Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (35)

Search Parameters:
Keywords = RNA replicon vaccines

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 1511 KB  
Review
The Interplay Between Therapeutic Self-Amplifying RNA and the Innate Immune System: Balancing Efficiency and Reactogenicity
by Dmitry Kunyk, Marina Plotnikova, Mikhail Bespalov, Daniil Shevyrev, Sergey Klotchenko, Roman Ivanov and Vasiliy Reshetnikov
Int. J. Mol. Sci. 2025, 26(18), 8986; https://doi.org/10.3390/ijms26188986 - 15 Sep 2025
Viewed by 1103
Abstract
Self-amplifying RNA (saRNA) is a promising platform for the production of vaccines, anti-tumor therapeutics, and gene therapy solutions. One of the advantages of the saRNA platform is the ability to use small doses of the therapeutic while maintaining prolonged expression of the target [...] Read more.
Self-amplifying RNA (saRNA) is a promising platform for the production of vaccines, anti-tumor therapeutics, and gene therapy solutions. One of the advantages of the saRNA platform is the ability to use small doses of the therapeutic while maintaining prolonged expression of the target protein. However, the presence of auxiliary sequences encoding non-structural alphavirus proteins, which facilitate the replication of saRNA in cells, necessitates a thorough assessment of the biosafety of this platform. In our review, we focus on saRNA functions in the context of its interaction with the innate immune system. Firstly, an analysis is conducted of the side effects of candidate saRNA therapeutics, as observed in preclinical and clinical trials. Then, the mechanisms underlying the function of saRNA products derived from various alphavirus genomes in cell systems are discussed, as well as the reasons for their reactogenicity. The key approaches to optimizing the saRNA platform, which are aimed at reducing the activation of the innate immune response and cytopathic effects, are described. To summarize, this review enables us to systematize our knowledge on the advantages and disadvantages of saRNA, as well as potential approaches to improving this platform in order to develop more effective and safer therapeutics. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

14 pages, 944 KB  
Article
A Novel, Safe, Non-Adjuvanted Alphavirus Replicon-Based Vaccine Expressing the Feline Leukemia Virus Envelope Protein Protects Against Virulent FeLV Challenge
by Kari Carritt, Randall Davis, Ken Stachura, Paige Crumley, Mark Mogler, Madeleine Stahl, Lijuan Deng, Zach Xu and Ian Tarpey
Vaccines 2025, 13(7), 697; https://doi.org/10.3390/vaccines13070697 - 27 Jun 2025
Viewed by 1865
Abstract
Background/Objectives: A number of different vaccines against feline leukemia virus (FeLV) are available; however, there is continuous debate regarding the efficacy advantages of adjuvanted vaccines versus the potential safety advantages of non-adjuvanted vaccines. Methods: For this reason, we developed a non-adjuvanted vaccine based [...] Read more.
Background/Objectives: A number of different vaccines against feline leukemia virus (FeLV) are available; however, there is continuous debate regarding the efficacy advantages of adjuvanted vaccines versus the potential safety advantages of non-adjuvanted vaccines. Methods: For this reason, we developed a non-adjuvanted vaccine based on a replicon RNA particle (RP) expressing the FeLV gp85 envelope protein, which possesses the safety of a non-adjuvanted vaccine while consistently providing high efficacy. Results: In two efficacy studies, a high-level of protection against virulent FeLV challenge was demonstrated with two doses given 3 weeks apart based on the prevention of FeLV p27 antigenemia. Furthermore, in both studies, we compared this novel vaccine against a non-adjuvanted, canarypox-vectored FeLV vaccine, demonstrating that none of the cats that received two doses of the RP-FeLV vaccine developed persistent antigenemia post-challenge. In comparison, of cats receiving the canarypox-vectored FeLV vaccine, three of seven (43%) became persistently antigenemic in one study, and three of ten (30%) became persistently antigenemic in the other study. In a field safety study using two commercial serials, safety of the RP-FeLV vaccine was demonstrated in over 800 cats receiving two doses of the vaccine. Conclusions: These data suggest that the RP-FeLV vaccine offers advantages over some current FeLV vaccines by combining the safety profile of a non-adjuvanted vaccine with the induction of a robust immune response demonstrated by some adjuvanted vaccines. Full article
(This article belongs to the Section Veterinary Vaccines)
Show Figures

