Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (294)

Search Parameters:
Keywords = non-replicating vaccines

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 4477 KB  
Article
Unraveling the Potential Antiviral Activity of Isoxazoline-Carbocyclic Monophosphate Nucleotides Against Echovirus 11
by Emilia Palazzotto, Valeria Stefanizzi, Floriana Bonura, Federica Cacioppo, Marco Leusciatti, Paolo Quadrelli, Annalisa Chianese, Carla Zannella, Anna De Filippis, Antonio Mastino, Francesca Marino Merlo and Simona De Grazia
Microorganisms 2025, 13(12), 2662; https://doi.org/10.3390/microorganisms13122662 - 23 Nov 2025
Viewed by 288
Abstract
From July 2022, a novel Echovirus 11 (E11) variant has been associated with severe neonatal infections and liver failure. Currently, there are no vaccines or antiviral options for the targeted treatment of non-polio enterovirus (EV) infections; therefore, anti-EV drugs are urgently needed. In [...] Read more.
From July 2022, a novel Echovirus 11 (E11) variant has been associated with severe neonatal infections and liver failure. Currently, there are no vaccines or antiviral options for the targeted treatment of non-polio enterovirus (EV) infections; therefore, anti-EV drugs are urgently needed. In this study, the putative anti-E11 activity of two isoxzoline-carbocyclic monophosphate nucleotides (4a and 4b) was evaluated in vitro by cytopathic effect (CPE) reduction in VERO 76 cells and qRT-PCR. Treatment with nucleotide 4a at 25 and 50 μM successfully diminished the CPE caused by E11 by 90% and 75%, respectively, and induced a reduction in viral RNA in the supernatant by 72% and 89%. In contrast, the treatment with 25 and 50 μM of 4b caused a minor inhibition of CPE (58 and 38%), and no significant E11 RNA level changes were observed. A time course viral progeny production assay was performed to assess the inhibitory effect of nucleotide 4a on E11 infection progression. Compared to the control, the treated group showed a significant drop in viral RNA levels, with reductions of 43% at 10 h, 65% at 24 h, and 96% at 48 h post-infection. The results showed the extensive antiviral properties of the monophosphate nucleotide 4a in vitro. Moreover, a retrospective molecular docking study strongly supports that nucleotide 4a is an RdRp inhibitor capable of decreasing E11 genome replication and virus particle formation in VERO 76 cells. Full article
(This article belongs to the Section Virology)
Show Figures

