[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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

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
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

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
remove_circle_outline

Search Results (2,657)

Search Parameters:
Keywords = vaccine design

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 1295 KiB  
Article
Dielectrically Monitored Flow Synthesis of Functional Vaccine Adjuvant Mixtures via Microwave-Assisted Catalytic Chain Transfer Processing
by Cordula S. Hege, Ian E. Hamilton, Adam A. Dundas, Kevin Adlington, Edward Walker, Helena Henke, Kaiyang Wang, Georgios Dimitrakis, Adam Buttress, Christopher Dodds, Christopher B. Fox and Derek J. Irvine
Chemistry 2025, 7(1), 10; https://doi.org/10.3390/chemistry7010010 - 17 Jan 2025
Viewed by 255
Abstract
A novel flow process to produce low-molecular-weight (Mwt) methacrylate oligomer mixtures that have potential as vaccine adjuvants and chain transfer agents (CTAs) is reported. The chemistry and process were designed to significantly reduce the number of stages required to manufacture methyl methacrylate oligomer-in-monomer [...] Read more.
A novel flow process to produce low-molecular-weight (Mwt) methacrylate oligomer mixtures that have potential as vaccine adjuvants and chain transfer agents (CTAs) is reported. The chemistry and process were designed to significantly reduce the number of stages required to manufacture methyl methacrylate oligomer-in-monomer mixtures with an oligomer Mwt range of dimers to pentamers and >50% conversion. Combining rapid in-flow, in situ catalytic chain transfer polymerization catalyst synthesis and volumetric microwave heating of the reaction medium resulted in catalyst flow synthesis being completed in <4 min, removing the need to pre-synthesize it. The steady-state operation was then successfully maintained with very low levels of external energy, as the process utilized the reaction exotherm. The microwave process outperformed a comparative conventionally heated system by delivering a 20% increase in process throughput with no change in final product quality or conversion. Additionally, combining flow and in situ catalyst processing enabled the use of a more oxidatively unstable catalyst. This allowed for in situ catalyst deactivation post-generation of the oligomers, such that residual catalyst did not need to be removed prior to preparing subsequent vaccine adjuvant or CTA screening formulations. Finally, dielectric property measurements were able to monitor the onset of reaction and steady-state operation. Full article
(This article belongs to the Section Molecular Organics)
Show Figures

Figure 1

Figure 1
<p>The plot of the experimentally recorded bulk reaction temperature against time for the MWH CCTP-catalyzed oligomerization of MMA for 3 reactions aiming for different target set-point reaction temperatures (i.e., 80, 90, and 100 °C).</p>
Full article ">Figure 2
<p>(<b>Top</b>) Comparison plot of reaction temperature and monomer conversion with time for continuous-flow CCTP-mediated oligomerization of MMA conducted with MWH (flow rate 150 g min<sup>−1</sup>) and CH (flow rate 120 g min<sup>−1</sup>). (<b>Bottom</b>) Plot of dynamic dielectric constant values with time. Values collected via in situ coaxial probe measurements of the continuous MWH production of CCTP oligomers at a mass flow rate of 150 g min<sup>−1</sup> with a target set-point temperature of 100 °C. Each data point represents a combined average of 3 measurements.</p>
Full article ">Figure 3
<p>Particle size of emulsified PMMA as measured by dynamic light scattering.</p>
Full article ">Scheme 1
<p>Schematic showing the dual-feed-vessel reactor layout for the flow manufacture of methyl methacrylate oligomers. A unique combination of rapid microwave heating (in cavity), catalytic polymerization control, and flow synthesis (in cooling coil) delivered the target products. Characterization of processing onset and yield was conducted in real-time by in situ dielectric measurements. DLS and post-process polymerization showed that the formulated MMA product solutions could be used directly to formulate stable emulsions of a target droplet size/stability and act as chain transfer agents, respectively. The figures associated with the final polymer structure, (i.e., 4–5) represent the expected degree of polymerization of the polymer product. These figures indicate that 4 to 5 repeat units are present in the final material.</p>
Full article ">
24 pages, 3443 KiB  
Article
Phenomenological Modeling of Antibody Response from Vaccine Strain Composition
by Victor Ovchinnikov and Martin Karplus
Antibodies 2025, 14(1), 6; https://doi.org/10.3390/antib14010006 - 16 Jan 2025
Viewed by 194
Abstract
The elicitation of broadly neutralizing antibodies (bnAbs) is a major goal of vaccine design for highly mutable pathogens, such as influenza, HIV, and coronavirus. Although many rational vaccine design strategies for eliciting bnAbs have been devised, their efficacies need to be evaluated in [...] Read more.
The elicitation of broadly neutralizing antibodies (bnAbs) is a major goal of vaccine design for highly mutable pathogens, such as influenza, HIV, and coronavirus. Although many rational vaccine design strategies for eliciting bnAbs have been devised, their efficacies need to be evaluated in preclinical animal models and in clinical trials. To improve outcomes for such vaccines, it would be useful to develop methods that can predict vaccine efficacies against arbitrary pathogen variants. As a step in this direction, here, we describe a simple biologically motivated model of antibody reactivity elicited by nanoparticle-based vaccines using only antigen amino acid sequences, parametrized with a small sample of experimental antibody binding data from influenza or SARS-CoV-2 nanoparticle vaccinations. Results: The model is able to recapitulate the experimental data to within experimental uncertainty, is relatively insensitive to the choice of the parametrization/training set, and provides qualitative predictions about the antigenic epitopes exploited by the vaccine, which are testable by experiment. For the mosaic nanoparticle vaccines considered here, model results suggest indirectly that the sera obtained from vaccinated mice contain bnAbs, rather than simply different strain-specific Abs. Although the present model was motivated by nanoparticle vaccines, we also apply it to a mutlivalent mRNA flu vaccination study, and demonstrate good recapitulation of experimental results. This suggests that the model formalism is, in principle, sufficiently flexible to accommodate different vaccination strategies. Finally, we show how the model could be used to rank the efficacies of vaccines with different antigen compositions. Conclusion: Overall, this study suggests that simple models of vaccine efficacy parametrized with modest amounts of experimental data could be used to compare the effectiveness of designed vaccines. Full article
17 pages, 1570 KiB  
Article
Archival and Newly Isolated Historical Bacillus anthracis Strains Populate the Deeper Phylogeny of the A.Br.075(Sterne) Clade
by Markus Antwerpen, Peter Braun, Wolfgang Beyer, Dirk Aldenkortt, Michael Seidel and Gregor Grass
Pathogens 2025, 14(1), 83; https://doi.org/10.3390/pathogens14010083 - 16 Jan 2025
Viewed by 358
Abstract
The anthrax pathogen Bacillus anthracis can remain dormant as spores in soil for many years. This applies to both natural foci and to sites of anthropogenic activity such as tanneries, abattoirs, or wool factories. The A.Br.075 (A-branch) clade (also known as A.Br.Sterne) is [...] Read more.
The anthrax pathogen Bacillus anthracis can remain dormant as spores in soil for many years. This applies to both natural foci and to sites of anthropogenic activity such as tanneries, abattoirs, or wool factories. The A.Br.075 (A-branch) clade (also known as A.Br.Sterne) is prominent not only because it comprises several outbreak strains but even more so because spore preparations of its namesake, the Sterne strain, are counted among the most utilized anthrax animal vaccines. In this study, we genome-sequenced and analyzed 56 additional B. anthracis isolates of the A.Br.075 clade. Four of these we recently retrieved from soil samples taken from a decades-long abandoned tannery. The other 52 strains originated from our archival collection from the 20th century. Notably, the extended phylogeny of the A.Br.075 clade indicated that many of the newly added chromosomes represent basal members, some of which are among the most basal strains from this lineage. Twelve new strains populate a very deep-branching lineage we have named A.Br.Ortho-Sterne (also known as A.Br.076). A further 11 isolates amend the clade named A.Br.Para-Sterne (A.Br.078). Finally, some of the terminal clusters of the clade named A.Br.Eu-Sterne appear to be replete with (near) identical isolates, possibly a result of widespread use of the Sterne vaccine and of its re-isolation from vaccination-related animal anthrax outbreaks. From the accrued new phylogenetic information, we designed and tested a variety of new SNP-PCR assays for rapid and facile genotyping of unassigned B. anthracis genomes. Lastly, the successful isolation of live B. anthracis from a long-abandoned tannery reemphasizes the need for continued risk awareness of such sites. Full article
(This article belongs to the Section Bacterial Pathogens)
Show Figures

