating the systemic delivery of oncolytics against brain tumors. So far, the direct lytic effects and immunomodulatory activities of many flaviviruses have been widely studied in experimental animal models across various types of tumors. In this review, we delve into the findings of these studies and contemplate the promising potential of flaviviruses in oncolytic therapies.
Collapse
Affiliation(s)
- Alina S. Nazarenko
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Mikhail F. Vorovitch
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Yulia K. Biryukova
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Nikolay B. Pestov
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Ekaterina A. Orlova
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Nickolai A. Barlev
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Nadezhda M. Kolyasnikova
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
| | - Aydar A. Ishmukhametov
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, Moscow 108819, Russia
- Institute of Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| |
Collapse
|
2
|
Lundstrom K. Application of DNA Replicons in Gene Therapy and Vaccine Development. Pharmaceutics 2023; 15:pharmaceutics15030947. [PMID: 36986808 PMCID: PMC10054396 DOI: 10.3390/pharmaceutics15030947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
DNA-based gene therapy and vaccine development has received plenty of attention lately. DNA replicons based on self-replicating RNA viruses such as alphaviruses and flaviviruses have been of particular interest due to the amplification of RNA transcripts leading to enhanced transgene expression in transfected host cells. Moreover, significantly reduced doses of DNA replicons compared to conventional DNA plasmids can elicit equivalent immune responses. DNA replicons have been evaluated in preclinical animal models for cancer immunotherapy and for vaccines against infectious diseases and various cancers. Strong immune responses and tumor regression have been obtained in rodent tumor models. Immunization with DNA replicons has provided robust immune responses and protection against challenges with pathogens and tumor cells. DNA replicon-based COVID-19 vaccines have shown positive results in preclinical animal models.
Collapse
|
3
|
Garcia G, Chakravarty N, Abu AE, Jeyachandran AV, Takano KA, Brown R, Morizono K, Arumugaswami V. Replication-Deficient Zika Vector-Based Vaccine Provides Maternal and Fetal Protection in Mouse Model. Microbiol Spectr 2022; 10:e0113722. [PMID: 36169338 PMCID: PMC9602260 DOI: 10.1128/spectrum.01137-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/28/2022] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne human pathogen, causes dire congenital brain developmental abnormalities in children of infected mothers. The global health crisis precipitated by this virus has led to a concerted effort to develop effective therapies and prophylactic measures although, unfortunately, not very successfully. The error-prone nature of RNA viral genome replication tends to promote evolution of novel viral strains, which could cause epidemics and pandemics. As such, our objective was to develop a safe and effective replication-deficient ZIKV vector-based vaccine candidate. We approached this by generating a ZIKV vector containing only the nonstructural (NS) 5'-untranslated (UTR)-NS-3' UTR sequences, with the structural proteins capsid (C), precursor membrane (prM), and envelope (E) (CprME) used as a packaging system. We efficiently packaged replication-deficient Zika vaccine particles in human producer cells and verified antigen expression in vitro. In vivo studies showed that, after inoculation in neonatal mice, the Zika vaccine candidate (ZVAX) was safe and did not produce any replication-competent revertant viruses. Immunization of adult, nonpregnant mice showed that ZVAX protected mice from lethal challenge by limiting viral replication. We then evaluated the safety and efficacy of ZVAX in pregnant mice, where it was shown to provide efficient maternal and fetal protection against Zika disease. Mass cytometry analysis showed that vaccinated pregnant animals had high levels of splenic CD8+ T cells and effector memory T cell responses with reduced proinflammatory cell responses, suggesting that endogenous expression of NS proteins by ZVAX induced cellular immunity against ZIKV NS proteins. We also investigated humoral immunity against ZIKV, which is potentially induced by viral proteins present in ZVAX virions. We found no significant difference in neutralizing antibody titer in vaccinated or unvaccinated challenged animals; therefore, it is likely that cellular immunity plays a major role in ZVAX-mediated protection against ZIKV infection. In conclusion, we demonstrated ZVAX as an effective inducer of protective immunity against ZIKV, which can be further evaluated for potential prophylactic application in humans. IMPORTANCE This research is important as it strives to address the critical need for effective prophylactic measures against the outbreak of Zika virus (ZIKV) and outlines an important vaccine technology that could potentially be used to induce immune responses against other pandemic-potential viruses.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Nikhil Chakravarty
- Department of Epidemiology, University of California, Los Angeles, Los Angeles, California, USA
| | - Angel Elma Abu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, USA
| | - Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Kari-Ann Takano
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Rebecca Brown
- Departments of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Kouki Morizono
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
- AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
4
|
Gutiérrez-Álvarez J, Honrubia JM, Sanz-Bravo A, González-Miranda E, Fernández-Delgado R, Rejas MT, Zúñiga S, Sola I, Enjuanes L. 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:e2111075118. [PMID: 34686605 PMCID: PMC8639359 DOI: 10.1073/pnas.2111075118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2021] [Indexed: 12/11/2022] Open
Abstract
Self-amplifying RNA replicons are promising platforms for vaccine generation. Their defects in one or more essential functions for viral replication, particle assembly, or dissemination make them highly safe as vaccines. We previously showed that the deletion of the envelope (E) gene from the Middle East respiratory syndrome coronavirus (MERS-CoV) produces a replication-competent propagation-defective RNA replicon (MERS-CoV-ΔE). Evaluation of this replicon in mice expressing human dipeptidyl peptidase 4, the virus receptor, showed that the single deletion of the E gene generated an attenuated mutant. The combined deletion of the E gene with accessory open reading frames (ORFs) 3, 4a, 4b, and 5 resulted in a highly attenuated propagation-defective RNA replicon (MERS-CoV-Δ[3,4a,4b,5,E]). This RNA replicon induced sterilizing immunity in mice after challenge with a lethal dose of a virulent MERS-CoV, as no histopathological damage or infectious virus was detected in the lungs of challenged mice. The four mutants lacking the E gene were genetically stable, did not recombine with the E gene provided in trans during their passage in cell culture, and showed a propagation-defective phenotype in vivo. In addition, immunization with MERS-CoV-Δ[3,4a,4b,5,E] induced significant levels of neutralizing antibodies, indicating that MERS-CoV RNA replicons are highly safe and promising vaccine candidates.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Viral/biosynthesis
- Coronavirus Infections/genetics
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/virology
- Defective Viruses/genetics
- Defective Viruses/immunology
- Female
- Gene Deletion
- Genes, env
- Humans
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Middle East Respiratory Syndrome Coronavirus/genetics
- Middle East Respiratory Syndrome Coronavirus/immunology
- Middle East Respiratory Syndrome Coronavirus/pathogenicity
- RNA, Viral/administration & dosage
- RNA, Viral/genetics
- RNA, Viral/immunology
- Replicon
- Vaccines, DNA
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Virulence/genetics
- Virulence/immunology
Collapse
Affiliation(s)
- J Gutiérrez-Álvarez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain
| | - J M Honrubia
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain
| | - A Sanz-Bravo
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain
| | - E González-Miranda
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain
| | - R Fernández-Delgado
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain
| | - M T Rejas
- Electron Microscopy Service, Centro de Biología Molecular "Severo Ochoa" (CBMSO-CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - S Zúñiga
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain
| | - I Sola
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain
| | - L Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Universidad Autónoma de Madrid 28049 Madrid, Spain;
| |
Collapse
|
5
|
Lundstrom K. Self-Replicating RNA Viruses for Vaccine Development against Infectious Diseases and Cancer. Vaccines (Basel) 2021; 9:1187. [PMID: 34696295 PMCID: PMC8541504 DOI: 10.3390/vaccines9101187] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 12/21/2022] Open
Abstract
Alphaviruses, flaviviruses, measles viruses and rhabdoviruses are enveloped single-stranded RNA viruses, which have been engineered for recombinant protein expression and vaccine development. Due to the presence of RNA-dependent RNA polymerase activity, subgenomic RNA can replicate close to 106 copies per cell for translation in the cytoplasm providing extreme transgene expression levels, which is why they are named self-replicating RNA viruses. Expression of surface proteins of pathogens causing infectious disease and tumor antigens provide the basis for vaccine development against infectious diseases and cancer. Self-replicating RNA viral vectors can be administered as replicon RNA at significantly lower doses than conventional mRNA, recombinant particles, or DNA plasmids. Self-replicating RNA viral vectors have been applied for vaccine development against influenza virus, HIV, hepatitis B virus, human papilloma virus, Ebola virus, etc., showing robust immune response and protection in animal models. Recently, paramyxovirus and rhabdovirus vector-based SARS-CoV-2 vaccines as well as RNA vaccines based on self-amplifying alphaviruses have been evaluated in clinical settings. Vaccines against various cancers such as brain, breast, lung, ovarian, prostate cancer and melanoma have also been developed. Clinical trials have shown good safety and target-specific immune responses. Ervebo, the VSV-based vaccine against Ebola virus disease has been approved for human use.