Figure 1

20 pages, 353 KB  
Review
Mpox: Exploring Epidemiology, Disease Outcomes, and Preventative Vaccination Among People with HIV During the Ongoing Outbreaks
by Chloe Orkin, Ralph Torgler, Rebecca Dawson, Ian W. Holloway and Christian Hoffmann
Viruses 2025, 17(12), 1526; https://doi.org/10.3390/v17121526 - 21 Nov 2025
Viewed by 509
Abstract
Human mpox, caused by the mpox virus, is a reemerging viral zoonosis that has gained global attention due to recent Clade IIb outbreaks outside of Africa, as well as ongoing Clade Ia and Ib outbreaks in the Democratic Republic of Congo (DRC) and [...] Read more.
Human mpox, caused by the mpox virus, is a reemerging viral zoonosis that has gained global attention due to recent Clade IIb outbreaks outside of Africa, as well as ongoing Clade Ia and Ib outbreaks in the Democratic Republic of Congo (DRC) and surrounding regions. Since the start of these outbreaks in 2022, approximately 160,000 people have been affected across more than 100 countries. People with human immunodeficiency virus (HIV; hereafter referred to as PWH) have been disproportionately affected, accounting for approximately 50% of all cases. Mpox is typically a self-limiting illness causing smallpox-like symptoms lasting 2–4 weeks, which can cause significant pain and morbidity. People with uncontrolled or advanced HIV face an elevated risk of severe mpox, secondary complications, and worse outcomes. Vaccination with second- and third-generation vaccinia-based smallpox vaccines has emerged as an important tool in mpox prevention, alongside behavioural modification to mitigate risk. However, only the third-generation, live-attenuated, non-replicating vaccine, modified vaccinia Ankara (MVA-BN [Bavarian Nordic]), is approved for use in PWH. Real-world estimates suggest that two doses of MVA-BN administered as pre-exposure prophylaxis confers vaccine effectiveness in the range of 66–90%. Additionally, MVA-BN has been widely demonstrated to have an acceptable safety profile. This narrative review explores the changing epidemiology, clinical manifestations, and outcomes of mpox in PWH. We also summarise evidence from the Clade IIb outbreaks on the effectiveness and safety of MVA-BN among PWH. Despite progress in our understanding, knowledge gaps persist regarding vaccine performance in individuals with advanced immunosuppression. Furthermore, due to the emergent nature of outbreaks in the DRC and surrounding areas, limited information is available regarding implications for PWH in the context of Clade Ia and Ib. We aim to provide healthcare providers, community stakeholders, and researchers with a foundational understanding of mpox in PWH and the role of MVA-BN in mpox prevention among this group, while highlighting areas of uncertainty. These insights may be helpful in the planning of future research and to inform strategies for the prevention and management of mpox among PWH, particularly those with advanced or uncontrolled HIV. Full article
24 pages, 608 KB  
Review
West Nile Virus: Insights into Microbiology, Epidemiology, and Clinical Burden
by Andrea Marino, Ermanno Vitale, Antonino Maniaci, Luigi La Via, Vittoria Moscatt, Serena Spampinato, Paola Senia, Emmanuele Venanzi Rullo, Vincenzo Restivo, Bruno Cacopardo and Giuseppe Nunnari
Acta Microbiol. Hell. 2025, 70(4), 44; https://doi.org/10.3390/amh70040044 - 8 Nov 2025
Viewed by 1033
Abstract
West Nile Virus (WNV), a mosquito-borne flavivirus first identified in Uganda in 1937, has emerged over the past quarter century as a major global public health threat. Since its introduction into North America in 1999, WNV has become the leading cause of arboviral [...] Read more.
West Nile Virus (WNV), a mosquito-borne flavivirus first identified in Uganda in 1937, has emerged over the past quarter century as a major global public health threat. Since its introduction into North America in 1999, WNV has become the leading cause of arboviral neuroinvasive disease, with recurrent outbreaks continuing across Europe, Africa, and the Americas. This review provides a comprehensive overview of the microbiology, epidemiology, and clinical impact of WNV. We discuss the molecular biology of the virus, highlighting its genomic organization, replication strategies, and the structural and non-structural proteins that underpin viral pathogenesis and immune evasion. The complex enzootic transmission cycle, involving Culex mosquitoes and diverse avian reservoir hosts, is examined alongside ecological and climatic determinants of viral amplification and spillover into humans and equines. The clinical spectrum of WNV infection is outlined, ranging from asymptomatic seroconversion to West Nile fever and life-threatening neuroinvasive disease, with particular emphasis on risk factors for severe outcomes in older and immunocompromised individuals. Current approaches to diagnosis, supportive management, and vector control are critically reviewed, while challenges in vaccine development and the absence of effective antiviral therapy are underscored. Finally, we address future research priorities, including therapeutic innovation, predictive outbreak modeling, and genomic surveillance of viral evolution. WNV exemplifies the dynamics of emerging zoonotic diseases, and its persistence underscores the necessity of a coordinated One Health approach integrating human, animal, and environmental health. Continued scientific advances and public health commitment remain essential to mitigate its enduring global impact. Full article
Show Figures