Figure 1

Figure 1
<p>Partial horizons of drillings for sample acquisition at a derelict tannery and isolates thereof. Partial horizons of boreholes from the waterworks area and tanning pits or tanning barrels are shown. (<b>A</b>) Samples #6 and #7 from 4.0–4.7 or 4.7–5.0 m depth, respectively, composed of backfill, organic matter (low density), and decomposed wood (blackish). (<b>B</b>) Sample #8 from 4.3–4.5 m depth composed of fill, organic matter, decomposed wood, and animal skins (dark brown). Indicated by red arrows are core sample numbers yielding <span class="html-italic">B. anthracis</span>-positive <span class="html-italic">plcR</span> PCR results (#6 and #8) and a sample that turned out negative upon further analysis (#7). (<b>C</b>) Designations of isolates retrieved from core samples #6 and #8.</p>
Full article ">Figure 2
<p>Schematic overview of the phylogeny of <span class="html-italic">B. anthracis</span>. Shown is a simplified phylogenetic tree focusing on the A-branch, drawn (not to scale) from data in [<a href="#B21-pathogens-14-00083" class="html-bibr">21</a>]. Colored in black are major canSNP clades of <span class="html-italic">B. anthracis</span>. Light green circles denote the position of canSNPs defining lineages of the new genomes sequenced in this study. Indicated in orange is the canSNP that leads to the reference (Ames ‘Ancestor’). The star designates the ancestral lineage of both A.Br.001 and A.Br.002 clades (identical to that shown in <a href="#pathogens-14-00083-f003" class="html-fig">Figure 3</a>).</p>
Full article ">Figure 3
<p>Maximum parsimony tree (MPT) of the canonical A.Br.075(Sterne) clade of <span class="html-italic">B. anthracis</span>. An MST was calculated from 957 chromosomal SNPs and visualized using Grapetree [<a href="#B38-pathogens-14-00083" class="html-bibr">38</a>] (<b>A</b>). Numerical SNP distances between nodes (chromosomes) and branching points are indicated at the branches (as grey numbers). Newly sequenced isolate names are displayed in light red, and sub-clade-defining SNPs in dark red (A.Br.Eu-Sterne), blue (A.Br.Ortho-Sterne), or green (A.Br.Para-Sterne) arrows with circles. Clusters of closely related isolates compiled as large circles in panel A are shown in detail in panels (<b>B</b>–<b>D</b>). Designations of additional informative SNPs are highlighted as numbers in boxes (respective chromosome positions are listed in <a href="#app1-pathogens-14-00083" class="html-app">Supplementary Table S3</a>). Filled, partially filled, and empty circles indicate the presence of both plasmids either pXO1 (filled left) or pXO2 (filled right) or no plasmid, respectively.</p>
Full article ">
7 pages, 186 KiB  
Commentary
Elegant and Innovative Recoding Strategies for Advancing Vaccine Development
by François Meurens, Fanny Renois and Uladzimir Karniychuk
Vaccines 2025, 13(1), 78; https://doi.org/10.3390/vaccines13010078 - 16 Jan 2025
Viewed by 310
Abstract
Recoding strategies have emerged as a promising approach for developing safer and more effective vaccines by altering the genetic structure of microorganisms, such as viruses, without changing their proteins. This method enhances vaccine safety and efficacy while minimizing the risk of reversion to [...] Read more.
Recoding strategies have emerged as a promising approach for developing safer and more effective vaccines by altering the genetic structure of microorganisms, such as viruses, without changing their proteins. This method enhances vaccine safety and efficacy while minimizing the risk of reversion to virulence. Recoding enhances the frequency of CpG dinucleotides, which in turn activates immune responses and ensures a strong attenuation of the pathogens. Recent advancements highlight synonymous recoding’s potential, offering improved genetic stability and immunogenicity compared to traditional methods. Live vaccines attenuated using classical methods pose a risk of reversion to virulence and can be time-consuming to produce. Synonymous recoding, involving numerous codon alterations, boosts safety and vaccine stability. One challenge is balancing attenuation with yield; however, innovations like Zinc-finger antiviral protein (ZAP) knockout cell lines can enhance vaccine production. Beyond viral vaccines, recoding can apply to bacterial vaccines, as exemplified by modified Escherichia coli and Streptococcus pneumoniae strains, which show reduced virulence. Despite promising results, challenges like ensuring genetic stability, high yield, and regulatory approval remain. Briefly, ongoing research aims to harness these innovations for comprehensive improvements in vaccine design and deployment. In this commentary, we sought to further engage the community’s interest in this elegant approach by briefly highlighting its main advantages, disadvantages, and future prospects. Full article
(This article belongs to the Section Attenuated/Inactivated/Live and Vectored Vaccines)
24 pages, 10552 KiB  
Review
Nano-Oncologic Vaccine for Boosting Cancer Immunotherapy: The Horizons in Cancer Treatment
by Chao Chen, Yue Xu, Hui Meng, Hongyi Bao, Yong Hu, Chunjian Li and Donglin Xia
Nanomaterials 2025, 15(2), 122; https://doi.org/10.3390/nano15020122 - 16 Jan 2025
Viewed by 281
Abstract
Nano-oncologic vaccines represent a groundbreaking approach in the field of cancer immunotherapy, leveraging the unique advantages of nanotechnology to enhance the effectiveness and specificity of cancer treatments. These vaccines utilize nanoscale carriers to deliver tumor-associated antigens and immunostimulatory adjuvants, facilitating targeted immune activation [...] Read more.
Nano-oncologic vaccines represent a groundbreaking approach in the field of cancer immunotherapy, leveraging the unique advantages of nanotechnology to enhance the effectiveness and specificity of cancer treatments. These vaccines utilize nanoscale carriers to deliver tumor-associated antigens and immunostimulatory adjuvants, facilitating targeted immune activation and promoting robust antitumor responses. By improving antigen presentation and localizing immune activation within the tumor microenvironment, nano-oncologic vaccines can significantly increase the efficacy of cancer immunotherapy, particularly when combined with other treatment modalities. This review highlights the mechanisms through which nano-oncologic vaccines operate, their potential to overcome existing limitations in cancer treatment, and ongoing advancements in design. Additionally, it discusses the targeted delivery approach, such as EPR effects, pH response, ultrasonic response, and magnetic response. The combination therapy effects with photothermal therapy, radiotherapy, or immune checkpoint inhibitors are also discussed. Overall, nano-oncologic vaccines hold great promise for changing the landscape of cancer treatment and advancing personalized medicine, paving the way for more effective therapeutic strategies tailored to individual patient needs. Full article