Collapse
|
6
|
Damase TR, Sukhovershin R, Boada C, Taraballi F, Pettigrew RI, Cooke JP. The Limitless Future of RNA Therapeutics. Front Bioeng Biotechnol 2021; 9:628137. [PMID: 33816449 PMCID: PMC8012680 DOI: 10.3389/fbioe.2021.628137] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Recent advances in the generation, purification and cellular delivery of RNA have enabled development of RNA-based therapeutics for a broad array of applications. RNA therapeutics comprise a rapidly expanding category of drugs that will change the standard of care for many diseases and actualize personalized medicine. These drugs are cost effective, relatively simple to manufacture, and can target previously undruggable pathways. It is a disruptive therapeutic technology, as small biotech startups, as well as academic groups, can rapidly develop new and personalized RNA constructs. In this review we discuss general concepts of different classes of RNA-based therapeutics, including antisense oligonucleotides, aptamers, small interfering RNAs, microRNAs, and messenger RNA. Furthermore, we provide an overview of the RNA-based therapies that are currently being evaluated in clinical trials or have already received regulatory approval. The challenges and advantages associated with use of RNA-based drugs are also discussed along with various approaches for RNA delivery. In addition, we introduce a new concept of hospital-based RNA therapeutics and share our experience with establishing such a platform at Houston Methodist Hospital.
Collapse
Affiliation(s)
- Tulsi Ram Damase
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Roman Sukhovershin
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Christian Boada
- Colleges of Medicine, Engineering, Texas A&M University and Houston Methodist Hospital, Houston, TX, United States
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Roderic I. Pettigrew
- Colleges of Medicine, Engineering, Texas A&M University and Houston Methodist Hospital, Houston, TX, United States
| | - John P. Cooke
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
7
|
Zhang C, Maruggi G, Shan H, Li J. Advances in mRNA Vaccines for Infectious Diseases. Front Immunol 2019; 10:594. [PMID: 30972078 PMCID: PMC6446947 DOI: 10.3389/fimmu.2019.00594] [Citation(s) in RCA: 414] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/05/2019] [Indexed: 12/19/2022] Open
Abstract
During the last two decades, there has been broad interest in RNA-based technologies for the development of prophylactic and therapeutic vaccines. Preclinical and clinical trials have shown that mRNA vaccines provide a safe and long-lasting immune response in animal models and humans. In this review, we summarize current research progress on mRNA vaccines, which have the potential to be quick-manufactured and to become powerful tools against infectious disease and we highlight the bright future of their design and applications.
Collapse
Affiliation(s)
- Cuiling Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | | | - Hu Shan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Junwei Li
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
8
|
Establishment and Application of Flavivirus Replicons. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1062:165-173. [PMID: 29845532 DOI: 10.1007/978-981-10-8727-1_12] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) are enveloped, positive-strand RNA viruses belonging to the genus Flavivirus in the family Flaviviridae. The genome of ~11 kb length encodes one long open reading frame flanked by a 5' and a 3' untranslated region (UTR). The 5' end is capped and the 3' end lacks a poly(A) tail. The encoded single polyprotein is cleaved co-and posttranslationally by cellular and viral proteases. The first one-third of the genome encodes the structural proteins (C-prM-E), whereas the nonstructural (NS) proteins NS1-NS2A-NS3-NS4A-2K-NS4B-NS5 are encoded by the remaining two-thirds of the genome.Research on flaviviruses was driven forward by the ability to produce recombinant viruses using reverse genetics technology. It is known that the purified RNA of flaviviruses is per se infectious, which allows initiation of a complete viral life cycle by transfecting the genomic RNA into susceptible cells. In 1989, the first infectious flavivirus RNA was transcribed from full-length cDNA templates of yellow fever virus (YFV) facilitating molecular genetic analyses of this virus. In addition to the production of infectious recombinant viruses, reverse genetics can also be used to establish non-infectious replicons. Replicons contain an in-frame deletion in the structural protein genes but still encode all nonstructural proteins and contain the UTRs necessary to mediate efficient replication, a factor that enables their analyses under Biosafety Level (BSL) 1 conditions. This is particularly important since many flaviviruses are BSL3 agents.The review will cover strategies for generating flavivirus replicons, including the establishment of bacteriophage (T7 or SP6) promoter-driven constructs as well as cytomegalovirus (CMV) promoter-driven constructs. Furthermore, different reporter replicons or replicons expressing selectable marker proteins will be outlined using examples of their application to answer basic questions of the flavivirus replication cycle, to select and test antiviral compounds or to produce virus replicon particles. The establishment and application of flavivirus replicons will further be exemplified by my own data using an established YFV reporter replicon to study the role of YFV NS2A in the viral life cycle. In addition, we established a reporter replicon of a novel insect-specific flavivirus, namely Niénokoué virus (NIEV), to define the barrier(s) involved in host range restriction.
Collapse
|
9
|
Hu P, Chen X, Huang L, Liu S, Zang F, Xing J, Zhang Y, Liang J, Zhang G, Liao M, Qi W. A highly pathogenic porcine reproductive and respiratory syndrome virus candidate vaccine based on Japanese encephalitis virus replicon system. PeerJ 2017; 5:e3514. [PMID: 28740748 PMCID: PMC5522605 DOI: 10.7717/peerj.3514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 06/07/2017] [Indexed: 01/19/2023] Open
Abstract
In the swine industry, porcine reproductive and respiratory syndrome (PRRS) is a highly contagious disease which causes heavy economic losses worldwide. Effective prevention and disease control is an important issue. In this study, we described the construction of a Japanese encephalitis virus (JEV) DNA-based replicon with a cytomegalovirus (CMV) promoter based on the genome of Japanese encephalitis live vaccine virus SA14-14-2, which is capable of offering a potentially novel way to develop and produce vaccines against a major pathogen of global health. This JEV DNA-based replicon contains a large deletion in the structural genes (C-prM-E). A PRRSV GP5/M was inserted into the deletion position of JEV DNA-based replicons to develop a chimeric replicon vaccine candidate for PRRSV. The results showed that BALB/c mice models with the replicon vaccines pJEV-REP-G-2A-M-IRES and pJEV-REP-G-2A-M stimulated antibody responses and induced a cellular immune response. Analysis of ELSA data showed that vaccination with the replicon vaccine expressing GP5/M induced a better antibodies response than traditional DNA vaccines. Therefore, the results suggested that this ectopic expression system based on JEV DNA-based replicons may represent a useful molecular platform for various biological applications, and the JEV DNA-based replicons expressing GP5/M can be further developed into a novel, safe vaccine candidate for PRRS.