Figure 1

19 pages, 3264 KB  
Article
Sindbis Virus Replicon-Based SARS-CoV-2 and Dengue Combined Vaccine Candidates Elicit Immune Responses and Provide Protective Immunity in Mice
by Yihan Zhu, Wenfeng He, Rui Hu, Xiahua Liu, Mengzhu Li and Yuan Liu
Vaccines 2024, 12(11), 1292; https://doi.org/10.3390/vaccines12111292 - 19 Nov 2024
Viewed by 2129
Abstract
Background/Objectives: Since its emergence in 2019, the rapid spread of SARS-CoV-2 led to the global pandemic. Recent large-scale dengue fever outbreaks overlapped with the COVID-19 pandemic, leading to increased cases of co-infection and posing severe public health risks. Accordingly, the development of [...] Read more.
Background/Objectives: Since its emergence in 2019, the rapid spread of SARS-CoV-2 led to the global pandemic. Recent large-scale dengue fever outbreaks overlapped with the COVID-19 pandemic, leading to increased cases of co-infection and posing severe public health risks. Accordingly, the development of effective combined SARS-CoV-2 and dengue virus (DENV) vaccines is necessary to control the spread and prevalence of both viruses. Methods: In this study, we designed Sindbis virus (SINV) replicon-based SARS-CoV-2 and DENV chimeric vaccines using two delivery strategies: DNA-launched self-replicating RNA replicon (DREP) and viral replicon particle (VRP) systems. Results: Cellular and animal experiments confirmed that the vaccines effectively produced viral proteins and elicited strong immunogenicity. These vaccines induced robust immune responses and neutralizing activity against live SARS-CoV-2, DENV1, and DENV2 viruses. In addition, passively transferred sera from BALB/c mice immunized with these vaccines into AG129 mice provided significant protection against lethal DENV2 challenge. The transferred sera protected the mice from physical symptoms, reduced viral loads in the kidney, spleen, liver, and intestine, and prevented DENV2-induced vascular leakage in these tissues. Conclusions: Therefore, combined vaccines based on the SINV replicon system are promising candidates for pandemic control. These results lay a foundation for further development of a safe and effective combination vaccine against SARS-CoV-2 and DENV. Full article
(This article belongs to the Section COVID-19 Vaccines and Vaccination)
Show Figures

Figure 1

14 pages, 501 KB  
Review
Self-Replicating Alphaviruses: From Pathogens to Therapeutic Agents
by Kenneth Lundstrom
Viruses 2024, 16(11), 1762; https://doi.org/10.3390/v16111762 - 12 Nov 2024
Cited by 3 | Viewed by 2707
Abstract
Alphaviruses are known for being model viruses for studying cellular functions related to viral infections but also for causing epidemics in different parts of the world. More recently, alphavirus-based expression systems have demonstrated efficacy as vaccines against infectious diseases and as therapeutic applications [...] Read more.
Alphaviruses are known for being model viruses for studying cellular functions related to viral infections but also for causing epidemics in different parts of the world. More recently, alphavirus-based expression systems have demonstrated efficacy as vaccines against infectious diseases and as therapeutic applications for different cancers. Point mutations in the non-structural alphaviral replicase genes have generated enhanced transgene expression and created temperature-sensitive expression vectors. The recently engineered trans-amplifying RNA system can provide higher translational efficiency and eliminate interference with cellular translation. The self-replicating feature of alphaviruses has provided the advantage of extremely high transgene expression of vaccine-related antigens and therapeutic anti-tumor and immunostimulatory genes, which has also permitted significantly reduced doses for prophylactic and therapeutic applications, potentially reducing adverse events. Furthermore, alphaviruses have shown favorable flexibility as they can be delivered as recombinant viral particles, RNA replicons, or DNA-replicon-based plasmids. In the context of infectious diseases, robust immune responses against the surface proteins of target agents have been observed along with protection against challenges with lethal doses of infectious agents in rodents and primates. Similarly, the expression of anti-tumor genes and immunostimulatory genes from alphavirus vectors has provided tumor growth inhibition, tumor regression, and cures in animal cancer models. Moreover, protection against tumor challenges has been observed. In clinical settings, patient benefits have been reported. Alphaviruses have also been considered for the treatment of neurological disorders due to their neurotrophic preference. Full article
(This article belongs to the Special Issue Self-Replicating RNA Viruses)
Show Figures

