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.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Apr 21;9(1):1747688.
doi: 10.1080/2162402X.2020.1747688. eCollection 2020.

CD19-targeting fusion protein combined with PD1 antibody enhances anti-tumor immunity in mouse models

Affiliations

CD19-targeting fusion protein combined with PD1 antibody enhances anti-tumor immunity in mouse models

Zhuangwei Lv et al. Oncoimmunology. .

Abstract

In our previous studies, using a B cell vaccine (scFv-Her2), the targeting of tumor-associated antigen Her2 (human epidermal growth factor receptor-2) to B cells via the anti-CD19 single chain variable fragment (scFv) was shown to augment tumor-specific immunity, which enhanced tumor control in the prophylactic and therapeutic setting. However, the fusion protein displayed limited activity against established tumors, and local relapses often occurred following scFv-Her2 treatment, indicating that scFv-Her2-induced responses are inadequate to maintain anti-tumor immunity. In this study, targeting the IV region (D4) of the extracellular region of Her2 to B cells via CD19 molecules (scFv-Her2D4) was found to enhance IFN-γ-producing-CD8+ T cell infiltration in tumor tissues and reduced the number of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). However, negative co-stimulatory molecules such as programmed cell death protein-1 (PD-1), CD160, and LAG-3 on T cells and programmed death protein ligand-1 (PD-L1) on tumor cells were upregulated in the tumor microenvironment after scFv-Her2D4 treatment. Further, anti-PD1 administration enhanced the efficacy of scFv-Her2D4 and anti-tumor immunity, as evidenced by the reversal of tumor-infiltrating CD8+ T cell exhaustion and the reduction of MDSCs and Treg cells, which suppress T cells and alter the tumor immune microenvironment. Moreover, combining this with anti-PD1 antibodies promoted complete tumor rejection. Our data provide evidence of a close interaction among tumor vaccines, T cells, and the PD-L1/PD-1 axis and establish a basis for the rational design of combination therapy with immune modulators and tumor vaccine therapy.

Keywords: B cell vaccine; CD19; anti-PD1; breast cancer; combination therapy; immunotherapy.