Figure 1

55 pages, 3869 KB  
Review
A Review of the Biology of Chikungunya Virus Highlighting the Development of Current Novel Therapeutic and Prevention Approaches
by Geovana Martelossi-Cebinelli, Jessica A. Carneiro, Kelly M. Yaekashi, Mariana M. Bertozzi, Beatriz H. S. Bianchini, Fernanda S. Rasquel-Oliveira, Camila Zanluca, Claudia N. Duarte dos Santos, Rachel Arredondo, Tiffani A. Blackburn, Rubia Casagrande and Waldiceu A. Verri
Pathogens 2025, 14(10), 1047; https://doi.org/10.3390/pathogens14101047 - 16 Oct 2025
Viewed by 3275
Abstract
Chikungunya virus (CHIKV) is an arthritogenic alphavirus transmitted primarily via Aedes aegypti and Aedes albopictus mosquitoes. Since its identification, CHIKV remained confined to parts of Africa and Asia until the early 2000s, when it expanded to other continents, causing epidemics. Structurally, it is [...] Read more.
Chikungunya virus (CHIKV) is an arthritogenic alphavirus transmitted primarily via Aedes aegypti and Aedes albopictus mosquitoes. Since its identification, CHIKV remained confined to parts of Africa and Asia until the early 2000s, when it expanded to other continents, causing epidemics. Structurally, it is an enveloped virus with a positive-single-stranded RNA genome, which encodes four non-structural proteins (nsP1-nsP4), responsible for viral replication, and five structural proteins (C, E3, E2, 6K, and E1), which form the capsid and envelope. Of these proteins, glycoproteins E1 and E2 are essential for cell recognition and membrane fusion, determining infectivity and viral tropism. CHIKV replication occurs in the cytosol of different cell types, triggering an intense inflammatory and immune response, which manifests clinically as Chikungunya fever (CHIKF). Despite its epidemiological impact, current treatment is limited to symptomatic approaches, including the use of analgesics and anti-inflammatories, as no specific antiviral therapies are available. In response, promising advances are being made, including the development of vaccines, targeted antivirals, and immunotherapies. This article aims to review the main aspects of viral biology, epidemiology, and immunopathogenesis of CHIKV infection, in addition to discussing the main advances in the development of new therapeutic approaches for its control. Full article
(This article belongs to the Special Issue Virus–Host Cell Interactions and Research of New Antivirals)
Show Figures

Figure 1

18 pages, 4107 KB  
Article
Intertypic Recombination Between Coxsackievirus A16 and Enterovirus A71 Structural and Non-Structural Genes Modulates Virulence and Protection Efficacy
by Hooi Yee Chang, Han Kang Tee, Kien Chai Ong, Kartini Jasni, Syahril Abdullah, I.-Ching Sam and Yoke Fun Chan
Vaccines 2025, 13(10), 1017; https://doi.org/10.3390/vaccines13101017 - 29 Sep 2025
Viewed by 859
Abstract
Background/Objectives: Enterovirus A71 (EV-A71) and coxsackievirus A16 (CVA16) are major causative agents of hand, foot and mouth disease (HFMD), often co-circulating and occasionally undergoing genetic recombination. While natural recombinants often involve genomic regions encoding non-structural proteins, their effects on replication and pathogenesis [...] Read more.
Background/Objectives: Enterovirus A71 (EV-A71) and coxsackievirus A16 (CVA16) are major causative agents of hand, foot and mouth disease (HFMD), often co-circulating and occasionally undergoing genetic recombination. While natural recombinants often involve genomic regions encoding non-structural proteins, their effects on replication and pathogenesis remain unclear. Methods: To address this, four chimera viruses (Chi-CCE, Chi-ECE, Chi-EEC, and Chi-CEC) were constructed with 5′UTR, capsid P1, and non-structural P2 and P3 genes, from CVA16 (denoted as C) or EV-A71 (denoted as E). These chimeras were tested for replication kinetics and cytopathic effects in rhabdomyosarcoma cells while in vivo virulence and protection efficacy were evaluated using a newborn BALB/c mouse model. Results: All chimeric viruses remained viable and exhibited higher replication than CVA16. In vivo, all chimeric viruses were avirulent except Chi-CCE and CVA16, which showed high virulence and viral titres in the brains and limbs of infected newborn mice. This suggests that 5′UTR and capsid P1 genes of CVA16 are critical genetic determinants of virulence. Notably, only the anti-inflammatory cytokine IL-10 was elevated, suggesting potential immune modulation during infection. Inactivated Chi-CCE immunisation conferred 100% protection against lethal CVA16 or mouse-adapted EV-A71 challenge revealing its potential as a bivalent vaccine candidate. Conclusions: Our study demonstrates that recombination between CVA16 and EV-A71 influences viral virulence and protection efficacy with implications for future development of multivalent vaccines. Full article
(This article belongs to the Section Vaccines Against Tropical and Other Infectious Diseases)
Show Figures