Collapse
Affiliation(s)
- Pingsheng Hu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaoming Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lihong Huang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shukai Liu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Fuyu Zang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jinchao Xing
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Youyue Zhang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiaqi Liang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guihong Zhang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonoses, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Ministry of Agriculture, Guangzhou, China
| | - Wenbao Qi
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonoses, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Ministry of Agriculture, Guangzhou, China
| |
Collapse
|
10
|
Mosquito cell-derived West Nile virus replicon particles mimic arbovirus inoculum and have reduced spread in mice. PLoS Negl Trop Dis 2017; 11:e0005394. [PMID: 28187142 PMCID: PMC5322982 DOI: 10.1371/journal.pntd.0005394] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/23/2017] [Accepted: 02/06/2017] [Indexed: 12/17/2022] Open
Abstract
Half of the human population is at risk of infection by an arthropod-borne virus. Many of these arboviruses, such as West Nile, dengue, and Zika viruses, infect humans by way of a bite from an infected mosquito. This infectious inoculum is insect cell-derived giving the virus particles distinct qualities not present in secondary infectious virus particles produced by infected vertebrate host cells. The insect cell-derived particles differ in the glycosylation of virus structural proteins and the lipid content of the envelope, as well as their induction of cytokines. Thus, in order to accurately mimic the inoculum delivered by arthropods, arboviruses should be derived from arthropod cells. Previous studies have packaged replicon genome in mammalian cells to produce replicon particles, which undergo only one round of infection, but no studies exist packaging replicon particles in mosquito cells. Here we optimized the packaging of West Nile virus replicon genome in mosquito cells and produced replicon particles at high concentration, allowing us to mimic mosquito cell-derived viral inoculum. These particles were mature with similar genome equivalents-to-infectious units as full-length West Nile virus. We then compared the mosquito cell-derived particles to mammalian cell-derived particles in mice. Both replicon particles infected skin at the inoculation site and the draining lymph node by 3 hours post-inoculation. The mammalian cell-derived replicon particles spread from the site of inoculation to the spleen and contralateral lymph nodes significantly more than the particles derived from mosquito cells. This in vivo difference in spread of West Nile replicons in the inoculum demonstrates the importance of using arthropod cell-derived particles to model early events in arboviral infection and highlights the value of these novel arthropod cell-derived replicon particles for studying the earliest virus-host interactions for arboviruses. Many emerging viruses of public health concern are arthropod-borne, including tick-borne encephalitis, dengue, Zika, chikungunya, and West Nile viruses. The arboviruses are maintained in nature via virus-specific transmission cycles, involving arthropod (e.g. mosquitos, midges, and ticks) and vertebrate animals (e.g. birds, humans, and livestock). Common to all transmission cycles is the requirement of the arbovirus to replicate in these very different hosts. Since viruses rely on the host cell machinery to produce progeny, the virus particles from these hosts can differ in viral protein glycosylation and lipid content. Thus, the viral inoculum deposited by an infected arthropod will have different properties than virus produced in vertebrate cells. We set out to study the early events of arbovirus infection in a vertebrate host, using the mosquito-borne West Nile virus as a model. Here, we are the first to describe a robust protocol to produce West Nile replicon particles from mosquito cells. Since replicon particles are restricted to a single round of infection, we were able to compare the tropism and spread of the inoculum in animals for mosquito cell- and mammalian cell-derived replicon particles. We found that West Nile replicon particles derived from mosquito cells were significantly reduced in spread to distant sites compared to those derived from mammalian cells. Our results suggest that studies on arbovirus pathogenesis should be conducted with arthropod cell-derived virus, especially for the study of early virus-host interactions.
Collapse
|
11
|
Replicon RNA Viral Vectors as Vaccines. Vaccines (Basel) 2016; 4:vaccines4040039. [PMID: 27827980 PMCID: PMC5192359 DOI: 10.3390/vaccines4040039] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/14/2016] [Accepted: 10/28/2016] [Indexed: 12/22/2022] Open
Abstract
Single-stranded RNA viruses of both positive and negative polarity have been used as vectors for vaccine development. In this context, alphaviruses, flaviviruses, measles virus and rhabdoviruses have been engineered for expression of surface protein genes and antigens. Administration of replicon RNA vectors has resulted in strong immune responses and generation of neutralizing antibodies in various animal models. Immunization of mice, chicken, pigs and primates with virus-like particles, naked RNA or layered DNA/RNA plasmids has provided protection against challenges with lethal doses of infectious agents and administered tumor cells. Both prophylactic and therapeutic efficacy has been achieved in cancer immunotherapy. Moreover, recombinant particles and replicon RNAs have been encapsulated by liposomes to improve delivery and targeting. Replicon RNA vectors have also been subjected to clinical trials. Overall, immunization with self-replicating RNA viruses provides high transient expression levels of antigens resulting in generation of neutralizing antibody responses and protection against lethal challenges under safe conditions.
Collapse
|
12
|
Abstract
RNA viruses are characterized by their efficient capacity to replicate at high levels in mammalian cells leading to high expression of foreign genes and making them attractive candidates for vectors engineered for vaccine development and gene therapy. Particularly, alphaviruses, flaviviruses, rhabdoviruses and measles viruses have been applied for immunization against infectious agents and tumors. Application of replicon RNA, DNA/RNA-layered vectors and replication-deficient viral particles have provided strong immune responses and protection against challenges with lethal doses of viral pathogens or tumor cells. Moreover, tumor regression has been obtained when RNA replicons have been administered in the form of RNA, DNA and viral particles, including replication-proficient oncolytic particles.
Collapse
|
13
|
Abstract
The advent of reverse genetic approaches to manipulate the genomes of both positive (+) and negative (-) sense RNA viruses allowed researchers to harness these genomes for basic research. Manipulation of positive sense RNA virus genomes occurred first largely because infectious RNA could be transcribed directly from cDNA versions of the RNA genomes. Manipulation of negative strand RNA virus genomes rapidly followed as more sophisticated approaches to provide RNA-dependent RNA polymerase complexes coupled with negative-strand RNA templates were developed. These advances have driven an explosion of RNA virus vaccine vector development. That is, development of approaches to exploit the basic replication and expression strategies of RNA viruses to produce vaccine antigens that have been engineered into their genomes. This study has led to significant preclinical testing of many RNA virus vectors against a wide range of pathogens as well as cancer targets. Multiple RNA virus vectors have advanced through preclinical testing to human clinical evaluation. This review will focus on RNA virus vectors designed to express heterologous genes that are packaged into viral particles and have progressed to clinical testing.
Collapse
Affiliation(s)
- Mark A Mogler
- Harrisvaccines, Inc., 1102 Southern Hills Drive, Suite 101, Ames, IA 50010, USA
| | | |
Collapse
|
14
|
Baz A, Jackson DC, Kienzle N, Kelso A. Memory cytolytic T-lymphocytes: induction, regulation and implications for vaccine design. Expert Rev Vaccines 2014; 4:711-23. [PMID: 16221072 DOI: 10.1586/14760584.4.5.711] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The design of vaccines that protect against intracellular infections or cancer remains a challenge. In many cases, immunity depends on the development of antigen-specific memory CD8+ T-cells that can express cytokines and kill antigen-bearing cells when they encounter the pathogen or tumor. Here, the authors review current understanding of the signals and cells that lead to memory CD8+ T-cell differentiation, the relationship between the primary CD8+ T-cell response and the memory response and the regulation of memory CD8+ T-cell survival and function. The implications of this new knowledge for vaccine design are discussed, and recent progress in the development of lipidated peptide vaccines as a promising approach for vaccination against intracellular infections and cancer is reviewed.
