[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
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

Search Results (4,273)

Search Parameters:
Keywords = monoclonal antibody

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 4428 KiB  
Article
Development of 111In-Labeled Monoclonal Antibodies Targeting SFTSV Structural Proteins for Molecular Imaging of SFTS Infectious Diseases by SPECT
by Takeshi Fuchigami, Mya Myat Ngwe Tun, Yusuke Tanahara, Kodai Nishi, Sakura Yoshida, Kazuma Ogawa, Morio Nakayama and Daisuke Hayasaka
Molecules 2025, 30(1), 38; https://doi.org/10.3390/molecules30010038 (registering DOI) - 26 Dec 2024
Abstract
No effective vaccines or treatments are currently available for severe fever with thrombocytopenia syndrome (SFTS), a fatal tick-borne infectious disease caused by the SFTS virus (SFTSV). This study evaluated the potential of 111In-labeled anti-SFTSV antibodies targeting SFTSV structural proteins as single-photon emission [...] Read more.
No effective vaccines or treatments are currently available for severe fever with thrombocytopenia syndrome (SFTS), a fatal tick-borne infectious disease caused by the SFTS virus (SFTSV). This study evaluated the potential of 111In-labeled anti-SFTSV antibodies targeting SFTSV structural proteins as single-photon emission computed tomography (SPECT) imaging agents for the selective visualization of SFTSV-infected sites. This study used nuclear medicine imaging to elucidate the pathology of SFTS and assess its therapeutic efficacy. Immunostaining experiments confirmed that the anti-SFTSV antibody (N-mAb), which targets the N protein, specifically accumulated in SFTSV-infected Vero E6 cells. 111In-labeled N-mAb was successfully prepared using a diethylenetriaminepentaacetic acid (DTPA) chelator, resulting in [111In]In-DTPA-N-mAb with high radiochemical purity exceeding 95% and a radiochemical yield of 55%. Cell-binding assays using SFTSV-infected Vero E6 cells demonstrated that [111In]In-DTPA-N-mAb binding was detectable even without membrane permeabilization, with the binding intensity correlating with infection levels. In vivo studies using SFTSV-infected A129 mice showed high spleen accumulation of [111In]In-DTPA-N-mAb (87.5% ID/g), consistent with SFTSV tropism, compared to 12.3% ID/g in mock-infected mice. SPECT/CT imaging clearly revealed high radioactivity in these regions. Although nonspecific accumulation was noted in the liver and spleen, this issue may be mitigated through antibody modifications such as fragmentation or PEGylation. Overall, [111In]In-DTPA-N-mAb is a promising imaging agent for non-invasive visualization of SFTSV-infected sites and may aid in elucidating SFTS pathology and assessing therapeutic efficacy. Full article
(This article belongs to the Special Issue New Insights into Radiopharmaceuticals)
Show Figures

Figure 1

Figure 1
<p>Fluorescence images of the SFTSV antibody targeting N protein (Primary Ab: 4A10) (<b>A</b>), Gn protein (2D4) (<b>B</b>), and Gc protein (3B4) (<b>C</b>) in Vero E6 cells infected with SFTSV at mock-infected or multiplicity of infection (MOI)-0.1. The secondary antibody was FITC-conjugated anti-mouse IgG (green), and DAPI was used for nuclear staining (blue). Scale bar = 100 µm.</p>
Full article ">Figure 2
<p>In vitro binding of <sup>111</sup>In-labeled IgG ([<sup>111</sup>In]In-DTPA-cIgG, [<sup>111</sup>In]In-DTPA-N-mAb) in SFTSV-infected (MOI-0.01, MOI-0.1) and mock-infected Vero E6 cells. Vero E6 cells were processed as only fixation (<b>A</b>) or fixation and permeabilization (<b>B</b>). ** <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 (two-way ANOVA with Tukey’s post hoc test). Values are mean ± SEM, n = 5–10.</p>
Full article ">Figure 3
<p>Biodistribution of [<sup>111</sup>In]In-DTPA-N-mAb in mock- or SFTSV-infected (3 days p.i.) A129 mice. [<sup>111</sup>In]In-DTPA-N-mAb was injected intravenously via the tail vein into the mock- or SFTSV- infected mice. To evaluate the biodistribution, after 24 h the mice were sacrificed, and the organs were dissected. * <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 for mock-infected mice vs. SFTSV-infected mice (multiple unpaired <span class="html-italic">t</span>-test). Data are represented as the percentage of injected dose (%ID)/g ± SD (n = 5–6).</p>
Full article ">Figure 4
<p>Representative axial (left panels), coronal (middle panels), and sagittal (right panels) SPECT/CT images of mock-infected (<b>A</b>) and SFTSV-infected (<b>B</b>) A129 mice 3 days after infection. SPECT/CT images were acquired 24 h after intravenous injection of [<sup>111</sup>In]In-DTPA-N-mAb. The arrows indicate the liver (yellow), spleen (red), and gastrointestinal tract (green).</p>
Full article ">Figure 5
<p>Analysis of the SPECT/CT images of [<sup>111</sup>In]In-DTPA-N-mAb in SFTSV-infected mouse using the CPR method. The cross-sectional image (<b>A</b>) was reconstructed along the red line in the SFTSV-infected mouse SPECT/CT imaging data (<b>B</b>).</p>
Full article ">Scheme 1
<p>Preparation of [<sup>111</sup>In]In-DTPA-cIgG and [<sup>111</sup>In]In-DTPA-N-mAb.</p>
Full article ">
17 pages, 3163 KiB  
Article
A Cost-Effective and Robust Cell-Based Bioassay Method for Evaluating the Bioactivity of Trastuzumab-like Antibodies
by Pooja Bharali, Subhash Chand and Harish Chander
Biomedicines 2025, 13(1), 23; https://doi.org/10.3390/biomedicines13010023 (registering DOI) - 26 Dec 2024
Abstract
Background/Objectives: Trastuzumab is an effective therapeutic intervention for treating HER2-positive breast cancers. The cost-effectiveness, global demand, and patent expiration of trastuzumab have led to the inflow of its biosimilars in the global market. With the rise of biosimilars in the biopharmaceutical market, it [...] Read more.
Background/Objectives: Trastuzumab is an effective therapeutic intervention for treating HER2-positive breast cancers. The cost-effectiveness, global demand, and patent expiration of trastuzumab have led to the inflow of its biosimilars in the global market. With the rise of biosimilars in the biopharmaceutical market, it has become crucial to ensure that the biosimilar is at par with the original monoclonal antibody (mAb)in terms of efficacy, safety, and quality. Bioassay is one of the critical quality attributes (CQAs), hence developing a reliable and robust bioassay is essential for the evaluation of their biological activity and the harmonization of the quality of these biologics, supporting their safe and effective use in clinical practice. Methods: The present study aimed to develop a robust cell-based bioassay to assess the bioactivity of trastuzumab and its biosimilars for quality control testing. For this purpose, molecular characterization of different HER2-positive breast cancer cell lines of SKBR3, BT474, MDA-MD-453, MDA-MB-175, MCF-7, and MDA-MB-231 was performed to select a suitable cell line for the cell-based bioassay. Results: The SKBR3 cell line was found to express the HER2 receptors significantly higher in comparison to the other cell lines, and it was thereby selected for further bioassay optimization. The biological activity of trastuzumab was determined using the inhibition of proliferation (IOP) assay on the SKBR3, which was optimized based on the parameters of cell seeding density, drug dilution range, and incubation time, and it was further validated as per the compendial guidelines and found valid for the parameters of specificity, accuracy (% relative bias = 0.0067%), precision (repeatability: % GCV = 1.21%), linearity (R2 = 0.99), and range (50% to 200%). Additionally, the biological activity of different trastuzumab biosimilars was assessed using the validated IOP assay and compared to the HER2 binding assay performed by flow cytometry. The biological activity of different trastuzumab biosimilars was found to be comparable to the WHO primary reference standard of trastuzumab in terms of its relative potency using the IOP assay and binding assay by flow cytometry. Conclusions: Thus, an economic and robust cell-based bioassay method was successfully developed to assess the bioactivity of trastuzumab and its biosimilars. Full article
(This article belongs to the Section Drug Discovery, Development and Delivery)
Show Figures