Figure 1

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 3014
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

22 pages, 2424 KB  
Review
Impact of Obesity on Immunity to the Influenza Virus: Gut Microbiota, Mechanisms, and Novel Therapeutic Strategies
by Xiaoyue Ji and Jing Sun
Diseases 2025, 13(8), 267; https://doi.org/10.3390/diseases13080267 - 19 Aug 2025
Viewed by 1875
Abstract
Obesity affects millions of individuals globally, and a deeper understanding of its associated physiological disturbances is essential for addressing key public health concerns. It has been demonstrated that the influenza virus possesses substantial global epidemic potential, with higher incidence rates observed in obese [...] Read more.
Obesity affects millions of individuals globally, and a deeper understanding of its associated physiological disturbances is essential for addressing key public health concerns. It has been demonstrated that the influenza virus possesses substantial global epidemic potential, with higher incidence rates observed in obese individuals and prolonged recovery times. Obese individuals exhibit impaired immune organ function, decreased immune cell activity, disrupted metabolism characterized by mitochondrial dysfunction, and an imbalance in gut microbiota associated with intestinal mucosal barrier damage. The gut microbiota and their metabolic composition in obese patients differ from those in non-obese individuals, potentially promoting viral replication and exacerbating disease severity. These factors collectively contribute to more severe tissue damage and heightened immune responses in obese patients during influenza infection. Therefore, understanding the impact of obesity on influenza virus infection dynamics enables the development of strategies promoting healthy lifestyles to manage body weight and enhance immunity against viral infections. Additionally, given that this special population may not respond optimally to antimicrobial drugs and vaccination, it is necessary to consider how treatment strategies for this group are managed. This review illustrates findings concerning the impact of obesity on the immune response to influenza virus infection, including potential underlying mechanisms. Full article
Show Figures

Figure 1

14 pages, 2017 KB  
Article
The S2 Glycoprotein Subunit Determines Intestinal Tropism in Infectious Bronchitis Virus
by Zhenkai Dai, Jing Zhang, Ying Huang, Benli Huang, Zhengzhong Xiao, Keyu Feng, Guanming Shao, Xinheng Zhang and Qingmei Xie
Microorganisms 2025, 13(8), 1918; https://doi.org/10.3390/microorganisms13081918 - 17 Aug 2025
Viewed by 737
Abstract
The molecular basis for the distinct intestinal tropism of infectious bronchitis virus (IBV) strains remains poorly understood. This study identifies the S2 subunit of the spike glycoprotein as the critical determinant conferring duodenal tropism to the IBV CSL strain. Comparative pathogenesis in specific-pathogen-free [...] Read more.
The molecular basis for the distinct intestinal tropism of infectious bronchitis virus (IBV) strains remains poorly understood. This study identifies the S2 subunit of the spike glycoprotein as the critical determinant conferring duodenal tropism to the IBV CSL strain. Comparative pathogenesis in specific-pathogen-free (SPF) chicks revealed that the CSL strain achieved significantly higher viral titers in the duodenum compared to strains D90, PYG QX1, and XXX QX5. This duodenal replication was associated with severe epithelial inflammation, characterized by upregulation of pro-inflammatory cytokines (IL-6, IL-17A, IL-22, TNF-α, IFN-β, IFN-γ) and disruption of barrier integrity via downregulation of tight junction proteins (Occludin, Claudin-1, ZO-1). Crucially, reverse genetics using the non-enterotropic D90 backbone demonstrated that recombinant viruses carrying the CSL-S2 gene (rD90-ΔS/CSL and rD90-ΔS2/CSL), but not those carrying CSL-S1 (rD90-ΔS1/CSL), replicated efficiently and induced inflammation in the duodenum, phenocopying wild-type CSL. In contrast, renal tropism was independent of the S2 subunit. These findings establish the S2 subunit as both necessary and sufficient for IBV duodenal tropism, uncoupling it from renal pathogenicity. This identifies S2 as a prime molecular target for developing next-generation vaccines against intestinal IBV pathotypes. Full article
(This article belongs to the Special Issue Animal Viral Infectious Diseases)
Show Figures