(This article belongs to the Special Issue Applications of Functional Nanomaterials in Biomedical Science)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram of the carrier-free nano-vaccines. (<b>A</b>) The diagram illustrates the cell-based fabrication of nano-vaccines by expressing a fusion protein containing the B-subunit of AB5 toxin and a trimer-forming peptide in vivo, adopted from ref. [<a href="#B35-nanomaterials-15-00122" class="html-bibr">35</a>] (WILEY). (<b>B</b>) Schematic illustration of the construction of CIRTAs@R848, reproduced with permission from ref. [<a href="#B61-nanomaterials-15-00122" class="html-bibr">61</a>] (Elsevier). (<b>C</b>) Schematic diagram of the potential mechanism by which CIRTAs@R848 inhibits resistant tumor growth. After subcutaneous injection, CIRTAs@R848 was taken up by DCs and delivered signals to T cells, which subsequently synergized with αPD-1 to kill GEM-resistant 4T1 tumor cells, reproduced with permission from ref. [<a href="#B61-nanomaterials-15-00122" class="html-bibr">61</a>] (Elsevier).</p>
Full article ">Figure 2
<p>Graphical depiction of cell-based nano-vaccines. (<b>A</b>) Schematic illustration of personalized DC-mimicking nano-vaccines for TAA presentation and cancer immunotherapy, adopted from ref. [<a href="#B63-nanomaterials-15-00122" class="html-bibr">63</a>] (WILEY). (<b>B</b>) Schematic illustration of remodeling tumor-associated neutrophils to enhance DC-based HCC neoantigen nano-vaccine efficiency, adopted from ref. [<a href="#B66-nanomaterials-15-00122" class="html-bibr">66</a>] (WILEY).</p>
Full article ">Figure 3
<p>Illustrative diagram of antigen-based nano-vaccines. (<b>A</b>) Schematic diagram of antigen-polymer conjugated as nano-vaccines to enhance antigen-specific immunity in vivo, reproduced from ref. [<a href="#B68-nanomaterials-15-00122" class="html-bibr">68</a>], published by ACS Nano. (<b>B</b>) Graphic depiction of the preparation procedure for nano-vaccine NP-TP1@M-M, reproduced with permission from ref. [<a href="#B70-nanomaterials-15-00122" class="html-bibr">70</a>] (Springer Materials).</p>
Full article ">Figure 4
<p>Partial examples of pH-responsive nano-vaccines. (<b>A</b>) pH-responsive multivesicular polymeric nano-vaccine for the co-delivery of STING agonists and neoantigens in combination tumor immunotherapy, adopted from ref. [<a href="#B82-nanomaterials-15-00122" class="html-bibr">82</a>] (WILEY). (<b>B</b>) The diagram illustrates the creation of ORP nanoparticles for immune activation as a nano-vaccine and their release from the nanocomposite gel triggered by ultrasound, reproduced from ref. [<a href="#B84-nanomaterials-15-00122" class="html-bibr">84</a>], published by <span class="html-italic">Nano Letters</span>. (<b>C</b>) Diagrammatic representation of sono-nano-vaccine structures and their therapeutic mechanisms activated by whole-body ultrasound irradiation in a free-field setting, adopted from ref. [<a href="#B85-nanomaterials-15-00122" class="html-bibr">85</a>] (WILEY). (<b>D</b>) The α-AP-fmNPs were formed by self-association between ICG, α-AP, and the iron oxide@phospholipid complexes, in which loaded BMDCs were injected into the hind-leg footpad and subjected to MPF treatment for the promotion of migration to PLN, reproduced from ref. [<a href="#B87-nanomaterials-15-00122" class="html-bibr">87</a>], published by <span class="html-italic">Theranostics</span>.</p>
Full article ">Figure 5
<p>Schematic diagram of combination therapy with nano-vaccines. (<b>A</b>) A photodynamic therapy-motivated nano-vaccine (Dex-HDL/ALA-Fe<sub>3</sub>O<sub>4</sub>) co-delivered 5-aminolevulinic acid and Fe<sub>3</sub>O<sub>4</sub> nanozyme. Upon laser irradiation, Dex-HDL/ALA-Fe<sub>3</sub>O<sub>4</sub> effectively generated ROS at the tumor site not only to induce the immunogenic cell death cascade but also to trigger the on-demand release of full types of tumor antigens, reproduced from ref. [<a href="#B71-nanomaterials-15-00122" class="html-bibr">71</a>], published by <span class="html-italic">Nano Letters</span>. (<b>B</b>) Schematic illustration of a positive tumoricidal immunity feedback loop excited by the DPMG NP-induced ISCVs in combination with a checkpoint inhibitor, reproduced with permission from ref. [<a href="#B40-nanomaterials-15-00122" class="html-bibr">40</a>], Elsevier. (<b>C</b>) Schematic illustration of the neoantigen screening process, the preparation of thiolated nano-vaccine, and the mechanisms for personalized cancer immunotherapy, adopted from ref. [<a href="#B41-nanomaterials-15-00122" class="html-bibr">41</a>] (WILEY).</p>
Full article ">Scheme 1
<p>Schematic illustration of nano-oncologic vaccine for cancer immunotherapy.</p>
Full article ">
18 pages, 1654 KiB  
Article
The Role of Dendritic Cells in Adaptive Immune Response Induced by OVA/PDDA Nanoparticles
by Daniele R. Pereira, Yunys Pérez-Betancourt, Bianca C. L. F. Távora, Geraldo S. Magalhães, Ana Maria Carmona-Ribeiro and Eliana L. Faquim-Mauro
Vaccines 2025, 13(1), 76; https://doi.org/10.3390/vaccines13010076 - 16 Jan 2025
Viewed by 262
Abstract
Background/Objective: Cationic polymers were shown to assemble with negatively charged proteins yielding nanoparticles (NPs). Poly-diallyl-dimethyl-ammonium chloride (PDDA) combined with ovalbumin (OVA) yielded a stable colloidal dispersion (OVA/PDDA-NPs) eliciting significant anti-OVA immune response. Dendritic cells (DCs), as sentinels of foreign antigens, exert a [...] Read more.
Background/Objective: Cationic polymers were shown to assemble with negatively charged proteins yielding nanoparticles (NPs). Poly-diallyl-dimethyl-ammonium chloride (PDDA) combined with ovalbumin (OVA) yielded a stable colloidal dispersion (OVA/PDDA-NPs) eliciting significant anti-OVA immune response. Dendritic cells (DCs), as sentinels of foreign antigens, exert a crucial role in the antigen-specific immune response. Here, we aimed to evaluate the involvement of DCs in the immune response induced by OVA/PDDA. Methods: In vivo experiments were used to assess the ability of OVA/PDDA-NPs to induce anti-OVA antibodies by ELISA, as well as plasma cells and memory B cells using flow cytometry. Additionally, DC migration to draining lymph nodes following OVA/PDDA-NP immunization was evaluated by flow cytometry. In vitro experiments using bone marrow-derived DCs (BM-DCs) were used to analyze the binding and uptake of OVA/PDDA-NPs, DC maturation status, and their antigen-presenting capacity. Results: Our data confirmed the potent effect of OVA/PDDA-NPs inducing anti-OVA IgG1 and IgG2a antibodies with increased CD19+CD138+ plasma cells and CD19+CD38+CD27+ memory cells in immunized mice. OVA/PDDA-NPs induced DC maturation and migration to draining lymph nodes. The in vitro results showed higher binding and the uptake of OVA/PDDA-NPs by BM-DCs. In addition, the NPs were able to induce the upregulation of costimulatory and MHC-II molecules on DCs, as well as TNF-α and IL-12 production. Higher OVA-specific T cell proliferation was promoted by BM-DCs incubated with OVA/PDDA-NPs. Conclusions: The data showed the central role of DCs in the induction of antigen-specific immune response by OVA-PDDA-NPs, thus proving that these NPs are a potent adjuvant for subunit vaccine design. Full article
(This article belongs to the Special Issue Vaccines Targeting Dendritic Cells)
Show Figures

