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

Journals

Article Types

Countries / Regions

Search Results (137)

Search Parameters:
Keywords = co-formulated antibodies

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 7666 KB  
Article
In-Situ Self-Assembling Oligomeric Collagen Scaffold Enhances Vaccine Retention and Vaccine-Induced Humoral Immunity
by Juan F. Hernandez-Franco, Sushma Gude, Rachel A. Morrison, Daniela Castillo Perez, Sherry L. Voytik-Harbin and Harm HogenEsch
Vaccines 2025, 13(11), 1146; https://doi.org/10.3390/vaccines13111146 - 8 Nov 2025
Viewed by 431
Abstract
Background/Objectives: Subunit vaccines composed of purified proteins and adjuvants offer excellent safety, but often generate short-lived immunity due to rapid antigen clearance and limited antigen-presenting cell engagement. Sustained, localized delivery of antigen and adjuvant may improve the magnitude and durability of the [...] Read more.
Background/Objectives: Subunit vaccines composed of purified proteins and adjuvants offer excellent safety, but often generate short-lived immunity due to rapid antigen clearance and limited antigen-presenting cell engagement. Sustained, localized delivery of antigen and adjuvant may improve the magnitude and durability of the immune response without compromising safety. This study evaluated an in-situ polymerizing type I oligomeric collagen (Oligomer) scaffold to localize antigen/adjuvant at the injection site and prolong antigen presentation. Methods: Mice were immunized intramuscularly with ovalbumin (OVA) and CpG oligonucleotide adjuvant delivered alone or co-formulated with Oligomer. Antibody response and inflammation at the injection site were assessed post-booster at early (Day 32) and late (Day 68) time points. Antigen retention and dendritic cell trafficking to draining lymph nodes were evaluated using fluorescently labeled OVA. Results: The Oligomer scaffold retained vaccine antigen at the injection site without eliciting a material-mediated foreign body response. Co-delivery of OVA and CpG within the scaffold enhanced germinal center activity, increased follicular helper T cells and germinal center B cells, and skewed CD4+ T cells toward a Th1 phenotype. Humoral responses were greater and more durable, with higher OVA-specific IgG, IgG1, and IgG2a titers and an increased number of bone marrow antibody-secreting cells persisting through Day 68. Antigen-positive dendritic cells, including both resident and migratory subsets, were elevated in draining lymph nodes, indicating enhanced antigen transport. No anti-mouse collagen I antibodies were detected, confirming the maintenance of collagen self-tolerance. Conclusions: The Oligomer delivery platform functioned as a localized, immunotolerant vaccine depot, sustaining antigen availability and immune cell engagement. This spatiotemporal control enhanced germinal center responses and generated a more robust, durable humoral immune response, supporting its potential to improve subunit vaccine efficacy while maintaining an excellent safety profile. Full article
(This article belongs to the Special Issue Vaccine Design and Development)
Show Figures

Figure 1

20 pages, 1726 KB  
Article
Heterologous SARS-CoV-2 Buccal Immunization with Oral Dissolving Films Generated a Strong Systemic and Mucosal Immunity in a Murine Model
by Tanisha Manoj Arte, Smital Patil, Emmanuel Adediran, Mahek Gulani, Amarae Ferguson, Sarthak Shah, Priyal Bagwe, Susu M. Zughaier and Martin J. D’Souza
Vaccines 2025, 13(11), 1105; https://doi.org/10.3390/vaccines13111105 - 29 Oct 2025
Viewed by 385
Abstract
Background: In response to the emergence of immune-evasive variants of SARS-CoV-2, this study explores a novel heterologous vaccination strategy using a microparticulate formulation approach that is delivered via oral dissolving film (ODF) formulations into the buccal cavity. Heterologous administration has the potential to [...] Read more.
Background: In response to the emergence of immune-evasive variants of SARS-CoV-2, this study explores a novel heterologous vaccination strategy using a microparticulate formulation approach that is delivered via oral dissolving film (ODF) formulations into the buccal cavity. Heterologous administration has the potential to generate cross-reactive antibodies, which can be especially beneficial against viruses with ever-mutating variants. Moreover, the microparticulate oral dissolving film-based vaccine approach is a non-invasive vaccine delivery platform. Methods: The vaccine design incorporated whole inactivated Delta and Omicron variants of the virus, administered at prime and booster doses, respectively, effectively encapsulated in a Poly(lactic-co-glycolic) acid (PLGA) polymer matrix, and adjuvanted with Alum to enhance immune activation. Following vaccination, serum, mucosal, and tissue samples were analyzed to evaluate humoral and cellular immune responses against the model antigen, as well as other variants such as Alpha and Beta variants, to understand the cross-reactive response. Result: In vitro evaluations confirmed the vaccine’s safety and its ability to stimulate immune responses. On administering microparticulate oral dissolving films to mice, whole inactivated delta and omicron variant-specific antibodies were observed in serum samples along with neutralizing titers in terminal week. The formulated vaccine showed significant secretory IgA antibody levels in mucosal samples. Moreover, CD4+ and CD8a cellular responses were observed in tissue samples of spleen and lymph nodes, along with antibodies (IgG, IgA, and IgM) detected in lung supernatant samples. Humoral and cellular cross-reactive antibodies were observed in the samples. Conclusions: This approach offers a promising platform for developing next-generation vaccines capable of inducing broad immunity. Full article
Show Figures