Figure 1

Figure 1
<p>Screening of HER2-positive cell line for bioassay. (<b>a</b>) Immunoblotting analysis of HER2 receptor protein isolated from 6 cell lines. (<b>b</b>) Bar diagram of relative mRNA level expression of HER2 of 6 different cell lines. MDA-MB-231 was taken as a negative control for the experiment. Each data point is the average of three replicates and the error bars represent standard error. For the statistical analysis, the levels of HER2 receptor expressions in the HER2-positive cell lines were compared to the HER2-negative cell line of MDA-MB-231. There was a statistically significant difference at * <span class="html-italic">p</span> &lt; 0.05. (<b>c</b>) Mean fluorescence intensity (MFI) measured by the flow cytometry of different cell lines when treated with the reference standard (NIBSC 19/108) to check the surface expression of HER2. Data are represented as the mean ± SD.</p>
Full article ">Figure 1 Cont.
<p>Screening of HER2-positive cell line for bioassay. (<b>a</b>) Immunoblotting analysis of HER2 receptor protein isolated from 6 cell lines. (<b>b</b>) Bar diagram of relative mRNA level expression of HER2 of 6 different cell lines. MDA-MB-231 was taken as a negative control for the experiment. Each data point is the average of three replicates and the error bars represent standard error. For the statistical analysis, the levels of HER2 receptor expressions in the HER2-positive cell lines were compared to the HER2-negative cell line of MDA-MB-231. There was a statistically significant difference at * <span class="html-italic">p</span> &lt; 0.05. (<b>c</b>) Mean fluorescence intensity (MFI) measured by the flow cytometry of different cell lines when treated with the reference standard (NIBSC 19/108) to check the surface expression of HER2. Data are represented as the mean ± SD.</p>
Full article ">Figure 2
<p>Optimization of the working dose concentration range. The 4-PL curve fitting for the trastuzumab reference standard (NIBSC-19/108) in different drug dose ranges: (<b>a</b>) 0.01–100 µg/mL; (<b>b</b>) 0.063–1.6 µg/mL; (<b>c</b>) 0.001–0.5 µg/mL; and (<b>d</b>) 0.004–0.25 µg/mL.</p>
Full article ">Figure 3
<p>Optimization of the seeding density. (<b>a</b>) 5 × 10<sup>3</sup>, (<b>b</b>) 1.0 × 10<sup>4</sup>, and (<b>c</b>) 2.0 × 10<sup>4</sup> cells per well were seeded to optimize the anti-proliferation assay for trastuzumab biosimilars. The 4-PL curves for the anti-HER2 reference standard (NIBSC-19/108) of different seeding densities.</p>
Full article ">Figure 4
<p>Optimization of the drug incubation time. 4-PL curves for the anti-HER2 reference standard (NIBSC 19/108) and trastuzumab biosimilar drug at different incubation times: (<b>a</b>) 96 h (4 days); (<b>b</b>) 120 h (5 days); and (<b>c</b>) 168 h (7 days).</p>
Full article ">Figure 5
<p>Specificity of the trastuzumab reference standard (NIBSC-19/108). The specificity of trastuzumab (NIBSC-19/108), where bevacizumab was used as a non-specific antibody, which did not show any dose response in the SKBR3 cell line.</p>
Full article ">Figure 6
<p>Linearity of the trastuzumab reference standard (NIBSC-19/108). Linearity of the inhibition of proliferation assay including the logarithmic relation between the observed and expected relative potencies.</p>
Full article ">Figure 7
<p>Comparing the anti-proliferative activity of trastuzumab biosimilars with the alamarBlue cell viability assay. (<b>a</b>,<b>b</b>) 4-PL curves for the anti-HER2 reference standard (NIBSC-19/108) and trastuzumab biosimilar drug were measured using the alamarBlue proliferation inhibition assay in the HER2+ breast cancer cell line of SKBR-3. Experiments were performed in triplicate and for 120 h of treatment. (<b>c</b>) % Relative potencies of four biosimilar drugs with respect to the trastuzumab reference standard (NIBSC-19/108). Data are represented as the mean ± SD.</p>
Full article ">Figure 8
<p>HER2 binding assay by flow cytometry: (<b>a</b>) 4-PL curve for anti-HER2 trastuzumab reference (NIBSC-19/108) and trastuzumab biosimilar drug measured by flow cytometry; (<b>b</b>) % Relative binding activity of trastuzumab/reference product and four trastuzumab biosimilars with the HER2 receptor; (<b>c</b>) % Relative potencies of four biosimilar drugs with respect to the trastuzumab reference standard (NIBSC-19/108). Data are represented as the mean ± SD. (<b>d</b>) % Relative potencies evaluated from the IOP and flow cytometry. Data are represented as the mean ± 2SD.</p>
Full article ">
12 pages, 3048 KiB  
Article
Development of an Enzyme-Linked Immunosorbent Assay Based on a Monoclonal Antibody for the Rapid Detection of Citrinin in Wine
by Xingdong Yang, Yang Qu, Chenchen Wang, Lihua Wu and Xiaofei Hu
Foods 2025, 14(1), 27; https://doi.org/10.3390/foods14010027 - 25 Dec 2024
Abstract
The ingestion of food contaminated with citrinin (CIT) poses a variety of health risks to humans and animals. The immunogens (CIT-COOH-BSA, CIT-H-BSA) and detection antigen (CIT-COOH-OVA, CIT-H-OVA) were synthesised using the active ester method (-COOH) and formaldehyde addition method (-H). A hybridoma cell [...] Read more.
The ingestion of food contaminated with citrinin (CIT) poses a variety of health risks to humans and animals. The immunogens (CIT-COOH-BSA, CIT-H-BSA) and detection antigen (CIT-COOH-OVA, CIT-H-OVA) were synthesised using the active ester method (-COOH) and formaldehyde addition method (-H). A hybridoma cell line (3G5) that secretes anti-CIT monoclonal antibodies (mAbs) was screened via CIT-H-BSA immunisation of mice, cell fusion, and ELISA screening technology. The cell line was injected intraperitoneally to prepare ascites. The reaction conditions for the indirect competitive ELISA (ic-ELISA) were optimised, and an ic-ELISA method for detecting CIT was preliminarily established. The results revealed that the IC50 of CIT from optimised ic-ELISA was 37 pg/mL, the linear detection range was 5.9~230 pg/mL, and the cross-reaction (CR) rate with other analogues was less than 0.01%. The intra-assay and interassay sample recovery rates of CIT were 84.7~92.0% and 83.6~91.6%, and the coefficients of variation (CVs) were less than 10%. The ic-ELISA of CIT established in this study was not significantly different from the HPLC results and is rapid, highly sensitive and strongly specific, providing technical support for the detection of CIT. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Synthesis route of the complete antigen (citrinin-COOH-BSA); (<b>B</b>) synthesis of the complete antigen (citrinin-H-BSA).</p>
Full article ">Figure 2
<p>Ultraviolet spectrum of citrinin-H-BSA. The solvent was PBS (pH 7.4).</p>
Full article ">Figure 3
<p>(<b>A</b>) Coating conditions of citrinin-H-OVA. (<b>B</b>) Reaction time of citrinin and anti-citrinin mAb. (<b>C</b>) Dilution ratio of the anti-mouse HRP-IgG. (<b>D</b>) Colour development time. “*”: the difference is significant (<span class="html-italic">p</span> &lt; 0.05), “**”: the difference is very significant (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>Standard curve of indirect competitive ELISA for anti-citrinin mAbs.</p>
Full article ">
29 pages, 9628 KiB  
Review
The Role of YY1 in the Regulation of LAG-3 Expression in CD8 T Cells and Immune Evasion in Cancer: Therapeutic Implications
by Adam Merenstein, Loiy Obeidat, Apostolos Zaravinos and Benjamin Bonavida
Cancers 2025, 17(1), 19; https://doi.org/10.3390/cancers17010019 - 25 Dec 2024
Abstract
The treatment of cancers with immunotherapies has yielded significant milestones in recent years. Amongst these immunotherapeutic strategies, the FDA has approved several checkpoint inhibitors (CPIs), primarily Anti-Programmed Death-1 (PD-1) and Programmed Death Ligand-1/2 (PDL-1/2) monoclonal antibodies, in the treatment of various cancers unresponsive [...] Read more.
The treatment of cancers with immunotherapies has yielded significant milestones in recent years. Amongst these immunotherapeutic strategies, the FDA has approved several checkpoint inhibitors (CPIs), primarily Anti-Programmed Death-1 (PD-1) and Programmed Death Ligand-1/2 (PDL-1/2) monoclonal antibodies, in the treatment of various cancers unresponsive to immune therapeutics. Such treatments resulted in significant clinical responses and the prolongation of survival in a subset of patients. However, not all patients responded to CPIs, due to various mechanisms of immune resistance. One such mechanism is that, in addition to PD-1 expression on CD8 T cells, other inhibitory receptors exist, such as Lymphocyte Activation Gene 3 (LAG-3), T cell Immunoglobulin Mucin 3 (TIM3), and T cell immunoreceptor with Ig and ITIM domains (TIGIT). These inhibitory receptors might be active in the presence of the above approved CPIs. Clearly, it is clinically challenging to block all such inhibitory receptors simultaneously using conventional antibodies. To circumvent this difficulty, we sought to target a potential transcription factor that may be involved in the molecular regulation of more than one inhibitory receptor. The transcription factor Yin Yang1 (YY1) was found to regulate the expression of PD-1, LAG-3, and TIM3. Therefore, we hypothesized that targeting YY1 in CD8 T cells should inhibit the expression of these receptors and, thus, prevent the inactivation of the anti-tumor CD8 T cells by these receptors, by corresponding ligands to tumor cells. This strategy should result in the prevention of immune evasion, leading to the inhibition of tumor growth. In addition, this strategy will be particularly effective in a subset of cancer patients who were unresponsive to approved CPIs. In this review, we discuss the regulation of LAG-3 by YY1 as proof of principle for the potential use of targeting YY1 as an alternative therapeutic approach to preventing the immune evasion of cancer. We present findings on the molecular regulations of both YY1 and LAG-3 expressions, the direct regulation of LAG-3 by YY1, the various approaches to targeting YY1 to evade immune evasion, and their clinical challenges. We also present bioinformatic analyses demonstrating the overexpression of LAG-3, YY1, and PD-L1 in various cancers, their associations with immune infiltrates, and the fact that when LAG-3 is hypermethylated in its promoter region it correlates with a better overall survival. Hence, targeting YY1 in CD8 T cells will result in restoring the anti-tumor immune response and tumor regression. Notably, in addition to the beneficial effects of targeting YY1 in CD8 T cells to inhibit the expression of inhibitory receptors, we also suggest targeting YY1 overexpressed in the tumor cells, which will also inhibit PD-L1 expression and other YY1-associated pro-tumorigenic activities. Full article
(This article belongs to the Special Issue Cancer Immunotherapy in Clinical and Translational Research)
Show Figures