Figure 1

15 pages, 2203 KB  
Article
A Lipid Nanoparticle-Formulated Self-Amplifying RNA Rift Valley Fever Vaccine Induces a Robust Humoral Immune Response in Mice
by Paul K. Kitandwe, Paul Rogers, Kai Hu, Owen Nayebare, Anna K. Blakney, Paul F. McKay, Pontiano Kaleebu and Robin J. Shattock
Vaccines 2024, 12(10), 1088; https://doi.org/10.3390/vaccines12101088 - 24 Sep 2024
Cited by 2 | Viewed by 3131
Abstract
Rift Valley fever (RVF) is a mosquito-borne viral zoonosis that causes high fetal and neonatal mortality rates in ruminants and sometimes severe to fatal complications like encephalitis and hemorrhagic fever in humans. There is no licensed RVF vaccine for human use while approved [...] Read more.
Rift Valley fever (RVF) is a mosquito-borne viral zoonosis that causes high fetal and neonatal mortality rates in ruminants and sometimes severe to fatal complications like encephalitis and hemorrhagic fever in humans. There is no licensed RVF vaccine for human use while approved livestock vaccines have suboptimal safety or efficacy. We designed self-amplifying RNA (saRNA) RVF vaccines and assessed their humoral immunogenicity in mice. Plasmid DNA encoding the Rift Valley fever virus (RVFV) medium (M) segment consensus sequence (WT consensus) and its derivatives mutated to enhance cell membrane expression of the viral surface glycoproteins n (Gn) and c (Gc) were assessed for in vitro expression. The WT consensus and best-expressing derivative (furin-T2A) were cloned into a Venezuelan equine encephalitis virus (VEEV) plasmid DNA replicon and in vitro transcribed into saRNA. The saRNA was formulated in lipid nanoparticles and its humoral immunogenicity in BALB/c mice was assessed. High quantities of dose-dependent RVFV Gn IgG antibodies were detected in the serum of all mice immunized with either WT consensus or furin-T2A saRNA RVF vaccines. Significant RVFV pseudovirus-neutralizing activity was induced in mice immunized with 1 µg or 10 µg of the WT consensus saRNA vaccine. The WT consensus saRNA RVF vaccine warrants further development. Full article
Show Figures

Figure 1

13 pages, 724 KB  
Article
Vaccine Platform Comparison: Protective Efficacy against Lethal Marburg Virus Challenge in the Hamster Model
by Kyle L. O’Donnell, Corey W. Henderson, Hanna Anhalt, Joan Fusco, Jesse H. Erasmus, Teresa Lambe and Andrea Marzi
Int. J. Mol. Sci. 2024, 25(15), 8516; https://doi.org/10.3390/ijms25158516 - 5 Aug 2024
Cited by 4 | Viewed by 2389
Abstract
Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 [...] Read more.
Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions. Full article
(This article belongs to the Special Issue Immunopathology, Vaccine Development and Treatment of Viruses)
Show Figures

Figure 1

17 pages, 1407 KB  
Review
Trans-Amplifying RNA: A Journey from Alphavirus Research to Future Vaccines
by Ayşegül Yıldız, Cristian Răileanu and Tim Beissert
Viruses 2024, 16(4), 503; https://doi.org/10.3390/v16040503 - 25 Mar 2024
Cited by 10 | Viewed by 7277
Abstract
Replicating RNA, including self-amplifying RNA (saRNA) and trans-amplifying RNA (taRNA), holds great potential for advancing the next generation of RNA-based vaccines. Unlike in vitro transcribed mRNA found in most current RNA vaccines, saRNA or taRNA can be massively replicated within cells in the [...] Read more.
Replicating RNA, including self-amplifying RNA (saRNA) and trans-amplifying RNA (taRNA), holds great potential for advancing the next generation of RNA-based vaccines. Unlike in vitro transcribed mRNA found in most current RNA vaccines, saRNA or taRNA can be massively replicated within cells in the presence of RNA-amplifying enzymes known as replicases. We recently demonstrated that this property could enhance immune responses with minimal injected RNA amounts. In saRNA-based vaccines, replicase and antigens are encoded on the same mRNA molecule, resulting in very long RNA sequences, which poses significant challenges in production, delivery, and stability. In taRNA-based vaccines, these challenges can be overcome by splitting the replication system into two parts: one that encodes replicase and the other that encodes a short antigen-encoding RNA called transreplicon. Here, we review the identification and use of transreplicon RNA in alphavirus research, with a focus on the development of novel taRNA technology as a state-of-the art vaccine platform. Additionally, we discuss remaining challenges essential to the clinical application and highlight the potential benefits related to the unique properties of this future vaccine platform. Full article
(This article belongs to the Special Issue Self-Replicating RNA Viruses)
Show Figures