Figure 1

16 pages, 2728 KB  
Article
Efficacy Evaluation of an E2 Subunit Vaccine Against Highly Virulent Classical Swine Fever Virus Strain
by Yu-Chieh Chen, Chi-Chih Chen, Wen-Bin Chung, Yen-Li Huang, Guan-Ming Ke and Hso-Chi Chaung
Vaccines 2025, 13(10), 1072; https://doi.org/10.3390/vaccines13101072 - 20 Oct 2025
Viewed by 420
Abstract
Background/Objectives: Classical swine fever (CSF) is listed by the World Organisation for Animal Health as a highly devastating and contagious pig disease, causing severe economic losses to the swine industry. In spite of the successful elimination of CSF in Taiwan, preparedness against [...] Read more.
Background/Objectives: Classical swine fever (CSF) is listed by the World Organisation for Animal Health as a highly devastating and contagious pig disease, causing severe economic losses to the swine industry. In spite of the successful elimination of CSF in Taiwan, preparedness against potential reintroduction remains essential. The live attenuated vaccines have been effective in disease control, but are not capable of a viable strategy that differentiates infected from vaccinated animals (DIVA). Subunit vaccines are recognized for their safety and ability to induce protective immunity against infectious diseases. Methods: In this study, the recombinant CSF virus (CSFV) E2 proteins were formulated with a CpG motif as an adjuvant to produce the E2-CpG subunit vaccine. Its efficiency in specific-pathogen-free (SPF) pigs was compared with a commercially available E2 subunit vaccine (Bayovac® CSF-E2; Bayer Taiwan Co., Ltd., Taipei City, Taiwan). Results: Significantly higher titers of anti-E2 antibodies were induced in pigs immunized with a single dose of the E2-CpG vaccine, particularly the reduced E-0.5A formulation, than those immunized with a dose of the commercialized E2 subunit vaccine adjusted to double dosage. This designed subunit vaccine showed high efficacy in protection against clinical symptoms and significant pathological alterations in pigs after a highly virulent CSFV (genotype 1.1) challenge. Viral shedding was not detected in vaccinated pigs before completion of the challenge study, and the viral load in their spleens remained undetectable. Conclusions: These results could support the potential of the E2-CpG vaccine as a cost-effective, single-dose subunit vaccine capable of inducing robust CSFV-specific immunity and providing 100% protection against lethal CSFV challenges. Full article
(This article belongs to the Special Issue Swine Vaccines and Vaccination)
Show Figures

Figure 1

22 pages, 1862 KB  
Article
Production of Clinical-Grade SARS-CoV-2 Spike Ferritin Nanoparticle Protein Immunogen by Transient Transfection
by Agnes Hajduczki, William C. Chang, Rafael De La Barrera, James F. Wood, Wei-Hung Chen, Elizabeth J. Martinez, Jaime L. Jensen, Rajeshwer S. Sankhala, Clayton Smith, Alexander Anderson, Elaine B. Morrison, Caroline E. Peterson, Phyllis A. Rees, Sandrine Soman, Caitlin Kuklis, Aslaa Ahmed, Jocelyn King, Farooq Nasar, Courtney Corbitt, Misook Choe, Paul V. Thomas, Michelle Zemil, Lindsay Wieczorek, Victoria R. Polonis, Helen M. Dooley, John R. Mascola, Natalie de Val, Gary R. Matyas, Mangala Rao, Gregory D. Gromowski, Kayvon Modjarrad, Sandhya Vasan, Jeffrey W. Froude, Nelson L. Michael, M. Gordon Joyce and Stasya Zarlingadd Show full author list remove Hide full author list
Vaccines 2025, 13(10), 1041; https://doi.org/10.3390/vaccines13101041 - 9 Oct 2025
Viewed by 1123
Abstract
Background/Objectives: In response to the COVID-19 pandemic, we developed a vaccine candidate against SARS-CoV-2. Spike Ferritin Nanoparticle (SpFN) comprises 24 identical prefusion-stabilized spike proteins anchored to a self-assembled nanoparticle. Organized along the three-fold axis of the ferritin particle, eight SARS-CoV-2 spike trimers [...] Read more.
Background/Objectives: In response to the COVID-19 pandemic, we developed a vaccine candidate against SARS-CoV-2. Spike Ferritin Nanoparticle (SpFN) comprises 24 identical prefusion-stabilized spike proteins anchored to a self-assembled nanoparticle. Organized along the three-fold axis of the ferritin particle, eight SARS-CoV-2 spike trimers are presented per nanoparticle. Methods: Here, we describe the CGMP processes for manufacturing SpFN using transient transfection of Expi293F cells. Results: The final yield of SpFN was ~10 mg per liter of media supernatant. The resulting protein is stable in cold storage for two years at −20 °C, as well as for a month at room temperature, and can withstand multiple freeze/thaw cycles. SpFN material produced using the CGMP protocols adjuvanted with Army Liposomal Formulation-QS-21 (ALFQ) elicited potent neutralizing antibodies against WA-1, Alpha, Beta, and Delta variants in mice as measured by a pseudovirus neutralization assay. Conclusions: This work demonstrates rapid development and scaled-up production of clinical-grade SARS-CoV-2 vaccine protein material, allowing permissive storage and transport conditions, and serves as a framework for recombinant protein production for future emergent pathogens. Full article
Show Figures