Figure 1

Figure 1
<p>YY1 gene structure. This figure provides a comprehensive depiction of the Yin Yang 1 (YY1) transcription factor, illustrating its key structural domains and functional regions. YY1 is 414 amino acids long and consists of three major domains. The transactivation domains have acidic domains of around 70 amino acids each. The histidine domains for activation of YY1 have 11 histidines in a row. The repression domains have a 32 amino acid-long Glycine Alanine Domain and a 25 amino acid-long MBTD1-binding domain. Finally, in the DNA-binding domains of YY1, there are four zinc finger domains. YY1 contributes to cancer progression and immune evasion in various ways. YY1 regulates the protein stability and expression of many different cancer-associated genes. YY1 also contributes to the upregulation or downregulation of various T cell stability and regulation processes, contributing to a much stronger immune evasion response. Prepared by BioRender, Inc. (Toronto, ON, Canada).</p>
Full article ">Figure 2
<p>The KIELLE domain in LAG-3. This figure depicts the structural organization of the LAG-3 protein, a key inhibitory receptor involved in the regulation of immune responses. The KIELLE domain, the CP domain, and the IgG domains. Created by BioRender, Inc.</p>
Full article ">Figure 3
<p>LAG-3 higher affinity for MHC-II. CD4 cells use four extracellular immunoglobulin superfamily-like domains (d1–d4). LAG-3 utilizes the extra loop with 30 amino acids in D1 to bind to MHC class II with greater affinity. Ligation of MHC class II, by antigen presenting cells or aberrantly by melanoma cells, with LAG-3 mediates an intrinsic negative inhibitory signal, in which the KIEELE motif in the cytoplasmic domain is indispensable. LAG-3 is highly glycosylated with LSECtin, expressed on melanoma cells, and Galectin-3 is expressed on stromal cells and CD8<sup>+</sup> T cells in the tumor microenvironment. This figure shows the interaction between LAG-3 and these three ligands and how it interacts with CD4 and CD8 T cells. Created by BioRender, Inc.</p>
Full article ">Figure 4
<p>Regulation of PD-1 and LAG-3 by YY1 in tumor-infiltrating CD8 T lymphocytes. This figure depicts p38MAPK/JNK/YY1/LAG-3-PD-1 pathway in tumor-infiltrating lymphocytes. MAP3K activation increases JNK and p38, leading to an increase in YY1 expression. This pathway, which drives YY1 expression, leads to YY1-mediated transcriptional PD-1 and LAG-3 upregulations. The anti-tumor CD8 T cells, expressing both PD-1 and LAG-3 inhibitory receptors, will bind the tumor target cells, leading to the inactivation of the CD8 T cells through their interactions with the PDL-1/2 and MHC-II, respectively. Thus, tumors escape via immune evasion and tumor growth. Created by BioRender, Inc.</p>
Full article ">Figure 5
<p>The expression of YY1, CD274 (PD-L1), and LAG-3 in pan-cancer using normalized and batch-corrected RSEM mRNA expression data for 14 TCGA cancer types paired with their normal tissue. (<b>a</b>) The bubble plot presents the fold change and FDR for gene expression across different cancer types, represented by bubble color and size. Rows indicate gene symbols, while columns correspond to selected cancer types. Bubble color transitions from purple to red, reflecting fold change (tumor vs. normal), and bubble size is proportional to FDR significance. (<b>b</b>,<b>c</b>) Boxplots display the expression levels of YY1, LAG-3, and CD274 between tumor and normal tissues across multiple cancers. A detailed example focusing on lung cancer is provided in panel (<b>c</b>), highlighting the differential expression patterns that may suggest varying roles in tumorigenesis.</p>
Full article ">Figure 6
<p>Correlation between YY1, CD274 and LAG-3 expression and immune cell infiltration in squamous cell lung carcinoma (LUSC) (<b>a</b>) and breast cancer (BRCA) (<b>b</b>). The Spearman’s test was used for correlations. The infiltrates of 24 immune cells were quantified using ImmuCellAI. Bubble size correlates with FDR significance. The black outline border indicates FDR ≤ 0.05. (<b>c</b>) Each gene’s mRNA expression was correlated with a specific immune cell’s infiltrates using scatter plots with a fitting line.</p>
Full article ">Figure 7
<p>Expression of YY1 and LAG-3 in five independent scRNA-seq datasets of breast cancer (GSE110686, GSE114727_10X, GSE114727_inDrop, GSE176078 and EMTAB8107). The global-scaling normalization method (‘NormalizeData’ function) in Seurat was used to scale the raw counts (UMI) in each cell to 10,000, and to log-transform the results. YY1 and LAG-3 expression levels were calculated in log2(TPM/10+1) values and displayed using UMAP.</p>
Full article ">Figure 8
<p>Expression of YY1 and LAG3 across different immune cells in breast cancer, using multiple GEO datasets. CD8<sup>+</sup> T cells express higher levels of YY1 compared to LAG-3.</p>
Full article ">Figure 9
<p>The GDC TCGA Breast Cancer (BRCA) dataset on the UCSC Xena browser was explored for LAG-3 and FGL1 methylation. (<b>a</b>) Electrophoresis result (2% agarose gel) of two methylation-specific PCR amplicons for LAG-3 and FGL1 (methylated and unmethylated DNA). (<b>b</b>) In silico analysis of LAG-3 promoter methylation using the beta values of specific markers (Illumina Human Methylation 450) shows that LAG-3 promoter is hypermethylated in breast cancer. (<b>c</b>) The Kaplan–Meier curves depict that breast cancer patients with LAG-3 hypermethylation (red curve) have better overall survival compared to those with LAG-3 hypomethylation (white curve) (<span class="html-italic">p</span> &lt; 0.05, Log-rank test). (<b>d</b>) In silico analysis shows no significant methylation levels in the promoter region of FGL1. (<b>e</b>) The Kaplan–Meier curves depict no difference in the overall survival between FLG1 hyper- and hypo-methylated breast cancer patients (<span class="html-italic">p</span> &gt; 0.05, Log-rank test). (<b>f</b>) Proposed model for LAG-3-expressing breast tumors (LAG-3 hyper-methylated), which could be targeted with Relatimab (anti-LAG-3) alone or in combination with anti-PD-1/PD-L1.</p>
Full article ">
56 pages, 2782 KiB  
Review
Plant-Derived Anti-Cancer Therapeutics and Biopharmaceuticals
by Ghyda Murad Hashim, Mehdi Shahgolzari, Kathleen Hefferon, Afagh Yavari and Srividhya Venkataraman
Bioengineering 2025, 12(1), 7; https://doi.org/10.3390/bioengineering12010007 - 25 Dec 2024
Abstract
In spite of significant advancements in diagnosis and treatment, cancer remains one of the major threats to human health due to its ability to cause disease with high morbidity and mortality. A multifactorial and multitargeted approach is required towards intervention of the multitude [...] Read more.
In spite of significant advancements in diagnosis and treatment, cancer remains one of the major threats to human health due to its ability to cause disease with high morbidity and mortality. A multifactorial and multitargeted approach is required towards intervention of the multitude of signaling pathways associated with carcinogenesis inclusive of angiogenesis and metastasis. In this context, plants provide an immense source of phytotherapeutics that show great promise as anticancer drugs. There is increasing epidemiological data indicating that diets rich in vegetables and fruits could decrease the risks of certain cancers. Several studies have proved that natural plant polyphenols, such as flavonoids, lignans, phenolic acids, alkaloids, phenylpropanoids, isoprenoids, terpenes, and stilbenes, could be used in anticancer prophylaxis and therapeutics by recruitment of mechanisms inclusive of antioxidant and anti-inflammatory activities and modulation of several molecular events associated with carcinogenesis. The current review discusses the anticancer activities of principal phytochemicals with focus on signaling circuits towards targeted cancer prophylaxis and therapy. Also addressed are plant-derived anti-cancer vaccines, nanoparticles, monoclonal antibodies, and immunotherapies. This review article brings to light the importance of plants and plant-based platforms as invaluable, low-cost sources of anti-cancer molecules of particular applicability in resource-poor developing countries. Full article
(This article belongs to the Section Biomedical Engineering and Biomaterials)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The chemical structures of some prominent natural alkaloids and their semisynthetic derivatives serve as effective agents in combating cancer. Reproduced from an open-access source Dhyani et al., 2022 [<a href="#B151-bioengineering-12-00007" class="html-bibr">151</a>].</p>
Full article ">Figure 2
<p>The phototherapy mechanism of action. In photodynamic therapy (PDT), photosensitizers (PS) absorb light, transitioning to an excited state. This leads to two pathways: PDT Type I, where the PS reacts with biomolecules to create reactive oxygen species (ROS), and PDT Type II, where the PS transfers energy directly to oxygen, producing ROS. ROS exhibits high oxidizing power, causing cytotoxic effects primarily near their site of generation due to their short lifespan. PS* refers to the photosensitizer’s excited state. Reproduced from an open access source Pivetta et al., 2021 [<a href="#B212-bioengineering-12-00007" class="html-bibr">212</a>].</p>
Full article ">Figure 3
<p>PVNPs as delivery therapeutic and imaging agents in cancer. (<b>A</b>) Tobacco mosaic virus (TMV) for the targeted delivery of cisplatin in Pt-resistant ovarian cancer cells [<a href="#B312-bioengineering-12-00007" class="html-bibr">312</a>] (Reprinted/Adapted with permission from [<a href="#B272-bioengineering-12-00007" class="html-bibr">272</a>] Copyright© 2018, American Chemical Society. (<b>B</b>) The preparative process for potato virus X (PVX)-HisTRAIL by coordinating the bond between a Ni-nitrilotriacetic (NTA) group on the virus; the His-tag at the N-terminus of HisTRAIL is shown with a purple triangle. Multivalent display of HisTRAIL on the elongated PVX particle permits proper binding on death receptors DR4/5 (the trimers with blue color) for activating the caspase-dependent apoptosis in cancerous cells [<a href="#B313-bioengineering-12-00007" class="html-bibr">313</a>] (Reprinted/Adapted with permission from [<a href="#B273-bioengineering-12-00007" class="html-bibr">273</a>] Copyright© 2019, American Chemical Society). (<b>C</b>) miR-181a is an important target for ovarian cancer therapy. qPCR data and cancer cell migration assays demonstrated higher knockdown efficacy when anti-miR-181a oligonucleotides were encapsulated and delivered using the VLPs resulting in reduced cancer cell invasiveness [<a href="#B314-bioengineering-12-00007" class="html-bibr">314</a>] [Adapted from open access source: 274 Citation needed]. (<b>D</b>) Schematic illustration of Gd-Cy5.5-PhMV-mPEG NPs for cancer imaging. In vivo NIR fluorescence images of PC-3 prostate tumors in athymic nude mice after the intravenous injection of Gd-Cy5.5-PhMV-DGEA [<a href="#B315-bioengineering-12-00007" class="html-bibr">315</a>] [Adapted from open access source 275: Citation needed].</p>
Full article ">Figure 4
<p>PVNPs in cancer immune and combinational therapy (<b>A</b>) Intratumoral administration of plant-derived Cowpea mosaic virus (CPMV) nanoparticles as an in situ vaccine overcomes the local immunosuppression and stimulates a potent anti-tumor response in several mouse cancer models and canine patients [<a href="#B349-bioengineering-12-00007" class="html-bibr">349</a>] (Adapted from open access source: 309, Citation needed). (<b>B</b>) The PhMV-based anti-HER2 vaccine PhMV-CH401, demonstrated efficacy as an anti-HER2 cancer vaccine. Our studies highlight that VLPs derived from PhMV are a promising platform to develop cancer vaccines [<a href="#B350-bioengineering-12-00007" class="html-bibr">350</a>] (Adapted from open access source: 310, Citation needed). (<b>C</b>) Schematic diagram of preparing CCMV VLPs containing ODN 1826 (CCMV-ODN1826) for cancer therapy [<a href="#B315-bioengineering-12-00007" class="html-bibr">315</a>] (Adapted from open access source: 275, Citation needed). (<b>D</b>) Photothermal immunotherapy of melanoma using TLR-7 agonist laden TMV with polydopamine coat [<a href="#B325-bioengineering-12-00007" class="html-bibr">325</a>]. (Adapted from open access source: 285, Citation needed). Statistical significance was measured by one-way ANOVA with Tukey’s 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; 0.001. ns refers to not significant.</p>