Figure 1

Figure 1
<p>Characterization of OVA/PDDA-NPs. (<b>A</b>) Hydrodynamic diameter of NPs and (<b>B</b>) protein dosage; (<b>A</b>) After OVA-NPs formation, the hydrodynamic diameter was analyzed by Dynamic Light Scattering (DLS) in ZetaPlus Analyzer. (<b>B</b>) OVA/water (100 μg/mL) or OVA/PDDA (100 μg/10 μg/mL) samples were subjected to centrifugation at 14,500 rpm/30 min. Then, the supernatants were collected, and the protein concentration was determined by the BCA method. The dotted line represents the limit of detection from the BSA standard curve. The results are expressed as the mean of the protein content of samples in quadruplicate ± SD.</p>
Full article ">Figure 2
<p>Potent effect of OVA/PDDA-NPs in induction of anti-OVA antibody production and B cell populations. Groups of BALB/c mice were immunized subcutaneously (s.c.) with 200 μL of OVA/Alum (100 μg/100 μg/mL) or OVA/PDDA (100 μg/10 μg/mL). As a control, mice received 200 μL of water. On the 21st day post-immunization, the mice received an antigenic booster, and on day 28, they were bled for antibody evaluation by ELISA. Their spleens were also collected to analyze the plasma cell (CD19<sup>+</sup>CD138<sup>+</sup>) and memory B (CD19<sup>+</sup>CD27<sup>+</sup>CD38<sup>+</sup>) cell populations by flow cytometry, as described in Materials and Methods. (<b>A</b>) Anti-OVA IgG1 (1/1280) and (<b>B</b>) anti-OVA IgG2a (1/20) antibodies in samples of individual mice/group. The results represent the optical density of the mean of the samples (n = 4) ± SD. (<b>C</b>) Number of plasma cells and (<b>D</b>) memory B cells in splenocyte suspensions of the individual mice/group (n = 4–5)/group ± SD. * <span class="html-italic">p</span> &lt; 0.01 of significance; *** <span class="html-italic">p</span> &lt; 0.001 of significance, **** <span class="html-italic">p</span> &lt; 0.0001 of significance.</p>
Full article ">Figure 3
<p>Effect of OVA/PDDA immunization in migration of CD11c<sup>+</sup> cells to draining lymph nodes and DCs maturation in vivo. Groups of BALB/c mice were immunized (s.c.) with 200 μL of OVA/Alum (100 μg/100 μg/mL) or OVA/PDDA (100 μg/10 μg/mL). As a control, mice received 200 μL of water. After 3 days of immunization, inguinal lymph nodes were obtained, and 1 × 10<sup>6</sup> cells were incubated with anti-CD11c, anti-CD80, anti-CD40, and anti-MHC II mAb labeled with fluorophores and analyzed by flow cytometry. (<b>A</b>) Number of CD11c<sup>+</sup> cells from mice groups immunized 3 days before; expression of (<b>B</b>) CD80, (<b>C</b>) CD40, and (<b>D</b>) MHC-II molecules on CD11c<sup>+</sup> cell population from mice groups immunized 3 days before. The analysis strategy is described in material and methods. The results represent the mean of the number of CD11c<sup>+</sup> cells from lymph node cell suspensions of individual mice/group (n = 4) ± SD. Mean of fluorescence intensity (MFI) of molecule expression on CD11c+ cells from individual mice/group (n = 4) ± SD. Statistical analysis was performed by one-way ANOVA with Tukey’s post-test. * <span class="html-italic">p</span> &lt; 0.01 of significance; ** <span class="html-italic">p</span> &lt; 0.05 of significance. Representative results from three independent experiments.</p>
Full article ">Figure 4
<p>Migration of CD11c<sup>+</sup> cells to draining lymph nodes induced by OVA/PDDA immunization 4 days before. Groups of BALB/c mice were immunized subcutaneously (s.c.) with 200 μL of OVA/Alum (100 μg/100 μg/mL) or OVA/PDDA (100 μg/10 μg/mL). As a control, mice received 200 μL of water. After 4 days of immunization, inguinal lymph nodes were obtained, and 1 × 10<sup>6</sup> cells were incubated with anti-CD11c, anti-CD80, anti-CD40, and anti-MHC II mAbs labeled with fluorophores and analyzed by flow cytometry. (<b>A</b>) Number of CD11c<sup>+</sup> cells from mice groups immunized 4 days before. Expression of (<b>B</b>) CD80, (<b>C</b>) CD40, and (<b>D</b>) MHC-II molecules on CD11c<sup>+</sup> cell population from mice groups immunized 4 days before. The analysis strategy is described in material and methods. The results represent the mean of the number of CD11c<sup>+</sup> cells from lymph node cell suspensions of individual mice/group (n = 4) ± SD. Mean of fluorescence intensity (MFI) of molecule expression on CD11c+ cells from individual mice/group (n = 4) ± SD. Statistical analysis was performed by one-way ANOVA with Tukey’s post-test. * <span class="html-italic">p</span> &lt; 0.01 of significance. Representative results from three independent experiments.</p>
Full article ">Figure 5
<p>Effect of OVA/PDDA-NPs on viability of BM-DCs in culture. The Presto Blue assay was performed in iBM-DCs cultures incubated with OVA/PDDA-NPs for 24 h in RPMI medium supplemented with 10% FBS or OPT-MEM. Fluorescence was determined by the means of the samples in quadruplicate ± SD. **** <span class="html-italic">p</span> &lt; 0.0001 of significance.</p>
Full article ">Figure 6
<p>Binding and uptake of OVA-FITC/PDDA and OVA-FITC by iBM-DCs. (<b>A</b>) iBM-DCs were incubated with OVA-FITC or OVA-FITC/PDDA for 30 min at 4 °C and (<b>B</b>) at 37 °C. The samples were analyzed by flow cytometry. Results are expressed as mean fluorescence intensity (MIF) of the samples in triplicate ± SD. * <span class="html-italic">p</span> &lt; 0.01 of significance; ** <span class="html-italic">p</span> &lt; 0.05 of significance; *** <span class="html-italic">p </span>&lt; 0.001 of significa and **** <span class="html-italic">p</span> &lt; 0.0001 of significance. Representative results of three independent experiments.</p>
Full article ">Figure 7
<p>Effect of OVA/PDDA-NPs in the expression of costimulatory and MHC-II on DC cultures. iBM-DCs (1 × 10<sup>6</sup>) were incubated for 18 h with OVA/LPS, OVA, and OVA/PDDA-NPs. After this, the DCs were incubated with anti-CD11c, anti-CD80, anti-CD86, anti-CD40, and anti-MHC II mAbs labeled with fluorophores and analyzed by flow cytometry. The analysis strategy is described in <a href="#app1-vaccines-13-00076" class="html-app">Supplementary Material</a>. The results represent the mean of fluorescence intensity of (<b>A</b>) CD40, (<b>B</b>) CD80, (<b>C</b>) CD86 and (<b>D</b>) MHC II expression on CD11c<sup>+</sup> cells in triplicate ± SD. * <span class="html-italic">p</span> &lt; 0.05 of significance; ** <span class="html-italic">p</span> &lt; 0.01 of significance; *** <span class="html-italic">p</span> &lt; 0.001 of significance. Representative results from three independent experiments.</p>
Full article ">Figure 8
<p>Cytokine production by DCs incubated with OVA/PDDA in vitro. iBM-DCs (1 × 10<sup>6</sup>) were incubated with OVA, OVA/LPS, or OVA/PDDA for 18 h. Then, the cell supernatants were collected to detect the cytokines using ELIS (<b>A</b>) IL-12, (<b>B</b>) TNF-alpha and (<b>C</b>) IL-6 production. The results represent the mean of the cytokine production in the samples in triplicate ± SD. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 of significance. Representative results from two independent experiments.</p>
Full article ">Figure 9
<p>DCs incubated with OVA/PDDA-NPs are able to induce proliferation of OVA-specific CD4+T cells in vitro. iDCs differentiated in vitro were incubated with OVA, OVA/LPS, and OVA/PDDA for 18 h. After this, the DCs were co-cultured with CD4<sup>+</sup> T cells purified from DO11.10 mice for 72 h. The proliferative response was evaluated using BrdU-ELISA assay. Results expressed as the mean of optical density obtained in samples in quadruplicate ± SD. ** <span class="html-italic">p</span> &lt; 0.01 and **** <span class="html-italic">p</span> &lt; 0.0001 of significance.</p>
Full article ">
25 pages, 6974 KiB  
Article
Do Activity Sensors Identify Physiological, Clinical and Behavioural Changes in Laying Hens Exposed to a Vaccine Challenge?
by Hyungwook Kang, Sarah Brocklehurst, Marie Haskell, Susan Jarvis and Victoria Sandilands
Animals 2025, 15(2), 205; https://doi.org/10.3390/ani15020205 - 14 Jan 2025
Viewed by 317
Abstract
This study aimed to identify if sensor technology could be used to detect sickness-type signs (caused by a live vaccine) in laying hens compared to physiological and clinical sign scoring and behaviour observation. The experiment comprised 5 replicate batches (4 hens and 12 [...] Read more.
This study aimed to identify if sensor technology could be used to detect sickness-type signs (caused by a live vaccine) in laying hens compared to physiological and clinical sign scoring and behaviour observation. The experiment comprised 5 replicate batches (4 hens and 12 days per batch) using previously non-vaccinated hens (n = 20). Hens were moved on day 1 to a large experimental room with various designated zones (e.g., litter, perches, nest box), where they wore two sensors (FitBark, TrackLab). Saline was applied using ocular and nasal drops on day 3 as a control. A live vaccine (Infectious Laryngotracheitis, ILT, vaccine), applied using the same method on day 6, was used to induce mild respiratory and other responses. Physiological and clinical signs, and behaviour from videos were also recorded by a single observer. There were significant changes in body weight (p < 0.001), feed intake (p = 0.031), cloacal temperature (p < 0.001) and three out of five clinical signs (ocular discharge (p < 0.001), conjunctivitis (p < 0.001) and depression (p = 0.009)) over days. A significant decrease (p < 0.001) in activity level (FitBark) and distance travelled (both sensors) were identified over the study days, and activity and distance travelled were highly significantly associated (p < 0.001) with total clinical scores, with hens showing reduced activity and distance travelled with worsening total clinical scores. With behaviour observations from videos, the proportions of sitting, foraging and feeding behaviours (p = 0.044, 0.036 and 0.004, respectively), the proportion of total visit duration to the litter zone (p < 0.001) and perch (p = 0.037) with TrackLab and the proportions of visit counts of hens in the litter zone (p = 0.012) from video scanning changed significantly with days. This study suggests that the vaccine challenge caused associated changes in clinical/physiological signs and activity/distance travelled data from the sensors. Sensors may have a role in detecting changes in activity and movement in individual hens indicative of health or welfare problems. Full article
(This article belongs to the Section Animal Welfare)
Show Figures