Figure 1

15 pages, 1390 KB  
Article
Polyphosphazene-Mediated Assembly of TLR4 and TLR7/8 Agonists Enables a Potent Nano-Adjuvant Delivery System for Hepatitis C Virus Vaccine Antigens
by Alexander K. Andrianov, Alexander Marin, Sarah Jeong, Liudmila Kulakova, Ananda Chowdhury, Raman Hlushko, Sayan Das, Francesca Moy, Eric A. Toth, Robert K. Ernst and Thomas R. Fuerst
Vaccines 2025, 13(10), 1012; https://doi.org/10.3390/vaccines13101012 - 28 Sep 2025
Viewed by 597
Abstract
Background: The quest for well-defined immunoadjuvants remains one of the highest priorities for the successful development of effective vaccines. Combination adjuvants, which are designed to integrate both the ability to activate a variety of immune mechanisms and synergistically improve the delivery of [...] Read more.
Background: The quest for well-defined immunoadjuvants remains one of the highest priorities for the successful development of effective vaccines. Combination adjuvants, which are designed to integrate both the ability to activate a variety of immune mechanisms and synergistically improve the delivery of vaccine components, are well-positioned to address the unmet needs. The development of a preventive vaccine against hepatitis C virus (HCV)—a major public health concern—is a particular instance in which the choice of the immunoadjuvant is of utmost importance. Methods: We assembled a lipid A Toll-like receptor 4 (TLR4) agonist BECC438 and TLR7/8 agonist resiquimod (R848) on a polyphosphazene macromolecule (PCPP) to create a nanoscale immunoadjuvant-vaccine delivery system: PCPP-R+BECC438. This aqueous-based system was formulated with the HCV sE2 antigen, and the resulting vaccine candidate was evaluated in vivo for the ability to induce immune responses. Results: Co-assembly of adjuvants resulted in a visually clear aqueous system of nanoscale dimensions, monomodal size distribution, and entropy-driven interactions between components. Intramuscular immunization of mice with HCV sE2 antigen formulated in a polyphosphazene-based nano-system induced ten-fold higher IgG and IgG2a titers than the antigen adjuvanted with BECC438 alone. PCPP-R+BECC438 formulated HCV sE2 also produced statistically significant improvements in IgG2a/IgG1 ratio and more robust HCVpp neutralization ID50 titers than control formulations. Conclusions: Polyphosphazene-assembled adjuvant nano-system promotes in vivo immune responses of enhanced quantity and quality of antibodies with increased potency of HCV neutralization. Full article
(This article belongs to the Section Hepatitis Virus Vaccines)
Show Figures