Full article ">
19 pages, 1149 KiB  
Article
Rat as a Predictive Model for Human Clearance and Bioavailability of Monoclonal Antibodies
by Jason D. Robarge, Kevin M. Budge, Lucy Her, Andrea M. Patterson and Patricia Brown-Augsburger
Antibodies 2025, 14(1), 2; https://doi.org/10.3390/antib14010002 - 24 Dec 2024
Abstract
Background: The prediction of human clearance (CL) and subcutaneous (SC) bioavailability is a critical aspect of monoclonal antibody (mAb) selection for clinical development. While monkeys are a well-accepted model for predicting human CL, other preclinical species have been less-thoroughly explored. Unlike CL, predicting [...] Read more.
Background: The prediction of human clearance (CL) and subcutaneous (SC) bioavailability is a critical aspect of monoclonal antibody (mAb) selection for clinical development. While monkeys are a well-accepted model for predicting human CL, other preclinical species have been less-thoroughly explored. Unlike CL, predicting the bioavailability of SC administered mAbs in humans remains challenging as contributing factors are not well understood, and preclinical models have not been systematically evaluated. Methods: Non-clinical and clinical pharmacokinetic (PK) parameters were mined from public and internal sources for rats, cynomolgus monkeys, and humans. Intravenous (IV) and SC PK was determined in Sprague Dawley rats for fourteen mAbs without existing PK data. Together, we obtained cross-species data for 25 mAbs to evaluate CL and SC bioavailability relationships among rats, monkeys, and humans. Results: Rat and monkey CL significantly correlated with human CL and supported the use of species-specific exponents for body-weight-based allometric scaling. Notably, rat SC bioavailability significantly correlated with human SC bioavailability, while monkey SC bioavailability did not. Bioavailability also correlated with clearance. Conclusions: The rat model enables an early assessment of mAb PK properties, allowing discrimination among molecules in the discovery pipeline and prediction of human PK. Importantly, rat SC bioavailability significantly correlated with human SC bioavailability, which has not been observed with other species. Rats are cost-effective and efficient relative to monkeys and provide a valuable tool for pharmacokinetic predictions in therapeutic antibody discovery. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Weight-normalized clearance (CL) relationships between humans and pre-clinical species. (<b>a</b>) CL in humans versus CL in monkeys for 22 mAbs; (<b>b</b>) CL in humans versus CL in rats for 23 mAbs. Solid black lines represent the unity line, while dashed lines represent 0.5 and 2-fold change from the unity line.</p>
Full article ">Figure 2
<p>Goodness-of-fit plots and scaled human clearance (CL) for monkey-to-human (<b>a</b>–<b>c</b>) and rat-to-human (<b>d</b>–<b>f</b>) allometric CL models. Observed versus predicted CL in monkeys (<b>a</b>) and humans (<b>b</b>) from the monkey-to-human model. (<b>c</b>) Observed human CL versus human CL scaled from monkeys using an allometric exponent of 0.84. Observed versus predicted CL in rats (<b>d</b>) and humans (<b>e</b>) from the rat-to-human model. (<b>f</b>) Observed human CL versus human CL scaled from rats using an allometric exponent of 0.92. Solid lines represent the unity line, while dashed lines represent 2-fold change in slope from the unity line.</p>
Full article ">Figure 3
<p>Subcutaneous bioavailability (SC%F) relationships between humans and pre-clinical species. (<b>A</b>) SC%F in humans versus SC%F in monkeys for 14 mAbs. (<b>B</b>) SC%F in humans versus SC%F in rats for 13 mAbs. Solid black lines represent the unity line, while dashed black lines represent a 1.5-fold change in slope from the unity line. Dashed blue lines and equations represent linear regression of the data points.</p>
Full article ">Figure 4
<p>Correlation analysis between weight-normalized clearance (CL) and subcutaneous bioavailability (SC%F) in (<b>A</b>) humans, (<b>B</b>) monkeys, and (<b>C</b>) rats. Linear regression lines and analyses are shown on each graph.</p>
Full article ">Figure A1
<p>(<b>a</b>) Weight-normalized clearance for all mAbs in the dataset across species. (<b>b</b>) Subcutaneous bioavailability (%) for all mAbs in the dataset across species. Data are presented as mean ± standard deviation.</p>
Full article ">
22 pages, 2121 KiB  
Review
Decoding NAD+ Metabolism in COVID-19: Implications for Immune Modulation and Therapy
by Shixu Song, Jialing Gan, Qiuyue Long, Zhancheng Gao and Yali Zheng
Vaccines 2025, 13(1), 1; https://doi.org/10.3390/vaccines13010001 - 24 Dec 2024
Abstract
The persistent threat of COVID-19, particularly with the emergence of new variants, underscores the urgency for innovative therapeutic strategies beyond conventional antiviral treatments. Current immunotherapies, including IL-6/IL-6R monoclonal antibodies and JAK inhibitors, exhibit suboptimal efficacy, necessitating alternative approaches. Our review delves into the [...] Read more.
The persistent threat of COVID-19, particularly with the emergence of new variants, underscores the urgency for innovative therapeutic strategies beyond conventional antiviral treatments. Current immunotherapies, including IL-6/IL-6R monoclonal antibodies and JAK inhibitors, exhibit suboptimal efficacy, necessitating alternative approaches. Our review delves into the significance of NAD+ metabolism in COVID-19 pathology, marked by decreased NAD+ levels and upregulated NAD+-consuming enzymes such as CD38 and poly (ADP-ribose) polymerases (PARPs). Recognizing NAD+’s pivotal role in energy metabolism and immune modulation, we propose modulating NAD+ homeostasis could bolster the host’s defensive capabilities against the virus. The article reviews the scientific rationale behind targeting NAD+ pathways for therapeutic benefit, utilizing strategies such as NAD+ precursor supplementation and enzyme inhibition to modulate immune function. While preliminary data are encouraging, the challenge lies in optimizing these interventions for clinical use. Future research should aim to unravel the intricate roles of key metabolites and enzymes in NAD+ metabolism and to elucidate their specific mechanisms of action. This will be essential for developing targeted NAD+ therapies, potentially transforming the management of COVID-19 and setting a precedent for addressing other infectious diseases. Full article
(This article belongs to the Special Issue Next-Generation Vaccine and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p><b>Overview of NAD+ biosynthesis and degradation pathways.</b> The upper panel presents three pathways involved in NAD+ biosynthesis: the de novo pathway, the Preiss-Handler pathway, and the salvage pathway. In the de novo pathway, tryptophan (Trp) is converted into quinolinic acid (QA), which is processed into nicotinic acid mononucleotide (NAMN) by QPRT, eventually leading to NAD+ synthesis. The Preiss-Handler pathway converts nicotinic acid (NA) into NAMN through NAPRT, with subsequent conversion into NAD+ via NMNAT and NADS enzymes. The salvage pathway involves nicotinamide riboside (NR) being converted to nicotinamide mononucleotide (NMN) by NRK, which is then transformed into NAD+ by NMNAT. The lower panel illustrates the degradation and utilization of NAD+ by enzymes like sirtuins (SIRTs), poly (ADP-ribose) polymerases (PARPs), and CD38. These enzymes regulate diverse cellular processes such as epigenetic modifications, DNA repair, immune response, and calcium signaling. NAD+ is hydrolyzed by these enzymes, producing nicotinamide (NAM) as a by-product, which can be recycled back into the salvage pathway via NAMPT.</p>
Full article ">Figure 2
<p><b>Impact of changes in NAD+ metabolism on the immune response in COVID-19 infection.</b> Upon entry of SARS-CoV-2 into host cells via the ACE2 receptor, the viral infection induces DNA damage that activates PARPs, leading to increased NAD+ consumption and further cellular damage, thereby compromising immune cell resilience. This NAD+ depletion reduces SIRT activity, impairing its anti-inflammatory regulatory function and triggering a surge of cytokines and chemokines that disrupt the innate immune balance. Concurrently, upregulation of CD38 expression exacerbates NAD+ depletion, promoting inflammation through calcium signaling to enhance innate immunity while converting extracellular NAD+ into adenosine, suppressing adaptive immune responses.</p>
Full article ">Figure 3
<p><b>CD38 and immune checkpoint expression patterns in T cell subsets of COVID-19 patients:</b> (<b>A</b>) Expression of CD38 and FOXP3 in different T cell subsets. (<b>B</b>) Expression of immune checkpoint molecules in different T cell subsets.</p>
Full article ">
13 pages, 4535 KiB  
Article
Tocilizumab in COVID-19: A Double-Edged Sword?
by Bartosz Kudliński, Jacek Zawadzki, Wiktoria Kulińska, Jagoda Kania, Magdalena Murkos, Marta Stolińska, Dominika Zgoła, Anna Noga and Paweł Nowak
Biomedicines 2024, 12(12), 2924; https://doi.org/10.3390/biomedicines12122924 - 23 Dec 2024
Abstract
Background/Objectives: SARS-CoV-2 was responsible for the global pandemic. Approximately 10–15% of patients with COVID-19 developed respiratory failure with adult acute respiratory distress syndrome (ARDS), which required treatment in the Intensive Care Unit (ICU). The cytokine storm observed in severe COVID-19 was frequently handled [...] Read more.
Background/Objectives: SARS-CoV-2 was responsible for the global pandemic. Approximately 10–15% of patients with COVID-19 developed respiratory failure with adult acute respiratory distress syndrome (ARDS), which required treatment in the Intensive Care Unit (ICU). The cytokine storm observed in severe COVID-19 was frequently handled with steroids. Synergically, tocilizumab, an anti-interleukin-6 receptor monoclonal antibody, gained popularity as a cytokine storm-suppressing agent. However, immunosuppression was proven to increase the predisposition to infections with resistant bacteria. Our study aimed to assess the relationship between positive tests for secondary infections and the survival of patients with severe COVID-19-attributed ARDS treated with immunosuppressive agents. Methods: This study included 342 patients qualified for the ICU and mechanical ventilation (MV). The patients were divided based on the type of immunomodulating therapy and the culture tests results. Results: The results showed the highest survival rate among patients <61 years, favoring the combined treatment (tocilizumab + steroids). Atrial fibrillation (AF) and coronary heart disease (CHD) correlated with a lower survival rate than other comorbidities. Tocilizumab was associated with an increased risk of positive pathogen cultures, which could potentially cause secondary infections; however, the survival rate among these patients was higher. Conclusions: MV and ICU procedures as well as the application of tocilizumab significantly decreased the mortality rate in patients with severe COVID-19-related ARDS. The suppression of cytokine storms played a crucial role in survival. Tocilizumab was found to be both efficient and safe despite the ‘side effect’ of the increased risk of positive results for secondary infections. Full article
Show Figures