Figure 1

19 pages, 2642 KB  
Article
Lipid Nanoparticle-Encapsulated TALEN-Encoding mRNA Inactivates Hepatitis B Virus Replication in Cultured Cells and Transgenic Mice
by Tiffany Smith, Prashika Singh, Ridhwaanah Bhana, Dylan Kairuz, Kristie Bloom, Mohube Betty Maepa, Abdullah Ely and Patrick Arbuthnot
Viruses 2025, 17(8), 1090; https://doi.org/10.3390/v17081090 - 7 Aug 2025
Viewed by 1716
Abstract
Chronic infection with the hepatitis B virus (HBV) results in over 1 million deaths annually. Although currently licensed treatments, including pegylated interferon-α and nucleoside/nucleotide analogs, can inhibit viral replication, they rarely eradicate covalently closed circular DNA (cccDNA) reservoirs. Moreover, vaccination does not offer [...] Read more.
Chronic infection with the hepatitis B virus (HBV) results in over 1 million deaths annually. Although currently licensed treatments, including pegylated interferon-α and nucleoside/nucleotide analogs, can inhibit viral replication, they rarely eradicate covalently closed circular DNA (cccDNA) reservoirs. Moreover, vaccination does not offer therapeutic benefit to already infected individuals or non-responders. Consequently, chronic infection is maintained by the persistence of cccDNA in infected hepatocytes. For this reason, novel therapeutic strategies that permanently inactivate cccDNA are a priority. Obligate heterodimeric transcription activator-like effector nucleases (TALENs) provide the precise gene-editing needed to disable cccDNA. To develop this strategy using a therapeutically relevant approach, TALEN-encoding mRNA targeting viral core and surface genes was synthesized using in vitro transcription with co-transcriptional capping. TALENs reduced hepatitis B surface antigen (HBsAg) by 80% in a liver-derived mammalian cell culture model of infection. In a stringent HBV transgenic murine model, a single dose of hepatotropic lipid nanoparticle-encapsulated TALEN mRNA lowered HBsAg by 63% and reduced viral particle equivalents by more than 99%, without evidence of toxicity. A surveyor assay demonstrated mean in vivo HBV DNA mutation rates of approximately 16% and 15% for Core and Surface TALENs, respectively. This study presents the first evidence of the therapeutic potential of TALEN-encoding mRNA to inactivate HBV replication permanently. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