Figure 1

15 pages, 1257 KB  
Article
Amino Compound-Synthesized Gold Nanoparticles for SARS-CoV-2 Antigen Delivery
by Layane Souza Rego, Marianna Teixeira Pinho Favaro, Monica Josiane Rodrigues-Jesus, Robert Andreata-Santos, Luiz Mário Ramos Janini, Marcelo Martins Seckler, Luis Carlos de Souza Ferreira and Adriano Rodrigues Azzoni
Pharmaceutics 2025, 17(9), 1211; https://doi.org/10.3390/pharmaceutics17091211 - 17 Sep 2025
Viewed by 599
Abstract
Background: Gold nanoparticles (AuNPs) are a promising platform for vaccine antigen delivery due to their ability to stimulate both innate and adaptive immune responses. These effects depend strongly on physicochemical properties such as size, polydispersity, morphology, and surface charge, which are in turn [...] Read more.
Background: Gold nanoparticles (AuNPs) are a promising platform for vaccine antigen delivery due to their ability to stimulate both innate and adaptive immune responses. These effects depend strongly on physicochemical properties such as size, polydispersity, morphology, and surface charge, which are in turn determined by the synthesis method. While amino acids are often used as capping agents for AuNPs, their direct use as both reducing and stabilizing agents has been rarely investigated. Objectives: This study aimed to establish an ultrasound-assisted method for synthesizing AuNPs using amino compounds as both reducing and stabilizing agents, and assess their physicochemical characteristics, antigen-binding capacity, and immunogenicity. Methods: AuNPs were synthesized using L-cysteine, L-arginine, and cysteamine as dual reducing/stabilizing agents under ultrasonic conditions. The nanoparticles were combined with a recombinant receptor-binding domain (RBD) of SARS-CoV-2 and evaluated in mice for their ability to induce antibody responses. Results: The synthesized AuNPs exhibited hydrodynamic diameters ranging from 6.3 to 12.4 nm and zeta potentials from −40.5 to +36.5 mV, depending on the amino compound used. All formulations elicited robust anti-RBD IgG responses, but virus neutralization activity varied significantly. Notably, AuNP–arginine induced the strongest neutralizing response despite lower adsorption capacity and stability, suggesting that epitope preservation and antigen presentation quality were more decisive than antigen density. Conclusions: These findings underscore the importance of nanoparticle design in optimizing antigen presentation and highlight the potential of amino compound-synthesized AuNPs as effective antigen delivery vehicles for future vaccine development. Full article
Show Figures

Graphical abstract

11 pages, 790 KB  
Article
Assessing the Impact of SARS-CoV-2 Spike Mutations on Antibody Binding: A Comparative Assessment of the Wuhan and JN.1 Variants’ Full-Length Spikes in a Multiplex Luminex Assay
by Gerald Waweru, Ruth Nyakundi, Bernadette Kutima, Sharon Owuor, Gloria Konyino, John Gitonga, Doreen Lugano, Angela Maina, Jennifer Musyoki, Lucy Ochola, Martin Omondi, Christopher K. Kariuki, Paul Ogongo, Christina Mwachari, Faiz Shee, Charles Agoti, Charles Sande, Sophie Uyoga, Eunice Kagucia, Ambrose Agweyu, Philip Bejon, J. Anthony G. Scott, George M. Warimwe, L. Isabella Ochola-Oyier and James Nyagwangeadd Show full author list remove Hide full author list
Viruses 2025, 17(9), 1248; https://doi.org/10.3390/v17091248 - 16 Sep 2025
Viewed by 948
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to evolve, with mutations leading to the emergence of new variants. JN.1, a subvariant of omicron BA.2.86, has demonstrated marked immune escape and is now included in updated vaccine formulations. While reduced sensitivity has been [...] Read more.
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to evolve, with mutations leading to the emergence of new variants. JN.1, a subvariant of omicron BA.2.86, has demonstrated marked immune escape and is now included in updated vaccine formulations. While reduced sensitivity has been reported for antibody assays using ancestral spike protein subunits to detect omicron-induced responses, the performance of full-length spike-based assays against omicron sublineages remains unclear. We aimed to compare the sensitivity of ELISA and Luminex assays using full-length spike proteins from the ancestral Wuhan strain and the JN.1 variant. Methods: Wuhan and JN.1 full-length spike protein constructs were designed and expressed in Expi293F mammalian cells. In-house ELISAs based on previously validated protocols were used to measure anti-spike IgG levels. Additionally, a Luminex-based assay for anti-spike antibody detection was developed and validated. Both assays were applied to the following sample groups: pre-pandemic samples (designated “gold standard negatives”); PCR confirmed 2020 positives (“gold standard wildtype positives”); PCR confirmed 2024 positives (“gold standard omicron positives”); 2022 vaccinated individuals with verbal confirmed infection (“gold standard hybrid positives”); and 2024 household samples (“unknowns”). Results: Wuhan spike protein showed a sensitivity of 100% (95% CI: 0.88–1.0) in detecting omicron-specific antibodies using gold standard omicron positives with JN.1 spike protein as a reference assay. Overall, across all samples, in ELISA, the Wuhan antigen had a sensitivity of 0.93 (95% CI: 0.89–0.95) and a specificity of 0.98 (95% CI: 0.94–0.99). The JN.1 antigen showed a sensitivity of 0.91 (95% CI: 0.87–0.94) and a specificity of 0.97 (95% CI: 0.93–0.99). In Luminex, sensitivity was 0.95 (95% CI: 0.91–0.97) for Wuhan and 0.94 (95% CI: 0.91–0.96) for JN.1. Specificity for both antigens in Luminex was 0.98 (95% CI: 0.94–0.99). Conclusions: Both ELISA and Luminex assays showed comparable sensitivity and specificity for both Wuhan and JN.1 antigens, indicating that mutations in the JN.1 variant do not significantly impact assay performance. This suggests preserved antigenic recognition across variants. Full article
(This article belongs to the Section Coronaviruses)
Show Figures