Figure 1

Figure 1
<p>The probability of survival of COVID-19 patients in the ICU in relation to sex, age, and therapy described by the Kaplan–Meyer curves: (<b>A</b>) Survival probability by sex, with a trend favoring females, though without sufficient statistical significance. (<b>B</b>) Survival probability by therapy, favoring tocilizumab + steroids with marginal statistical significance (<span class="html-italic">p</span> = 0.051). (<b>C</b>) Survival probability by age, with a trend favoring younger patients, which is statistically significant (<span class="html-italic">p</span> &lt; 0.001). Age (years); types of therapy used: steroids, tocilizumab + steroids.</p>
Full article ">Figure 2
<p>Age comparison to treatment methods described by the Kaplan–Meyer curves. The differences between the curves are statistically significant (<span class="html-italic">p</span> &lt; 0.05). Regardless of the therapy used, younger patients have a higher survival chance than older ones. Tocilizumab combined with steroids appears to improve outcomes mostly in middle-aged patients (41–60 years), but the effect is less pronounced in the oldest cohort (≥61 years).</p>
Full article ">Figure 3
<p>Survival probability of patients with comorbidities such as AF and CHD described by the Kaplan–Meyer curves. The groups of patients suffering from the diseases are smaller in size, though there is a high statistical difference in survival probability favoring patients without CHD (<b>B</b>) and AF (<b>A</b>) in severe COVID-19 (respectively, <span class="html-italic">p</span> = 0.025, <span class="html-italic">p</span> = 0.008). AF—atrial fibrillation, CHD—coronary heart disease.</p>
Full article ">Figure 4
<p>Quality chart of patients showing proportions of age, presence of AF, CHD, and mortality with regard to applied therapy. The therapy including tocilizumab + steroids was more frequently used in patients &lt;40 y.o., without AF and/or CHD. The difference between the therapies (toci + steroids vs. steroids) was greater in patients who died (29.3% vs. 70.7%) compared to patients who survived (42.9% vs. 57.1%). AF—atrial fibrillation, CHD—coronary heart disease.</p>
Full article ">Figure 5
<p>Potential secondary infections in all patients described by the Kaplan–Meyer curves: (<b>A</b>) The survival probability regarding NDM infection favoring patients with NDM, which is statistically significant (<span class="html-italic">p</span> &lt; 0.0001). The statistical significance is marginal (<span class="html-italic">p</span> = 0.051) in the case of <span class="html-italic">A. baumanii</span> (<b>B</b>) with the same trend, though there is no statistical significance in mortality prediction in the case of VRE and GRE (<b>C</b>,<b>D</b>). The patients with NDM are also the most proportional compared to the patients without secondary infection regarding numbers, meaning it was the most popular SI. NDM—<span class="html-italic">Klebsiella pneumoniae</span> New Delhi metallo-β-lactamase-resistant, VRE—Vancomycin-resistant Enterococcus, <span class="html-italic">A. baumanii—Acinetobacter baumanii</span>, and GRE—Glycopeptide-resistant Enterococcus.</p>
Full article ">Figure 6
<p>Probability of survival in potential secondary infections with <span class="html-italic">A. baumanii</span> and NDM and the therapy used, described by the Kaplan–Meyer curves. There is a statistical significance in the case of mortality prediction in NDM (<b>B</b>) and <span class="html-italic">A. baumanii</span> (<b>A</b>) in regard to the therapy used (<span class="html-italic">p</span> &lt; 0.009, <span class="html-italic">p</span> &lt; 0.02), favoring patients treated with tocilizumab + steroids and having NDM (+) or <span class="html-italic">A. baumanii</span> (+). On the contrary, patients treated with steroids only, with no secondary infections, were more likely to die. NDM—<span class="html-italic">Klebsiella pneumoniae</span> New Delhi metallo-β-lactamase-resistant, and <span class="html-italic">A. baumanii—Acinetobacter baumanii.</span></p>
Full article ">
16 pages, 1017 KiB  
Article
Vedolizumab Clearance as a Surrogate Marker for Remission in Inflammatory Bowel Disease Patients: Insights from Real-World Pharmacokinetics
by Srđan Marković, Đorđe Kralj, Petar Svorcan, Tamara Knežević Ivanovski, Olga Odanović, Sanja Obradović, Ana Homšek, Marija Jovanović, Rada Savić and Katarina M. Vučićević
Pharmaceutics 2024, 16(12), 1629; https://doi.org/10.3390/pharmaceutics16121629 - 23 Dec 2024
Abstract
Background/Objectives: Vedolizumab (VDZ) is approved in the treatment of patients with moderate to severe ulcerative colitis (UC) or Crohn’s disease (CD). VDZ exhibits considerable variability in its pharmacokinetic (PK) profile, and its exposure-response relationship is not yet fully understood. The aim was to [...] Read more.
Background/Objectives: Vedolizumab (VDZ) is approved in the treatment of patients with moderate to severe ulcerative colitis (UC) or Crohn’s disease (CD). VDZ exhibits considerable variability in its pharmacokinetic (PK) profile, and its exposure-response relationship is not yet fully understood. The aim was to investigate the variability in VDZ trough levels and PK parameters, to assess the relationship between VDZ PK and biochemical response, as well as clinical and endoscopic outcomes. Methods: We included 61 UC and 45 CD patients. Patients’ data and trough VDZ concentrations were retrospectively obtained. Population PK analysis was performed using non-linear mixed-effects modelling with NONMEM (version 7.5). Graphs and statistical analyses were performed using R (version 4.1.3). Results: In total, 116 trough VDZ concentrations from 106 patients were described by a two-compartment model. For a typical patient, clearance (CL) was estimated at 0.159 L/day, while in patients previously treated with anti-TNFα agents, VDZ CL increased by 26.4% on average. In univariate binary logistic regression, VDZ trough concentration was not statistically significant predictor of remission, whereas CL was. Moreover, combined CL and faecal calprotectin (FCP) were a statistically significant predictors of remission. The hazard ratio (HR) for CL above 0.1886 L/day was 0.35 (p = 0.05) and for FCP below 250 µg/g was 2.66 (p = 0.02) in a time-to-event analysis. Conclusions: Our population PK model incorporates the effect of prior anti-TNFα agents on CL, suggesting its association with more severe forms of IBD. VDZ CL emerged as a more robust and clinically relevant predictor of remission in IBD patients than trough concentration. Full article
(This article belongs to the Special Issue Population Pharmacokinetics and Its Clinical Applications)
Show Figures