18 pages, 2125 KB  
Article
A Replication-Defective Myxoma Virus Inducing Pro-Inflammatory Responses as Monotherapy and an Adjuvant to Chemo- and DC Immuno-Therapy for Ovarian Cancer
by Martin J. Cannon and Jia Liu
Viruses 2025, 17(8), 1058; https://doi.org/10.3390/v17081058 - 29 Jul 2025
Viewed by 1078
Abstract
Myxoma virus (MYXV), a rabbit-specific poxvirus and non-pathogenic in humans and mice, is an excellent candidate oncolytic virus for cancer therapy. MYXV also has immunotherapeutic benefits. In ovarian cancer (OC), immunosuppressive tumor-associated macrophages (TAMs) are key to inhibiting antitumor immunity while hindering therapeutic [...] Read more.
Myxoma virus (MYXV), a rabbit-specific poxvirus and non-pathogenic in humans and mice, is an excellent candidate oncolytic virus for cancer therapy. MYXV also has immunotherapeutic benefits. In ovarian cancer (OC), immunosuppressive tumor-associated macrophages (TAMs) are key to inhibiting antitumor immunity while hindering therapeutic benefit by chemotherapy and dendritic cell (DC) vaccine. Because MYXV favors binding/entry of macrophages/monocytes, we examined the therapeutic potential of MYXV against TAMs. We found previously that a replication-defective MYXV with targeted deletion of an essential gene, M062R, designated ΔM062R MYXV, activated both the host DNA sensing pathway and the SAMD9 pathway. Treatment with ΔM062R confers therapeutic benefit comparable to that of wild-type replicating MYXV in preclinical models. Here we found that ΔM062R MYXV, when integrated with cisplatin and DC immunotherapy, further improved treatment benefit, likely through promoting tumor antigen-specific T cell function. Moreover, we also tested ΔM062R MYXV in targeting human immunosuppressive TAMs from OC patient ascites in a co-culture system. We found that ΔM062R treatment subverted the immunosuppressive properties of TAMs and elevated the avidity of cytokine production in tumor antigen-specific CD4+ T cells. Overall, ΔM062R presents a promising immunotherapeutic platform as a beneficial adjuvant to chemotherapy and DC vaccine. Full article
(This article belongs to the Special Issue Women in Virology 2025)
Show Figures

Figure 1

18 pages, 25244 KB  
Article
The Procaine-Based ProcCluster® Impedes the Second Envelopment Process of Herpes Simplex Virus Type 1
by Johannes Jungwirth, Lisa Siegert, Lena Gauthier, Andreas Henke, Oliver H. Krämer, Beatrice Engert and Christina Ehrhardt
Int. J. Mol. Sci. 2025, 26(15), 7185; https://doi.org/10.3390/ijms26157185 - 25 Jul 2025
Viewed by 1019
Abstract
Herpes simplex virus type 1 (HSV-1) has a global prevalence of 64%. Established antiviral drugs, such as acyclovir (ACV), have been successfully used over the past decades. However, due to growing viral resistance against approved antivirals and the lack of effective vaccines, new [...] Read more.
Herpes simplex virus type 1 (HSV-1) has a global prevalence of 64%. Established antiviral drugs, such as acyclovir (ACV), have been successfully used over the past decades. However, due to growing viral resistance against approved antivirals and the lack of effective vaccines, new concepts are essential to target HSV-1 infections. Here, we present data on the inhibitory effect of the procaine-based substance ProcCluster® (PC) in reducing HSV-1 replication in vitro. Non-toxic PC concentrations significantly decreased HSV-1 replication in infected cells. Immunofluorescence microscopy revealed an accumulation of viral proteins in early and recycling endosomes, resulting in reduced viral release. The combination of PC with ACV resulted in an enhanced antiviral effect. Based on these results, PC alone, as well as in combination with ACV, appears to be a promising substance with antiviral potential against HSV-1 infections. Full article
Show Figures

Graphical abstract

10 pages, 1139 KB  
Article
Efficacy of PCV2 Vaccination Under Natural Conditions: A Longitudinal Study Using PCR and Virus Isolation
by Eugene Mazimpaka, Rissar Siringo Ringo, Tasuku Hirooka and Tamaki Okabayashi
Vet. Sci. 2025, 12(6), 575; https://doi.org/10.3390/vetsci12060575 - 11 Jun 2025
Viewed by 1169
Abstract
Porcine circovirus type 2 (PCV2) is the main cause of porcine circovirus-associated disease (PCVAD). Despite the widespread use of anti-PCV2 vaccines, their efficacy varies, influenced by co-infection and evaluation methods. This study assessed the efficacy of Ingelvac CircoFLEX® PCV2 vaccine under natural [...] Read more.
Porcine circovirus type 2 (PCV2) is the main cause of porcine circovirus-associated disease (PCVAD). Despite the widespread use of anti-PCV2 vaccines, their efficacy varies, influenced by co-infection and evaluation methods. This study assessed the efficacy of Ingelvac CircoFLEX® PCV2 vaccine under natural conditions. One hundred serum samples were collected from vaccinated and non-vaccinated piglets aged 21 to 173 days. PCR and antibody positivity rates did not show significant differences between the two groups, but PCV2 gene load at 91 days was significantly lower (p = 0.0095) in the vaccinated group. Anti-PCV2 antibody titers were also significantly lower in the vaccinated group at 91, 145, and 173 days (p < 0.0001). PCV2 was isolated from 50% of piglets in the non-vaccinated group (50%), compared with none (0%) in the vaccinated group, suggesting that PCV2 gene load in the non-vaccinated group did not correlate with viremia. Both groups were positive for antibodies to porcine reproductive and respiratory syndrome virus (PRRSV) at 63 days, prior to the surge in PCV2 gene load, suggesting PRRSV may enhance PCV2 replication. These findings highlight that while the vaccine reduced PCVAD damage, evaluation should incorporate methods such as virus isolation instead of relying solely on PCR. Full article
(This article belongs to the Special Issue Prevention and Control of Swine Infectious Diseases: 2nd Edition)
Show Figures