Figure 1

21 pages, 2522 KB  
Article
Epitope Profiling of SARS-CoV-2 Spike Antigen Provides a Novel Strategy for Developing ELISAs Specific for Different Spike Protein Variants in Bivalent Vaccine Formulations
by Luciano Ettorre, Trevor Williams, Camille Houy, Shaolong Zhu, Michael Kishko, Ali Azizi, Andrew D. James, Beata Gajewska and Jason Szeto
Vaccines 2025, 13(8), 794; https://doi.org/10.3390/vaccines13080794 - 26 Jul 2025
Viewed by 881
Abstract
Background/Objectives: An initial COVID-19 candidate vaccine containing a purified ancestral SARS-CoV-2 spike antigen was characterized with an ELISA using recombinant monoclonal antibodies (mAbs) generated against this variant. Upon the emergence of a new Beta (B.1.351) spike variant early in the pandemic, the assessment [...] Read more.
Background/Objectives: An initial COVID-19 candidate vaccine containing a purified ancestral SARS-CoV-2 spike antigen was characterized with an ELISA using recombinant monoclonal antibodies (mAbs) generated against this variant. Upon the emergence of a new Beta (B.1.351) spike variant early in the pandemic, the assessment of a bivalent vaccine containing ancestral and Beta spike antigens began. Due to accelerated project timelines, mAbs generated specifically against the Beta spike antigen were not available at the time to address assay development and vaccine testing requirements. Methods: Using only the initial mAb panel raised against the ancestral spike antigen, an epitope-blocking ELISA strategy was developed to independently measure Beta spike antigen in bivalent vaccine formulations. To facilitate this, epitope profiling of spike antigens from both ancestral and Beta variants was performed with biolayer interferometry and hydrogen–deuterium exchange mass spectrometry using the original panel of mAbs. Results: The resulting blocking ELISA was precise and specific for the Beta spike antigen and detected the expected amount of this antigen in bivalent vaccine formulations. The specific amount of ancestral spike protein in the bivalent vaccine was also confirmed using the original ELISA developed at the onset of the pandemic. Conclusions: This epitope-blocking strategy helped to overcome key reagent availability issues and could be applied to other projects involving related proteins. Full article
(This article belongs to the Special Issue New Approaches to Vaccine Development and Delivery)
Show Figures

Figure 1

26 pages, 542 KB  
Review
Challenges to the Effectiveness and Immunogenicity of COVID-19 Vaccines: A Narrative Review with a Systematic Approach
by Alexander A. Soldatov, Nickolay A. Kryuchkov, Dmitry V. Gorenkov, Zhanna I. Avdeeva, Oxana A. Svitich and Sergey Soshnikov
Vaccines 2025, 13(8), 789; https://doi.org/10.3390/vaccines13080789 - 24 Jul 2025
Viewed by 3505
Abstract
The COVID-19 pandemic accelerated the rapid development and distribution of various vaccine platforms, resulting in a significant reduction in disease severity, hospitalizations, and mortality. However, persistent challenges remain concerning the durability and breadth of vaccine-induced protection, especially in the face of emerging SARS-CoV-2 [...] Read more.
The COVID-19 pandemic accelerated the rapid development and distribution of various vaccine platforms, resulting in a significant reduction in disease severity, hospitalizations, and mortality. However, persistent challenges remain concerning the durability and breadth of vaccine-induced protection, especially in the face of emerging SARS-CoV-2 variants. This review aimed to evaluate the factors influencing the immunogenicity and effectiveness of COVID-19 vaccines to inform future vaccine advancement strategies. A narrative review with systematic approach was conducted following PRISMA guidelines for narrative review. Literature was sourced from databases including PubMed, Embase, and Web of Science for studies published between December 2019 and May 2025. Encompassed studies assessed vaccine efficacy, immunogenicity, and safety across various populations and vaccine platforms. Data were collected qualitatively, with quantitative data from reviews highlighted where available. We have uncovered a decline in vaccine efficacy over time and weakened protection against novel variants such as Delta and Omicron. Booster doses, specifically heterologous regimens, improved immunogenicity and increased protection. Vaccine-induced neutralizing antibody titers have been found to correlate with clinical protection, although the long-term correlates of immunity remain poorly defined. The induction of IgG4 antibodies after repeated mRNA vaccinations raised concerns about potential modulation of the immune response. COVID-19 vaccines have contributed significantly to pandemic control; however, their efficacy is limited by the evolution of the virus and declining immunity. Forthcoming vaccine strategies should focus on broad-spectrum, variant-adapted formulations and defining robust comparisons of protection. Recognizing the immunological basis of vaccine response, including the role of specific antibody subclasses, is fundamental for optimizing long-term protection. Full article
(This article belongs to the Section COVID-19 Vaccines and Vaccination)
Show Figures