PubMed Disclaimer

Figures

Figure 1.
Figure 1.
Generation and characterization of anti-CD19 scFv fusion protein. (a, b) Lysates of OrigamiB(DE3)pLysS cells transfected with expression plasmids for CD19-scFv-D4, CD19–scFv, and her-2/neu D4. After transfection, the proteins were purified, and western blotting was performed using Abs against His-Tag (a) and her-2/neu (b).(c) For in vitro B cell binding, splenocytes were incubated with biotin-labeled scFv, D4, and scFv–D4, followed by anti-mouse B220 and streptavidin. Cells were washed and assessed by flow cytometry. For in vivo binding, biotin-labeled proteins were i.v. injected into mice. Peripheral blood was drawn 10 min after injection. Cells were stained with anti-mouse B220 and streptavidin and assessed by flow cytometry.(d) B cells were stimulated with fusion proteins, as indicated, for 24 h. Surface molecules CD80, CD86, and MHCII were assessed by flow cytometry. Data shown are representative of three independent experiments. B cells were stimulated with fusion proteins. A summary of CD80, CD86, and MHCII mean fluorescence intensity (MFI) is shown in the bar graph. ***p < .0001, *p < .05.
Figure 2.
Figure 2.
CD19-mediated Ag targeting of B cells induces antibody and T cell responses. (a, b) Mice (n = 5) were immunized with scFv, D4, or scFv–D4 (50 μg per mouse) three times at 1-week intervals; mice were bled at day 21. The sera were assessed for her2/neu–specific Abs by ELISA (a). Anti-her2/neu-specific Ab competitive inhibition assay showing that sera (1:10) from mice immunized with scFv-D4 are capable of inhibiting Herceptin-mediated binding. Percentage of inhibition is shown (b).(c) BALB/c mice (n = 3) were immunized with scFv, D4, or scFv–D4 three times at 1-week intervals. Splenocytes were harvested at day 21 and stimulated with scFv–D4 for 3 d. Assessment of intracellular production of IFN-γ by CD4+ and CD8+ T cells (c).(d) Splenocytes from immunized mice with scFv-D4 were harvested, the B cells were removed by CD19 beads, and then splenocytes with or without B cells (B cell-depleted) were stimulated with scFv–D4 for 3 d. Intracellular production of IFN-γ by CD4+ T cells is shown (d).(e) Graph showing body weight of BALB/c mice (n = 5) as a measure of systemic fusion protein toxicity.Data are representative of three experiments. Error bars represent standard error of the mean **p < .01, *p < .05.
Figure 3.
Figure 3.
Targeting Ags to B cells induces anti-tumor activity in a 4T1/E2 breast cancer model. (a) Schematic representation of subcutaneous (s.c.) 4T1/E2 tumor cell treatments applied to BALB/c mice.(b, c) BALB/c mice (n = 7) were challenged s.c. with 106 4T1/E2 tumor cells. The mice were treated with scFv, D4, or scFv–D4 three times at 1-week intervals when the tumor size (diameter) reached 3–5 mm. Tumor growth (b) and survival (c) were recorded.(d, e) BALB/c mice (n = 5) with established 4T1/E2 tumors were treated with scFv-D4, as described in Figure 3(a). Tumors were isolated on day 26, and immune cells were analyzed by flow cytometry. Shown are the numbers of tumor-infiltrating (d) CD45+CD3+CD8+ T cells and (e) CD45+ CD11b+Gr1+ cells in the different treatment groups.(f–h) Tumors were isolated on day 26, and tumor-infiltrating lymphocytes (TILs) from the tumors were stimulated with PMA and ionomycin in the presence of brefeldin A for 4 h. Intracellular production of IFN-γ by CD4+ (f) and CD8+ T cells (g) is shown. The percentage of myeloid-derived suppressor cells (MDSCs) among TILs was analyzed by flow cytometry (h).Data are representative of three experiments. Error bars represent standard error of the mean. **p < .01, *p < .05.
Figure 4.
Figure 4.
Targeting Ags to B cells promotes PD1 and PDL1 expression in the tumor microenvironment. (a, b) BALB/c mice (n = 3) were immunized with scFv, D4, or scFv–D4 three times at 1-week intervals. Splenocytes from immunized mice were stimulated with scFv–D4 (20 μg/mL) for 3 d. PD1 expression on the surface of CD4+ (a) and CD8+ T cells (b) is shown.(c,d) BALB/c mice (n = 5) with established 4T1/E2 tumors were treated with scFv-D4 as described in Figure 3(a). Tumors were isolated on day 26, and tumor-infiltrating lymphocytes (TILs) from the tumors were harvested. PD1 expression on the surface of CD4+ (c) and CD8+ T cells (d) is shown.(e, f) BALB/c mice (n = 5) with established 4T1/E2 tumors were treated with PBS, scFv, D4, or scFv-D4. Tumors were isolated on day 26, and PD-L1 expression on the surface of tumor cells (CD45 cells) and CD45+ cells were assessed by flow cytometry.(g, h) 4T1/E2 cells and CT26/E2 cells were stimulated with mouse IFN-γ (20 ng/mL), and then surface PD-L1 expression was assessed.Data are representative of three experiments. Error bars represent standard error of the mean. ***p < .0001, **p < .01,*p < .05.