Figure 1

23 pages, 865 KB  
Review
RNA Combined with Nanoformulation to Advance Therapeutic Technologies
by Eduarda Santos Lima, Déborah dos Santos, Atena Liriel Souza, Maria Eduarda Macedo, Mariana Evangelista Bandeira, Sérgio Santos Silva Junior, Bianca Sampaio Dotto Fiuza, Vinicius Pinto Costa Rocha, Larissa Moraes dos Santos Fonseca, Danielle Devequi Gomes Nunes, Katharine Valéria Saraiva Hodel and Bruna Aparecida Souza Machado
Pharmaceuticals 2023, 16(12), 1634; https://doi.org/10.3390/ph16121634 - 21 Nov 2023
Cited by 11 | Viewed by 3545
Abstract
Nucleic acid-based therapies have the potential to address numerous diseases that pose significant challenges to more traditional methods. RNA-based therapies have emerged as a promising avenue, utilizing nanoformulation treatments to target a range of pathologies. Nanoformulation offers several advantages compared to other treatment [...] Read more.
Nucleic acid-based therapies have the potential to address numerous diseases that pose significant challenges to more traditional methods. RNA-based therapies have emerged as a promising avenue, utilizing nanoformulation treatments to target a range of pathologies. Nanoformulation offers several advantages compared to other treatment modalities, including targeted delivery, low toxicity, and bioactivity suitable for drug loading. At present, various types of nanoformulations are available, such as liposomes, polymeric nanoparticles (NPs), magnetic NPs, nanoshells, and solid lipid nanoparticles (SLNs). RNA-based therapy utilizes intracellular gene nanoparticles with messenger RNA (mRNA) emerging prominently in cancer therapy and immunotechnology against infectious diseases. The approval of mRNA-based technology opens doors for future technological advancements, particularly self-amplifying replicon RNA (repRNA). RepRNA is a novel platform in gene therapy, comprising viral RNA with a unique molecular property that enables the amplification of all encoded genetic information countless times. As a result, repRNA-based therapies have achieved significant levels of gene expression. In this context, the primary objective of this study is to furnish a comprehensive review of repRNA and its applications in nanoformulation treatments, with a specific focus on encapsulated nanoparticles. The overarching goal is to provide an extensive overview of the use of repRNA in conjunction with nanoformulations across a range of treatments and therapies. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

