Acute myeloid leukemia (AML) is a high-risk disease with a poor prognosis, particularly in elderly patients. Because current AML treatment relies primarily on untargeted therapies with severe side effects that limit patient eligibility, identification of novel therapeutic AML targets is highly desired. We recently described AT1413, an antibody produced by donor B cells of a patient with AML cured after allogeneic hematopoietic stem cell transplantation. AT1413 binds CD43s, a unique sialylated epitope on CD43, which is weakly expressed on normal myeloid cells and overexpressed on AML cells. Because of its selectivity for AML cells, we considered CD43s as a target for a bispecific T-cell–engaging antibody (bTCE) and generated a bTCE by coupling AT1413 to two T-cell–targeting fragments using chemo-enzymatic linkage. In vitro, AT1413 bTCE efficiently induced T-cell–mediated cytotoxicity toward different AML cell lines and patient-derived AML blasts, whereas endothelial cells with low binding capacity for AT1413 remained unaffected. In the presence of AML cells, AT1413 bTCE induced upregulation of T-cell activation markers, cytokine release, and T-cell proliferation. AT1413 bTCE was also effective in vivo. Mice either coinjected with human peripheral blood mononuclear cells or engrafted with human hematopoietic stem cells [human immune system (HIS) mice] were inoculated with an AML cell line or patient-derived primary AML blasts. AT1413 bTCE treatment strongly inhibited tumor growth and, in HIS mice, had minimal effects on normal human hematopoietic cells. Taken together, our results indicate that CD43s is a promising target for T-cell–engaging antibodies and that AT1413 holds therapeutic potential in a bTCE-format.

Significance:

These findings offer preclinical evidence for the therapeutic potential of a bTCE antibody that targets a sialylated epitope on CD43 in AML.

Acute myeloid leukemia (AML) is a high-risk hematologic malignancy, with high mortality rates especially in elderly patients (1, 2). With a median age of 68 years at diagnosis, AML is predominantly a disease of the elderly population (3). For most AML subtypes, current standard of care has not improved over the past three decades and still relies on chemotherapy and allogeneic hematopoietic stem cell transplantation (HSCT; ref. 4). The severe side effects and comorbidities of these treatments limit their applicability especially in older patients (>60 years) and/or patients with poor physical fitness (5, 6). This poses an unmet medical need and highlights the need for better tolerated, broadly applicable AML treatments. To develop new therapeutic options for patients with AML, the identification of novel AML-specific targets is essential.

Earlier, we have examined whether patients with AML successfully treated with allogeneic HSCT generate AML-specific antibodies (7–9). Allogeneic HSCT can evoke an immunotherapeutic graft-versus-leukemia reaction, which includes a B-cell response and the generation of tumor-specific antibodies (10). From a number of allogeneic HSCT–treated patients with AML in durable remission we immortalized B cells, to generate clonal lines of donor-derived B cells, which produce antibodies reacting with autologous and allogeneic AML tumor samples (11, 12).

One of these antibodies is AT1413, which recognizes a unique sialylated epitope on CD43 (CD43s) and binds to all AML and myelodysplastic syndrome (MDS) blast samples that we tested (n = 80) including the patient's own blasts (8). These samples represent all World Health Organization 2008 types of AML and MDS. AT1413 also binds to healthy granulocytes and monocytes, but to a lower extend compared with AML or MDS blasts from the same donor, and weakly binds endothelial cells. No binding to other normal cells was detected in an IHC screen containing samples of multiple tissues (8).

AT1413 induces antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in vitro and inhibited growth of an AML cell line in an in vivo mouse model of AML, suggesting that AT1413 may have impacted the favorable clinical outcome of the patient it was isolated from.

Because of the preferential reactivity of AT1413 with AML cells, we reasoned that a bispecific T-cell–engaging antibody (bTCE) based on AT1413 could have increased cytotoxic potency and therapeutic potential. Many bTCEs are in clinical development, and two bTCEs have reached market approval (13). One of these, blinatumomab, is used to treat patients with relapsed or refractory B-cell precursor acute lymphatic leukemia and is undergoing clinical testing for other types of leukemia. Hence, the bTCE concept is clinically validated for hematologic malignancies (14, 15).