Collapse
Affiliation(s)
- Adriana Baz
- Cooperative Research Centre for Vaccine Technology, Queensland Institute of Medical Research, Brisbane, Australia.
| | | | | | | |
Collapse
|
15
|
Mukherjee S, Pierson TC, Dowd KA. Pseudo-infectious reporter virus particles for measuring antibody-mediated neutralization and enhancement of dengue virus infection. Methods Mol Biol 2014; 1138:75-97. [PMID: 24696332 DOI: 10.1007/978-1-4939-0348-1_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This chapter outlines methods for the production of dengue virus (DENV) reporter virus particles (RVPs) and their use in assays that measure antibody-mediated neutralization and enhancement of DENV infection. RVPs are pseudo-infectious virions produced by complementation of a self-replicating flavivirus replicon with the DENV structural genes in trans. RVPs harvested from transfected cells are capable of only a single round of infection and encapsidate replicon RNA that encodes a reporter gene used to enumerate infected cells. RVPs may be produced using the structural genes of different DENV serotypes, genotypes, and mutants by changing plasmids used for complementation. Further modifications are possible including generating RVPs with varying levels of uncleaved prM protein, which resemble either the immature or mature form of the virus. Neutralization potency is measured by incubating RVPs with serial dilutions of antibody, followed by infection of target cells that express DENV attachment factors. Enhancement of infection is measured similarly using Fc receptor-expressing cells capable of internalizing antibody-virus complexes.
Collapse
Affiliation(s)
- Swati Mukherjee
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
16
|
Fournier C, Duverlie G, Castelain S. Are trans-complementation systems suitable for hepatitis C virus life cycle studies? J Viral Hepat 2013; 20:225-33. [PMID: 23490366 PMCID: PMC7167126 DOI: 10.1111/jvh.12069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 11/01/2012] [Indexed: 01/28/2023]
Abstract
Complementation is a naturally occurring genetic mechanism that has been studied for a number of plus-strand RNA viruses. Although trans-complementation is well documented for Flaviviridae family viruses, the first such system for hepatitis C virus (HCV) was only described in 2005. Since then, the development of a number of HCV trans-complementation models has improved our knowledge of HCV protein functions and interactions, genome replication and viral particle assembly. These models have also been used to produce defective viruses and so improvements are necessary for vaccine assays. This review provides an update on HCV trans-complementation systems, the viral mechanisms studied therewith and the production and characterization of trans-encapsidated particles.
Collapse
Affiliation(s)
- C. Fournier
- EA4294 Unité de Virologie Clinique et FondamentaleUniversité de Picardie Jules VerneAmiensFrance,Laboratoire de VirologieCentre Hospitalier Universitaire d'AmiensAmiensFrance
| | - G. Duverlie
- EA4294 Unité de Virologie Clinique et FondamentaleUniversité de Picardie Jules VerneAmiensFrance,Laboratoire de VirologieCentre Hospitalier Universitaire d'AmiensAmiensFrance
| | - S. Castelain
- EA4294 Unité de Virologie Clinique et FondamentaleUniversité de Picardie Jules VerneAmiensFrance,Laboratoire de VirologieCentre Hospitalier Universitaire d'AmiensAmiensFrance
| |
Collapse
|
17
|
Ulmer JB, Mason PW, Geall A, Mandl CW. RNA-based vaccines. Vaccine 2012; 30:4414-8. [DOI: 10.1016/j.vaccine.2012.04.060] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/10/2012] [Accepted: 04/18/2012] [Indexed: 02/07/2023]
|
18
|
Reynard O, Mokhonov V, Mokhonova E, Leung J, Page A, Mateo M, Pyankova O, Georges-Courbot MC, Raoul H, Khromykh AA, Volchkov VE. Kunjin virus replicon-based vaccines expressing Ebola virus glycoprotein GP protect the guinea pig against lethal Ebola virus infection. J Infect Dis 2011; 204 Suppl 3:S1060-5. [PMID: 21987742 DOI: 10.1093/infdis/jir347] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pre- or postexposure treatments against the filoviral hemorrhagic fevers are currently not available for human use. We evaluated, in a guinea pig model, the immunogenic potential of Kunjin virus (KUN)-derived replicons as a vaccine candidate against Ebola virus (EBOV). Virus like particles (VLPs) containing KUN replicons expressing EBOV wild-type glycoprotein GP, membrane anchor-truncated GP (GP/Ctr), and mutated GP (D637L) with enhanced shedding capacity were generated and assayed for their protective efficacy. Immunization with KUN VLPs expressing full-length wild-type and D637L-mutated GPs but not membrane anchor-truncated GP induced dose-dependent protection against a challenge of a lethal dose of recombinant guinea pig-adapted EBOV. The surviving animals showed complete clearance of the virus. Our results demonstrate the potential for KUN replicon vectors as vaccine candidates against EBOV infection.
Collapse
Affiliation(s)
- O Reynard
- Filovirus Laboratory, INSERM U758, Human Virology Department, Claude Bernard University Lyon-1, Université de Lyon, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Dendritic cell-specific antigen delivery by coronavirus vaccine vectors induces long-lasting protective antiviral and antitumor immunity. mBio 2010; 1. [PMID: 20844609 PMCID: PMC2939679 DOI: 10.1128/mbio.00171-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 08/16/2010] [Indexed: 11/20/2022] Open
Abstract
Efficient vaccination against infectious agents and tumors depends on specific antigen targeting to dendritic cells (DCs). We report here that biosafe coronavirus-based vaccine vectors facilitate delivery of multiple antigens and immunostimulatory cytokines to professional antigen-presenting cells in vitro and in vivo. Vaccine vectors based on heavily attenuated murine coronavirus genomes were generated to express epitopes from the lymphocytic choriomeningitis virus glycoprotein, or human Melan-A, in combination with the immunostimulatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF). These vectors selectively targeted DCs in vitro and in vivo resulting in vector-mediated antigen expression and efficient maturation of DCs. Single application of only low vector doses elicited strong and long-lasting cytotoxic T-cell responses, providing protective antiviral and antitumor immunity. Furthermore, human DCs transduced with Melan-A-recombinant human coronavirus 229E efficiently activated tumor-specific CD8+ T cells. Taken together, this novel vaccine platform is well suited to deliver antigens and immunostimulatory cytokines to DCs and to initiate and maintain protective immunity. Vaccination against infectious agents has protected many individuals from severe disease. In addition, prophylactic and, most likely, also therapeutic vaccination against tumors will save millions from metastatic disease. This study describes a novel vaccine approach that facilitates delivery of viral or tumor antigens to dendritic cells in vivo. Concomitant immunostimulation via the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) was achieved through delivery by the same viral vector. Single immunization with only low doses of coronavirus-based vaccine vectors was sufficient to elicit (i) vigorous expansion and optimal differentiation of CD8+ T cells, (ii) protective and long-lasting antiviral immunity, and (iii) prophylactic and therapeutic tumor immunity. Moreover, highly efficient antigen delivery to human DCs with recombinant human coronavirus 229E and specific stimulation of human CD8+ T cells revealed that this approach is exceptionally well suited for translation into human vaccine studies.
