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
[Preprint]. 2024 Aug 1:rs.3.rs-4792057.
doi: 10.21203/rs.3.rs-4792057/v1.

Bi-specific antibody engagers for cancer immunotherapy

Affiliations

Bi-specific antibody engagers for cancer immunotherapy

Hidde Ploegh et al. Res Sq. .

Update in

Abstract

Bispecific antibody engagers are fusion proteins composed of a nanobody that recognizes immunoglobulin kappa light chains ( VHH kappa ) and a nanobody that recognizes either CTLA-4 or PD-L1. These fusions show strong antitumor activity in mice through recruitment of polyclonal immunoglobulins independently of specificity or isotype. In the MC38 mouse model of colorectal carcinoma, the anti-CTLA-4 VHH-VHH kappa conjugate eradicates tumors and reduces the number of intratumoral regulatory T cells. The anti-PD-L1 VHH-VHH kappa conjugate is less effective in the MC38 model, whilst still outperforming an antibody of similar specificity. The potency of the anti-PD-L1 VHH-VHH kappa conjugate was strongly enhanced by installation of the cytotoxic drug maytansine or a STING agonist. The ability of such fusions to engage the Fc-mediated functions of all immunoglobulin isotypes is an appealing strategy to further improve on the efficacy of immune checkpoint blockade, commonly delivered as a monoclonal immunoglobulin of a single defined isotype.

PubMed Disclaimer

Conflict of interest statement

Declarations Competing interests X.L., and H.L.P. have filed a patent covering the technologies described in this publication (International Publication Number: WO2023141500A2). The other authors declare no competing interests. Additional Declarations: Yes there is potential Competing Interest. X.L., and H.L.P. have filed a patent covering the technologies described in this publication (International Publication Number: WO2023141500A2). The other authors declare no competing interests.