It has been suggested that the target of a bTCE needs to be located sufficiently close to the cell membrane, to form a stable cytolytic synapse between the target tumor cell and the T cell (16). AT1413 binds a sialylated epitope in the extracellular domain of CD43 (8). Being highly glycosylated, this extracellular domain is described to protrude away from the plasma membrane in a rod-like manner with a length of about 45 nm (17). While other CD43-targeting antibodies bind to membrane-distal CD43 regions, the CD43s epitope was mapped within a membrane-proximal part of the extracellular domain (8). This further encouraged us to generate a bTCE against CD43s as this favorable location of the CD43s epitope will likely support the bTCE's efficacy and potency (16).

To prepare bTCEs, antibodies or antibody fragments have successfully been equipped with single-chain variable fragments (scFv) derived from established CD3ϵ-binding antibodies (18–21). To convert AT1413 into a bTCE, we chose to chemo-enzymatically link a CD3ϵ-binding scFv, derived from the antibody UCHT1 (22), to the C-termini of the AT1413 IgG heavy chains in a C-to-C orientation. C-to-C fusion of the IgG and UCHT1 scFv is achieved by combining sortase transpeptidation and click chemistry (23–25). To abolish interaction with other non-T-cell immune effector cells, we removed Fc-gamma receptor (FcγR) interaction sites by introducing two point mutations G236R and L328R into the AT1413 heavy chains (AT1413-Fc0), as described previously (26). The resulting conjugation product is a bispecific antibody, in which two CD3ϵ-binding scFv are coupled to one IgG molecule. Here, we describe the generation of an AT1413 bTCE and demonstrate that this bTCE efficiently induces T-cell–mediated lysis of CD43s-expressing AML cells in vitro and in vivo.

Protein expression and purification

S. aureus sortase A (residues 82–249) with an N-terminal His6-tag was expressed and purified as described previously (27). The E. coli strain expressing S. aureus sortase A was a gift from Hidde Ploegh (Harvard Medical School, Boston, MA).

C-terminally LPETGGH6-tagged (tag sequence: GGGGSLPETGGHHHHHH) antibodies were cloned, expressed, and purified as described previously (24).

The UCHT1 scFv sequence was derived from the antibody UCHT1 (22, 28), by fusing the UCHT1 heavy and light chain variable regions, with the heavy chain variable domain oriented N-terminal to the light chain variable domain and a (G4S)3-linker sequence in between both domains, and adding a C-terminal LPETGGH6-tag. The construct was cloned into a pXC19 vector (Lonza), using synthetic codon-optimized open reading frames (GeneArt) encoding the human VK3 leader sequence. LPETGGH6-tagged UCHT1 scFv was stably expressed in CHO-GS cells and purified by immobilized metal affinity chromatography (IMAC) and size exclusion chromatography (SEC). IMAC was conducted on HisTrap excel and SEC on HiLoad Superdex 200 16/600 (GE Healthcare) columns using an ÄKTA Explorer 10S System (GE Healthcare).

Generation, purification, and stability of bTCE

AT1413 bTCE and AT1002 bTCE molecules were generated and purified as described previously (25).

bTCE stability was assessed by incubation of 0.1 μmol/L bTCE in PBS or IMDM + 8% FBS at 37°C for up to 21 days followed by SDS-PAGE and flow cytometric analysis.

Primary cell isolation and cell culture

Study protocols were approved by the institutional Medical Ethical Committee of Amsterdam UMC, location AMC (Amsterdam, the Netherlands). Human peripheral blood mononuclear cells (PBMC) were obtained from buffy coats of healthy blood donors (Sanquin) and AML blasts from peripheral blood (PB) or bone marrow (BM) of patients with AML by ficoll density gradient centrifugation, after obtaining written informed consent, and cryopreserved or used directly.

