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
. 2019 Nov 26;10(1):5387.
doi: 10.1038/s41467-019-13196-0.

On-target restoration of a split T cell-engaging antibody for precision immunotherapy

Affiliations

On-target restoration of a split T cell-engaging antibody for precision immunotherapy

Agnes Banaszek et al. Nat Commun. .

Abstract

T cell-engaging immunotherapies are changing the landscape of current cancer care. However, suitable target antigens are scarce, restricting these strategies to very few tumor types. Here, we report on a T cell-engaging antibody derivative that comes in two complementary halves and addresses antigen combinations instead of single molecules. Each half, now coined hemibody, contains an antigen-specific single-chain variable fragment (scFv) fused to either the variable light (VL) or variable heavy (VH) chain domain of an anti-CD3 antibody. When the two hemibodies simultaneously bind their respective antigens on a single cell, they align and reconstitute the original CD3-binding site to engage T cells. Employing preclinical models for aggressive leukemia and breast cancer, we show that by the combinatorial nature of this approach, T lymphocytes exclusively eliminate dual antigen-positive cells while sparing single positive bystanders. This allows for precision targeting of cancers not amenable to current immunotherapies.

PubMed Disclaimer

Conflict of interest statement

G.S. is listed as inventor on patent WO 2013/104804, which is licensed to T.G.P.B. and G.S./Cherry Biolabs. All other authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Combinatorial immunotherapy by hemibodies. a Binding of two hemibodies to their respective antigens on a target cell, each consisting of a single-chain variable fragment (scFv) fused to the variable light (VL) or variable heavy chain domain (VH) of a CD3-specific antibody, enables VH/VL association and the reconstitution of a functional CD3-binding site to engage T cells. b Allogeneic mismatch transplantation model: The hematopoietic system of a HLA-A2-positive leukemia patient is replaced by blood stem cells of an HLA-A2-negative donor. Hemibodies directed against HLA-A2 and the pan-hematopoietic marker CD45 tag the patient’s dual antigen-positive diseased hematopoiesis for lysis by T cells, while sparing single antigen-positive healthy tissues and donor-derived blood cells.
Fig. 2
Fig. 2
Dual antigen-restricted reconstitution of a functional CD3-binding site. a VLαCD3-scFvαHLA-A2 (green curve) and VHαCD3-scFvαCD45 (black curve) hemibodies were isolated by immobilized metal ion affinity chromatography (IMAC) and size-exclusion chromatography (SEC) and analyzed by FPLC at 1 mg/ml. V0 void volume/aggregates. Typical elution profiles of the hemibodies, the bispecific BiTE construct (scFvαCD3-FvαHLA-A2, red curve on top), and a mixture of the two hemibodies (blue curve, bottom) are shown for comparison. b HLA-A2/CD45 dual-positive THP-1 acute myeloid leukemia cells were co-incubated with HLA-A2-negative donor peripheral blood mononuclear cells (PBMCs) and constructs as indicated. T cell engagement was assessed by interferon-γ (IFN-γ) and interleukin-2 (IL-2) secretion and target cell lysis. The bispecific HLA-A2 × CD3 BiTE was used as a positive control. Data represent the mean values with standard deviations (±SD) from triplicate wells from three independent experiments; the effector to target cell ratio (E:T) was 5:1. c Binding of hemibodies (6 nM each) to THP-1 cells was competitively blocked by molar excess of scFv specific for CD45 (left) or HLA-A2 (right). Stimulation of donor PBMCs was assessed by IL-2 secretion at E:T = 2:1. Data represent the mean from two replicate wells from three experiments that yielded similar results. d Dual antigen-positive THP-1 leukemia, single-positve RAJI Burkitt lymphoma, or HT1080 fibrosarcoma cells were loaded with the hemibody pair (blue bars) or BiTEs (red bars), washed twice, and incubated with donor PBMCs as indicated at a E:T of 2:1. Secretion of IL-2 was assessed by ELISA. Data represent the mean (±SD) from two replicate wells from three experiments. e HLA-A2-positive (red bars) and HLA-A2-negative (blue bars) PBMNC were incubated with constructs as indicated (10 nM each). % apoptosis was assessed after annexinV staining by flow cytometry techniques as a measure of fratricide reaction. Data are representative for three independent experiments yielding comparable results.
Fig. 3
Fig. 3