Figures

Figure 1
Figure 1. Recruitment of polyclonal immunoglobulins using nanobody-based immune checkpoint inhibitor-VHHkappa conjugates: proposed mechanism of action.
(A) Mechanism of action for VHHkappa-conjugated anti-CTLA-4 VHH (H11) and anti-PD-L1 VHH (A12) as anti-cancer immunotherapies: (1) the H11-VHHkappa or A12-VHHkappa conjugate binds to kappa light chains of polyclonal immunoglobulins, regardless of specificity or isotype, thereby prolonging the circulatory half-life of the conjugates; (2) the H11-VHHkappa conjugate recruits immunoglobulins to mediate Fc receptor-dependent depletion of CTLA-4-positive T regulatory cells; (3) the A12-VHHkappa conjugate can be further modified for delivery of drugs to the tumor microenvironment by targeting PD-L1, overexpressed by tumor and myeloid cells. (B) The triglycine-modified therapeutic payload is site-specifically attached to the C-terminus of A12-VHHkappa conjugate through a sortase reaction. These payloads include a near-infrared dye imaging agent, cytotoxic drugs, and immunostimulatory drugs.
Figure 2
Figure 2. Affinity, specificity and antibody-recruiting capability of the anti-CTLA-4 VHH (H11)-VHHkappa and the anti-PD-L1 VHH (A12)-VHHkappa conjugates.
(A) Saturation binding curves of biotinylated H11, A12, H11-VHHkappa conjugate, and A12-VHHkappa conjugate to murine CTLA-4 and PD-L1 (Data represent mean ± SD, n = 3). (B) Saturation binding curves of biotinylated H11-VHHkappa and A12-VHHkappa conjugates to murine polyclonal IgG (Data represent mean ± SD, n = 3). (C) Flow cytometry confirms that the A12-VHHkappa conjugate can recruit phycoerythrin (PE)-conjugated mouse IgG, IgM, and IgA, to PD-L1-positive B16-F10 cells. (D) Flow cytometry of MC38 tumor cells, isolated from tumor-bearing C57BL/6 mice and stained with H11-Cy5, reveals CTLA-4 expression on T regulatory cells within the tumor, with only marginal staining observed on splenic T cells. (E) Flow cytometry of cells recovered from PD-L1-positive and PD-L1 knockout MC38 tumors in C57BL/6 mice stained with A12-Cy5 confirms PD-L1 staining on wild-type MC38 cells (CD45-) and infiltrating CD11b+ immune cells (CD45+).
Figure 3
Figure 3. The anti-CTLA-4 VHH (H11)-VHHkappa conjugate shows better in vivo antitumor activity than the anti CTLA-4 antibody 9H10 or H11 VHH alone.
(A) Tumor growth and survival curves for MC38 tumor-bearing mice (n = 6) treated intraperitoneally with the indicated VHHs at 5 mg/kg on day 1 post-tumor injection, followed by 3 times a week for 3 weeks as indicated by the arrows. † Mouse died from an unknown cause. (B) Tumor growth and survival curves for B16-F10 tumor-bearing mice (n = 6) treated intraperitoneally with the indicated VHHs at 5 mg/kg on day 1 post-tumor injection, followed by 3 times a week for 3 weeks as indicated by the arrows. Mice were vaccinated with GM-CSF–secreting B16 cells (GVAX) on day 0. † Mice were sacrificed due to severe ulceration of their tumors. (C) Changes in T cell populations after treatment with the (H11)-VHHkappa conjugate. MC38 tumor-bearing mice were treated intraperitoneally with the indicated VHHs at 5 mg/kg on day 8, 10, 12, and 14 post-tumor injection. Single cell suspensions from tumors, spleens, and draining lymph nodes were analyzed by flow cytometry on day 15 post-tumor injection. The experiment was performed twice, and the data were combined (Data represent mean ± SD, n= 7, unpaired t test). (D) Changes in regulatory T cell populations after treatment with the H11-VHHkappa conjugate in wild-type, Fc receptor common gamma chain knockout mice, and complement-deficient mice. MC38 tumor-bearing mice were treated intraperitoneally with the indicated VHHs at 5 mg/kg on day 8, 10, 12, and 14 post-tumor injection. Single cell suspensions from tumors and spleens were analyzed by flow cytometry on day 15 post-tumor injection (Data represent mean ± SD, n= 4, unpaired t test). (E) Half-life measurements of 89Zr-labelled H11-VHHkappa conjugate and 89Zr-labelled H11 in mice (Data represent mean ± SD, n = 3). (F) Tumor growth and survival curves for MC38 tumor-bearing mice (n = 6) treated intravenously with the indicated VHHs at 5 mg/kg or anti-CTLA-4 antibody 9H10 at 12.5 mg/kg (equimolar) on day 7, 10, and 13 post-tumor injection as indicated by the arrows. † Mouse was sacrificed due to severe ulceration of its tumor.
Figure 4
Figure 4. Whole-body imaging and biodistribution of near-infrared dye-labelled anti-PD-L1 VHH (A12)-VHHkappaconjugate in MC38 tumor-bearing mice show its PD-L1-dependent accumulation in the tumor.
(A) Structure of near-infrared (NIR) dye (IRDye800CW)-labelled A12-VHHkappa conjugate. (B) Timeline of the imaging experiment. Wild-type and PD-L1 knockout MC38 tumor-bearing mice (n = 3) were treated intravenously with the indicated NIR dye-labelled VHHs (1 nmol) on day 14 post-tumor injection. Imaging was done 24 h later. (C) Whole-body imaging. Left: comparison of various NIR dye-labelled VHHs in tumor-bearing mice; right: individual images of mice from the same treatment group. SD36 is a VHH that recognizes influenza virus hemagglutinin (specificity control). Where indicated (30x), a 30-fold molar excess of unlabeled imaging agent was given as a competitor. (D) Left: Ex vivo NIR imaging of tumors from the mice treated with the indicated NIR dye-labelled VHHs; Right: quantification of the NIR dye-labelled VHHs accumulated in the tumors (Data represent mean ± SD, n = 3, unpaired t test). (E) Left: Ex vivo NIR imaging of organs from mice treated with the indicated NIR dye-labelled VHHs (from top to bottom: tumor, lung, heart, liver, stomach, intestine, spleen, and kidneys); Right: quantification of accumulation of the NIR dye-labelled VHHs (Data represent mean ± SD, n = 3, unpaired t test).