For cytotoxicity assays, PBMCs and AML blasts were thawed and rested in IMDM Medium (Gibco) supplemented with 8% FBS (Gibco) and 100 IU/mL penicillin and 100 μg/mL streptomycin (PS; Roche) overnight.

CD8-positive T cells for T-cell cultures were isolated from PBMCs by FACS on a FACSAria System (BD Biosciences). T cells were expanded in the presence of 1 eq irradiated (25 Gy) JY cells and 10 eq irradiated (100 Gy) PBMCs with 0.5 μmol/L phytohaemagglutinin (BioTrading) in Yssel medium (29) containing 1% human serum (ATCC), 0.1 nmol/L recombinant human IL2 (Sigma), and PS. Medium was renewed every 3–4 days and fresh IL2 was added.

Cell lines were obtained from DSMZ (SH-2, Molm13, BV173, and Kasumi3) or ATCC (THP-1, HL-60, Jurkat, and U266) and endothelial cells from Lonza (HAEC and HUVEC) and were maintained as recommended by the supplier. Jurkat TCRα−/− cells were a gift from Ton Schumacher (Netherlands Cancer Institute, Amsterdam, the Netherlands). Cell lines and endothelial cells were tested monthly for Mycoplasma by PCR. Cell lines were authenticated using short tandem repeat analysis.

Flow cytometry

Flow cytometry was conducted on a FACS Canto II (BD Biosciences) or a FACS LSR Fortessa X-20 (BD Biosciences). Data were processed with FlowJo 10.1 (FlowJo, LLC) and Prism 8.0.1 (GraphPad Software, Inc.).

FACS binding assays were carried out in 384-well plates in 1% BSA (Roche) in PBS (Gibco) at 4°C. Cells were harvested, washed, and incubated with antibody, scFv, or bTCE at 12,000 cells per well for 30 minutes. After washing, surface bound antibody and bTCE were detected with AF647 goat anti-human IgG H+L (Thermo Fisher Scientific) and UCHT1 scFv with AF647 anti-PentaHis (Qiagen) by incubating for 30 minutes in the dark. Cells were washed and resuspended in 10 nmol/L 4′,6-diamidino-2-fenylindole (DAPI; Sigma) directly before cytometric analysis.

In vitro cytotoxicity assays

Cytotoxicity was assessed by a modified Calcein AM retention assay adjusted for T-cell engagement (30). PBMCs or T-cell–depleted PBMCs were added at an effector-to-target (E:T) ratio of 10:1 (unless otherwise indicated) to target cells before incubation at 37°C and 5 % CO2 for 6–44 hours.

Whole-blood assays were performed likewise, but by adding PB from healthy volunteers instead of PBMCs to target cells before incubating for 5 hours at 37°C and 5 % CO2. Leukocytes were stained with anti-human CD66b PE and CD14 APC-Cy7 (BioLegend) and live cells with an eFluor660 Viability Marker (eBioscience). Red blood cells were lysed by adding BD FACS Lysing Solution (BD Biosciences), incubating for 15 minutes at 22°C, centrifuging, discarding supernatant, and repeating this step once. Accudrop beads were added and samples analyzed as above.

T-cell activation assays

AML target cells were allowed to opsonize AT1413 bTCE or control as during cytotoxicity assays and were incubated with PBMCs (E:T ratio 10:1 or 2:1) or T cells (E:T ratio of 1:1), for 24–48 hours at 37°C and 5% CO2. T cells were stained with anti-human CD4 PE-Cy7, CD8a APC-Cy7, CD25 APC, CD45RO FITC (all BioLegend), CD69 PE/APC (BioLegend/BD Biosciences), CD107a APC (BD Biosciences), and/or CD197 (CCR7) PE (BD Biosciences) for 30 minutes in the dark or during incubation (CD107a). Cells were washed, resuspended in 10 nmol/L DAPI, and analyzed by flow cytometry. T cells were selected as DAPI-negative, gated for CD4- or CD8-positive, where applicable for CD45RO- and CD197-positive or -negative, and for CD69-, CD25-, or CD107a-positive cells. Statistical testing was performed with Prism 8.0.1.