Figure 1

21 pages, 3415 KB  
Article
SARS-CoV-2 RBD Scaffolded by AP205 or TIP60 Nanoparticles and Delivered as mRNA Elicits Robust Neutralizing Antibody Responses
by Johnathan D. Guest, Yi Zhang, Daniel Flores, Emily Atkins, Kuishu Ren, Yingyun Cai, Kim Rosenthal, Zimeng Wang, Kihwan Kim, Charles Chen, Richard Roque, Bei Cheng, Marianna Yanez Arteta, Liping Zhou, Jason Laliberte and Joseph R. Francica
Vaccines 2025, 13(8), 778; https://doi.org/10.3390/vaccines13080778 - 22 Jul 2025
Viewed by 2302
Abstract
Background/Objectives: SARS-CoV-2 vaccine candidates comprising the receptor binding domain (RBD) of the spike protein have been shown to confer protection against infection. Previous research evaluating vaccine candidates with SARS-CoV-2 RBD fused to ferritin (RBD-ferritin) and other scaffolds suggested that multimeric assemblies of RBD [...] Read more.
Background/Objectives: SARS-CoV-2 vaccine candidates comprising the receptor binding domain (RBD) of the spike protein have been shown to confer protection against infection. Previous research evaluating vaccine candidates with SARS-CoV-2 RBD fused to ferritin (RBD-ferritin) and other scaffolds suggested that multimeric assemblies of RBD can enhance antigen presentation to improve the potency and breadth of immune responses. Though RBDs directly fused to a self-assembling scaffold can be delivered as messenger RNA (mRNA) formulated with lipid nanoparticles (LNPs), reports of SARS-CoV-2 vaccine candidates that combine these approaches remain scarce. Methods: Here, we designed RBD fused to AP205 or TIP60 self-assembling nanoparticles following a search of available structures focused on several scaffold properties. RBD-AP205 and RBD-TIP60 were tested for antigenicity following transfection and for immunogenicity and neutralization potency when delivered as mRNA in mice, with RBD-ferritin as a direct comparator. Results: All scaffolded RBD constructs were readily secreted to transfection supernatant and showed antigenicity in ELISA, though clear heterogeneity in assembly was observed. RBD-AP205 and RBD-TIP60 also exhibited robust antibody binding and neutralization titers in mice that were comparable to those elicited by RBD-ferritin or a full-length membrane-bound spike. Conclusions: These data suggest that AP205 and TIP60 can present RBD as effectively as ferritin and induce similar immune responses. By describing additional scaffolds for multimeric display that accommodate mRNA delivery platforms, this work can provide new tools for future vaccine design efforts. Full article
(This article belongs to the Section COVID-19 Vaccines and Vaccination)
Show Figures

Figure 1

17 pages, 2846 KB  
Article
IL-24 Is a Promising Molecular Adjuvant for Enhancing Protective Immunity Induced by DNA Vaccination Against Toxoplasma gondii
by Bohuai Xu, Xiuqiang Zhang, Yaowen Wang and Jia Chen
Microorganisms 2025, 13(7), 1661; https://doi.org/10.3390/microorganisms13071661 - 14 Jul 2025
Viewed by 664
Abstract
Toxoplasma gondii, a parasitic protozoan, causes zoonotic infections with severe health impacts in humans and warm-blooded animals, underscoring the urgent need for effective vaccines to control these infections. In this study, a DNA vaccine encoding TgROP5, TgROP18, TgGRA7, TgGRA15, and TgMIC6 was [...] Read more.
Toxoplasma gondii, a parasitic protozoan, causes zoonotic infections with severe health impacts in humans and warm-blooded animals, underscoring the urgent need for effective vaccines to control these infections. In this study, a DNA vaccine encoding TgROP5, TgROP18, TgGRA7, TgGRA15, and TgMIC6 was formulated using the eukaryotic expression vector pVAX I. IL-24 was delivered as a molecular adjuvant using plasmid pVAX-IL-24. BALB/c, C57BL/6, and Kunming mouse strains received the DNA immunization, after which antibody levels, cytokine production, and lymphocyte surface markers were analyzed to assess immune responses. Additionally, survival rates and brain cyst counts were measured 1 to 2 months post-vaccination in experimental models of toxoplasmosis. As a result, compared to controls, the DNA vaccine cocktail significantly increased serum IgG levels, Th1 cytokine production, and proportions of CD4+/CD8+ T cells, leading to extended survival and reduced brain cyst counts post-challenge with T. gondii ME49. Furthermore, the five-gene DNA vaccine cocktail conferred greater protection compared to single-gene immunizations. Co-administration of IL-24 significantly enhanced the immune efficacy of the multi-gene DNA vaccination. Our findings suggest that IL-24 is an effective molecular adjuvant, enhancing the protective immunity of DNA vaccines against T. gondii, supporting its potential role in vaccine strategies targeting other apicomplexan parasites. Full article
(This article belongs to the Topic Advances in Infectious and Parasitic Diseases of Animals)
Show Figures