Biochemical basis for hemibody complementation. a A plasmon resonance chip was coated with VLαCD3-scFvαHLA-A2. VHαCD3-scFvαCD45 was used as an analyte at 100, 200, 400, 800, 1600 (nM) as indicated. Association contact time was 300 s, dissociation contact time 3.600 s at a flow rate of 30 μl/min, data represent one out of three experiments. b FCS autocorrelation functions, normalized to the number of molecules, recorded for 1 nM VLαCD3-scFvαHLA-A2 alone (dark red) and for the same sample in presence of 1 µM VHαCD3-scFvαCD45 (light red) or 100 nM HLA-A2 antigen (blue). Curves are fitted using data derived from three independent experiments c Top: Equilibrium binding study of scFv-GpL (Gaussia princeps luciferase) fusion proteins specific for CD45, HLA-A2, or CD3 to the respective antigens on 105 Jurkat (CD3+, CD45+) and U266 cells (HLA-A2+, CD45+). Specific binding (triangles, solid line) was calculated as the difference of total (circles, dashed line) and non-specific binding (squares, dashed line) determined by using an irrelevant scFv-GpL fusion protein, HLA-A2-negative KMS-12-BM cells or CD3-negative U266 cells as indicated. Bottom: For heterologous competition analysis, cells were incubated with scFvCD45-GpL (2 nM), scFvHLA-A2-GpL (2 nM), or scFvCD3-GpL (4 nM) and the indicated concentrations of the hemibodies or the bispecific BiTE construct. IC50 values were determined and used to calculate the Ki of CD45-, HLA-A2-, and CD3-specific scFv domains by help of the previously determined KD-values of the scFv-GpL fusion proteins. Data show one out of three experiments yielding comparable results. d scFvCD3 or isolated VHCD3 and VLCD3 domains were fused to GpL. No specific binding of individual or combined constructs to CD3-positive Jurkat cells were detected as assessed by cell-bound luciferase activity. Data show one out of three experiments yielding comparable results. e Preservation of T cell triggering activity after incubation of hemibodies and BiTEs in human serum for 72 h at 37 °C. Read out was specific lysis at E:T of 5:1. Data represent the mean (±SD) from three replicate wells from one out of three experiments yielding comparable results. f In vivo serum half-lives after intravenous injection of 8 µg of a hemibody or BiTE construct into Balb/c mice. Values are deduced from >40 mice as determined by ELISA.
Fig. 4
Fig. 4
High precision cancer cell targeting in vivo. Immune deficient mice (6 per group) were challenged with 1 × 106 luciferase-positive THP-1 (CD45 and HLA-A2 positive) tumor cells intravenously (i.v.) on day 1. HLA-A2 negative memory CD4 and CD8 donor T lymphocytes were added i.v. on day 1, 22, and 28. After tumor engraftment on day 7, mice were treated subcutaneously with either saline (PBS), individual hemibodies addressing CD45 or HLA-A2 antigens, the combination of both hemibodies, or the HLA-A2 targeting BiTE control with a starting dose of 2 µg/mouse per day for 1 week, followed by 8 µg/mouse per day at distant sites until day 39. a Tumor burden of luciferase-positive THP-1 cells were assessed on a weekly basis by IVIS Lumina XR Real-Time Bioluminescence Imaging and b survival was monitored daily until day 110. Significance was determined by the Kaplan–Meier estimator; p = 0.0017 for the hemibody combination versus VLαCD3-scFvαHLA-A2, p = 0.0005 for the hemibody combination versus VHαCD3-scFvαCD45. No significant difference for the hemibody combination versus the bispecific BiTE control, p = 0.6670. The data represent one experiment, statistical considerations on effect and group size are provided in Supplementary Table 1.
Fig. 5
Fig. 5
Absence of bystander cytotoxicity and translation into solid tumor model. a Intracellular caspase-3 activation as a measure of apoptosis in HLA-A2/CD45 dual-positive THP-1 (upper panels) and CD45-positive but HLA-A2-negative bystander cells (bottom panels) was assessed by flow cytometry in vitro after co-culture with donor PBMCs and CD45 or HLA-A2 targeting hemibodies as indicated (3 nM). The HLA-A2 specific BiTE was used as a positive control (left panel); data represent one out of two experiments that yielded similar results. b Luciferase-expressing dual antigen-positive THP-1 and single antigen-positive RAJI cells were injected subcutaneously in the left and right thigh, respectively. Donor T cells were injected i.v. at day 1 and saline (PBS), paired hemibodies and the BiTE control (8 µg/mouse) were administered s.c. daily until day 7. Data are representative for two independent experiments. c Immune deficient mice were challenged i.v. with luciferase-expressing human breast cancer cells MDA-MB-231, which co-express EGFR and Her2/neu antigens. After engraftment of lung metastases on day 3, PBMCs from a healthy donor were administered i.v. followed by s.c. injection of a buffer control (saline), a CD3 × EGFR BiTE or the combination of EGFR and Her2/neu-specific hemibodies. Tumor burden was visualized by IVIS Lumina XR Real-Time Bioluminescence Imaging on days 1 and 8. One in vivo experiment out of two is shown. For in vitro analyses, data represent means (±SD) of triplicate wells from at least two independent experiments, E:T = 10:1.

References

    1. Kaplon H, Reichert JM. Antibodies to watch in 2019. MAbs. 2019;11:219–238. doi: 10.1080/19420862.2018.1556465. - DOI - PMC - PubMed
    1. Weiner JG. Building better monoclonal antibody-based therapeutics. Nat. Rev. Cancer. 2015;15:361–370. doi: 10.1038/nrc3930. - DOI - PMC - PubMed
    1. Hansel TT, et al. The safety and side effects of monoclonal antibodies. Nat. Rev. Drug Discov. 2010;9:325–338. doi: 10.1038/nrd3003. - DOI - PubMed
    1. Morgan RA. Risky business: target choice in adoptive therapy. Blood. 2013;122:3392–3394. doi: 10.1182/blood-2013-09-527622. - DOI - PMC - PubMed
    1. Swerdlow, S. H. et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, 4th edn (IARC, Lyon, 2008).

Publication types

MeSH terms

LinkOut - more resources