Figure 1

27 pages, 1879 KB  
Article
Syndemic Synergy of HPV, HIV, and HSV-2 for Oncogenic HPV Replication in Female Sex Workers
by Jonathan Muwonga Tukisadila, Ralph-Sydney Mboumba Bouassa, Serge Tonen-Wolyec, Hugues Loemba, Jeremie Muwonga and Laurent Belec
Trop. Med. Infect. Dis. 2025, 10(6), 157; https://doi.org/10.3390/tropicalmed10060157 - 7 Jun 2025
Cited by 1 | Viewed by 2205
Abstract
Background: Female sex workers (FSWs) in sub-Saharan Africa bear a disproportionate burden of sexually transmitted infections, including HIV, high-risk HPV (HR-HPV), and herpes simplex virus type 2 (HSV-2). This study evaluated possible association between HR-HPV, HIV, and HSV-2 among FSWs in the Democratic [...] Read more.
Background: Female sex workers (FSWs) in sub-Saharan Africa bear a disproportionate burden of sexually transmitted infections, including HIV, high-risk HPV (HR-HPV), and herpes simplex virus type 2 (HSV-2). This study evaluated possible association between HR-HPV, HIV, and HSV-2 among FSWs in the Democratic Republic of the Congo. Methods: A cross-sectional study was conducted among 432 FSWs (mean age, 28.1 years) recruited via respondent-driven sampling. Genital self-sampling using the V-Veil UP2™ device was performed, followed by HPV genotyping and quantification by multiplex PCR, and HSV-2 DNA detection by PCR. Results: Among 415 participants, HR-HPV prevalence was 36.9%, with HPV-52 (14.9%), HPV-58 (10.1%), and HPV-16 (6.5%) as leading genotypes. Overall, 89% of HR-HPV-positive women harbored genotypes covered by Gardasil-9®. Co-infection with HIV and HSV-2 significantly increased HPV prevalence, genotype diversity, and viral load. Notably, HSV-2 positivity was the sole independent predictor of elevated replication of HR-HPV (p < 0.001), vaccine HR-HPV (p < 0.001), and non-vaccine HR-HPV (p < 0.021). Conclusions: FSWs exhibit a high burden of HR-HPV, shaped by co-infections with HIV and HSV-2. HSV-2 independently drives HR-HPV replication, highlighting its role in HPV persistence and cervical cancer risk. Integrated HSV-2 detection and Gardasil-9® vaccination should be prioritized in cervical cancer elimination strategies targeting high-risk populations in sub-Saharan Africa. Full article
(This article belongs to the Special Issue HIV Testing, Prevention and Care Interventions, 2nd Edition)
Show Figures