Figure 1

17 pages, 2146 KB  
Article
Development of an Effective Single-Dose PCV2/CSFV Bivalent Subunit Vaccine Against Classical Swine Fever Virus and Porcine Circovirus Type 2
by Yu-Chieh Chen, Wen-Bin Chung, Hso-Chi Chaung, Yen-Li Huang, Chi-Chih Chen and Guan-Ming Ke
Vaccines 2025, 13(7), 736; https://doi.org/10.3390/vaccines13070736 - 8 Jul 2025
Viewed by 988
Abstract
Background/Objectives: Porcine Circovirus Type 2 (PCV2) impairs pigs’ immune systems and increases susceptibility to co-infections, including Classical Swine Fever (CSF), a highly contagious disease listed by the World Organisation for Animal Health (WOAH) as notifiable. Therefore, swine operations in CSF-endemic regions are [...] Read more.
Background/Objectives: Porcine Circovirus Type 2 (PCV2) impairs pigs’ immune systems and increases susceptibility to co-infections, including Classical Swine Fever (CSF), a highly contagious disease listed by the World Organisation for Animal Health (WOAH) as notifiable. Therefore, swine operations in CSF-endemic regions are encouraged to immunize piglets with both PCV2 and CSFV vaccinations. Currently, there is no commercially available bivalent vaccine for PCV2/CSFV. Methods: In this study, a total of twenty 4-week-old SPF pigs were administered our formulated PCV2/CSFV bivalent subunit vaccine, containing soluble CSFV-E2 (50 µg) and PCV2-ORF2 (100 µg) antigens with a porcine-specific CpG adjuvant. After 4 weeks of vaccination, all pigs were evaluated for efficacy against PCV2 and CSFV. Results: Pigs were only immunized once and showed significantly increased neutralizing or ELISA antibody titers against both viruses four weeks post-vaccination. After viral challenges, vaccinated pigs displayed no clinical signs or lesions and had markedly reduced CSFV and PCV2 viral loads in the serum and tissues compared to controls. Conclusions: These results demonstrate that a single dose of the PCV2/CSFV bivalent subunit vaccine is safe and effective in young pigs, induces strong antibody responses, and suppresses viral replication, making it a promising tool for swine disease control and cost-effective vaccination strategies. Full article
(This article belongs to the Special Issue Vaccination Against Major Respiratory Pathogens in Livestock Farming)
Show Figures

Figure 1

21 pages, 939 KB  
Review
Kidney Involvement in SARS-CoV-2 Infection: Peritoneal Dialysis as the Preferred Modality
by Marko Baralić, Nikola Stojanović, Selena Gajić, Aleksandar Sič, Aarish Manzar, Ana Bontić, Jelena Pavlović, Mateja N. Bojić and Aleksandra Kezić
Vaccines 2025, 13(7), 723; https://doi.org/10.3390/vaccines13070723 - 2 Jul 2025
Viewed by 937
Abstract
Patients undergoing peritoneal dialysis (PD) represent a uniquely vulnerable population due to intrinsic immunological dysfunction and a high prevalence of comorbid conditions. This review examines the complex interplay between natural and vaccine-induced immune responses to SARS-CoV-2 in this group, focusing on viral entry, [...] Read more.
Patients undergoing peritoneal dialysis (PD) represent a uniquely vulnerable population due to intrinsic immunological dysfunction and a high prevalence of comorbid conditions. This review examines the complex interplay between natural and vaccine-induced immune responses to SARS-CoV-2 in this group, focusing on viral entry, immune activation, and immune evasion mechanisms. Particular attention is given to the impaired cellular and humoral responses seen in PD patients, including reduced T-cell function, diminished antibody production, and abnormal cytokine signaling, all of which contribute to an elevated risk of severe COVID-19 outcomes. The immunogenicity and clinical efficacy of various vaccine platforms, including inactivated, vector-based, and mRNA formulations, are critically assessed, with an emphasis on the role of booster doses in enhancing protection amid waning immunity and evolving viral variants. Furthermore, the review highlights the advantages of PD as a home-based modality that is compatible with telemedicine and may reduce the risk of viral exposure. These insights underscore the importance of developing individualized vaccination strategies, maintaining close immunological surveillance, and implementing innovative dialysis care approaches to improve clinical outcomes during the ongoing pandemic and future public health crises. Tailored booster strategies and telemedicine-integrated care models are essential for improving outcomes in this high-risk population. Full article
(This article belongs to the Special Issue Immune Responses in Patients with Chronic Disease After Vaccination)
Show Figures