Collapse
|
20
|
Japanese encephalitis virus-based replicon RNAs/particles as an expression system for HIV-1 Pr55 Gag that is capable of producing virus-like particles. Virus Res 2009; 144:298-305. [PMID: 19406175 DOI: 10.1016/j.virusres.2009.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2008] [Revised: 04/16/2009] [Accepted: 04/20/2009] [Indexed: 11/23/2022]
Abstract
Ectopic expression of the structural protein Pr55(Gag) of HIV-1 has been limited by the presence of inhibitory sequences in the gag coding region that must normally be counteracted by HIV-1 Rev and RRE. Here, we describe a cytoplasmic RNA replicon based on the RNA genome of Japanese encephalitis virus (JEV) that is capable of expressing HIV-1 gag without requiring Rev/RRE. This replicon system was constructed by deleting all three JEV structural protein-coding regions (C, prM, and E) from the 5'-proximal region of the genome and simultaneously inserting an HIV-1 gag expression cassette driven by the internal ribosome entry site of encephalomyocarditis virus into the 3'-proximal noncoding region of the genome. Transfection of this JEV replicon RNA led to expression of Pr55(Gag) in the absence of Rev/RRE in the cytoplasm of hamster BHK-21, human HeLa, and mouse NIH/3T3 cells. Production of the Pr55(Gag) derived from this JEV replicon RNA appeared to be increased by approximately 3-fold when compared to that based on an alphavirus replicon RNA. Biochemical and morphological analyses demonstrated that the Pr55(Gag) proteins were released into the culture medium in the form of virus-like particles. We also observed that the JEV replicon RNAs expressing the Pr55(Gag) could be encapsidated into single-round infectious JEV replicon particles when transfected into a stable packaging cell line that provided the three JEV structural proteins in trans. This ectopic expression of the HIV-1 Pr55(Gag) by JEV-based replicon RNAs/particles in diverse cell types may represent a useful molecular platform for various biological applications in medicine and industry.
Collapse
|
21
|
Ishii K, Murakami K, Hmwe SS, Zhang B, Li J, Shirakura M, Morikawa K, Suzuki R, Miyamura T, Wakita T, Suzuki T. Trans-encapsidation of hepatitis C virus subgenomic replicon RNA with viral structure proteins. Biochem Biophys Res Commun 2008; 371:446-50. [PMID: 18445476 DOI: 10.1016/j.bbrc.2008.04.110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 04/02/2008] [Indexed: 01/30/2023]
Abstract
A trans-packaging system for hepatitis C virus (HCV) subgenomic replicon RNAs was developed. HCV subgenomic replicon was efficiently encapsidated by the HCV structural proteins that were stably expressed in trans under the control of a mammalian promoter. Infectious HCV-like particles (HCV-LPs), established a single-round infection, were produced and released into culture medium in titers of up to 10(3) focus forming units/ml. Expression of NS2 protein with structural proteins (core, E1, E2, and p7) was shown to be critical for the infectivity of HCV-LPs. Anti-CD81 treatment decreased the number of infected cells, suggesting that HCV-LPs infected cells in a CD81-dependent manner. The packaging cell line should be useful both for the production of single-round infectious HCV-LPs to elucidate the mechanisms of HCV assembly, particle formation and infection to host cells, and for the development of HCV replicon-based vaccines.
Collapse
Affiliation(s)
- Koji Ishii
- Department of Virology II, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Anraku I, Mokhonov VV, Rattanasena P, Mokhonova EI, Leung J, Pijlman G, Cara A, Schroder WA, Khromykh AA, Suhrbier A. Kunjin replicon-based simian immunodeficiency virus gag vaccines. Vaccine 2008; 26:3268-76. [PMID: 18462846 PMCID: PMC7115363 DOI: 10.1016/j.vaccine.2008.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 03/18/2008] [Accepted: 04/03/2008] [Indexed: 12/15/2022]
Abstract
An RNA-based, non-cytopathic replicon vector system, based on the flavivirus Kunjin, has shown considerable promise as a new vaccine delivery system. Here we describe the testing in mice of four different SIVmac239 gag vaccines delivered by Kunjin replicon virus-like-particles. The four vaccines encoded the wild type gag gene, an RNA-optimised gag gene, a codon-optimised gag gene and a modified gag-pol gene construct. The vaccines behaved quite differently for induction of effector memory and central memory responses, for mediation of protection, and with respect to insert stability, with the SIV gag-pol vaccine providing the optimal performance. These results illustrate that for an RNA-based vector the RNA sequence of the antigen can have profound and unforeseen consequences on vaccine behaviour.
Collapse
Affiliation(s)
- Itaru Anraku
- Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kent SJ, De Rose R, Mokhonov VV, Mokhonova EI, Fernandez CS, Alcantara S, Rollman E, Mason RD, Loh L, Peut V, Reece JC, Wang XJ, Wilson KM, Suhrbier A, Khromykh A. Evaluation of recombinant Kunjin replicon SIV vaccines for protective efficacy in macaques. Virology 2008; 374:528-34. [PMID: 18272194 DOI: 10.1016/j.virol.2008.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2007] [Revised: 01/02/2008] [Accepted: 01/05/2008] [Indexed: 01/03/2023]
Abstract
Persistent gag-specific T cell immunity would be a useful component of an effective HIV vaccine. The Flavivirus Kunjin replicon was previously engineered to persistently express HIV gag and was shown to induce protective responses in mice. We evaluated Kunjin replicon virus-like-particles expressing SIVgag-pol in pigtail macaques. Kunjin-specific antibodies were induced, but no SIV-specific T cell immunity were detected. Following SIVmac251 challenge, there was no difference in SIV viremia or retention of CD4 T cells between Kunjin-SIVgag-pol vaccine immunized animals and controls. An amnestic SIV gag-specific CD8 T cell response associated with control of viremia was observed in 1 of 6 immunized animals. Refinements of this vector system and optimization of the immunization doses, routes, and schedules are required prior to clinical trials.
Collapse
Affiliation(s)
- Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, 3010, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
RNA replicons represent potential vaccine delivery vehicles, but are considered too unstable for such use. This study examined the recovery, integrity and function of in vitro transcribed replicon RNA encoding hepatitis C virus (HCV) proteins. To remove residual template DNA, the RNA was digested with TURBO DNase followed by RNeasy DNase set and purified through an RNeasy column. The RNA was freeze-dried in distilled water or trehalose, stored under nitrogen gas for up to 10 months and analyzed at different time points. The recovery of RNA stored at < or = 4 degrees C that was freeze-dried in distilled water varied between 66% to zero of that recovered from RNA freeze-dried in 10% trehalose, a figure that depended on the duration of storage. In contrast, the recovery of the RNA stored in trehalose was consistently high for all time points. After recovery, both RNAs were translationally competent and expressed high levels of proteins after transfection, although the level of expression from the trehalose-stored RNA was consistently higher. Thus the addition of trehalose permitted stable storage of functional RNA at 4 degrees C for up to 10 months and this permits the development of RNA vaccines, even in developing countries where only minimum storage conditions (e.g., 4 degrees C) can be achieved.
