This site needs JavaScript to work properly. Please enable it to take advantage of the complete set of features!
Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

NIH NLM Logo
Log in
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Oct;148(10):2743-2757.
doi: 10.1007/s00432-022-04036-8. Epub 2022 May 12.

Attenuated Toxoplasma gondii enhances the antitumor efficacy of anti-PD1 antibody by altering the tumor microenvironment in a pancreatic cancer mouse model

Affiliations

Attenuated Toxoplasma gondii enhances the antitumor efficacy of anti-PD1 antibody by altering the tumor microenvironment in a pancreatic cancer mouse model

Said Ahmed Bahwal et al. J Cancer Res Clin Oncol. 2022 Oct.

Abstract

Purpose: To investigate whether attenuated Toxoplasma is efficacious against solid tumors of pancreatic cancer and whether attenuated Toxoplasma improves the antitumor activity of αPD-1 antibody on pancreatic cancer.

Methods: The therapeutic effects of attenuated Toxoplasma NRTUA strain monotherapy and combination therapy of NRTUA with anti-PD-1 antibody on PDAC tumor volume and tumor weight of Pan02 tumor-bearing mice were investigated. We characterized the effects of combination therapy of NRTUA with anti-PD-1 antibody on tumor-infiltrating lymphocytes and tumor-specific IFN-γ by using immunohistochemistry, flow cytometry and ELISA. The antitumor mechanisms of combination therapy of NRTUA with anti-PD-1 antibody were investigated via depletion of CD8+ T cells and IL-12.

Results: NRTUA strain treatment inhibited tumor growth in a subcutaneous mouse model of PDAC through activating dendritic cells and increasing CD8+ T cell infiltration in the tumor microenvironment. More importantly, combination therapy of NRTUA with anti-PD-1 antibody elicited a significant antitumor immune response and synergistically controlled tumor growth in Pan02 tumor-bearing mice. Specifically, the combination treatment led to elevation of CD8+ T cell infiltration mediated by dendritic cell-secreted IL-12 and to tumor-specific IFN-γ production in the PDAC tumor microenvironment. Also, the combination treatment markedly reduced the immunosuppressive myeloid-derived suppressor cell population in PDAC mice.

Conclusion: These findings could provide a novel immunotherapy approach to treating solid tumors of PDAC and overcoming resistance to anti-PD-1 agents in PDAC tumors.

Keywords: Dendritic cells; Immunotherapy; Programmed cell death 1 receptor; Toxoplasma gondii; Vaccination.