Figure 1

24 pages, 5700 KB  
Review
Mpox 2022 to 2025 Update: A Comprehensive Review on Its Complications, Transmission, Diagnosis, and Treatment
by Rajesh Yadav, Anis Ahmad Chaudhary, Ujjwal Srivastava, Saurabh Gupta, Sarvesh Rustagi, Hassan Ahmed Rudayni, Vivek Kumar Kashyap and Sanjay Kumar
Viruses 2025, 17(6), 753; https://doi.org/10.3390/v17060753 - 25 May 2025
Cited by 9 | Viewed by 14466
Abstract
Monkeypox virus (Mpox virus) is a zoonotic orthopoxvirus that has gained increased global attention due to recent outbreaks. The current review reports the latest update of Mpox cases from 25 February 2022 to 29 April 2025. It also evaluates the possible major complications [...] Read more.
Monkeypox virus (Mpox virus) is a zoonotic orthopoxvirus that has gained increased global attention due to recent outbreaks. The current review reports the latest update of Mpox cases from 25 February 2022 to 29 April 2025. It also evaluates the possible major complications in human life caused by Mpox. In early 2022, more than 40 countries reported Mpox outbreaks. As of 12 June 2024, the global case count for the 2022–2023 Mpox outbreak was 97,281 confirmed cases, in 118 countries. The World Health Organization (WHO) declared the Mpox virus, a zoonotic disease, a public health emergency of international concern (PHEIC) on 14 August 2024. Mpox symptoms include fever, headache, muscle pain, and face-to-body rashes. The review also highlights Mpox virus replication, genomics, pathology, transmission, diagnosis, and antiviral therapies. The 2022 outbreak is also discussed in detail. The coinfection of HIV in patients infected with Mpox is also discussed. The evolving Mpox epidemiology has raised concerns about the disease’s increasing spread in non-endemic countries, emphasizing the urgent need for control and prevention. The discussion on preventive measures, including vaccination, suggests that cross-protection against Mpox may be possible using orthopoxvirus-specific antibodies. Although there are no specific antiviral drugs available, certain drugs, such as tecovirimat, cidofovir, and ribavirin, are worth considering. Full article
Show Figures

Figure 1

17 pages, 842 KB  
Review
Recombinant Sendai Virus Vectors as Novel Vaccine Candidates Against Animal Viruses
by Álex Gómez and Ramsés Reina
Viruses 2025, 17(5), 737; https://doi.org/10.3390/v17050737 - 21 May 2025
Cited by 1 | Viewed by 1766
Abstract
Vaccination plays a pivotal role in the control and prevention of animal infectious diseases. However, no efficient and safe universal vaccines are currently registered for major pathogens such as influenza A virus, foot-and-mouth disease virus (FMDV), simian immunodeficiency virus (SIV), and small ruminant [...] Read more.
Vaccination plays a pivotal role in the control and prevention of animal infectious diseases. However, no efficient and safe universal vaccines are currently registered for major pathogens such as influenza A virus, foot-and-mouth disease virus (FMDV), simian immunodeficiency virus (SIV), and small ruminant lentiviruses (SRLV). Here, we review the development of Sendai virus (SeV) vectors as a promising vaccine platform for animal diseases. Recombinant SeV vectors (rSeVv) possess several key features that make them highly suitable for developing vaccination strategies: (1) SeV has exclusively cytoplasmic replication cycle, therefore incapable of transforming host cells by integrating into the cellular genome, (2) rSeVv can accommodate large foreign gene/s inserts (~5 kb) with strong but adjustable transgene expression, (3) can be propagated to high titers in both embryonated chicken eggs and mammalian cell lines, (4) exhibits potent infectivity across a broad range of mammalian cells from different animals species, (5) undergo transient replication in the upper and lower respiratory tracts of non-natural hosts, (6) has not been associated with disease in pigs, non-humans primates, and small ruminants, ensuring a favorable safety profile, and (7) induce a robust innate and cellular immune responses. Preclinical and clinical studies using rSeVv-based vaccines against influenza A virus, FMDV, SIV, and SRLV have yielded promising results. Therefore, this review highlights the potential of rSeVv-based vaccine platforms as a valuable strategy for combating animal viruses. Full article
(This article belongs to the Special Issue Advances in Endemic and Emerging Viral Diseases in Livestock)
Show Figures

Figure 1

Back to TopTop