Collapse
|
25
|
Jin H, Xiao W, Xiao C, Yu Y, Kang Y, Du X, Wei X, Wang B. Protective Immune Responses against Foot-and-Mouth Disease Virus by Vaccination with a DNA Vaccine Expressing Virus-Like Particles. Viral Immunol 2007; 20:429-40. [DOI: 10.1089/vim.2007.0031] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Huali Jin
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Wang Xiao
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Chong Xiao
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yang Yu
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Youmin Kang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Xiaogang Du
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Xuefeng Wei
- Inner Mongolia Factory for Biological Products, Jinyu Group Corporation, Huhhot, Inner Mongolia, China
| | - Bin Wang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, China
| |
Collapse
|
26
|
Lai CY, Hu HP, King CC, Wang WK. Incorporation of dengue virus replicon into virus-like particles by a cell line stably expressing precursor membrane and envelope proteins of dengue virus type 2. J Biomed Sci 2007; 15:15-27. [PMID: 17768670 DOI: 10.1007/s11373-007-9204-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 08/08/2007] [Indexed: 11/26/2022] Open
Abstract
While virus-like particles (VLPs) containing subgenomic replicons, which can transduce replicons into target cells efficiently for studying viral replication and vectors of gene therapy and vaccine, have been established for several flaviviruses, none has been reported for the four serotypes of dengue virus, the causal agent of the most important arboviral diseases in this century. In this study, we successfully established a cell line stably expressing the precursor membrane/envelope (PrM/E) proteins of dengue virus type 2 (DENV2), which can package a DENV2 replicon with deletion of PrM/E genes and produce single-round infectious VLPs. Moreover, it can package a similar replicon of different serotype, dengue virus type 4, and produce infectious chimeric VLPs. To our knowledge, this study reports for the first time replicon-containing VLPs of dengue virus. Moreover, this convenient system has potential as a valuable tool to study encapsidation of dengue virus and to develop novel chimeric VLPs containing dengue virus replicon as vaccine in the future.
Collapse
Affiliation(s)
- Chih-Yun Lai
- Institute of Microbiology, College of Medicine, National Taiwan University, No.1 Sec.1 Jen-Ai Rd, Taipei, Taiwan
| | | | | | | |
Collapse
|
27
|
Pijlman GP, Suhrbier A, Khromykh AA. Kunjin virus replicons: an RNA-based, non-cytopathic viral vector system for protein production, vaccine and gene therapy applications. Expert Opin Biol Ther 2006; 6:135-45. [PMID: 16436039 DOI: 10.1517/14712598.6.2.135] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The application of viral vectors for gene expression and delivery is rapidly evolving, with several entering clinical trials. However, a number of issues, including safety, gene expression levels, cell selectivity and antivector immunity, are driving the search for new vector systems. A number of replicon-based vectors derived from positive-strand RNA viruses have recently been developed, and this paper reviews the current knowledge on the first flavivirus replicon system, which is based on the Australian flavivirus Kunjin (KUN). Like most replicon systems, KUN replicons can be delivered as DNA, RNA or virus-like particles, they replicate their RNA in the cytoplasm and direct prolonged high-level gene expression. However, unlike most alphavirus replicon systems, KUN replicons are non-cytopathic, with transfected cells able to divide, allowing the establishment of cell lines stably expressing replicon RNA and heterologous genes. As vaccine vectors KUN replicons can induce potent, long-lived, protective, immunogen-specific CD8+ T cell immunity, a feature potentially related to extended production of antigen and double-stranded RNA-induced 'danger signals'. The identification of KUN replicon mutants that induce increased levels of IFN-alpha/beta has also spawned investigation of KUN replicons for use in cancer gene therapy. The unique characteristics of KUN replicons may thus make them suitable for specific protein production, vaccine and gene therapy applications.
Collapse
Affiliation(s)
- Gorben P Pijlman
- University of Queensland, School of Molecular and Microbial Sciences, MBS Bld 76, St. Lucia, 4072 QLD, Australia
| | | | | |
Collapse
|
28
|
Fayzulin R, Scholle F, Petrakova O, Frolov I, Mason PW. Evaluation of replicative capacity and genetic stability of West Nile virus replicons using highly efficient packaging cell lines. Virology 2006; 351:196-209. [PMID: 16647099 DOI: 10.1016/j.virol.2006.02.036] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Revised: 02/23/2006] [Accepted: 02/25/2006] [Indexed: 11/16/2022]
Abstract
A stable cell system for high-efficiency packaging of West Nile virus (WNV) subgenomic replicons into virus-like particles (VLPs) was developed. VLPs could be propagated on these packaging cells and produced infectious foci similar to foci produced by WNV. Focus size correlated with the replicative capacity of WNV replicons, indicating that genome copy number, rather than amount of trans-complementing structural proteins, was rate-limiting in packaging of VLPs. Comparison of VLP production from replicon genomes encoding partial or complete C genes indicated that portions of C downstream of the cyclization sequence could improve genome replication or that cis expression of C could enhance packaging. Interestingly, a rapid loss of replicon-encoded reporter gene activity was detected within two serial passages of reporter gene-containing VLPs. The loss of reporter activity correlated with gene deletion and better VLP growth, indicating a powerful selection pressure for WNV genomes lacking reporter genes.
Collapse
Affiliation(s)
- Rafik Fayzulin
- Department of Pathology, 3.206B Mary Moody Northen Pavilion, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0436, USA
| | | | | | | | | |
Collapse
|
29
|
Stone GW, Barzee S, Snarsky V, Kee K, Spina CA, Yu XF, Kornbluth RS. Multimeric soluble CD40 ligand and GITR ligand as adjuvants for human immunodeficiency virus DNA vaccines. J Virol 2006; 80:1762-72. [PMID: 16439533 PMCID: PMC1367159 DOI: 10.1128/jvi.80.4.1762-1772.2006] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
For use in humans, human immunodeficiency virus (HIV) DNA vaccines may need to include immunostimulatory adjuvant molecules. CD40 ligand (CD40L), a member of the tumor necrosis factor (TNF) superfamily (TNFSF), is one candidate adjuvant, but it has been difficult to use because it is normally expressed as a trimeric membrane molecule. Soluble trimeric forms of CD40L have been produced, but in vitro data indicate that multimeric, many-trimer forms of soluble CD40L are more active. This multimerization requirement was evaluated in mice using plasmids that encoded either 1-trimer, 2-trimer, or 4-trimer soluble forms of CD40L. Fusion with the body of Acrp30 was used to produce the 2-trimer form, and fusion with the body of surfactant protein D was used to produce the 4-trimer form. Using plasmids for secreted HIV-1 antigens Gag and Env, soluble CD40L was active as an adjuvant in direct proportion to the valence of the trimers (1 < 2 < 4). These CD40L-augmented DNA vaccines elicited strong CD8(+) T-cell responses but did not elicit significant CD4(+) T-cell or antibody responses. To test the applicability of the multimeric fusion protein approach to other TNFSFs, a 4-trimer construct for the ligand of glucocorticoid-induced TNF family-related receptor (GITR) was also prepared. Multimeric soluble GITR ligand (GITRL) augmented the CD8(+) T-cell, CD4(+) T-cell, and antibody responses to DNA vaccination. In summary, multimeric CD40L and GITRL are new adjuvants for DNA vaccines. Plasmids for expressing multimeric TNFSF fusion proteins permit the rapid testing of TNFSF molecules in vivo.
Collapse
Affiliation(s)
- Geoffrey W Stone
- Department of Medicine-0679, Stein Clinical Sciences Bldg., Room 304, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0679, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Yoshii K, Hayasaka D, Goto A, Kawakami K, Kariwa H, Takashima I. Packaging the replicon RNA of the Far-Eastern subtype of tick-borne encephalitis virus into single-round infectious particles: development of a heterologous gene delivery system. Vaccine 2005; 23:3946-56. [PMID: 15917116 DOI: 10.1016/j.vaccine.2005.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2004] [Revised: 01/31/2005] [Accepted: 03/02/2005] [Indexed: 10/25/2022]
Abstract
The sub-genomic replicon of tick-borne encephalitis (TBE) virus (Far-Eastern subtype) was packaged into infectious particles by providing the viral structural proteins in trans. Sequential transfection of TBE replicon RNA and a plasmid that expressed the structural proteins led to the secretion of infectious particles that contained TBE replicon RNA. The secreted particles had single-round infectivity, which was inhibited by TBE virus-neutralizing antibody. The physical structure of the particles was almost identical to that of infectious virions, and the packaged replicon RNA showed no recombination with the mRNAs of the viral structural proteins. Furthermore, heterologous genes were successfully delivered and expressed by packaging TBE replicon RNA with inserted GFP and Neo genes. This replicon packaging system may be a useful tool for the molecular study of the TBE virus genome packaging mechanism, and for the development of vaccine delivery systems.
