By Drug Type

Vaccine

Biocytogen has developed various HLA-humanized models and established state-of-the-art in vivo and in vitro platforms. These models enable mouse antigen-presenting cells (APCs) to present and recognize peptide epitopes similar or identical to those in humans, accelerating the development of tumor vaccines.

on this page

  • Introduction
  • Preclinical Capabilities for Tumor Vaccines Evaluation
  • Case Studies for Tumor Vaccines Evaluation
  • Related Model List

Publication

    Introduction

    The tumor antigens can be prepared as vaccine formulations by artificially synthesizing various forms of them, such as tumor cells obtained from patients, tumor-related proteins or peptides, and genes that express tumor antigens, and then combining them with adjuvants or using dendritic cells as delivery carriers. Once the vaccine enters the body, the tumor antigens are engulfed by DC cells and then processed and presented to T cells, activating the antigen-specific cytotoxic T cells to kill the tumor. The most ideal tumor-specific antigen is the neoantigen, which is generated by gene mutations in tumor cells and is not expressed in normal tissues. It can be presented on the cell surface and recognized by T cells after binding to MHC molecules.

    Vaccines have evolved from purely preventive measures to therapeutic applications, with types ranging from traditional protein and peptide vaccines to advanced mRNA vaccines. Biocytogen has developed various immunogenicity evaluation and tumor efficacy models based on HLA-humanized mice and HLA-humanized tumor cell lines. These models enable mouse antigen-presenting cells (APCs) to present and recognize peptide epitopes similar or identical to those in humans, accelerating the development of tumor vaccines. We also have established state-of-the-art in vivo and in vitro platforms to comprehensively evaluate the immunogenicity, efficacy, mechanism of action, and safety of vaccines.

    Preclinical Capabilities for Tumor Vaccines Evaluation
    Immunogenicity validation Efficacy validation MOA exploration
    • Ex Vitro:
      • Antigen-specific ELISpot analysis
      • Antigen-specific T cells detection
      • Titration of neutralizing antibodies
      • ADCC assay
    • In Vivo:
      • Wild type C57BL/6 mice
      • HLA humanized mice
      • Immune reconstitution models
    • Ex/In Vitro:
      • Antigen-specific ELISpot analysis
      • Antigen-specific T cells detection
      • Antigen-specific CTL analysis
      • Titration of neutralizing antibodies
      • ADCC assay
    • In Vivo:
      • Syngeneic tumor models
      • Xenograft models
    • In Vitro:
      • Antigen transduction and expression
      • DC/immune cell activation
    • Ex Vitro:
      • TILs analysis (FACS, pathology)
      • Cytokine release (ELISA, MSD)
    • In Vivo:
      • Biodistribution
    Case Studies for Tumor Vaccines Evaluation

    Animal models for vaccines evaluation:

    Syngeneic models:

    • C57BL/6 + TAA humanized cell lines
    • HLA humanized mice + HLA/target humanized cell lines
      • B-HLA-A2.1 mice
      • B-hCD3E&HLA-A2.1 mice
      • B-hCD3E&h4-1BB&HLA-A2.1 mice
      • B-HLA-A24.2 mice
      • B-HLA-A11.1 mic

    Xenograft models:

    • HLA humanized B-NDG mice + human cell lines
    • HLA humanized B-NDG mice + CD34+ HSC/PBMC immune reconstitution + human cell lines
      • B-NDG HLA-A2.1
      • B-NDG hB2M&HLA-A2.1 mice plus
      • B-NDG hB2M&HLA-A11.1 mice
    Case Study 1: Immunogenicity testing and in vivo efficacy evaluation of mRNA vaccines using HLA-humanized mice
    Immunogenicity testing of mRNA vaccines (NY-ESO-1)

    Figure 1. Detection of mRNA vaccine-induced immune responses in B-HLA-A2.1 mice. Female B-HLA-A2.1 mice at the age of 6–8 weeks were divided into PBS group and mRNA vaccine group (n = 6), and then inoculated PBS or vaccines at the inside muscle of both legs. One week after the last immunization, mice were sacrificed. The splenocytes were extracted, stimulated with individual peptide or target-unrelated polypeptide as negative control (NC), and then measured for IFN-γ secretion and antigen-specific T cells. (A) Experimental scheme. (B) No significant difference in body weight among groups. Values are expressed as mean ± SEM.