Figure 1

Figure 1
<p>Boxplot of vedolizumab (VDZ) trough concentrations in patients receiving 300 mg as a 30-min infusion during the induction period and every 4, 6 or 8 weeks onwards. The dot represents the mean value, the central line within each box represents the median, and the edges of the box denote the interquartile range (IQR). The whiskers extend to values within 1.5 times the IQR and individual points are outliers beyond this range (* <span class="html-italic">p</span> ˂ 0.05, ** <span class="html-italic">p</span> ˂ 0.01, *** <span class="html-italic">p</span> ˂ 0.001).</p>
Full article ">Figure 2
<p>Distribution of individual vedolizumab (VDZ) clearance (CL) estimated from the final population pharmacokinetic (PopPK) model in patients with and without prior anti-TNFα therapy. The dot represents the mean value, the central line within each box represents the median CL, and the edges of the box denote the interquartile range (IQR). The whiskers extend to values within 1.5 times the IQR and individual points are outliers beyond this range.</p>
Full article ">Figure 3
<p>Predicted probability of (<b>A</b>) clinical and (<b>B</b>) endoscopic remission with vedolizumab (VDZ) clearance (CL) based on faecal calprotectin (FCP) values.</p>
Full article ">Figure 4
<p>(<b>A</b>) Time to endoscopic remission based on vedolizumab (VDZ) clearance (CL) and faecal calprotectin (FCP) categories. (<b>B</b>) Forest plot illustrates the estimated effects of VDZ CL and FCP on endoscopic remission where each horizontal line represents a 95% confidence interval (CI).</p>
Full article ">
14 pages, 2047 KiB  
Article
Enhancement of Human Immunodeficiency Virus-Specific CD8+ T Cell Responses with TIGIT Blockade Involves Trogocytosis
by Nazanin Ghasemi, Kayla A. Holder, Danielle P. Ings and Michael D. Grant
Pathogens 2024, 13(12), 1137; https://doi.org/10.3390/pathogens13121137 - 23 Dec 2024
Abstract
Natural killer (NK) and CD8+ T cell function is compromised in human immunodeficiency virus type 1 (HIV-1) infection by increased expression of inhibitory receptors such as TIGIT (T cell immunoreceptor with Ig and ITIM domains). Blocking inhibitory receptors or their ligands with [...] Read more.
Natural killer (NK) and CD8+ T cell function is compromised in human immunodeficiency virus type 1 (HIV-1) infection by increased expression of inhibitory receptors such as TIGIT (T cell immunoreceptor with Ig and ITIM domains). Blocking inhibitory receptors or their ligands with monoclonal antibodies (mAb) has potential to improve antiviral immunity in general and facilitate HIV eradication strategies. We assessed the impact of TIGIT engagement and blockade on cytotoxicity, degranulation, and interferon-gamma (IFN-γ) production by CD8+ T cells from persons living with HIV (PLWH). The effect of TIGIT engagement on non-specific anti-CD3-redirected cytotoxicity was assessed in redirected cytotoxicity assays, and the effect of TIGIT blockade on HIV-specific CD8+ T cell responses was assessed by flow cytometry. In 14/19 cases where peripheral blood mononuclear cells (PBMC) mediated >10% redirected cytotoxicity, TIGIT engagement reduced the level of cytotoxicity to <90% of control values. We selected PLWH with >1000 HIV Gag or Nef-specific IFN-γ spot forming cells per million PBMC to quantify the effects of TIGIT blockade on HIV-specific CD8+ T cell responses by flow cytometry. Cell surface TIGIT expression decreased on CD8+ T cells from 23/40 PLWH following TIGIT blockade and this loss was associated with increased anti-TIGIT mAb fluorescence on monocytes. In total, 6 of these 23 PLWH had enhanced HIV-specific CD8+ T cell degranulation and IFN-γ production with TIGIT blockade, compared to 0/17 with no decrease in cell surface TIGIT expression. Reduced CD8+ T cell TIGIT expression with TIGIT blockade involved trogocytosis by circulating monocytes, suggesting that an effector monocyte population and intact fragment crystallizable (Fc) functions are required for mAb-based TIGIT blockade to effectively enhance HIV-specific CD8+ T cell responses. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Figure 1

Figure 1
<p>Effect of TIGIT engagement on T cell-mediated cytotoxicity. (<b>a</b>) Diagram illustrating strategy employed to enact TIGIT engagement on freshly isolated T cells triggered by anti-CD3 to lyse P815 cells (created with BioRender.com). (<b>b</b>) Comparison of anti-CD3-triggered lysis of P815 targets by PBMC from PLWH in the presence of anti-TIGIT or isotype control. Cases where specific lysis was reduced by &gt;1/10 of baseline values by TIGIT engagement compared to the isotype control are shown with red lines and the probability of a significant reduction in specific lysis for the overall group following TIGIT engagement was calculated. (** <span class="html-italic">p</span> &lt; 0.01, Student’s paired <span class="html-italic">t</span> test).</p>
Full article ">Figure 2
<p>Impact of TIGIT blockade on CD8<sup>+</sup> T cell TIGIT expression. To assess the effects of TIGIT blockade on HIV-specific CD8<sup>+</sup> T cell function, PBMC were labeled with fluorescence conjugated anti-TIGIT mAb for 30 min before 5 h incubation with HIV Gag or Nef peptides. Additional labeled anti-TIGIT mAb was added after the 5 h incubation period with cell surface staining for CD3, CD4, and CD8. Our gating strategy for analysis of CD8<sup>+</sup> T cell TIGIT expression is shown (<b>a</b>–<b>c</b>) with representative results in (<b>d</b>,<b>e</b>). Summary results for 23 subjects losing &gt;1/10th of TIGIT expression from their CD8<sup>+</sup> T cells following TIGIT blockade are shown graphically in (<b>f</b>).</p>
Full article ">Figure 3
<p>Flow cytometry analysis of HIV-specific CD8<sup>+</sup> T cell activation and effect of TIGIT blockade. The gating strategy for analyzing effects of TIGIT blockade on IFN-γ production and CD107a expression by CD8<sup>+</sup> T cells stimulated with HIV Gag and Nef peptides is shown with a representative example of an HIV-specific CD8<sup>+</sup> T cell response enhanced by TIGIT blockade (<b>a</b>–<b>f</b>). Summary graphs of effect of TIGIT blockade on (<b>g</b>) IFN-γ production and (<b>h</b>) CD107a expression by CD8<sup>+</sup> T cells from responders to TIGIT blockade with an increase &gt;10% above that seen with isotype control treatment.</p>
Full article ">Figure 4
<p>Detection of monocyte-mediated trogocytosis of TIGIT from CD8<sup>+</sup> T cells. After five-hour incubation of PBMC with either fluorescent anti-TIGIT mAb or isotype control, anti-TIGIT, and isotype control-treated cells were surface stained with fluorescent anti-TIGIT mAb and monocytes gated for analysis as shown in (<b>a</b>,<b>b</b>). A representative example of the gain in anti-TIGIT mAb fluorescence on monocytes following TIGIT blockade is shown in (<b>c</b>,<b>d</b>). Letters A, B, and C within the flow cytometry plot frames refer to the gated population analyzed and the percentages above markers in (<b>c</b>,<b>d</b>) indicate the percent of cells in that gate positive for anti-TIGIT fluorescence. Monocyte identity was confirmed by 0% CD3 expression and &gt;90% CD14 expression on cells gated in (<b>b</b>). (<b>e</b>) Summary graph of the results obtained with PBMC from eight PLWH responders to TIGIT blockade in terms of loss of CD8<sup>+</sup> T cell TIGIT expression. (<b>f</b>) TIGIT blockade was carried out in PBMC separated from untreated PBMC with a semi-permeable membrane with changes in anti-TIGIT mAb fluorescence in the monocyte population shown after 5 h incubation with or without cell contact allowed.</p>
Full article ">
11 pages, 755 KiB  
Article
Limited Efficacy of Anti-EGFR Monoclonal Antibodies in Colorectal Cancer Patients with Rare RAS Variants: Analysis of the C-CAT Database
by Shuhei Suzuki, Yosuke Saito, Koki Saito, Yuta Yamada, Koshi Takahashi, Ryosuke Kumanishi, Tadahisa Fukui and Takashi Yoshioka
Curr. Issues Mol. Biol. 2024, 46(12), 14476-14486; https://doi.org/10.3390/cimb46120869 - 23 Dec 2024
Abstract
Epidermal growth factor receptor (EGFR) inhibition is crucial in treating RAS wild-type metastatic colorectal cancer, yet current testing methods may miss rare RAS variants affecting treatment efficacy. We analyzed 4122 colorectal cancer patients receiving anti-EGFR antibodies from the Center for Cancer Genomics and [...] Read more.
Epidermal growth factor receptor (EGFR) inhibition is crucial in treating RAS wild-type metastatic colorectal cancer, yet current testing methods may miss rare RAS variants affecting treatment efficacy. We analyzed 4122 colorectal cancer patients receiving anti-EGFR antibodies from the Center for Cancer Genomics and Advanced Therapeutics database, identifying 54 patients (1.3%) with rare RAS variants undetectable by standard testing. These patients showed significantly lower response rates to anti-EGFR therapy (28.3%) compared to RAS wild-type cases (44.6%, p = 0.003). Disease control rates were also lower in rare variant cases (60.9%) versus wild-type cases (80.0%). Most common rare variants included KRAS Q22K, A59E, and A11_G12insGA. Comprehensive genomic profiling revealed additional alterations in TP53 (90.7%), APC (87.0%), and non-V600E BRAF mutations (25.9%). Our findings suggest that rare RAS variants predict poor anti-EGFR therapy response, highlighting the potential benefit of comprehensive genomic profiling before treatment initiation. This study provides real-world evidence supporting the clinical relevance of rare RAS variants in treatment decision-making for colorectal cancer. Future studies should focus on developing cost-effective comprehensive testing strategies and evaluating alternative treatment approaches for patients with rare RAS variants. Full article
Show Figures