17 pages, 2859 KB  
Article
Establishment of an Antiplasmodial Vaccine Based on PfRH5-Encoding RNA Replicons Stabilized by Cationic Liposomes
by Wesley L. Fotoran, Jamile Ramos da Silva, Christiane Glitz, Luís Carlos de Souza Ferreira and Gerhard Wunderlich
Pharmaceutics 2023, 15(4), 1223; https://doi.org/10.3390/pharmaceutics15041223 - 12 Apr 2023
Cited by 8 | Viewed by 3375
Abstract
Background: Nucleic acid-based vaccines have been studied for the past four decades, but the approval of the first messenger RNA (mRNA) vaccines during the COVID-19 pandemic opened renewed perspectives for the development of similar vaccines against different infectious diseases. Presently available mRNA vaccines [...] Read more.
Background: Nucleic acid-based vaccines have been studied for the past four decades, but the approval of the first messenger RNA (mRNA) vaccines during the COVID-19 pandemic opened renewed perspectives for the development of similar vaccines against different infectious diseases. Presently available mRNA vaccines are based on non-replicative mRNA, which contains modified nucleosides encased in lipid vesicles, allowing for entry into the host cell cytoplasm, and reducing inflammatory reactions. An alternative immunization strategy employs self-amplifying mRNA (samRNA) derived from alphaviruses, but lacks viral structural genes. Once incorporated into ionizable lipid shells, these vaccines lead to enhanced gene expression, and lower mRNA doses are required to induce protective immune responses. In the present study, we tested a samRNA vaccine formulation based on the SP6 Venezuelan equine encephalitis (VEE) vector incorporated into cationic liposomes (dimethyldioctadecyl ammonium bromide and a cholesterol derivative). Three vaccines were generated that encoded two reporter genes (GFP and nanoLuc) and the Plasmodium falciparum reticulocyte binding protein homologue 5 (PfRH5). Methods: Transfection assays were performed using Vero and HEK293T cells, and the mice were immunized via the intradermal route using a tattooing device. Results: The liposome–replicon complexes showed high transfection efficiencies with in vitro cultured cells, whereas tattooing immunization with GFP-encoding replicons demonstrated gene expression in mouse skin up to 48 h after immunization. Mice immunized with liposomal PfRH5-encoding RNA replicons elicited antibodies that recognized the native protein expressed in P. falciparum schizont extracts, and inhibited the growth of the parasite in vitro. Conclusion: Intradermal delivery of cationic lipid-encapsulated samRNA constructs is a feasible approach for developing future malaria vaccines. Full article
Show Figures

Figure 1

12 pages, 779 KB  
Review
Application of DNA Replicons in Gene Therapy and Vaccine Development
by Kenneth Lundstrom
Pharmaceutics 2023, 15(3), 947; https://doi.org/10.3390/pharmaceutics15030947 - 15 Mar 2023
Cited by 7 | Viewed by 3830
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue Plasmid DNA for Gene Therapy and DNA Vaccine Applications)
Show Figures

Figure 1

17 pages, 3063 KB  
Article
Efficient CRISPR-Cas13d-Based Antiviral Strategy to Combat SARS-CoV-2
by Mouraya Hussein, Zaria Andrade dos Ramos, Monique A. Vink, Pascal Kroon, Zhenghao Yu, Luis Enjuanes, Sonia Zuñiga, Ben Berkhout and Elena Herrera-Carrillo
Viruses 2023, 15(3), 686; https://doi.org/10.3390/v15030686 - 6 Mar 2023
Cited by 14 | Viewed by 4378
Abstract
The current SARS-CoV-2 pandemic forms a major global health burden. Although protective vaccines are available, concerns remain as new virus variants continue to appear. CRISPR-based gene-editing approaches offer an attractive therapeutic strategy as the CRISPR-RNA (crRNA) can be adjusted rapidly to accommodate a [...] Read more.
The current SARS-CoV-2 pandemic forms a major global health burden. Although protective vaccines are available, concerns remain as new virus variants continue to appear. CRISPR-based gene-editing approaches offer an attractive therapeutic strategy as the CRISPR-RNA (crRNA) can be adjusted rapidly to accommodate a new viral genome sequence. This study aimed at using the RNA-targeting CRISPR-Cas13d system to attack highly conserved sequences in the viral RNA genome, thereby preparing for future zoonotic outbreaks of other coronaviruses. We designed 29 crRNAs targeting highly conserved sequences along the complete SARS-CoV-2 genome. Several crRNAs demonstrated efficient silencing of a reporter with the matching viral target sequence and efficient inhibition of a SARS-CoV-2 replicon. The crRNAs that suppress SARS-CoV-2 were also able to suppress SARS-CoV, thus demonstrating the breadth of this antiviral strategy. Strikingly, we observed that only crRNAs directed against the plus-genomic RNA demonstrated antiviral activity in the replicon assay, in contrast to those that bind the minus-genomic RNA, the replication intermediate. These results point to a major difference in the vulnerability and biology of the +RNA versus −RNA strands of the SARS-CoV-2 genome and provide important insights for the design of RNA-targeting antivirals. Full article
(This article belongs to the Special Issue Advances in Antiviral Agents against SARS-CoV-2 and Its Variants)
Show Figures