    Figure 2. Detection of mRNA vaccine-induced immune responses in B-HLA-A2.1 mice by IFN-γ ELISpot assay. (A) Scheme of vaccination and testing. Male B-HLA-A2.1 mice at the age of 9–10 weeks were divided into PBS group, LNP group and LNP-mRNA group (n = 3), and then inoculated PBS, LNP or LNP-mRNA at the inside muscle of both legs. Mice were vaccinated with PBS, LNP or LNP-mRNA one time. One week after the immunization, mice were sacrificed. The splenocytes were extracted, stimulated with individual peptide, or no peptide as negative control, or PMA/Ionomycin as positive control, and then measured for IFN-γ secretion. (B) Representative results showing stimulation of splenocytes harvested from immunized mice with negative control, or peptide vaccines, or positive control in duplicates. (C) Summary of results. Values are expressed as mean ± SEM. NC: negative control. PC: positive control. (Experimental No.: 24P037503)

    Figure 3. Detection of mRNA vaccine-induced immune responses in B-HLA-A2.1 mice by IFN-γ ELISpot assay. (A) Scheme of vaccination and testing. Male B-HLA-A2.1 mice at the age of 9–10 weeks were divided into PBS group, LNP group and LNP-mRNA group (n = 3), and then inoculated PBS, LNP or LNP-mRNA at the inside muscle of both legs. Mice were vaccinated with PBS, LNP or LNP-mRNA three times at 1-week interval. One week after the last immunization, mice were sacrificed. The splenocytes were extracted, stimulated with individual peptide, or no peptide as negative control, or PMA/Ionomycin as positive control, and then measured for IFN-γ secretion. (B) Representative results showing stimulation of splenocytes harvested from immunized mice with negative control, or peptide vaccines, or positive control in duplicates. (C) Summary of results. Values are expressed as mean ± SEM. NC: negative control. PC: positive control. (Experimental No.: 24P037503)

    B-HLA-A2.1 mice + B-HLA-A2.1/NY-ESO-1 MC38 models for mRNA vaccines evaluation

    Figure 1. Antitumor activity of LNP-mRNA against syngeneic tumors. (A) Experimental scheme. (B) Antitumor activity of treatment with LNP or LNP-mRNA. B-HLA-A2.1 mice (n = 6 /group) were vaccinated with PBS, LNP or LNP-mRNA (10 μg/mouse). B-HLA-A2.1/hNY-ESO-1 MC38 cells were inoculated into the right flank of the mice. (C) Body weight changes during treatment. As shown in panel B, LNP-mRNA was efficacious in controlling tumor growth in B-HLA-A2.1 mice. (D) Antitumor activity of mRNA against syngeneic tumors. B-HLA-A2.1/hNY-ESO-1 MC38 tumor cells growth of individual mice. Values are expressed as mean ± SEM. (Experimental No.: 24P037502)

    Figure 2. (A and B) Detection of vaccine-induced immune responses in B-HLA-A2.1 mice by IFN-γ ELISpot assay. NC: negative control. PC: positive control. (C and D) LDH release assay for cytotoxicity of CTLs from B-HLA-A2.1 mice immunized with PBS, LNP or LNP-mRNA against the B-HLA-A2.1/hNY-ESO-1 MC38 cell line. Cytotoxic activities of CTLs isolated from the splenocytes of immunized mice against NY-ESO-1 peptides pulsed B-HLA-A2.1/hNY-ESO-1 MC38 cell line were detected by LDH release assay at effector-to-target ratio of 50:1 or 100:1 with two different peptide concentrations, 10μg or 50μg. Values are expressed as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Dunnett's multiple comparisons test for each treatment group compared to PBS group **p<0.01, ***p<0.001, ****p<0.0001.

    Figure 3. Vaccination mRNA vaccine generates specific effector CD8+ T cells in spleens. Spleens from B-HLA-A2.1/hNY-ESO-1 MC38 tumor-bearing mice that were immunized with the PBS, LNP, LNP-mRNA vaccines. (A) Analysis of CD8+T, CD4+ T and Tregs in the spleens determined by the flow cytometric assay. For spleen T cells and CD8+T cells, the percentage (in CD45+ cells) were significantly elevated. mRNA vaccine generated frequencies of tetramer+ CD8+ T cells at approximately 50% of total CD8+ T cells in spleens. The CD8+ T cells had an obviously lower frequency in the naïve and central memory and were mainly localized in the effector memory. (B) Intracellular cytokine staining of splenocytes isolated from immunized mice, stimulated with the NY-ESO-1 peptide. The mIFN-γ were mainly secreted by CD8+ T cells but not CD4+ T cells. Values are expressed as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Dunnett's multiple comparisons test for each treatment group compared to G1. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.

    Figure 4. LNP-mRNA enhances beneficial repertoire of anti-tumor T cells. Tumors from B-HLA-A2.1/hNY-ESO-1 MC38 tumor-bearing mice that were immunized with the PBS, empty LNP or LNP-mRNA. (A) Analysis of CD8+T, CD4+ T and Tregs in the tumors determined by the flow cytometric assay. For tumor infiltrated T and CD8+T cells, the percentage (in CD45+ cells) were significantly elevated. In contrast, the frequencies of Tregs was significantly diminished in LNP-mRNA group. Tumor-infiltrated CD8+ T cells had an obviously lower frequency in the naïve and the effector memory (EM) CD8+T cells was increased significantly. (B) Intracellular cytokine staining of Tils isolated from immunized mice, stimulated with the NY-ESO-1 peptide. The mIFN-γ in Both CD8+ T cells and CD4+T cells produced mIFN-γ. Values are expressed as mean ± SEM. Statistical analysis was performed using one-way ANOVA followed by Dunnett's multiple comparisons test for each treatment group compared to G1. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.