Figure 1

Figure 1
<p>The floor plan of the experimental room and the furniture included there. The number of each item is given in brackets. (Figure is not to scale).</p>
Full article ">Figure 2
<p>Mean (±SD) values of (<b>a</b>) feed intake (kg) rescaled to show per hen per day, (<b>b</b>) mean individual body weight, and (<b>c</b>) mean cloacal temperature. Days on which saline and live vaccine were administered to birds are indicated.</p>
Full article ">Figure 3
<p>Mean (±SD) clinical sign scores of (<b>a</b>) ocular discharge (OD), conjunctivitis (CON), mean sum scores of all five clinical signs (TCS) and (<b>b</b>) abnormal breathing (AB), depression (DEP), nasal discharge (ND). Days on which saline and live vaccine were administered to birds are indicated.</p>
Full article ">Figure 4
<p>(<b>a</b>) Mean (±SD) activity level (in ‘Bark points’) recorded by FitBark and (<b>b</b>) mean (±SD) distance travelled (in metres) recorded by FitBark and TrackLab. Days on which saline and live vaccine were administered to birds are indicated.</p>
Full article ">Figure 5
<p>Mean (±SD) proportions of active behaviours, sitting, foraging, and feeding. The days on which saline and a live vaccine were administered to birds are indicated.</p>
Full article ">Figure 6
<p>Mean (±SD) proportions of time spent in the (<b>a</b>) litter zone and (<b>b</b>) at the perch, according to TrackLab. The days on which saline and a live vaccine were administered to birds are indicated.</p>
Full article ">Figure 7
<p>The mean (±SD) proportions of visit counts of birds in the litter zone by video scanning. The days on which saline and a live vaccine were administered to birds are indicated.</p>
Full article ">Figure 8
<p>A scatterplot matrix between variables, coloured by days 1–12 (colour scales for day: <span class="html-fig-inline" id="animals-15-00205-i001"><img alt="Animals 15 00205 i001" src="/animals/animals-15-00205/article_deploy/html/images/animals-15-00205-i001.png"/></span>). The curves on the diagonal show the distribution for each variable on each day. Pearson’s correlation coefficients are shown in the top right between pairs of variables, over all days and on each day. The variables shown in the figure are the mean cloacal temperature (temp), total score of clinical signs (TCS), mean activity level from FitBark (act_fb), mean distance travelled from TrackLab (dt_tl), proportions of active behaviour (active), feeding (feed), foraging (forage) behaviour, proportions of counts in the litter zone (lit_vo) from video scans and proportions of total visit duration to the litter zone (lit_tl) from Tracklab.</p>
Full article ">
13 pages, 209 KiB  
Article
Seasonal Influenza Vaccination Uptake Among Australian Healthcare Professionals: An Archetype for Success
by Caroline M. Hall, Anthony Cotton, Adrian Webster, Mary Bushell and Holly L. Northam
Vaccines 2025, 13(1), 71; https://doi.org/10.3390/vaccines13010071 - 14 Jan 2025
Viewed by 441
Abstract
Background/Objectives: Qualitative research suggests there may be identifiable characteristics that form a health professional (HCP) archetype associated with habitual seasonal influenza vaccination (SIV). However, the validity of this archetype requires further investigation, ideally within a theoretical framework that can elucidate this association and [...] Read more.
Background/Objectives: Qualitative research suggests there may be identifiable characteristics that form a health professional (HCP) archetype associated with habitual seasonal influenza vaccination (SIV). However, the validity of this archetype requires further investigation, ideally within a theoretical framework that can elucidate this association and its generalisability to other vaccines. This study aims to confirm key HCP archetype characteristics associated with SIV, as informed by prior qualitative research findings, and test the generalisability of the association between this archetype and SIV to COVID-19 vaccine acceptance. Method: A cross-sectional survey was designed and distributed to an Australian HCP sample consisting of practicing nurses, midwives, pharmacists, and medical practitioners. The anonymous online survey measured key characteristics that predict vaccination behaviour and intention. Results: Most participants (n = 173) demonstrated habitual SIV behaviour (77.91%) associated with the intention to vaccinate in the future. Survey findings supported the HCP archetype, as key constructs were associated with vaccination intention and behaviour, including heightened professional responsibility, vaccine confidence, and protection of self and patients. Furthermore, results suggested progressing vaccination intention to behaviour, overcoming vaccine complacency, is possible through the provision of free, accessible vaccination services. These critical factors were broadly generalisable to the COVID-19 vaccine. Conclusions: A vaccination-positive HCP archetype, supported by access to free, convenient vaccination services, was associated with the likelihood of future vaccination behaviour, including in future pandemic response scenarios. However, it will be important to ensure that HCP vaccine knowledge gaps are minimised to enhance trust in this cohort to enable broad success. Full article
21 pages, 3533 KiB  
Article
T4 Phage Displaying Dual Antigen Clusters Against H3N2 Influenza Virus Infection
by Shenglong Liu, Mengzhou Lin and Xin Zhou
Vaccines 2025, 13(1), 70; https://doi.org/10.3390/vaccines13010070 - 13 Jan 2025
Viewed by 539
Abstract
Background: The current H3N2 influenza subunit vaccine exhibits weak immunogenicity, which limits its effectiveness in preventing and controlling influenza virus infections. Methods: In this study, we aimed to develop a T4 phage-based nanovaccine designed to enhance the immunogenicity of two antigens by displaying [...] Read more.
Background: The current H3N2 influenza subunit vaccine exhibits weak immunogenicity, which limits its effectiveness in preventing and controlling influenza virus infections. Methods: In this study, we aimed to develop a T4 phage-based nanovaccine designed to enhance the immunogenicity of two antigens by displaying the HA1 and M2e antigens of the H3N2 influenza virus on each phage nanoparticle. Specifically, we fused the Soc protein with the HA1 antigen and the Hoc protein with the M2e antigen, assembling them onto a T4 phage that lacks Soc and Hoc proteins (SocHocT4), thereby constructing a nanovaccine that concurrently presents both HA1 and M2e antigens. Results: The analysis of the optical density of the target protein bands indicated that each particle could display approximately 179 HA1 and 68 M2e antigen molecules. Additionally, animal experiments demonstrated that this nanoparticle vaccine displaying dual antigen clusters induced a stronger specific immune response, higher antibody titers, a more balanced Th1/Th2 immune response, and enhanced CD4+ and CD8+ T cell effects compared to immunization with HA1 and M2e antigen molecules alone. Importantly, mice immunized with the T4 phage displaying dual antigen clusters achieved full protection (100% protection) against the H3N2 influenza virus, highlighting its robust protective efficacy. Conclusions: In summary, our findings indicate that particles based on a T4 phage displaying antigen clusters exhibit ideal immunogenicity and protective effects, providing a promising strategy for the development of subunit vaccines against various viruses beyond influenza. Full article
(This article belongs to the Special Issue Next-Generation Vaccines for Animal Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Construction of the nanovaccine. (<b>A</b>) Expression and purification of Soc-HA1, M2e-Hoc, and HA1 proteins. The black arrows indicate flexible linkers between the two proteins (M: marker). (<b>B</b>) Purification and TEM characterization of Soc<sup>−</sup>Hoc<sup>−</sup>T4 phage. The red circle denotes the purified Soc<sup>−</sup>Hoc<sup>−</sup>T4 phage, while the arrow indicates the characterization of this Soc<sup>−</sup>Hoc<sup>−</sup>T4 phage under TEM. (<b>C</b>) Validation of in vitro assembly of T4@Soc-HA1 nanoparticles; confirmed using ELISA and Western blot analysis (Lane M: marker, Lane 1: Soc<sup>−</sup>Hoc<sup>−</sup>T4 phage, Lane 2: T4@Soc-HA1 particles assembled and centrifuged pellet, Lane 3: supernatant after washing and centrifugation). (<b>D</b>) Validation of in vitro assembly of T4@M2e-Hoc nanoparticles; confirmed using ELISA and Western blot analysis (Lane M: marker, Lane 1: Soc<sup>−</sup>Hoc<sup>−</sup>T4 phage, Lane 2: T4@M2e-Hoc particles assembled and centrifuged pellet, Lane 3: supernatant after washing and centrifugation).</p>
Full article ">Figure 2
<p>HA1/M2e-specific humoral immune responses. (<b>A</b>) Immunization regimen. (<b>B</b>) HA1-specific IgG titers measured by ELISA. (<b>C</b>) M2e-specific IgG titers measured by ELISA. (<b>D</b>) Ratio of HA1-specific IgG2a to IgG1 in serum. (<b>E</b>) Ratio of M2e-specific IgG2a to IgG1 in serum. (<b>F</b>) HAI antibody titers in serum. Data are represented as mean ± S.D. ***, **** and ns indicate <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.0001 and not statistically significant, respectively (ANOVA).</p>
Full article ">Figure 3
<p>HA1/M2e-specific cellular immune responses. Mice were immunized according to the regimen shown in <a href="#vaccines-13-00070-f002" class="html-fig">Figure 2</a>A, and splenocytes were isolated on day 14 after the final immunization. (<b>A</b>) Splenocytes isolated from immunized mice were stained with anti-CD3, CD4, and CD8a antibodies and analyzed by flow cytometry. (<b>B</b>) Percentage of CD3<sup>+</sup>CD4<sup>+</sup> T cells within the splenocyte population. (<b>C</b>) Percentage of CD3<sup>+</sup>CD8<sup>+</sup> T cells within the splenocyte population. (<b>D</b>) Levels of IL-4 in the culture supernatant of splenocytes. (<b>E</b>) Levels of IFN-γ in the culture supernatant of splenocytes. Data are represented as mean ± S.D. **, ***, **** and ns indicate <span class="html-italic">p</span> &lt; 0.01, <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.0001 and not statistically significant, respectively (ANOVA).</p>
Full article ">Figure 4
<p>Evaluation of nanovaccine protection efficacy. Mice (<span class="html-italic">n</span> = 6) were subjected to a viral challenge two weeks after the final immunization. (<b>A</b>) Changes in mice body weight over 14 days post-challenge. (<b>B</b>) Survival rates of mice over 14 days post-challenge. (<b>C</b>) Lung viral loads 5 days post-challenge. (<b>D</b>) Histopathological analysis of the lungs from virus-challenged mice (<span class="html-italic">n</span> = 3), which were immunized using the same regimen as described above. Mice were euthanized, and lung sections were prepared as described in the Materials and Methods. Images represent typical results for each group, including multifocal necrosis in lung tissue (black arrows), numerous fibroblasts (light green arrows), a heavy infiltration of lymphocytes and granulocytes (dark red arrows), slight granulocyte infiltration in the alveolar walls (red arrows), abundant lymphocytes, granulocytes, and macrophages within the alveoli (light blue arrows), slight bronchiolar epithelial cell necrosis (gray arrows), necrotic cell debris and eosinophilic material (brown arrows), slight perivascular edema (blue arrows), leukocytes occluding the lumen (purple arrows), necrotic cell debris (yellow arrows), detached epithelial cells (green arrows), a small amount of perivascular lymphocytic infiltration is observed in a ring-like pattern (white arrows), and mild hemorrhage (light purple arrows). The data were analyzed using the Mantel–Cox test. *, ***, **** and ns indicate <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">p</span> &lt; 0.0001 and not statistically significant, respectively (ANOVA).</p>
Full article ">Scheme 1
<p>Schematic diagram of T4 phage-based nanovaccine displaying dual antigen clusters. The HA1 and M2e antigens of the H3N2 influenza virus were fused with the Soc and Hoc protein to generate the Soc-HA1 and M2e-Hoc fusion proteins, respectively. These two fusion proteins were co-incubated in vitro with the Soc<sup>−</sup>Hoc<sup>−</sup>T4, resulting in the formation of the T4@Soc-HA1@M2e-Hoc nanovaccine.</p>
Full article ">
12 pages, 2225 KiB  
Brief Report
Development and Evaluation of the Immunogenic Potential of an Unmodified Nucleoside mRNA Vaccine for Herpes Zoster
by Shun Zhang, Xiaojie Wang, Tongyi Zhao, Chen Yang and Lulu Huang
Vaccines 2025, 13(1), 68; https://doi.org/10.3390/vaccines13010068 - 13 Jan 2025
Viewed by 500
Abstract
Background/Objectives: Approved mRNA vaccines commonly use sequences modified with pseudouridine to enhance translation efficiency and mRNA stability. However, this modification can result in ribosomal frameshifts, reduced immunogenicity, and higher production costs. This study aimed to explore the potential of unmodified mRNA sequences for [...] Read more.
Background/Objectives: Approved mRNA vaccines commonly use sequences modified with pseudouridine to enhance translation efficiency and mRNA stability. However, this modification can result in ribosomal frameshifts, reduced immunogenicity, and higher production costs. This study aimed to explore the potential of unmodified mRNA sequences for varicella-zoster virus (VZV) and evaluate whether codon optimization could overcome the limitations of pseudouridine modification. Methods: We utilized artificial intelligence (AI) to design several unmodified gE mRNA sequences for VZV, considering factors such as codon preference and secondary structure. The optimized mRNA sequences were assessed for protein expression levels in vitro and were subsequently used to develop a vaccine, named Vac07, encapsulated in a lipid nanoparticle (LNP) delivery system. The immunogenicity of Vac07 was evaluated in mice. Results: Codon-optimized mRNA sequences showed significantly higher protein expression levels in vitro compared to wild-type (WT) sequences. Vaccination with Vac07 demonstrated immunogenicity in mice that was comparable to, or even superior to, the licensed Shingrix vaccine, characterized by a stronger Th1-biased antibody response and a slightly more robust Th1-type cellular response. Conclusions: Codon-optimized unmodified mRNA sequences may also represent a viable approach for mRNA vaccine development. These optimized sequences have the potential to lower production costs while possibly enhancing the immunogenicity of mRNA vaccines. Vac07, developed using this method, shows promise as a potentially more efficient and cost-effective mRNA vaccine candidate for VZV. Full article
(This article belongs to the Special Issue Evaluating the Immune Response to RNA Vaccine)
Show Figures