PubMed Disclaimer

Conflict of interest statement

The author(s) declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
NRTUA vaccination effectively controls tumor growth in a Pan02 subcutaneous tumor model. A Six to 8-week-old female C57BL/6 J mice were subcutaneously inoculated with 1 ×ばつ 106 Pan02 cells. Mice were randomly divided into 4 groups when tumor volume reached 50 mm3. Each group of mice was treated with a relevant treatment according to the schematic diagram above. Tumor growth was measured with digital calipers every 3 days. B Left: representative images of tumors separated from each group at the experimental endpoint. Middle: change in tumor volume over time in each administration group. Right: corresponding tumor weight at the experimental endpoint (n = 4). C CD8+ tumor-infiltrating T cells were evaluated through IHC. Representative CD8 immunohistochemically stained sections and semiquantitative density results are shown, scale bar: 50 μm. Tumor growth curve (B, middle panel) was analyzed using a two-way ANOVA with a Bonferroni multiple comparison test. Tumor weight (B, right panel) and CD8+ T cell infiltration (C) were analyzed using one-way ANOVA. Data represented as mean ± SD. *P < 0.05; ***P < 0.001; ****P < 0.0001; n.s. not significant
Fig. 2
Fig. 2
NRTUA activates invaded DCs in the tumor microenvironment and increases PD-1 expression on tumor-infiltrating CD8+ T cells. A Dendritic cells from NRTUA treated and untreated Pan02 tumor–bearing mice were analyzed 1 day after treatment for expression of CD80 and CD86 (mean fluorescence index, MFI). NRTUA-invaded cells were tracked using CFSE-labeled NRTUA parasites. B Bone marrow cells were isolated from 4- to 6-week-old mice and differentiated into DCs by GM-CSF in vitro. BMDCs were cocultured with NRTUA (MOI = 1:1) for 24 h. Total mRNA was extracted from DCs, and reverse transcribed into cDNA. The levels of IL12 gene expression were detected by RT-qPCR. Results were normalized to the level of GAPDH mRNA and presented as fold increase in mRNA level over the untreated control that was arbitrarily set as 1. C Protein expression in immature DCs (iDCs) and NRTUA-infected DCs (NRTUA-DC) was analyzed by immunoblotting using antibodies against MYD88, NF-kB and GAPDH (loading control). D Supernatants collected from iDCs and DCs were cocultured with NRTUA for 24 h, washed, and added to the lower chambers of transwell plates. Medium only was used as a control. Anti-IL-12 or corresponding isotype (rabbit-IgG) control antibody was added as indicated. CD8+ T cells were added to the upper chamber and cells were incubated for 24 h. Cells in the lower chambers were harvested and stained with PE-conjugated anti-CD8 and analyzed by flow cytometry. E The intra-tumoral CD8+ T cells were gated and tested for expression of PD-1 by flow cytometry. Data in (A, B and E) were analyzed by student’s t test. Data in (D) was analyzed using one-way ANOVA tests. Data represented as mean ± SD. **P < 0.01; ***P < 0.001; ****P < 0.0001; n.s. not significant
Fig. 3
Fig. 3
Combination treatment with NRTUA and αPD-1 antibody enhances the inhibition of subcutaneous Pan02 tumor growth via increasing infiltration of CD8+ T cells in the TME. A Each group of mice was treated with a relevant treatment according to the schematic diagram above. B Left: representative images of tumors separated from each group at the experimental endpoint. Middle: change in tumor volume over time in each administration group. Right: corresponding tumor weight at the experimental endpoint. C CD8a tumor-infiltrating T cells in the TME were evaluated through IHC. Representative CD8α immunohistochemically stained sections and semi-quantitative density results are shown, scale bar: 50 μm. D Schematic representation of the treatment schedule for the CD8+ T cell depletion experiment. C Left: representative images of tumors separated from each group at the experimental endpoint. Middle: change in tumor volume over time in each administration group. Right: corresponding tumor weight at the experimental endpoint, (n = 4). F CD8a tumor-infiltrating T cells in the TME were evaluated through IHC. Representative CD8α immunohistochemically stained sections and semi-quantitative density results are shown, scale bar: 50 μm. Tumor growth curves were analyzed using a two-way ANOVA with a Bonferroni multiple comparison test. Tumor weight and CD8+ T cell infiltration data were analyzed by using one-way ANOVA. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 4
Fig. 4
NRTUA combined with PD-1 blockade enhances tumor-specific IFN-γ production in CD8+ T cells within the PDAC TME. A CD8+ T cells were isolated and purified from tumors of Pan02 tumor-bearing mice. Tumor-bearing mice were treated with IgG, NRTUA, αPD-1, or NRTUA + αPD-1 therapy as indicated. The percentage of IFNγ producing CD8+ T cells amongst all CD8+ T cells within the tumor-infiltrating lymphocytes is shown. ELISA assays were performed using autologous irradiated Pan02 tumor cells as antigenic targets for CD8+ T cells isolated from (B) tumor-infiltrating lymphocytes and (C) the spleen. Each experimental group consisted of 5 mice, pooled, and analyzed individually in triplicates. Data were analyzed using one-way ANOVA. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; n.s. not significant
Fig. 5
Fig. 5
Antitumor efficacy of combined treatment requires IL-12 production for CD8+ T cell activation. A The IL-12 secreting DC subpopulation in the TME was quantified by flow cytometry analysis. B Schematic representation of the treatment schedule for the IL12p75 depletion experiment. C Left: representative images of tumors separated from each group at the experimental endpoint. Middle: change in tumor volume over time in each administration group. Right: corresponding tumor weight at the experimental endpoint, (n = 4). D CD8a tumor-infiltrating T cells were evaluated through IHC. Representative CD8α immunohistochemically stained sections and semiquantitative density results are shown, scale bar: 50 μm. Tumor growth curve (B) was analyzed by two-way ANOVA with a Bonferroni multiple comparison test. Data in (A, D) were analyzed by one-way ANOVA test. Data represented as mean ± SD. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; n.s. not significant
Fig. 6
Fig. 6
Combination therapy with NRTUA and PD-1 blockade reduces the MDSC population in the PDAC TME. MDSC subpopulations PMN-MDSCs (B) M-MDSCs (C) and in the TME were determined by flow cytometry (n = 4). Polymorphonuclear MDSCs are CD11b+Ly6CloLy6Ghi, whereas monocytic MDSCs are CD11b+Ly6ChiLy6Glo. Data were analyzed by one-way ANOVA. n.s. non significance, *P < 0.05, ***P < 0.001. PMN-MDSCs polymorphonuclear myeloid-derived suppressor cells, M-MDSCs monocytic myeloid-derived suppressor cells

References

    1. Agrawal SR, Singh V, Ingale S et al (2014) Toxoplasmosis of spinal cord in acquired immunodeficiency syndrome patient presenting as paraparesis: a rare entity. J Glob Infect Dis 6:178–181. 10.4103/0974-777X.145248 - PMC - PubMed
    1. Akiyama Y, Nonomura C, Kondou R et al (2016) Immunological effects of the anti-programmed death-1 antibody on human peripheral blood mononuclear cells. Int J Oncol 49:1099–1107. 10.3892/ijo.2016.3586 - PubMed
    1. Atkins MB, Robertson MJ, Gordon M et al (1997) Phase I evaluation of intravenous recombinant human interleukin 12 in patients with advanced malignancies. Clin Cancer Res 3:409–417 - PubMed
    1. Baird JR, Fox BA, Sanders KL et al (2013a) Avirulent Toxoplasmagondii generates therapeutic antitumor immunity by reversing immunosuppression in the ovarian cancer microenvironment. Cancer Res 73:3842–3851. 10.1158/0008-5472.CAN-12-1974 - PMC - PubMed
    1. Baird JR, Byrne KT, Lizotte PH et al (2013b) Immune-mediated regression of established B16F10 melanoma by intratumoral injection of attenuated Toxoplasmagondii protects against rechallenge. J Immunol 190:469–478. 10.4049/jimmunol.1201209 - PMC - PubMed
Cite

AltStyle によって変換されたページ (->オリジナル) /