Collapse
Affiliation(s)
- Kentarou Yoshii
- Laboratory of Public Health, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18 Nishi-9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Tscharke DC, Suhrbier A. From mice to humans – murine intelligence for human CD8+T cell vaccine design. Expert Opin Biol Ther 2005; 5:263-71. [PMID: 15757387 DOI: 10.1517/14712598.5.2.263] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
There has been considerable progress in the design of vaccines capable of safely and effectively inducing CD8(+) T cells for prophylaxis and treatment of chronic infectious diseases and cancer. Much of what is known about CD8(+) T cell-mediated immunity has come from pioneering work in mice; this broad overview discusses recent work in mouse systems where lessons may be drawn for human vaccine development. The areas highlighted include antivector immunity, immunodominance, dendritic cell biology and targeting, the role of Toll-like receptors and their exploitation by novel adjuvants, the role of CD4(+) T cell help, regulatory T cells and, finally, some comments on the different requirements of prophylactic versus therapeutic vaccines.
Collapse
Affiliation(s)
- David C Tscharke
- Queensland Institute of Medical Research, EBVBiology, P.O. Royal Brisbane Hospital, Brisbane, QLD 4029, Australia.
| | | |
Collapse
|
32
|
Gehrke R, Heinz FX, Davis NL, Mandl CW. Heterologous gene expression by infectious and replicon vectors derived from tick-borne encephalitis virus and direct comparison of this flavivirus system with an alphavirus replicon. J Gen Virol 2005; 86:1045-1053. [PMID: 15784898 DOI: 10.1099/vir.0.80677-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The flavivirus tick-borne encephaltis virus (TBEV) was established as a vector system for heterologous gene expression. The variable region of the genomic 3′ non-coding region was replaced by an expression cassette consisting of the reporter gene enhanced green fluorescent protein (EGFP) under the translational control of an internal ribosomal entry site element, both in the context of an infectious virus genome and of a replicon lacking the genes of the surface proteins prM/M and E. The expression level and the stability of expression were measured by fluorescence-activated cell-sorting analysis and compared to an established alphavirus replicon vector derived from Venezuelan equine encephaltis virus (VEEV), expressing EGFP under the control of its natural subgenomic promoter. On the first day, the alphavirus replicon exhibited an approximately 180-fold higher expression level than the flavivirus replicon, but this difference decreased to about 20- and 10-fold on days 2 and 3, respectively. Four to six days post-transfection, foreign gene expression by the VEEV replicon vanished almost completely, due to extensive cell killing. In contrast, in the case of the TBEV replicon, the percentage of positive cells and the amount of EGFP expression exhibited only a moderate decline over a time period of almost 4 weeks. The infectious TBEV vector expressed less EGFP than the TBEV replicon at all times. Significant expression from the infectious vector was maintained for four cell-culture passages. The results indicate that the VEEV vector is superior with respect to achieving high expression levels, but the TBEV system may be advantageous for applications that require a moderate, but more enduring, gene expression.
Collapse
Affiliation(s)
- Rainer Gehrke
- Institute of Virology, Medical University of Vienna, Kinderspitalgasse 15, A-1095 Vienna, Austria
| | - Franz X Heinz
- Institute of Virology, Medical University of Vienna, Kinderspitalgasse 15, A-1095 Vienna, Austria
| | - Nancy L Davis
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christian W Mandl
- Institute of Virology, Medical University of Vienna, Kinderspitalgasse 15, A-1095 Vienna, Austria
| |
Collapse
|
33
|
Jones CT, Patkar CG, Kuhn RJ. Construction and applications of yellow fever virus replicons. Virology 2005; 331:247-59. [PMID: 15629769 DOI: 10.1016/j.virol.2004.10.034] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2004] [Revised: 06/28/2004] [Accepted: 10/01/2004] [Indexed: 11/18/2022]
Abstract
Subgenomic replicons of yellow fever virus (YFV) were constructed to allow expression of heterologous reporter genes in a replication-dependent manner. Expression of the antibiotic resistance gene neomycin phosphotransferase II (Neo) from one of these YFV replicons allowed selection of a stable population of cells (BHK-REP cells) in which the YFV replicon persistently replicated. BHK-REP cells were successfully used to trans-complement replication-defective YFV replicons harboring large internal deletions within either the NS1 or NS3 proteins. Although replicons with large deletions in either NS1 or NS3 were trans-complemented in BHK-REP, replicons that contained deletions of NS3 were trans-complemented at lower levels. In addition, replicons that retained the N-terminal protease domain of NS3 in cis were trans-complemented with higher efficiency than replicons in which both the protease and helicase domains of NS3 were deleted. To study packaging of YFV replicons, Sindbis replicons were constructed that expressed the YFV structural proteins in trans. Using these Sindbis replicons, both replication-competent and trans-complemented, replication-defective YFV replicons could be packaged into pseudo-infectious particles (PIPs). Although these results eliminate a potential role of either NS1 or full-length NS3 in cis for packaging and assembly of the flavivirus virion, they do not preclude the possibility that these proteins may act in trans during these processes.
Collapse
Affiliation(s)
- Christopher T Jones
- Department of Biological Sciences, Purdue University, 915 W. State Street, West Lafayette, IN 47907-2054, USA
| | | | | |
Collapse
|
34
|
Malkevitch NV, Robert-Guroff M. A call for replicating vector prime-protein boost strategies in HIV vaccine design. Expert Rev Vaccines 2005; 3:S105-17. [PMID: 15285710 DOI: 10.1586/14760584.3.4.s105] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A key challenge to HIV vaccine development is the integration of HIV proviral DNA into the host genome upon infection. Therefore, an optimal vaccine should block infection within hours of viral exposure, providing 'sterilizing immunity' at mucosal sites and in blood via potent, broadly reactive antibody to the HIV envelope glycoprotein. This is difficult due to the envelope's conformational complexity and sequence diversity. Antibodies that do not completely prevent infection nevertheless could reduce the viral infectious burden, allowing strong cellular immunity to control viremia, delay disease progression and prevent viral transmission, while also providing help for T- and B-cell responses. Rapidly responsive, potent, persistent immunity might best be achieved using prime-boost strategies incorporating a replicating vector and an optimally designed envelope subunit.