Figure 5
Figure 5. The anti-PD-L1 VHH (A12)-VHHkappa conjugate has superior in vivo antitumor activity compared to anti-PD-L1 antibody 10F.9G2 or A12 VHH alone.
(A) Tumor growth and survival curves for MC38 tumor-bearing mice (n = 6) treated intraperitoneally with the indicated VHHs at 5 mg/kg on day 1 post-tumor injection, followed by 3 times a week for 3 weeks as indicated by the arrows. (B) Tumor growth and survival curves for B16-F10 tumor-bearing mice (n = 6) treated intraperitoneally with the indicated VHHs at 5 mg/kg on day 1 post-tumor injection, followed by 3 times a week for 3 weeks, as indicated by the arrows. Mice were vaccinated with GM-CSF-secreting B16 cells (GVAX) on day 0. (C) Half-life measurements of 89Zr-labelled A12-VHHkappa conjugate and 89Zr-labelled A12 in mice (Data represent mean ± SD, n = 3). (D) Tumor growth and survival curves for wild-type and PD-L1 knockout MC38 tumor-bearing mice (n = 5) treated intraperitoneally with the indicated VHHs at 5 mg/kg on day 1 post-tumor injection, followed by three times a week for 3 weeks as indicated by the arrows.
Figure 6
Figure 6. Targeted delivery of the cytotoxicity drug maytansine enhances antitumor activity of the anti-PD-L1 VHH (A12)-VHHkappa conjugate.
(A) Tumor growth and survival curves for MC38 tumor-bearing mice (n = 6) treated intravenously with A12-VHHkappa-DM1 or A12-VHHkappa-DM4 at 5 mg/kg on day 7, 10,13, and 17 post-tumor injection as indicated by the arrows. (B) Tumor growth and survival curves for MC38 tumor-bearing mice (n = 6) treated intravenously with indicated VHH-DM4 conjugates at 5 mg/kg on day 7, 10,13, and 17 post-tumor injection as indicated by the arrows. (C) Body weight curves for the mice used in panel B (Data represent mean ± SD, n = 6). (D) Percentage of live cells and CD11b+ lymphocytes harvested from the tumors in mice treated with A12-VHHkappa-DM4 or A12-VHHkappa conjugate. MC38 tumor-bearing mice were treated intravenously with the indicated VHH conjugates at 5 mg/kg on day 11 and 13 post-tumor injection (Data represent mean ± SD, n = 3 or 4, unpaired t test). (E) Tumor growth and survival curves for B16-F10 tumor-bearing mice (n = 5) treated intravenously with indicated VHH-DM4 conjugates at 5 mg/kg on day 3, 6, 9, and 13 post-tumor injection as indicated by the arrows.
Figure 7
Figure 7. Targeted delivery of a STING agonist enhances antitumor activity of the anti-PD-L1 VHH (A12)-VHHkappaconjugate.
(A) Tumor growth and survival curves for MC38 tumor-bearing mice (n = 6) treated intravenously with A12-VHHkappa-STING agonist or A12-VHHkappa-conjugate plus free STING agonist (equimolar amount) at 5 mg/kg on day 7, 10, and 14 post-tumor injection as indicated by the arrows. (B) Body weight curves for the mice used in panel A (Data represent mean ± SD, n = 6). (C) Percentage of activated (CD69-positive) CD4+ and CD8+ T cells harvested from the tumors in mice treated with A12-VHHkappa-STING agonist or A12-VHHkappa conjugates. MC38 tumor-bearing mice were treated intravenously with the indicated VHH conjugates at 5 mg/kg on day 11 and 13 post-tumor injection. Single cell suspensions from tumors were analyzed by flow cytometry on day 15 post-tumor injection (Data represent mean ± SD, n = 3 or 4, unpaired t test). (D) Tumor growth and survival curves for B16-F10 tumor-bearing mice (n = 5) treated intravenously with A12-VHHkappa-STING agonist or A12-VHHkappa-conjugate plus free STING agonist (equimolar amount) on day 3, 7,11, and 15 post-tumor injection as indicated by the arrows. (E) Tumor growth and survival curves for MC38 tumor-bearing mice (n = 6) treated intravenously with A12-VHHkappa-STING agonist or A12-VHHkappa-conjugate plus free STING agonist (equimolar amount) at 10 mg/kg on day 7, 10, and 14 post-tumor injection as indicated by the arrows.

References

    1. Jiang X.-R. et al. Advances in the assessment and control of the effector functions of therapeutic antibodies. Nature reviews Drug discovery 10, 101–111 (2011). - PubMed
    1. Park H. I., Yoon H. W. & Jung S. T. The highly evolvable antibody Fc domain. Trends in biotechnology 34, 895–908 (2016). - PubMed
    1. Liu R., Oldham R. J., Teal E., Beers S. A. & Cragg M. S. Fc-engineering for modulated effector functions—improving antibodies for cancer treatment. Antibodies 9, 64 (2020). - PMC - PubMed
    1. Kretschmer A., Schwanbeck R., Valerius T. & Rösner T. Antibody isotypes for tumor immunotherapy. Transfusion Medicine and Hemotherapy 44, 320–326 (2017). - PMC - PubMed
    1. Mukai K., Tsai M., Starkl P., Marichal T. & Galli S. J. in Seminars in immunopathology. 581–603 (Springer; ). - PMC - PubMed

Publication types

LinkOut - more resources