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
. 2021 Jun;9(6):e002484.
doi: 10.1136/jitc-2021-002484.

Direct and indirect immune effects of CMP-001, a virus-like particle containing a TLR9 agonist

Affiliations

Direct and indirect immune effects of CMP-001, a virus-like particle containing a TLR9 agonist

Shakoora A Sabree et al. J Immunother Cancer. 2021 Jun.

Abstract

Background: CMP-001, also known as vidutolimod, is a virus-like particle containing a TLR9 agonist that is showing promise in early clinical trials. Our group previously demonstrated that the immunostimulatory effects of CMP-001 are dependent on an anti-Qβ antibody response which results in opsonization of CMP-001 and uptake by plasmacytoid dendritic cells (pDCs) that then produce interferon (IFN)-α. IFN-α then leads to an antitumor T-cell response that is responsible for the in vivo efficacy of CMP-001. Here, we explore mechanisms by which the initial effects of CMP-001 on pDCs activate other cells that can contribute to development of an antitumor T-cell response.

Methods: Uptake of CMP-001 by various peripheral blood mononuclear cell (PBMC) populations and response to anti-Qβ-coated CMP-001 were evaluated by flow cytometry and single-cell RNA sequencing. Purified monocytes were treated with anti-Qβ-coated CMP-001 or recombinant IFN-α to evaluate direct and secondary effects of anti-Qβ-coated CMP-001 on monocytes.

Results: Monocytes had the highest per cell uptake of anti-Qβ-coated CMP-001 with lower levels of uptake by pDCs and other cell types. Treatment of PBMCs with anti-Qβ-coated CMP-001 induced upregulation of IFN-responsive genes including CXCL10, PDL1, and indoleamine-2,3-dioxygenase (IDO) expression by monocytes. Most of the impact of anti-Qβ-coated CMP-001 on monocytes was indirect and mediated by IFN-α, but uptake of anti-Qβ-coated CMP-001 altered the monocytic response to IFN-α and resulted in enhanced expression of PDL1, IDO, and CD80 and suppressed expression of CXCL10. These changes included an enhanced ability to induce autologous CD4 T-cell proliferation.

Conclusions: Anti-Qβ-coated CMP-001 induces IFN-α production by pDCs which has secondary effects on a variety of cells including monocytes. Uptake of anti-Qβ-coated CMP-001 by monocytes alters their response to IFN-α, resulting in enhanced expression of PDL1, IDO and CD80 and suppressed expression of CXCL10. Despite aspects of an immunosuppressive phenotype, these monocytes demonstrated increased ability to augment autologous CD4 T-cell proliferation. These findings shed light on the complexity of the mechanism of action of anti-Qβ-coated CMP-001 and provide insight into pathways that may be targeted to further enhance the efficacy of this novel approach to immunotherapy.

Keywords: immunotherapy; tumor microenvironment.

PubMed Disclaimer

Conflict of interest statement

Competing interests: SEB holds stock options in Checkmate Pharmaceuticals. GJW received research funding from Checkmate Pharmaceuticals. All of the other authors declare no competing interests.