IFNγ ELISA

AML target cells were incubated with AT1413 bTCE or control and T cells for 48 hours as for T-cell activation assays. Cell-free supernatants were collected and diluted 4–10× with ELISA/ELISPOT Diluent (Thermo Fisher Scientific). ELISAs were performed with a Human IFN gamma ELISA Ready-SET-Go! Kit (Thermo Fisher Scientific) in a 384-well format. Absorbance at 450 nm was monitored with an EnVision 2104 Multilabel Reader (PerkinElmer).

Proliferation assays

Cultured T cells were washed with PBS, stained with 5 μmol/L Celltrace CFSE in PBS at 37°C and 5% CO2 for 20 minutes and washed with IMDM medium + 8% FBS. In 96-well U-bottom plates, 25,000 target cells per well were preincubated with AT1413 bTCE or control in IMDM medium + 8% FBS for 15 minutes at 22°C. T cells were added (E:T ratio 1:1) and samples were incubated for 5 days at 37°C and 5% CO2; then stained with APC-Cy7 anti-human CD8a as described for T-cell–activation assays, washed, resuspended in 10 nmol/L DAPI, and analyzed by flow cytometry.

In vivo experiments

Ethical consideration.

All animal protocols were carried out in accordance with Dutch and European laws. Experiments were approved by the AMC Animal Experiments Commission (Dierexperimentencommissie) and conducted according to AMC's institutional guidelines.

Generation of human immune system mice.

Human immune system (HIS) mice were generated using NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG; The Jackson laboratory) mice. Briefly, NSG newborns before 5 days of age were irradiated with 1 Gy using a Cs137 source and injected within 24 hours with 5 × 104 LinCD34+CD38 human stem cells obtained from fetal liver.

Efficacy studies against SH-2.

At 7–8 weeks, 12 female NSG mice (19.9 ± 1.5 g) were injected intravenously in the tail vein with 10 × 106 luciferase/ZsGreen-labeled SH-2 cells in 100 μL of PBS (day 0). Starting after 15 days, tumor growth was assessed weekly by bioluminescence imaging (BLI) using a Photon Imager (Biospace Lab). Mice were injected intraperitoneally with VivoGlo Luciferin, In Vivo Grade (3.75 mg, Promega) 15 minutes before image acquisition. During the acquisition, the mice were anesthetized using Isoflurane and maintained on a thermally controlled pad. Mice were weighted twice per week. Mice losing more than 15 % of their maximum weight were humanely sacrificed.

At day 19, mice were randomized and allocated to groups of 6 mice each. Mice were injected intravenously with 5 × 106 PBMC and directly given the first dose of 40 μg bTCE in 100 μL PBS i.p. Subsequent bTCE doses were biweekly injected intravenously from day 22 to day 33. Blood samples were collected from the submandibular vein. Mice were sacrificed on day 36 or day 40 by cervical dislocation after deep isoflurane anesthesia. The drug injection and measurements were performed by technicians blinded to the group allocation. Tumor growth inhibition was calculated as the median of tumor size of the AT1413 bTCE–treated group divided by the one from the control group at the end of the experiment.

Studies with HIS-NSG mice were conducted in the same way, but without PBMC injection and with the following variations. At 12–13 weeks of age, 7 female HIS-NSG mice (20.3 ± 1.4 g) were injected intravenously with 8 × 106 luciferase/ZsGreen-labeled SH-2 cells. Weekly BLI was started on day 12. Mice were treated biweekly with 40 μg of bTCE (4 mice AT1413 bTCE group, 3 mice control group) from day 23 to 40 after tumor engraftment. Blood samples were collected on day 30, day 40, and at sacrifice (day 43). Statistical testing was performed in Prism 8.0.1.