Figure 1

13 pages, 501 KB  
Review
Therapeutic Applications for Oncolytic Self-Replicating RNA Viruses
by Kenneth Lundstrom
Int. J. Mol. Sci. 2022, 23(24), 15622; https://doi.org/10.3390/ijms232415622 - 9 Dec 2022
Cited by 12 | Viewed by 3830
Abstract
Self-replicating RNA viruses have become attractive delivery vehicles for therapeutic applications. They are easy to handle, can be rapidly produced in large quantities, and can be delivered as recombinant viral particles, naked or nanoparticle-encapsulated RNA, or plasmid DNA-based vectors. The self-replication of RNA [...] Read more.
Self-replicating RNA viruses have become attractive delivery vehicles for therapeutic applications. They are easy to handle, can be rapidly produced in large quantities, and can be delivered as recombinant viral particles, naked or nanoparticle-encapsulated RNA, or plasmid DNA-based vectors. The self-replication of RNA in infected host cells provides the means for generating much higher transgene expression levels and the possibility to apply substantially reduced amounts of RNA to achieve similar expression levels or immune responses compared to conventional synthetic mRNA. Alphaviruses and flaviviruses, possessing a single-stranded RNA genome of positive polarity, as well as measles viruses and rhabdoviruses with a negative-stranded RNA genome, have frequently been utilized for therapeutic applications. Both naturally and engineered oncolytic self-replicating RNA viruses providing specific replication in tumor cells have been evaluated for cancer therapy. Therapeutic efficacy has been demonstrated in animal models. Furthermore, the safe application of oncolytic viruses has been confirmed in clinical trials. Multiple myeloma patients treated with an oncolytic measles virus (MV-NIS) resulted in increased T-cell responses against the measles virus and several tumor-associated antigen responses and complete remission in one patient. Furthermore, MV-CEA administration to patients with ovarian cancer resulted in a stable disease and more than doubled the median overall survival. Full article
Show Figures

Figure 1

13 pages, 1021 KB  
Review
Alphaviruses in Immunotherapy and Anticancer Therapy
by Kenneth Lundstrom
Biomedicines 2022, 10(9), 2263; https://doi.org/10.3390/biomedicines10092263 - 13 Sep 2022
Cited by 9 | Viewed by 3128
Abstract
Alphaviruses have been engineered as expression vectors for vaccine development and gene therapy. Due to the feature of RNA self-replication, alphaviruses can provide exceptional direct cytoplasmic expression of transgenes based on the delivery of recombinant particles, naked or nanoparticle-encapsulated RNA or plasmid-based DNA [...] Read more.
Alphaviruses have been engineered as expression vectors for vaccine development and gene therapy. Due to the feature of RNA self-replication, alphaviruses can provide exceptional direct cytoplasmic expression of transgenes based on the delivery of recombinant particles, naked or nanoparticle-encapsulated RNA or plasmid-based DNA replicons. Alphavirus vectors have been utilized for the expression of various antigens targeting different types of cancers, and cytotoxic and antitumor genes. The most common alphavirus vectors are based on the Semliki Forest virus, Sindbis virus and Venezuelan equine encephalitis virus, but the oncolytic M1 alphavirus has also been used. Delivery of immunostimulatory cytokine genes has been the basis for immunotherapy demonstrating efficacy in different animal tumor models for brain, breast, cervical, colon, lung, ovarian, pancreatic, prostate and skin cancers. Typically, therapeutic effects including tumor regression, tumor eradication and complete cure as well as protection against tumor challenges have been observed. Alphavirus vectors have also been subjected to clinical evaluations. For example, therapeutic responses in all cervical cancer patients treated with an alphavirus vector expressing the human papilloma virus E6 and E7 envelope proteins have been achieved. Full article
(This article belongs to the Special Issue Oncolytic Viruses as a Novel Form of Immunotherapy for Cancer III)
Show Figures