Collapse
Affiliation(s)
- Nina V Malkevitch
- Section on Immune Biology of Retroviral Infection, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892 5065, USA.
| | | |
Collapse
|
35
|
Liu WJ, Chen HB, Wang XJ, Huang H, Khromykh AA. Analysis of adaptive mutations in Kunjin virus replicon RNA reveals a novel role for the flavivirus nonstructural protein NS2A in inhibition of beta interferon promoter-driven transcription. J Virol 2004; 78:12225-35. [PMID: 15507609 PMCID: PMC525072 DOI: 10.1128/jvi.78.22.12225-12235.2004] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The establishment of persistent noncytopathic replication by replicon RNAs of a number of positive-strand RNA viruses usually leads to generation of adaptive mutations in nonstructural genes. Some of these adaptive mutations (e.g., in hepatitis C virus) increase the ability of RNA replication to resist the antiviral action of alpha/beta interferon (IFN-alpha/beta); others (e.g., in Sindbis virus) may also lead to more efficient IFN production. Using puromycin-selectable Kunjin virus (KUN) replicon RNA, we identified two adaptive mutations in the NS2A gene (producing Ala30-to-Pro and Asn101-to-Asp mutations in the gene product; for simplicity, these will be referred to hereafter as Ala30-to-Pro and Asn101-to-Asp mutations) that, when introduced individually or together into the original wild-type (wt) replicon RNA, resulted in approximately 15- to 50-fold more efficient establishment of persistent replication in hamster (BHK21) and human (HEK293 and HEp-2) cell lines. Transfection with a reporter plasmid carrying the luciferase gene under the control of the IFN-beta promoter resulted in approximately 6- to 7-fold-higher luciferase expression in HEp-2 cells stably expressing KUN replicon RNA with an Ala30-to-Pro mutation in the NS2A gene compared to that observed in HEp-2 cells stably expressing KUN replicon RNA with the wt NS2A gene. Moreover, cotransfection of plasmids expressing individual wt or Ala30-to-Pro-mutated NS2A genes with the IFN-beta promoter reporter plasmid, followed by infection with Semliki Forest virus to activate IFN-beta promoter-driven transcription, showed approximately 7-fold inhibition of luciferase expression by the wt but not by the Ala30-to-Pro-mutated NS2A protein. The results show for the first time a role for the flavivirus nonstructural protein NS2A in inhibition of IFN-beta promoter-driven transcription and identify a single-amino-acid mutation in NS2A that dramatically reduces this inhibitory activity. The findings determine a new function for NS2A in virus-host interactions, extend the range of KUN replicon vectors for noncytopathic gene expression, and identify NS2A as a new target for attenuation in the development of live flavivirus vaccines.
Collapse
Affiliation(s)
- Wen Jun Liu
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital, Brisbane, Queensland 4029, Australia
| | | | | | | | | |
Collapse
|
36
|
Herd KA, Harvey T, Khromykh AA, Tindle RW. Recombinant Kunjin virus replicon vaccines induce protective T-cell immunity against human papillomavirus 16 E7-expressing tumour. Virology 2004; 319:237-48. [PMID: 14980484 DOI: 10.1016/j.virol.2003.10.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Revised: 08/28/2003] [Accepted: 10/23/2003] [Indexed: 10/26/2022]
Abstract
The persistence of the E7 oncoprotein in transformed cells in human papillomavirus (HPV)-associated cervical cancer provides a tumour-specific antigen to which immunotherapeutic strategies may be directed. Self-replicating RNA (replicon) vaccine vectors derived from the flavivirus Kunjin (KUN) have recently been reported to induce T-cell immunity. Here, we report that inclusion of a CTL epitope of HPV16 E7 protein into a polyepitope encoded by a KUN vector induced E7-directed T-cell responses and protected mice against challenge with an E7-expressing epithelial tumour. We found replicon RNA packaged into virus-like particles to be more effective than naked replicon RNA or plasmid DNA constructed to allow replicon RNA transcription in vivo. Protective immunity was induced although the E7 CTL epitope was subdominant in the context of other CTL epitopes in the polyepitope. The results demonstrate the efficacy of the KUN replicon vector system for inducing protective immunity directed towards a virally encoded human tumour-specific antigen, and for inducing multi-epitopic CTL responses.
Collapse
Affiliation(s)
- Karen A Herd
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital, and Clinical Medical Virology Centre, University of Queensland, Brisbane, Australia
| | | | | | | |
Collapse
|
37
|
Abstract
The Kunjin virus (KUNV) has provided a useful laboratory model for Flavivirus RNA replication. The synthesis of progeny RNA(+) strands occurs via asymmetric and semiconservative replication on a template of recycling double-stranded RNA (dsRna) or replicative form (RF). Kinetics of viral RNA synthesis indicated a cycle period of about 15 min during which, on average, a single nascent RNA (+) strand displaces the pre-existing RNA(+) strand in the replicative intermediate. Data on the composition of the replication complex (RC) in KUNV-infected cells were obtained from several sources, including analyses of the partially-purified still active RC, immunogold labeling of cryosections using monospecific antibodies to the nonstructural proteins and to the dsRNA, radioimmunoprecipitations of cell lysates using antibodies to dsRNA and to an RC-associated cell marker, and pull-down assays of cell lysates using fusion proteins GST-NS2A and GST-NS4A. These results yeilded a consensus composition of NS1, NS2A, NS3, NS4A, and NS5 strongly associated with the dsRNA template. The RC was located in induced membranes described as vesicle packets. The RNA-dependent RNA polymerase activity late in infection did not require continuing protein synthesis. Replication of genomic RNA was completely dependent on the presence of conserved complementary or cyclization sequences near the 5' and 3' ends. Assembly of the RC during translation in cis and the relationships, particularly those of NS1 and NS5 among the components, were deduced from an extensive set of complementation experiments in trans involving mutations/deletions in all the nonstructural proteins and use of KUN or alphahavirus replicons as helpers. The KUN replicon has found useful applications also as a noncytopathic vector for the continuing expression of foreign genes, delivered either as packaged RNA or as plasmid DNA.
Collapse
Affiliation(s)
- Edwin G Westaway
- Clinical Medical Virology Center-University of Queensland, Sir Albert Sakzewski Virus Research Center, Royal Children's Hospital, Herston, Brisbane, Australia
| | | | | |
Collapse
|
38
|
Harvey TJ, Liu WJ, Wang XJ, Linedale R, Jacobs M, Davidson A, Le TTT, Anraku I, Suhrbier A, Shi PY, Khromykh AA. Tetracycline-inducible packaging cell line for production of flavivirus replicon particles. J Virol 2004; 78:531-8. [PMID: 14671135 PMCID: PMC303381 DOI: 10.1128/jvi.78.1.531-538.2004] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2003] [Accepted: 09/19/2003] [Indexed: 01/23/2023] Open
Abstract
We have previously developed replicon vectors derived from the Australian flavivirus Kunjin that have a unique noncytopathic nature and have been shown to direct prolonged high-level expression of encoded heterologous genes in vitro and in vivo and to induce strong and long-lasting immune responses to encoded immunogens in mice. To facilitate further applications of these vectors in the form of virus-like particles (VLPs), we have now generated a stable BHK packaging cell line, tetKUNCprME, carrying a Kunjin structural gene cassette under the control of a tetracycline-inducible promoter. Withdrawal of tetracycline from the medium resulted in production of Kunjin structural proteins that were capable of packaging transfected and self-amplified Kunjin replicon RNA into the secreted VLPs at titers of up to 1.6 x 10(9) VLPs per ml. Furthermore, secreted KUN replicon VLPs from tetKUNCprME cells could be harvested continuously for as long as 10 days after RNA transfection, producing a total yield of more than 10(10) VLPs per 10(6) transfected cells. Passaging of VLPs on Vero cells or intracerebral injection into 2- to 4-day-old suckling mice illustrated the complete absence of any infectious Kunjin virus. tetKUNCprME cells were also capable of packaging replicon RNA from closely and distantly related flaviviruses, West Nile virus and dengue virus type 2, respectively. The utility of high-titer KUN replicon VLPs was demonstrated by showing increasing CD8(+)-T-cell responses to encoded foreign protein with increasing doses of KUN VLPs. A single dose of 2.5 x 10(7) VLPs carrying the human respiratory syncytial virus M2 gene induced 1,400 CD8 T cells per 10(6) splenocytes in an ex vivo gamma interferon enzyme-linked immunospot assay. The packaging cell line thus represents a significant advance in the development of the noncytopathic Kunjin virus replicon-based gene expression system and may be widely applicable to the basic studies of flavivirus RNA packaging and virus assembly as well as to the development of gene expression systems based on replicons from different flaviviruses.
Collapse
Affiliation(s)
- Tracey J Harvey
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|