Figures

Figure 1
Figure 1
Uptake of fluorescently labeled CMP-001 by various immune cell subsets. Unfractionated human peripheral blood mononuclear cells (A) or monocytes polarized into classical M1 (B) and M2 (C) macrophages were treated with Cy5.5-labeled CMP-001 plus or minus anti-Qβ for 1 hour. Cells were stained for unique surface markers and analyzed via flow cytometry. Data are representative of two to three individual experiments. NK, natural killer.
Figure 2
Figure 2
scRNA-seq of PBMCs treated with anti-Qβ-coated CMP-001 demonstrates upregulation of genes in monocytes that can impact on T-cell function, including those in the IFN pathway. scRNA-seq was performed on unfractionated normal donor PBMCs treated with and without anti-Qβ-coated CMP-001. Data from one of two similar experiments are shown. (A) Clustering of merged datasets. (B) Clustering of treated cells. (C) Clustering of untreated cells. (D) Violin plots of classic cell-type surface markers. (E) Volcano plot of differentially expressed genes in monocytes (cluster 5) following anti-Qβ-coated CMP-001 treatment. For most genes, treatment resulted in positive logFCs. (F) Ingenuity pathway analysis of potential upstream regulators of differentially expressed genes in monocytes following anti-Qβ-coated CMP-001 treatment. (G) Ingenuity pathway analysis of upregulated signaling pathways in monocytes following anti-Qβ-coated CMP-001 treatment. UMAP plots of CXCL10 (H), IDO1 (IDO) (I), CD274 (PDL1) (J), and CD80 (K) untreated (left) and treated with anti-Qβ-coated CMP-001 (right). IRF, interferon regulatory factor; IFN, interferon; PBMC, peripheral blood mononuclear cell; scRNA-seq, single-cell RNA sequencing; UMAP, uniform manifold approximation and projection; IDO, indoleamine-2,3-dioxygenase.
Figure 3
Figure 3
Anti-Qβ-coated CMP-001-mediated induction of CXCL10 is dependent on monocytes and pDC-derived type I IFN. Various cell populations were treated with CMP-001 with and without anti-Qβ for 24 hours and supernatant evaluated for CXCL10, IFN-α, and IFN-β. (A) CXCL10 production in response to anti-Qβ-coated CMP-001 by PBMCs with and without monocyte depletion. (B) CXCL10 production by monocytes combined with pDCs in response to anti-Qβ-coated CMP-001. (C) CXCL10 production in response to anti-Qβ-coated CMP-001 by monocytes plus pDCs when cells were placed on the same (–) or opposite (+) sides of a transwell. (D) Production of IFN-α by pDCs treated with anti-Qβ-coated CMP-001. (E) Production of IFN-β by pDCs treated with anti-Qβ-coated CMP-001. (F) Impact of antibody blocking of IFN-α on CXCL10 production by PBMCs treated with anti-Qβ-coated CMP-001. (G) Impact of antibody blocking of IFNAR2 on CXCL10 production by PBMCs treated with anti-Qβ-coated CMP-001. Graphs depict means±SEM. Statistical significance was determined using two-way ANOVA with multiple comparisons test (A), one-way ANOVA with multiple comparisons test (B, C), and paired t-test (D–G) with alpha=0.05 (n=3–6). ANOVA, analysis of variance; IFN, interferon; ns, not significant; PBMC, peripheral blood mononuclear cell; pDC, plasmacytoid dendritic cell.
Figure 4
Figure 4
Anti-Qβ-coated CMP-001 enhanced expression of IDO and PDL1 by monocytes is dependent on type I IFN. Unfractionated PBMCs from healthy donors were treated with CMP-001 with and without anti-Qβ, and expression of cytoplasmic IDO and surface PDL1 by monocytes was determined by flow cytometry. (A, D) Expression of IDO and PDL1 induced by CMP-001 with and without anti-Qβ. (B, E) Impact of antibody blocking of IFNAR2 on expression of IDO and PDL1 induced by anti-Qβ-coated CMP-001. (C, F) Expression of IDO and PDL1 induced by recombinant IFN-α. Graphs illustrate expression (MFI) of IDO and PDL1 on Zombie Aqua negative CD14+ cells. Statistical significance was determined using one-way analysis of variance with multiple comparisons test (A, D) and paired t-test (B, C, E, F) with alpha=0.05 (n=6). IFN, interferon; MFI, median fluorescence intensity; ns, not significant; PBMC, peripheral blood mononuclear cell; IDO, indoleamine-2,3-dioxygenase.
Figure 5
Figure 5
Phagocytosis by monocytes of antibody-coated particles alters their response to IFN-α. Monocytes from healthy donors were treated with recombinant IFN-α plus or minus anti-Qβ-coated CMP-001, anti-Qβ-coated methylated CMP-001 (mCMP-001), G10 (soluble TLR9 agonist) or IgG +protein L beads for 18–20 hours. Expression of cytoplasmic IDO and CXCL10 and surface PDL1 was determined by flow cytometry. (A, E, I) Impact of recombinant IFN-α on IDO, PDL1, and CXCL10 expression by monocytes. (B, F, J) Impact of anti-Qβ-coated CMP-001 on IDO, PDL1, and CXCL10 expression by monocytes in response to IFN-α. (C, G, K) Impact of anti-Qβ-coated CMP-001, anti-Qβ-coated mCMP-001, or G10 on IDO, PDL1, and CXCL10 expression by monocytes in response to IFN-α. (D, H, L) Impact of IgG+ protein L beads on IDO, PDL1, and CXCL10 expression by monocytes in response to IFN-α. Statistical significance was determined using one-way analysis of variance with multiple comparisons test and paired t-test with alpha=0.05 (n=6–10). IFN, interferon; IDO, indoleamine-2,3-dioxygenase; MFI, median fluorescence intensity; ns, not significant.
Figure 6
Figure 6
Impact of CD32a blockade on uptake of anti-Qβ-coated CMP-001 by monocytes and concomitant response to IFN-α. (A) Monocytes from healthy donors were pretreated anti-CD32a or isotype control for 15 min followed by treatment with 10 μg/mL Cy5.5-labeled CMP-001 plus or minus anti-Qβ. Data are representative of two independent experiments. (B–D) Monocytes from healthy donors were pretreated with anti-CD32a or isotype control for 15 min followed by treatment with recombinant IFN-α and anti-Qβ-coated CMP-001 for 18–20 hours. Expression of PDL1, CXCL10, and IDO were determined by flow cytometry. (E) Monocytes from healthy donors were treated with 10 μg/mL anti-CD32a, anti-CD64, or isotype control (in the absence of particles) for 18–20 hours. Expression of IDO was determined by flow cytometry. Statistical significance was determined using paired t-test (B–D) and two-way analysis of variance (E) with alpha=0.05 (n=3–7). IFN, interferon; IDO, indoleamine-2,3-dioxygenase; MFI, median fluorescence intensity; ns, not significant.
Figure 7
Figure 7
Monocytes treated with anti-Qβ-coated CMP-001 and IFN-α upregulate CD80 expression and induce autologous CD4 T-cell proliferation. (A) Monocytes from healthy donors were treated with recombinant IFN-α plus or minus anti-Qβ-coated CMP-001 for 18–20 hours. CD80 surface expression was determined by flow cytometry. (B) Monocytes from healthy donors were treated with recombinant IFN-α plus or minus anti-Qβ-coated CMP-001 for 18–20 hours. The next day, autologous CFSE-labeled T cells were added to treated monocytes at a ratio of 1:10 (monocyte:T cell), and percent of CFSE-diluted CD3+ T cells was determined after 4 days as a measure of T-cell proliferation. (C) Purified CFSE-labeled T cells were treated directly with recombinant IFN-α plus or minus anti-Qβ-coated CMP-001 in the absence of monocytes, and percent of CFSE-diluted CD3+ T cells was determined after 4 days as a measure of T-cell proliferation. (D) Representative histograms of CFSE-diluted CD3+ T cells. (E, F) Phenotypical characterization of proliferated CD3+ T cells into CD4 and CD8 subsets. Statistical significance was determined using one-way analysis of variance with multiple comparisons test with alpha=0.05 (n=3–11). IFN, interferon; MFI, median fluorescence intensity; ns, not significant.