Figure 1

Figure 1
<p>Design and translation of mRNAs into VZV gE protein. (<b>a</b>) mRNA sequences with different designs are shown, along with their respective CAI and MFE values. Additional relevant parameters are listed on the right. (<b>b</b>,<b>c</b>) gE mRNAs were transfected into HEK-293T or DC2.4 cells using jetMessenger. Cells were collected at 24 or 48 h post transfection and analyzed for gE antigen expression by flow cytometry. Data from two independent experiments are shown. CPB, codon pair bias; TIE, translation initiation efficiency.</p>
Full article ">Figure 2
<p>Evaluation of antibody and memory B cell responses induced by Vac07 and Shingrix. (<b>a</b>) Schematic representation of the formulation procedure for the VZV mRNA vaccine. (<b>b</b>) Experimental design: C57BL/6 mice were immunized i.m. with escalating doses of the VZV mRNA vaccine Vac07 (n = 6), 0.1 human dose of Shingrix (n = 5), or PBS (n = 4). Blood samples were collected at specified time points to measure antibody levels, while spleens and dLNs were harvested 28 days post booster for further analysis. (<b>c</b>) Anti-gE IgG titers were measured by ELISA, and the endpoint titers are shown. (<b>d</b>) Anti-gE IgG1 and IgG2c titers at day 28 were measured by ELISA, and the endpoint titers are shown. (<b>e</b>) The ratio of IgG2c/IgG1 is shown. (<b>f</b>) Gating strategy used for identifying class-switched gE-binding MBC. Data from one representative animal is shown. The gating strategy used was as follows: First, 1 million cells were stained, and IgM and IgD antibodies were used to exclude cells that had not undergone antibody class-switching. CD19 and CD45/B220 were then used to label memory B cells. Finally, gE protein conjugated with two fluorophores (APC and BV421) was used to label the class-switched memory B cells specific to gE binding. The numbers in the figure represent the percentage of each cell population relative to the parent gating step. The right panel shows the data summary results. (<b>g</b>) Frequencies of class-switched (IgD<sup>-</sup>IgM<sup>-</sup>) gE-specific MBCs in dLNs and spleens were analyzed by flow cytometry. The left panel shows the flow cytometry representative plots, with each plot displaying a cell count of 3000. The numbers in the figure represent the frequency of class-switched gE-binding MBCs. Data are shown as mean ± SEM. One-way ANOVA with multiple comparisons tests was used for the analysis of statistical significance. ns, indicating no significant difference; * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001.</p>
Full article ">Figure 3
<p>Evaluation of T cell responses induced by Vac07 and Shingrix. C57BL/6 mice were i.m. immunized with escalating doses of Vac07 or 0.1 human dose of Shingrix on day 0 and day 14. Spleens were harvested 7 days (<b>a</b>) or 28 days (<b>b</b>–<b>f</b>) after the boost. (<b>a</b>) Splenocytes were stimulated with 2 μg/mL gE antigen for 20 h. Frequencies of IFN-γ- or IL-2-secreting T cells were assessed by ELISpot. (<b>b</b>) Splenocytes were stimulated with or without 2 μg/mL of gE antigen for 20 h. The frequency of AIM<sup>+</sup>CD4<sup>+</sup> T cells was measured by flow cytometry. The left panel shows the flow cytometry representative plots, with each plot displaying a cell count of 20,000. The numbers in the figure represent the frequency of AIM<sup>+</sup> CD4<sup>+</sup> T cells. The right panel shows the data summary results. (<b>c</b>) Splenocytes were stimulated with or without 2 μg/mL gE antigen for 8 h in the presence of Brefeldin A. The gating strategy used to analyze IFN-γ-, IL-2-, TNF-, or IL-21-producing CD4<sup>+</sup> T cells in the spleens of mice is shown. Data from one representative animal are presented. The gating strategy used was as follows: First, 1 million cells were stained, and CD4<sup>+</sup> T cells were identified using CD3 and CD4 antibodies. CD44 and CD62L antibodies were then used to label memory T cells. Finally, cytokine-specific memory T cells were identified using four cytokine antibodies (IFN-γ, IL-2, TNF, IL-21). The numbers in the figure represent the percentage of each cell population relative to the parent gating step. The right panel shows the data summary results. (<b>d</b>) Frequencies of IFN-γ-, IL-2-, TNF-, or IL-21-secreting CD4<sup>+</sup> T cells were determined by flow cytometry. (<b>e</b>) AIM<sup>+</sup> Tfh cells were determined by flow cytometry. (f) The ICOS expression on OX40<sup>+</sup>CD137<sup>+</sup>CXCR5<sup>+</sup>CD4<sup>+</sup> Tfh cells was evaluated. MFI value is shown. Data represent mean ± SEM. One-way ANOVA with multiple comparisons tests was used for the analysis of statistical significance. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001.**** <span class="html-italic">p</span> ≤ 0.0001.</p>
Full article ">
19 pages, 337 KiB  
Review
Live Plague Vaccine Development: Past, Present, and Future
by Andrey P. Anisimov, Anastasia S. Vagaiskaya, Alexandra S. Trunyakova and Svetlana V. Dentovskaya
Vaccines 2025, 13(1), 66; https://doi.org/10.3390/vaccines13010066 - 13 Jan 2025
Viewed by 609
Abstract
During the last 100 years, vaccine development has evolved from an empirical approach to one of the more rational vaccine designs where the careful selection of antigens and adjuvants is key to the desired efficacy for challenging pathogens and/or challenging populations. To improve [...] Read more.
During the last 100 years, vaccine development has evolved from an empirical approach to one of the more rational vaccine designs where the careful selection of antigens and adjuvants is key to the desired efficacy for challenging pathogens and/or challenging populations. To improve immunogenicity while maintaining a favorable reactogenicity and safety profile, modern vaccine design must consider factors beyond the choice of target antigen alone. With new vaccine technologies currently emerging, it will be possible to custom-design vaccines for optimal efficacy in groups of people with different responses to vaccination. It should be noted that after a fairly long period of overwhelming dominance of papers devoted to subunit plague vaccines, materials devoted to the development of live plague vaccines have increasingly been published. In this review, we present our opinion on reasonable tactics for the development and application of live, safe, and protective human plague vaccines causing an enhanced duration of protection and breadth of action against various virulent strains in vaccination studies representing different ages, genders, and nucleotide polymorphisms of the genes responsible for immune response. Full article
(This article belongs to the Section Attenuated/Inactivated/Live and Vectored Vaccines)
18 pages, 467 KiB  
Systematic Review
Multidimensional Demographic Analyses of COVID-19 Vaccine Inequality in the United States: A Systematic Review
by Seyed M. Karimi, Sirajum Munira Khan, Mana Moghadami, Md Yasin Ali Parh, Shaminul H. Shakib, Hamid Zarei, Sepideh Poursafargholi and Bert B. Little
Healthcare 2025, 13(2), 139; https://doi.org/10.3390/healthcare13020139 - 13 Jan 2025
Viewed by 359
Abstract
Background: COVID-19 vaccination uptake is associated with demographic characteristics such as age, sex, and ethnicity-race in the United States (U.S.). Prior research predominantly analyzed COVID-19 vaccination uptake unidimensionally, limiting insights into multidimensional demographic inequalities. Multidimensional studies provide a closer insight into vaccination inequality [...] Read more.
Background: COVID-19 vaccination uptake is associated with demographic characteristics such as age, sex, and ethnicity-race in the United States (U.S.). Prior research predominantly analyzed COVID-19 vaccination uptake unidimensionally, limiting insights into multidimensional demographic inequalities. Multidimensional studies provide a closer insight into vaccination inequality and assist in designing more effective vaccination strategies. Objectives: Review descriptive studies of the COVID-19 vaccination uptake across combinations of at least two of the three key demographic characteristics: age, sex, and ethnicity-race in the U.S. Methods: A systematic review was performed using the Joanna Briggs Institute methodology and adhering to the PRISMA-ScR principles for reporting. Six impartial reviewers examined all of the papers. The data were obtained using a tailored data extraction template. Results: A total of 2793 records were initially downloaded, 461 of them were dropped for duplication, and 2332 were reviewed. Based on the title and abstract reviews, 2115 records were excluded. After reviewing the full text of the remaining records, 212 more records were excluded. The remaining six records were reviewed to identify and compare their population, study period, data, the studied dose number, methodology, and results. Conclusions: Multidimensional COVID-19 vaccine uptake analyses are rare and mostly focused on the dose-one vaccination. Improving researchers’ access to immunization registry data while preserving data security is a prerequisite for such analyses. Full article
(This article belongs to the Section Coronaviruses (CoV) and COVID-19 Pandemic)
Show Figures