Figure 1

15 pages, 3695 KB  
Article
A Bivalent Trans-Amplifying RNA Vaccine Candidate Induces Potent Chikungunya and Ross River Virus Specific Immune Responses
by Christin Schmidt, Florian D. Hastert, Julia Gerbeth, Tim Beissert, Ugur Sahin, Mario Perkovic and Barbara S. Schnierle
Vaccines 2022, 10(9), 1374; https://doi.org/10.3390/vaccines10091374 - 23 Aug 2022
Cited by 17 | Viewed by 4637
Abstract
Alphaviruses such as the human pathogenic chikungunya virus (CHIKV) and Ross River virus (RRV) can cause explosive outbreaks raising public health concerns. However, no vaccine or specific antiviral treatment is yet available. We recently established a CHIKV vaccine candidate based on trans-amplifying RNA [...] Read more.
Alphaviruses such as the human pathogenic chikungunya virus (CHIKV) and Ross River virus (RRV) can cause explosive outbreaks raising public health concerns. However, no vaccine or specific antiviral treatment is yet available. We recently established a CHIKV vaccine candidate based on trans-amplifying RNA (taRNA). This novel system consists of a replicase-encoding mRNA and a trans-replicon (TR) RNA encoding the antigen. The TR-RNA is amplified by the replicase in situ. We were interested in determining whether multiple TR-RNAs can be amplified in parallel and if, thus, a multivalent vaccine candidate can be generated. In vitro, we observed an efficient amplification of two TR-RNAs, encoding for the CHIKV and the RRV envelope proteins, by the replicase, which resulted in a high antigen expression. Vaccination of BALB/c mice with the two TR-RNAs induced CHIKV- and RRV-specific humoral and cellular immune responses. However, antibody titers and neutralization capacity were higher after immunization with a single TR-RNA. In contrast, alphavirus-specific T cell responses were equally potent after the bivalent vaccination. These data show the proof-of-principle that the taRNA system can be used to generate multivalent vaccines; however, further optimizations will be needed for clinical application. Full article
(This article belongs to the Special Issue mRNA Vaccine Development against Tropical Diseases)
Show Figures

Figure 1

13 pages, 3956 KB  
Article
Evaluation of Potential Anti-Hepatitis A Virus 3C Protease Inhibitors Using Molecular Docking
by Reina Sasaki-Tanaka, Kalyan C. Nagulapalli Venkata, Hiroaki Okamoto, Mitsuhiko Moriyama and Tatsuo Kanda
Int. J. Mol. Sci. 2022, 23(11), 6044; https://doi.org/10.3390/ijms23116044 - 27 May 2022
Cited by 8 | Viewed by 3121
Abstract
Hepatitis A virus (HAV) infection is a major cause of acute hepatitis worldwide and occasionally causes acute liver failure and can lead to death in the absence of liver transplantation. Although HAV vaccination is available, the prevalence of HAV vaccination is not adequate [...] Read more.
Hepatitis A virus (HAV) infection is a major cause of acute hepatitis worldwide and occasionally causes acute liver failure and can lead to death in the absence of liver transplantation. Although HAV vaccination is available, the prevalence of HAV vaccination is not adequate in some countries. Additionally, the improvements in public health reduced our immunity to HAV infection. These situations motivated us to develop potentially new anti-HAV therapeutic options. We carried out the in silico screening of anti-HAV compounds targeting the 3C protease enzyme using the Schrodinger Modeling software from the antiviral library of 25,000 compounds to evaluate anti-HAV 3C protease inhibitors. Additionally, in vitro studies were introduced to examine the inhibitory effects of HAV subgenomic replicon replication and HAV HA11-1299 genotype IIIA replication in hepatoma cell lines using luciferase assays and real-time RT-PCR. In silico studies enabled us to identify five lead candidates with optimal binding interactions in the active site of the target HAV 3C protease using the Schrodinger Glide program. In vitro studies substantiated our hypothesis from in silico findings. One of our lead compounds, Z10325150, showed 47% inhibitory effects on HAV genotype IB subgenomic replicon replication and 36% inhibitory effects on HAV genotype IIIA HA11-1299 replication in human hepatoma cell lines, with no cytotoxic effects at concentrations of 100 μg/mL. The effects of the combination therapy of Z10325150 and RNA-dependent RNA polymerase inhibitor, favipiravir on HAV genotype IB HM175 subgenomic replicon replication and HAV genotype IIIA HA11-1299 replication showed 64% and 48% inhibitory effects of HAV subgenomic replicon and HAV replication, respectively. We identified the HAV 3C protease inhibitor Z10325150 through in silico screening and confirmed the HAV replication inhibitory activity in human hepatocytes. Z10325150 may offer the potential for a useful HAV inhibitor in severe hepatitis A. Full article
(This article belongs to the Special Issue Hepatitis Virus Infection and Molecular Research 2022)
Show Figures

Figure 1

Back to TopTop