Figure 5.
Figure 5.
Combined treatment with scFv-D4 and anti-PD-1 induces immune cell infiltration and shows remarkable anti-tumor activity in a 4T1/E2 breast cancer model. (a) Schematic representation of subcutaneous (s.c.) 4T1/E2 breast cancer cell treatments applied to BALB/c mice.(b, c) BALB/c mice (n = 7) were challenged s.c. with 106 4T1/E2 tumor cells. The mice were treated, as described in Figure 5(a), when the tumor size (diameter) reached 3–5 mm. Tumor growth (b) and survival (c) were recorded.(d–i) BALB/c mice (n = 5 with established 4T1/E2 tumors were treated as described in Figure 5(a). Tumors were isolated on day 34, and the immune cells were analyzed by flow cytometry. Shown are the numbers of tumor-infiltrating (d) CD45+CD3+CD8+ T cells and (e) CD45+CD11b+Gr1+ cells in the different treatment groups. The frequencies of CD44+CD127+ (f), LAG3+CD160+ (g), and PD1+ (h) cells among CD8+T-cell subsets, as well as PD1+ (i) cells among CD4+T-cell subsets, are shown.Data are representative of three experiments. Error bars represent standard error of the mean. *p < .05
Figure 6.
Figure 6.
scFv-D4 and anti-PD1 combination therapy enhances T cell responses in a 4T1/E2 tumor model. BALB/c mice (n = 5) with established 4T1/E2 tumors were treated as described in Figure 5(a). Tumors were isolated on day 34, and the immune cells were analyzed by flow cytometry. Tumor-infiltrating lymphocytes (TILs) from the tumors were stimulated with PMA and ionomycin in the presence of brefeldin A for 4 h. The percentages of IFN-γ-producing CD4+ (a), Foxp3+ CD4+ (b), gzmb-producing CD4+ (c), IFN-γ-producing CD8+ (d), gzmB-producing CD8+ (e), and TNF-α-producing CD8+ T cells (f) among TILs are shown.Data are representative of three experiments. Error bars represent standard error of the mean. *p < .05
Figure 7.
Figure 7.
Treatment with fusion protein scFv–D4 and anti-PD1 induces significant anti-tumor effects in a CT26/E2 tumor model. (a) Schematic representation of subcutaneous (s.c.) CT26/E2 colon cancer cell treatments applied to BALB/c mice.(b) BALB/c mice (n = 7) were challenged s.c. with 106 CT26/E2 tumor cells. The mice were treated, as described in Figure 5(a), when the tumor size (diameter) reached 3–5 mm. Tumor growth (b) was recorded(c–f) BALB/c mice (n = 5) with established CT26/E2 tumors were treated as described in Figure 6(a). Tumors were isolated on day 34, and the immune cells were analyzed by flow cytometry. (c–e) Tumor-infiltrating lymphocytes (TILs) from the tumors were stimulated with PMA and ionomycin in the presence of brefeldin A for 4 h. Intracellular production of IFN-γ by CD4+, CD8+, and gzmB+ CD8+ T cells is shown. The numbers of tumor-infiltrating myeloid-derived suppressor cells (MDSCs) are shown (f).Data are representative of three experiments. Error bars represent standard error of the mean. Data are representative of three experiments. *p < .05.

Similar articles

Cited by

References

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A.. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–15. doi:10.3322/caac.21492. - DOI - PubMed
    1. Force J, Leal J, McArthur HL. Checkpoint blockade strategies in the treatment of breast cancer: where we are and where we are heading. Curr Treat Options Oncol. 2019;20:35. doi:10.1007/s11864-019-0634-5. - DOI - PubMed
    1. Takegawa N, Yonesaka K. HER2 as an emerging oncotarget for colorectal cancer treatment after failure of anti-epidermal growth factor receptor therapy. Clin Colorectal Cancer. 2017;16:247–251. doi:10.1016/j.clcc.2017.03.001. - DOI - PubMed
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–296. doi:10.1146/annurev.immunol.25.022106.141609. - DOI - PMC - PubMed
    1. Jenne CN, Kubes P. Immune surveillance by the liver. Nat Immunol. 2013;14:996–1006. doi:10.1038/ni.2691. - DOI - PubMed

Publication types