Figure 1

20 pages, 1556 KB  
Article
Engineered PAM-SPION Nanoclusters for Enhanced Cancer Therapy: Integrating Magnetic Targeting with pH-Responsive Drug Release
by Dimitra Tzavara, Konstantina Papadia, Argiris Kolokithas-Ntoukas, Sophia G. Antimisiaris and Athanasios Skouras
Molecules 2025, 30(13), 2785; https://doi.org/10.3390/molecules30132785 - 28 Jun 2025
Cited by 1 | Viewed by 961
Abstract
Background: Nanomedicine approaches for cancer therapy face significant challenges, including a poor tumor accumulation, limited therapeutic efficacy, and systemic toxicity. We hypothesized that controlling the clustering of poly(acrylic acid-co-maleic acid) (PAM)-coated superparamagnetic iron oxide nanoparticles (SPIONs) would enhance their magnetic properties for improved [...] Read more.
Background: Nanomedicine approaches for cancer therapy face significant challenges, including a poor tumor accumulation, limited therapeutic efficacy, and systemic toxicity. We hypothesized that controlling the clustering of poly(acrylic acid-co-maleic acid) (PAM)-coated superparamagnetic iron oxide nanoparticles (SPIONs) would enhance their magnetic properties for improved targeting, while enabling a pH-responsive drug release in tumor microenvironments. Methods: PAM-stabilized SPION clusters were synthesized via arrested precipitation, characterized for physicochemical and magnetic properties, and evaluated for doxorubicin loading and pH-dependent release. A dual targeting approach combining antibody conjugation with magnetic guidance was assessed in cellular models, including a novel alternating magnetic field (AMF) pre-treatment protocol. Results: PAM-SPION clusters demonstrated controlled size distributions (60–100 nm), excellent colloidal stability, and enhanced magnetic properties, particularly for larger crystallites (13 nm). The formulations exhibited a pH-responsive drug release (8.5% at pH 7.4 vs. 14.3% at pH 6.5) and a significant enhancement of AMF-triggered release (17.5%). The dual targeting approach achieved an 8-fold increased cellular uptake compared to non-targeted formulations. Most notably, the novel AMF pre-treatment protocol demonstrated an 87% improved therapeutic efficacy compared to conventional post-treatment applications. Conclusions: The integration of targeting antibodies, magnetic guidance, and a pH-responsive PAM coating creates a versatile theranostic platform with significantly enhanced drug delivery capabilities. The unexpected synergistic effect of the AMF pre-treatment represents a promising new approach for improving the therapeutic efficacy of nanoparticle-based cancer treatments. Full article
Show Figures

Figure 1

17 pages, 1808 KB  
Article
Impact of B18R-Encoding Messenger Ribonucleic Acid Co-Delivery on Neutralizing Antibody Production in Self-Amplifying Messenger Ribonucleic Acid Vaccines
by Yutao Wang, Lei Li, Min Liang, Gan Liu and Yinying Lu
Vaccines 2025, 13(5), 537; https://doi.org/10.3390/vaccines13050537 - 18 May 2025
Viewed by 1244
Abstract
Objectives: The COVID-19 pandemic has brought mRNA vaccines to the forefront due to their widespread use. In this study, we explored the potential advantages of the self-amplifying mRNA (saRNA) vaccine over conventional mRNA vaccines. Methods: Initially, we optimized lipid nanoparticle formulations [...] Read more.
Objectives: The COVID-19 pandemic has brought mRNA vaccines to the forefront due to their widespread use. In this study, we explored the potential advantages of the self-amplifying mRNA (saRNA) vaccine over conventional mRNA vaccines. Methods: Initially, we optimized lipid nanoparticle formulations and employed dT20 affinity chromatography purification to improve the intracellular expression of saRNA. Subsequently, we demonstrated that saRNA exhibited sustained expression for up to one month, both in vitro and in vivo, in contrast to mRNA. Finally, we developed a saRNA-based COVID-19 vaccine and achieved superior immune protection in mice compared to mRNA vaccine by co-delivering the B18R-encoding mRNA. Results: The co-delivery of B18R-mRNA with the saRNA vaccine significantly enhanced neutralizing antibody responses, outperforming those induced by the mRNA vaccine alone. This co-delivery strategy effectively regulated the early innate immune activation triggered by saRNA, facilitating a more robust adaptive immune response. Conclusions: The optimization strategies we used in this study highlight the potential of saRNA vaccines to offer stronger and more durable immune protection. The insights gained from this study not only promote the advancement of saRNA vaccine development but also provide practical guidance for their broader application in the fight against infectious diseases. Full article
Show Figures

Figure 1

Back to TopTop