Similar articles

Cited by

References

    1. Hammerich L, Bhardwaj N, Kohrt HE, et al. . In situ vaccination for the treatment of cancer. Immunotherapy 2016;8:315–30. 10.2217/imt.15.120 - DOI - PubMed
    1. Cheng Y, Lemke-Miltner CD, Wongpattaraworakul W, et al. . In situ immunization of a TLR9 agonist virus-like particle enhances anti-PD1 therapy. J Immunother Cancer 2020;8:e000940. 10.1136/jitc-2020-000940 - DOI - PMC - PubMed
    1. Lemke-Miltner CD, Blackwell SE, Yin C, et al. . Antibody opsonization of a TLR9 Agonist-Containing virus-like particle enhances in situ immunization. J Immunol 2020;204:1386–94. 10.4049/jimmunol.1900742 - DOI - PMC - PubMed
    1. Miller AM, Lemke-Miltner CD, Blackwell S, et al. . Intraperitoneal CMP-001: a novel immunotherapy for treating peritoneal carcinomatosis of gastrointestinal and pancreaticobiliary cancer. Ann Surg Oncol 2021;28:1187–97. 10.1245/s10434-020-08591-7 - DOI - PMC - PubMed
    1. Weiner GJ. Cpg oligodeoxynucleotide-based therapy of lymphoid malignancies. Adv Drug Deliv Rev 2009;61:263–7. 10.1016/j.addr.2008.12.006 - DOI - PubMed

Publication types

MeSH terms