Figure 1

Figure 1
<p>Genomic alterations and anti-EGFR antibodies (Upper, cetuximab; Lower, Panitumumab) treatment responses of rare RAS variants cases registered in the Center for Cancer Genomics and Advanced Therapeutics. TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated.</p>
Full article ">Figure 1 Cont.
<p>Genomic alterations and anti-EGFR antibodies (Upper, cetuximab; Lower, Panitumumab) treatment responses of rare RAS variants cases registered in the Center for Cancer Genomics and Advanced Therapeutics. TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated.</p>
Full article ">Figure 1 Cont.
<p>Genomic alterations and anti-EGFR antibodies (Upper, cetuximab; Lower, Panitumumab) treatment responses of rare RAS variants cases registered in the Center for Cancer Genomics and Advanced Therapeutics. TMB: Tumor Mutational Burden; PR: Partial Response; SD: Stable Disease; PD: Progressive Disease; NE: not evaluated.</p>
Full article ">
12 pages, 2608 KiB  
Article
Construction of an Integration Vector with a Chimeric Signal Peptide for the Expression of Monoclonal Antibodies in Mammalian Cells
by Valentina S. Nesmeyanova, Daniil V. Shanshin, Denis E. Murashkin and Dmitriy N. Shcherbakov
Curr. Issues Mol. Biol. 2024, 46(12), 14464-14475; https://doi.org/10.3390/cimb46120868 - 22 Dec 2024
Viewed by 287
Abstract
Antibodies are complex protein structures, and producing them using eukaryotic expression systems presents significant challenges. One frequently overlooked aspect of expression vectors is the nucleotide sequence encoding the signal peptide, which plays a pivotal role in facilitating the secretion of recombinant proteins. This [...] Read more.
Antibodies are complex protein structures, and producing them using eukaryotic expression systems presents significant challenges. One frequently overlooked aspect of expression vectors is the nucleotide sequence encoding the signal peptide, which plays a pivotal role in facilitating the secretion of recombinant proteins. This study presents the development of an integrative vector, pVEAL3, for expressing full-length recombinant monoclonal antibodies in mammalian cells. The vector features a distinctive nucleotide sequence that encodes an artificial chimeric signal peptide with the following amino acid sequence: MMRTLILAVLLVYFCATVHC. Additionally, the vector incorporates several regulatory elements to enhance antibody expression, including the Gaussia luciferase signal sequence, internal ribosome entry site (IRES), P2A peptide, and a furin cleavage site. These elements coordinate to regulate the synthesis levels of the antibody chains. The analysis of clones obtained via transfection with the developed vector showed that over 95% of them secreted antibodies at levels significantly higher than those of the control. The immunochemical analysis of the chimeric antibody produced by the CHO-K1-10H10ch cell line confirmed the preservation of its functional activity. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram of the integration plasmid vector pVEAL3-10H10ch. 5′_SB and 3′_SB—transposase SB100X binding sites; CMV promoter—CMV promoter region; 176—nucleotide sequence encoding a hybrid signal peptide from luciferase (<span class="html-italic">Cypridina noctiluca</span>) and fibroin (<span class="html-italic">Dendrolimus spectabilis</span>), facilitating protein export from the cell; EMCV IRES—internal ribosome entry site; PuroR—nucleotide sequence encoding resistance to the antibiotic puromycin; SV40 poly (A) signal—nucleotide sequence stabilizing mRNA transcripts through polyadenylation; KanR—nucleotide sequence encoding resistance to the antibiotic kanamycin; ori—origin of replication. The complete nucleotide sequence of the developed vector is presented in <a href="#app1-cimb-46-00868" class="html-app">Figure S1</a>.</p>
Full article ">Figure 2
<p>Schematic representation of the expression cassette. Key design elements are highlighted in color. Key elements of the construct: 176—nucleotide sequence encoding a hybrid signal peptide from luciferase (<span class="html-italic">Cypridina noctiluca</span>) and fibroin (<span class="html-italic">Dendrolimus spectabilis</span>), facilitating protein export from the cell; Furin—nucleotide sequence encoding the proteolytic site for the cellular protease furin; P2A—nucleotide sequence encoding the self-cleaving P2A peptide; GL—nucleotide sequence encoding the Gaussia luciferase signal sequence, facilitating protein export from the cell; EMCV IRES—internal ribosome entry site. Additional elements: CMV promoter—CMV promoter region; Puro—nucleotide sequence encoding the antibiotic resistance factor for puromycin.</p>
Full article ">Figure 3
<p>Amino acid sequence of the chimeric signal peptide 176.</p>
Full article ">Figure 4
<p>Optical density (OD) values of culture supernatant samples from 10H10ch antibody clones. The positive control, with a murine 10H10 antibody, had an OD of 1.4. The negative control, with a casein protein, had an OD of 0.04.</p>
Full article ">Figure 5
<p>Electrophoresis in 15% SDS-PAGE. Lane 1—recombinant 10H10ch antibody under denaturing conditions (concentration ~10 µg/well); lane 2—murine 10H10 antibody under denaturing conditions (concentration ~20 µg/well); lane 3—molecular weight markers (250–10 kDa). The recombinant 10H10ch antibody was isolated using affinity chromatography. The murine 10H10 was isolated using a caprylic acid purification protocol from mouse ascites fluid.</p>
Full article ">Figure 6
<p>The results of the interaction of the recombinant 10H10ch and murine 10H10 antibodies with recombinant fragments of flavivirus envelope proteins are as follows: TEF1 represents the 1 + 2 domains of the E protein from tick-borne encephalitis virus; ZEF1 denotes the 1 + 2 domains of the E protein from Zika virus; WEF1 refers to the 1 + 2 domains of the E protein from West Nile virus; DEF1 comprises the 1 + 2 domains of the E protein from Dengue virus; and TNS1 indicates the non-structural protein 1 from tick-borne encephalitis virus.</p>
Full article ">
10 pages, 484 KiB  
Communication
Safety Concerns in Neurological Clinical Trials: A Challenge That the FDA Must Resolve
by Sarfaraz K. Niazi
Biomedicines 2024, 12(12), 2918; https://doi.org/10.3390/biomedicines12122918 - 22 Dec 2024
Viewed by 226
Abstract
Background: Monoclonal antibodies approved by the FDA, lecanemab, donanemab, and aducanumab, are failing to meet the expected efficacy to treat early Alzheimer’s disease, and aducanumab has been recalled. Methods: Recently, it was reported that the clinical trials of these antibodies may have [...] Read more.
Background: Monoclonal antibodies approved by the FDA, lecanemab, donanemab, and aducanumab, are failing to meet the expected efficacy to treat early Alzheimer’s disease, and aducanumab has been recalled. Methods: Recently, it was reported that the clinical trials of these antibodies may have violated patient’s rights and subjected them to high, likely lethal risk. The challenge with developing antibodies to treat neurological disorders is their poor blood–brain barrier (BBB) penetration if the antibody must enter the brain, resulting in almost negligible brain bioavailability, requiring high dosing that can be toxic. Results: The reported efficacy of these drugs should also be reviewed, considering the placebo effects, since all antibodies have shown severe side effects that are not prevented by the placebo responses. In this critical and urgent advice to the FDA, I am suggesting a guideline amendment to all clinical trials requiring proof of sufficient brain bioavailability at the site of action, where it is known. Conclusions: For antibodies to cross the blood–brain barrier, there are proven options such as conjugating with transferrin protein, making clinical trials in its absence more questionable. Full article
(This article belongs to the Special Issue Biomedical and Biochemical Basis of Neurodegenerative Diseases)
Show Figures

Figure 1

Figure 1
<p>Binding properties ΔG (kcal mol<sup>−1</sup>) of antibodies with amyloid-beta with transferrin conjugation and small and long linkers (created using <a href="https://www.cgl.ucsf.edu/chimera/" target="_blank">https://www.cgl.ucsf.edu/chimera/</a>; <a href="https://alphafoldserver.com/about" target="_blank">https://alphafoldserver.com/about</a>; and <a href="https://rascar.science.uu.nl/haddock2.4/" target="_blank">https://rascar.science.uu.nl/haddock2.4/</a>, accessed on 25 October 2024). The Gibbs free energy change (ΔG\Delta GΔG) in binding studies is closely related to the interactions between Interacting Components (ICs) and Non-Interacting Species (NIS), playing a critical role in determining the strength and stability of molecular interactions. Interacting Components (ICs) are species such as molecules or ions that engage in specific interactions like binding, forming complexes, or undergoing reactions. Electrostatic interactions, hydrogen bonds, van der Waals forces, and hydrophobic effects drive these interactions. When ICs bind to each other, they form a complex, and the ΔG\Delta GΔG of this binding process reflects the stability of the complex. A negative ΔG\Delta GΔG indicates a spontaneous and favorable interaction, meaning that the ICs have a strong affinity for each other and form a stable complex. The more negative the ΔG\Delta GΔG, the stronger the interaction between the ICs, such as in the case of charged–charged ICs, which often exhibit powerful interactions due to electrostatic forces, leading to a more negative ΔG\Delta GΔG. Charged–polar ICs, which exhibit a combination of electrostatic and dipole interactions, generally result in moderately negative ΔG\Delta GΔG. In contrast, polar–polar ICs, driven by hydrogen bonding and dipole–dipole interactions, also contribute to a moderately negative ΔG\Delta GΔG. Apolar–apolar ICs, driven by hydrophobic interactions, usually result in weaker (less negative) ΔG\Delta GΔG values than polar or charged interactions. The magnitude of ΔG\Delta GΔG for ICs reflects the strength and nature of their interactions, with stronger binding (more negative ΔG\Delta GΔG) leading to more stable complexes. This is particularly important in applications such as drug design, where optimizing binding affinity is crucial.</p>
Full article ">
23 pages, 29492 KiB  
Article
Suppression of Pathological Allergen-Specific B Cells by Protein-Engineered Molecules in a Mouse Model of Chronic House Dust Mite Allergy
by Nikola Ralchev, Silviya Bradyanova, Nikola Kerekov, Andrey Tchorbanov and Nikolina Mihaylova
Int. J. Mol. Sci. 2024, 25(24), 13661; https://doi.org/10.3390/ijms252413661 - 20 Dec 2024
Viewed by 254
Abstract
Der p1 is one of the major allergens causing house dust mite (HDM) allergy. Pathological Der p1-specific B cells play a key role in allergic inflammation as producers of allergen-specific antibodies. Crosslinking the inhibitory FcγRIIb with the B cell receptor triggers a high-affinity [...] Read more.
Der p1 is one of the major allergens causing house dust mite (HDM) allergy. Pathological Der p1-specific B cells play a key role in allergic inflammation as producers of allergen-specific antibodies. Crosslinking the inhibitory FcγRIIb with the B cell receptor triggers a high-affinity suppressive signal in B cells. Selective elimination of allergen-specific cells could potentially be achieved by administering chimeric molecules that combine, through protein engineering, the FcγRIIb-targeting monoclonal 2.4G2 antibody with the epitope-carrying Dp52–71 peptides from Der p1. We tested this hypothesis, in a chronic mouse model of HDM allergy induced in BalB/c mice, using FACS and ELISA assays, along with histopathological and correlational analyses. Dp52–71chimera treatment of HDM-challenged mice led to a decrease in serum anti-HDM IgG1 antibodies, a reduction in BALF β-hexosaminidase levels, a lowered number of SiglecFhigh CD11clow eosinophils, and an improved lung PAS score. Furthermore, we observed overexpression of FcγRIIb on the surface of CD19 cells in the lungs of HDM-challenged animals, which negatively correlated with the levels of lung alveolar macrophages, neutrophils, and BALF IL-13. Taken together, these results suggest that the use of FcγRIIb overexpression, combined with the expansion of chimeric protein technology to include more epitopes, could improve the outcome of inflammation. Full article
Show Figures