Figure 1

Figure 1
<p>Study selection flow chart.</p>
Full article ">
15 pages, 7711 KiB  
Article
Neo-BCV: A Novel Bacterial Liquid Complex Vaccine for Enhancing Dendritic Cell-Mediated Immune Responses Against Lung Cancer
by Zilong Zhu, Zhuze Chu, Fei Fei, Chenxi Wu, Zhengyue Fei, Yuxia Sun, Yun Chen and Peihua Lu
Vaccines 2025, 13(1), 64; https://doi.org/10.3390/vaccines13010064 - 13 Jan 2025
Viewed by 462
Abstract
Background: In the past decade, immunotherapy has become a major choice for the treatment of lung cancer, yet its therapeutic efficacy is still relatively limited due to the various immune escape mechanisms of tumors. Based on this, we introduce Neo-BCV, a novel bacterial [...] Read more.
Background: In the past decade, immunotherapy has become a major choice for the treatment of lung cancer, yet its therapeutic efficacy is still relatively limited due to the various immune escape mechanisms of tumors. Based on this, we introduce Neo-BCV, a novel bacterial composite vaccine designed to enhance immune responses against lung cancer. Methods: We investigated the immune enhancing effect of Neo-BCV through in vivo and in vitro experiments, including flow cytometry, RNA-seq, and Western blot. Results: We have demonstrated that Neo-BCV can promote Dendritic cells (DCs) maturation and induce DCs differentiation into pro-inflammatory subgroups, significantly enhancing cytotoxic T lymphocyte (CTL)-mediated anti-tumor responses. Transcriptome sequencing revealed that Neo-BCV exerts its effects by specifically inhibiting the JAK2-STAT3 signaling pathway, a crucial regulator of cancer progression, metabolism, and inflammation. Moreover, Neo-BCV significantly improved the immune microenvironment in both tumor and spleen tissues without inducing notable toxic effects in major organs. Conclusions: These findings highlight Neo-BCV’s potential as a safe and effective therapeutic strategy, offering a novel avenue for clinical translation in lung cancer immunotherapy. Full article
Show Figures

Figure 1

Figure 1
<p>The effect of Neo-BCV on BMDC maturation in vitro. (<b>A</b>) Morphology of BMDCs under light microscope. (<b>B</b>) Flow cytometry was used to detect the expression levels of CD86 and MHC-II on the surface of DCs, along with a semi-quantitative statistical chart. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Neo-BCV vaccine induces tumor-specific immune responses in mice. (<b>A</b>) Neo-BCV experimental treatment protocol; (<b>B</b>) Images of mouse tumor tissues after treatment; (<b>C</b>) Tumor growth curves in mice; (<b>D</b>) Tumor tissue weights in mice after treatment. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Immune response induced by Neo-BCV in the tumor. (<b>A</b>) Flow cytometry analysis of CD8<sup>+</sup> T and CD4<sup>+</sup> T lymphocyte expression in tumor tissues and semi-quantitative statistical charts. (<b>B</b>) Flow cytometry analysis of cDC1, CD103<sup>+</sup> DC, and CD83<sup>+</sup> DC expression in tumor tissues and semi-quantitative statistical charts. (<b>C</b>) Flow cytometry analysis of cytotoxic T lymphocyte perforin and granzyme B expression in tumor tissues and semi-quantitative statistical charts. n = 6, data are presented as mean ± standard deviation ( <math display="inline"><semantics> <mrow> <mover accent="true"> <mrow> <mi mathvariant="normal">x</mi> </mrow> <mo>¯</mo> </mover> </mrow> </semantics></math> ± s). Inter-group comparisons were analyzed using Student’s <span class="html-italic">t</span>-test. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001; ns indicates no significant difference.</p>
Full article ">Figure 4
<p>Immune markers induced by Neo-BCV in the spleen. (<b>A</b>) Flow cytometry analysis of DC expression in spleen tissues and semi-quantitative statistical charts. (<b>B</b>,<b>C</b>) Expression levels of CCR7, CD44, CD69, CD83, CD86, and Ki-67 in the spleen of the PBS and Neo-BCV-treated groups, along with semi-quantitative statistical charts. n = 6; staining intensity was used as the analysis metric, and statistical comparisons were made using the average OD value of the positive regions. Data are presented as mean ± standard deviation ( <math display="inline"><semantics> <mrow> <mover accent="true"> <mrow> <mi mathvariant="normal">x</mi> </mrow> <mo>¯</mo> </mover> </mrow> </semantics></math> ± s). Inter-group comparisons were analyzed using Student’s <span class="html-italic">t</span>-test; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001; ns indicates no significant difference.</p>
Full article ">Figure 5
<p>Tumor tissue transcriptomics.(<b>A</b>,<b>B</b>) Differential Gene Expression. (<b>A</b>) Volcano plot of differential genes, where the x-axis represents the fold change in gene or transcript expression between the two samples, and the y-axis represents the statistical significance of the differential expression, indicated by the <span class="html-italic">p</span>-value. (<b>B</b>) Clustering analysis of differential genes, with the color in the figure representing the expression level of that gene in this group of samples; red indicates high expression, while blue indicates low expression. (<b>C</b>,<b>D</b>) Enrichment Analysis of Differential Genes. (<b>C</b>) GO enrichment analysis, where each node represents a GO term, and the color intensity indicates the enrichment level; darker colors represent higher enrichment levels, with each node displaying the name of the GO term and the <span class="html-italic">p</span>-value from the enrichment analysis. (<b>D</b>) KEGG enrichment analysis, where the x-axis represents the enrichment factor and the y-axis represents the functional pathways enriched in the KEGG pathway. (<b>E</b>,<b>F</b>) Representative Western blot images and semi-quantitative statistical charts of the phosphorylation and total protein levels of STAT3 and JAK2 in tumor tissues. **** <span class="html-italic">p</span> &lt; 0.0001. The original Western blot figures can be found in <a href="#app1-vaccines-13-00064" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 6
<p>Safety considerations of Neo-BCV. (<b>A</b>) Body weight change curves of mice in different treatment groups. (<b>B</b>) H&amp;E staining of major organs (lung, liver, kidney, and heart) after two weeks of different treatments (scale bar = 100 μm). ns indicates no significant difference.</p>
Full article ">Figure 7
<p>Summary diagram. Neo-BCV mediates CTL anti-tumor immune responses by activating /dendritic cells (DCs). Simultaneously, it exerts anti-tumor effects through the inhibition of the JAK2-STAT3 signaling pathway.</p>
Full article ">
17 pages, 1111 KiB  
Article
The Role of Partnerships in Supporting COVID-19 Vaccine Uptake Among Migrants: A Qualitative Case Study from Tamil Nadu and Punjab, India
by Ankita Meghani, Bharathi Palanisamy, Sunita Singh, Tanya Singh, Natasha Kanagat, Anil Gupta, Kapil Singh and Gopal Krishna Soni
Vaccines 2025, 13(1), 62; https://doi.org/10.3390/vaccines13010062 - 12 Jan 2025
Viewed by 535
Abstract
Background: During the COVID-19 pandemic, migrant populations remained under-immunized due to limited access to health care, language barriers, and vaccine hesitancy. The USAID-funded MOMENTUM Routine Immunization Transformation and Equity project supported the government in collaborating with various local health and non-health partners to [...] Read more.
Background: During the COVID-19 pandemic, migrant populations remained under-immunized due to limited access to health care, language barriers, and vaccine hesitancy. The USAID-funded MOMENTUM Routine Immunization Transformation and Equity project supported the government in collaborating with various local health and non-health partners to identify and vaccinate migrants. This case study examines the roles of project partners and the strategies each entity implemented to increase COVID-19 vaccine uptake among migrants, as well as the perceptions regarding the effectiveness of these strategies. Methods: We designed a qualitative explanatory case study guided by the Behavioral and Social Drivers framework and RE-AIM implementation science frameworks. We conducted 31 focus group discussions and 50 in-depth interviews with migrants, project partners, community leaders, and government stakeholders in Tamil Nadu and Punjab. Results: In both states, partnerships with health departments, private employers, and community-based organizations were essential for identifying and vaccinating un- and under-vaccinated migrant groups. In Tamil Nadu, collaboration with the Department of Labor and mobile medical units facilitated vaccination camps at construction sites. In Punjab, religious institutions organized sessions at places of worship, and the Border Security Force enabled health workers to reach migrants living near the border. In both states, key strategies—involving influencers to discuss the importance of vaccine safety and value, bringing vaccination services to migrants’ workplaces and homes at flexible times and mandating workplace vaccination to encourage vaccination—shifted perceptions towards vaccination and increased vaccine uptake among migrants. Conclusions: The strategies and partnerships identified in this study highlight the broader implications for future public health interventions, demonstrating that collaboration with the private sector and faith-based organizations can enhance routine immunization efforts, particularly when localized to organizations that understand community needs and can address specific barriers and motivators. Full article
(This article belongs to the Special Issue Vaccines and Vaccinations in the Pandemic Period)
Show Figures