Figure 1

Figure 1
<p>The Dp52–71chimera binds to the FcγRIIb receptor on murine B cells and is recognized by epitope-specific serum IgG1 antibodies. (<b>A</b>) Dp52–71 chimera and irrelevant chimera binding on the surface of CD19 and CD3 cells was proved by FACS analysis. Splenocytes from healthy and HDM-challenged mice were incubated with both chimeras and secondary incubated with FITC-conjugated anti-rat IgG (left part). Summarized graph for the median fluorescent intensity (MFI) of anti-rat–FITC antibody fluorescence (right part). (<b>B</b>) Dp52–71 chimera competes with commercial 2.4G2-FITC antibody for the same receptor. The same splenocytes were pre-incubated with Dp52–71 chimera, irrelevant chimera, and pure 2.4G2 antibody, and secondary incubated with 2.4G2-FITC antibody (left part). Gated CD19 and CD3 cells were analyzed by FACS (left part). Summarized data of the 2.4G2-FITC binding to the FcγRIIb receptor on B cells (right part). (<b>C</b>). Dose-dependent inhibition of the 2.4G2-FITC binding to the FcγRIIb receptor. (<b>D</b>). Peptide recognition on the Dp52–71 chimera by serum IgG1 antibodies from HDM + Alum-sensitized mice analyzed by ELISA. Data are represented as mean ± SD of at least 3 mice per group. The differences between the groups were evaluated using a two-way ANOVA following Tukey’s multiple comparisons 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. Data are representative of at least 5 independent experiments.</p>
Full article ">Figure 2
<p>Scheme of the chronic HDM allergy model and treatment schedule (<b>A</b>). Serum levels of anti-HDM IgG (<b>B</b>), IgG1 (<b>C</b>), IgM (<b>D</b>), IgA (<b>E</b>), IgE (<b>G</b>), and total IgE antibodies (<b>F</b>) in healthy mice and HDM-challenged mice treated with PBS, Dp52–71 chimera, or irrelevant chimera, measured by ELISA. Data are shown as mean ± SD of 8–9 mice per group. <span class="html-italic">p</span> values are calculated using a one-way ANOVA following Tukey’s multiple comparisons test; * <span class="html-italic">p</span> &lt; 0.05; **** <span class="html-italic">p</span> &lt; 0.0001. Data are representative of at least 3 independent experiments.</p>
Full article ">Figure 3
<p>Protein analysis of BAL fluid performed by ELISA. BALF levels of total protein (<b>A</b>), β-hexosaminidase activity (<b>B</b>), IL-5 (<b>C</b>), IL-13 (<b>D</b>), HDM-specific IgG (<b>E</b>), IgG1 (<b>F</b>), IgM (<b>G</b>), IgA (<b>H</b>), and total IgE (<b>I</b>) were investigated in all groups. Data are shown as mean ± SD of 6–9 mice per group. The differences between the groups were evaluated using a one-way ANOVA, following Tukey’s multiple comparisons test; <span class="html-italic">p</span> values are indicated on the graphs: * <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; **** <span class="html-italic">p</span> &lt; 0.0001. Data are representative of at least 3 independent experiments.</p>
Full article ">Figure 4
<p>Total cells (per milliliter) isolated from spleen (<b>A</b>), lungs (<b>B</b>), and BAL (<b>C</b>). Differential staining of cells in BAL, defining macrophages (<b>D</b>,<b>H</b>), neutrophils (<b>E</b>,<b>I</b>), lymphocytes (<b>F</b>,<b>J</b>), and eosinophils (<b>G</b>,<b>K</b>), and expressed as percentages and cell count per ml of recovered BAL liquid. Data are shown as mean ± SD of 8–9 mice per group. The differences between the groups were evaluated using a one-way ANOVA followed by Tukey’s multiple comparisons test, or a Kruskal–Wallis test followed by Dunn’s multiple comparisons test, depending on the normality of the data; <span class="html-italic">p</span> values are indicated on the graphs: * <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; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Phenotyping of immune cells in the lungs by FACS analyses. (<b>A</b>) Characterization of myeloid cell types—alveolar macrophages (<b>I</b>), SiglecF<sup>high</sup> CD11c<sup>low</sup> (<b>III</b>), SiglecF<sup>med</sup> CD11c− (<b>IV</b>) and total eosinophils (<b>II</b>), and neutrophils (<b>V</b>)—presented as the percentage of the parent population. (<b>B</b>) Analysis of B and antibody-secreting cells—percentages of CD19 cells (<b>I</b>), CD19 CD80 cells (<b>II</b>), CD19 IgE cells (<b>III</b>), CD138 cells (<b>IV</b>), plasma cells (<b>V</b>), plasmablasts (<b>VI</b>), and CD32 mean fluorescent intensity (MFI) of CD19 (<b>VII</b>) and CD19 IgE cells (<b>VIII</b>). (<b>C</b>) Phenotyping of T cells—CD3 cells (<b>I</b>), CD3 CD4 cells (<b>II</b>), CD3 CD8 cells (<b>III</b>), CD4 CD69 − CD25 + cells (<b>IV</b>), CD4 CD69 + CD25 + cells (<b>V</b>), CD4 CD69 + CD25 − cells (<b>VI</b>), CD8 CD69 − CD25 + cells (<b>VII</b>), CD8 CD69 + CD25 − cells (<b>VIII</b>), and CD4 CD69 + CD25 + cells (<b>IX</b>). Data are shown as mean ± SD of 8–9 mice per group. The differences between the groups were evaluated using a one-way ANOVA followed by Tukey’s multiple comparisons, or a Kruskal–Wallis test followed by Dunn’s multiple comparisons test, depending on the normality of the data; <span class="html-italic">p</span> values are indicated on the graphs: * <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. Data are representative of at least 4 independent experiments.</p>
Full article ">Figure 6
<p>Histological analysis of lung pathology. Representative images and histological score of perivascular (<b>A</b>,<b>D</b>) and peribronchial (<b>B</b>,<b>E</b>) inflammation of H&amp;E-stained lung tissue. Periodic acid-Schiff (PAS) score (<b>F</b>) and representative images for mucus production (<b>C</b>). Scale bars, 250 µm. Data are shown as mean ± SD of 8–9 mice per group. <span class="html-italic">p</span> values were calculated using a one-way ANOVA followed by Tukey’s multiple comparisons test; <span class="html-italic">p</span> values are indicated on the graphs: *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001. Data are representative of at least 4 independent experiments.</p>
Full article ">Figure 7
<p>Correlation analysis of HDM allergy group. Correlation analysis of serum levels of anti-HDM IgG1 (<b>A</b>) and CD32 expression of B cells in the lungs (<b>C</b>) versus other parameters. Pearson (rp) and Spearman (rs) correlation coefficients and <span class="html-italic">p</span> values for Spearman (ps) and Pearson (pp) correlation analysis are indicated above each figure. (<b>B</b>) Correlation matrix of key immunological parameters achieved through Spearman’s 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; 0.001).</p>
Full article ">
16 pages, 5476 KiB  
Article
Development of a Colloidal Gold Immunochromatographic Assay Strip Using a Monoclonal Antibody for the Rapid Detection of Ofloxacin
by Xiaolan Li, Jin Huang, Na Li, Mahmoud Salah, Shuoning Guan, Wenwen Pan, Ziyi Wang, Xinghua Zhou and Yun Wang
Foods 2024, 13(24), 4137; https://doi.org/10.3390/foods13244137 - 20 Dec 2024
Viewed by 227
Abstract
The livestock industry uses ofloxacin, an antibiotic, to prevent several animal diseases; however, the overdose of ofloxacin used in animal farming treatments may appear in food products and cause some adverse human health effects. Hence, there is an immediate need to develop a [...] Read more.
The livestock industry uses ofloxacin, an antibiotic, to prevent several animal diseases; however, the overdose of ofloxacin used in animal farming treatments may appear in food products and cause some adverse human health effects. Hence, there is an immediate need to develop a method suitable for on site large-scale detection of ofloxacin residues in animal-derived foods. This study aimed to prepare a monoclonal antibody with high sensitivity and affinity for ofloxacin by re-synthesizing the ofloxacin hapten and synthesizing the corresponding complete antigen. The IC50 of the enzyme-linked immunosorbent assay (ic-ELISA) was 0.13 ng/mL, and the detection limit was 0.033 ng/mL. The visual detection limit of the established colloidal gold immunochromatographic test strip, for the visual detection of actual samples, was 1 ng/g. In summary, this work establishes a rapid detection method of ofloxacin residues on the basis of colloidal gold immunochromatography that is suitable for actual detection. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of OFL.</p>
Full article ">Figure 2
<p>Modification of the hapten of OFL.</p>
Full article ">Figure 3
<p>Preparation of immunogen and coating antigen ((<b>a</b>) OFL immunogen; (<b>b</b>) OFL coating antigen).</p>
Full article ">Figure 4
<p>The basic principle of the colloidal gold immunochromatographic assay: (<b>A</b>) structure and (<b>B</b>) principle.</p>
Full article ">Figure 5
<p>Mass spectrometry of the modified OFL.</p>
Full article ">Figure 6
<p>The <sup>1</sup>H NMR spectrum for the modified OFL.</p>
Full article ">Figure 7
<p>UV–vis spectrum of OFL immunogen (<b>a</b>) and coating antigen (<b>b</b>).</p>
Full article ">Figure 8
<p>Protein standard curve.</p>
Full article ">Figure 9
<p>The screening results of hybridoma cells of OFL.</p>
Full article ">Figure 10
<p>Subtype determination of mAb.</p>
Full article ">Figure 11
<p>Affinity constant result of OFL antibody.</p>
Full article ">Figure 12
<p>The optimization results of ELISA working conditions: blocking conditions (<b>a</b>), competition time (<b>b</b>), NaCl content (<b>c</b>), and pH (<b>d</b>).</p>
Full article ">Figure 13
<p>The standard inhibition curves of OFL.</p>
Full article ">Figure 14
<p>The characteristic images of colloidal gold particles: (<b>a</b>) UV–visible spectra, and (<b>b</b>) transmission electron microscopy images.</p>
Full article ">Figure 15
<p>The images of colloidal gold immunochromatographic strip tests for OFL in (<b>a</b>) pork, (<b>b</b>) fish, and (<b>c</b>) chicken samples. (Note: the additional amounts of OFL standard in these samples are 1: 0 ng/g, 2: 1 ng/g, 3: 2 ng/g, 4: 4 ng/g).</p>
Full article ">
Back to TopTop