Figure 1

Figure 1
<p>India’s approach to COVID-19 vaccine administration [<a href="#B19-vaccines-13-00062" class="html-bibr">19</a>,<a href="#B20-vaccines-13-00062" class="html-bibr">20</a>,<a href="#B21-vaccines-13-00062" class="html-bibr">21</a>,<a href="#B22-vaccines-13-00062" class="html-bibr">22</a>,<a href="#B23-vaccines-13-00062" class="html-bibr">23</a>,<a href="#B24-vaccines-13-00062" class="html-bibr">24</a>].</p>
Full article ">Figure 2
<p>Strategies developed by partners to increase COVID-19 vaccine uptake among migrants. <sup>1</sup> Employers of brick kilns, construction sites, hotels, and fertilizer and agricultural companies.</p>
Full article ">
13 pages, 3243 KiB  
Article
Genetically Engineered Bacterial Ghosts as Vaccine Candidates Against Klebsiella pneumoniae Infection
by Svetlana V. Dentovskaya, Anastasia S. Vagaiskaya, Alexandra S. Trunyakova, Alena S. Kartseva, Tatiana A. Ivashchenko, Vladimir N. Gerasimov, Mikhail E. Platonov, Victoria V. Firstova and Andrey P. Anisimov
Vaccines 2025, 13(1), 59; https://doi.org/10.3390/vaccines13010059 - 10 Jan 2025
Viewed by 462
Abstract
Background/Objectives Bacterial ghosts (BGs), non-living empty envelopes of bacteria, are produced either through genetic engineering or chemical treatment of bacteria, retaining the shape of their parent cells. BGs are considered vaccine candidates, promising delivery systems, and vaccine adjuvants. The practical use of BGs [...] Read more.
Background/Objectives Bacterial ghosts (BGs), non-living empty envelopes of bacteria, are produced either through genetic engineering or chemical treatment of bacteria, retaining the shape of their parent cells. BGs are considered vaccine candidates, promising delivery systems, and vaccine adjuvants. The practical use of BGs in vaccine development for humans is limited because of concerns about the preservation of viable bacteria in BGs. Methods: To increase the efficiency of Klebsiella pneumoniae BG formation and, accordingly, to ensure maximum killing of bacteria, we exploited previously designed plasmids with the lysis gene E from bacteriophage φX174 or with holin–endolysin systems of λ or L-413C phages. Previously, this kit made it possible to generate bacterial cells of Yersinia pestis with varying degrees of hydrolysis and variable protective activity. Results: In the current study, we showed that co-expression of the holin and endolysin genes from the L-413C phage elicited more rapid and efficient K. pneumoniae lysis than lysis mediated by only single gene E or the low functioning holin–endolysin system of λ phage. The introduction of alternative lysing factors into K. pneumoniae cells instead of the E protein leads to the loss of the murein skeleton. The resulting frameless cell envelops are more reminiscent of bacterial sacs or bacterial skins than BGs. Although such structures are less naive than classical bacterial ghosts, they provide effective protection against infection by a hypervirulent strain of K. pneumoniae and can be recommended as candidate vaccines. For our vaccine candidate generated using the O1:K2 hypervirulent K. pneumoniae strain, both safety and immunogenicity aspects were evaluated. Humoral and cellular immune responses were significantly increased in mice that were intraperitoneally immunized compared with subcutaneously vaccinated animals (p < 0.05). Conclusions: Therefore, this study presents novel perspectives for future research on K. pneumoniae ghost vaccines. Full article
(This article belongs to the Section Vaccines against Infectious Diseases)
Show Figures

Figure 1

Figure 1
<p>Preparation of <span class="html-italic">K. pneumoniae</span> KPI1627 BGs. Growth and lysis were monitored by measuring OD<sub>550</sub> (<b>A</b>) and the determination of the number of CFU (<b>B</b>). The data are presented as the mean ± s.d. of three samples.</p>
Full article ">Figure 2
<p>Transmission electron micrographs of <span class="html-italic">K. pneumoniae</span> strains: (<b>A</b>) KPI1627, (<b>B</b>) KPI1627/pEYR’-E, (<b>C</b>) KPI1627/pEYR’-S-R-Rz, (<b>D</b>) KPI1627/pEYR’-E-S-R-Rz, (<b>E</b>) KPI1627/pEYR’-Y-K, (<b>F</b>) KPI1627/pEYR’-E-Y-K. The bar represents 1 μm (<b>A</b>,<b>C</b>–<b>F</b>) or 500 nm (<b>B</b>).</p>
Full article ">Figure 3
<p>Antibody response in sera of mice immunized s.c. and i.p. with KPI-Y-K and PBS. #—<span class="html-italic">p</span> &gt; 0.05; *—<span class="html-italic">p</span> &lt; 0.05; **—<span class="html-italic">p</span> &lt; 0.005; ****—<span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Specific IFN-γ, IL-6, and TNF-α levels of splenic lymphocytes from immunized mice. * <span class="html-italic">p</span> &lt; 0.05 vs. PBS group.</p>
Full article ">Figure 5
<p>The expression levels of CD69 on CD3<sup>+</sup>CD4<sup>+</sup>, CD3<sup>+</sup>CD8<sup>+</sup>, and CD19<sup>+</sup> cell subsets of splenic lymphocytes from immunized mice. The splenic lymphocytes of mice were separated 28 days after the first immunization, and corresponding BGs were used as immunogens. Following a 48-h incubation period, lymphocytes were harvested and subjected to flow cytometry analysis. ** <span class="html-italic">p</span> &lt; 0.005 vs. PBS group. Graphs and histograms show the distribution of CD69 expression in the lymphocyte subsets.</p>
Full article ">Figure 6
<p>Protection of <span class="html-italic">KP</span>-BGs against a lethal challenge with the wild-type <span class="html-italic">K. pneumoniae</span> KPI1627 strain. Mice were subjected to i.p. and s.c. immunization with KPI-YK BGs at day 0 and boosted twice at 10 and 20 days. Ten days after the last immunization, 10 mice from each group were challenged i.p. with 10<sup>4</sup> CFUs of <span class="html-italic">K. pneumoniae</span> KPI1627 (5000 LD<sub>50</sub>). **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
Back to TopTop