Efficacy studies against primary AML samples.

At 4–7 weeks, 12 females NOD.Cg-Prkdcscid Il2rgtm1Sug Tg(SV40/HTLV-IL3,CSF2)10-7Jic/JicTac (hGM-CSF/hIL-3 NOG; Taconic; 16.2 g ± 0.9 g) were injected intravenously with AML blasts isolated from the PB of a patient with AML (BL059); T cells were depleted using a CD3 MicroBeads MACS Kit (Miltenyi Biotech) and 1.2 × 106 cells/mouse were injected intravenously. The engraftment of the AML cells was followed through time by blood sampling and FACS analysis for CD45+CD33+ cells. When AML blasts could be reliably detected in the blood (≥1% of live cells), mice were injected with PBMC and treated as described above.

AT1413 bTCE is assembled by sortase-catalyzed transpeptidation and click chemistry

C-to-C fusion of AT1413-Fc0 and UCHT1 scFv into a bTCE-format was accomplished by the combination of sortase transpeptidation and click chemistry (Fig. 1A; refs. 23–25). Methyltetrazine (MeTz) and trans-Cyclooctene (TCO) were used as complementary click chemistry handles (Fig. 1B; ref. 31). To prepare AT1413 bTCE, AT1413-Fc0-MeTz was incubated with UCHT1-TCO; the reaction product was purified by SEC (Supplementary Fig. S1A).

Figure 1.

Schematic representation of AT1413 bTCE generation. A, Schematic representation of the bTCE-format. B, Reaction scheme of the bTCE synthesis involving (1) the incorporation of tetrazine/TCO by sortase-catalyzed transpeptidation and (2) a tetrazine-TCO ligation.

Figure 1.

Schematic representation of AT1413 bTCE generation. A, Schematic representation of the bTCE-format. B, Reaction scheme of the bTCE synthesis involving (1) the incorporation of tetrazine/TCO by sortase-catalyzed transpeptidation and (2) a tetrazine-TCO ligation.

Close modal

Coupling of AT1413-Fc0-MeTz and UCHT1-TCO scFv by TCO-tetrazine cycloaddition proceeded nearly quantitatively as indicated by reducing and nonreducing SDS-PAGE (Supplementary Fig. S1B). The integrities of AT1413 bTCE, as well as its components, AT1413-Fc0 and UCHT1 scFv, were maintained for up to 21 days when incubated in PBS at 37°C (Supplementary Fig. S2).

Analogously, a control bTCE was prepared from the antibody AT1002 that recognizes an irrelevant target (influenza group 2 HA-protein; ref. 24).

AT1413 bTCE binds AML and T-cell receptor complex–expressing cells

To confirm that AT1413 bTCE retained dual binding capacity to CD43s- and CD3ϵ-expressing cells, binding to the SH-2 AML and Jurkat T-cell lines was compared with that of bTCE components, AT1413-Fc0 and UCHT1 scFv, by flow cytometry. Comparison of median fluorescence intensity signals for AT1413-Fc0 and AT1413 bTCE on SH-2 cells indicated only a slight signal reduction for the latter (Supplementary Fig. S3). Both, AT1413 bTCE and the control bTCE, bound Jurkat cells with similar half-maximal concentrations as unmodified UCHT1 scFv. TCRα−/− Jurkat cells, devoid of CD3ϵ surface expression, were used to confirm binding specificity toward CD3ϵ as part of the TCR complex. Complete lack of bTCE and UCHT1 scFv binding to TCRα−/− Jurkat cells was observed.

bTCE binding to SH-2 and Jurkat cells was maintained for up to 21 days of incubation at 37°C in PBS or IMDM medium (Supplementary Fig. S4).

The binding of AT1413 bTCE on SH-2 and Jurkat cells indicates that the bTCE retained the binding capacities of both components, AT1413-Fc0 IgG and UCHT1 scFv.

AML cell lines and primary AML, but not primary endothelial cells, are lysed by AT1413 bTCE–redirected T cells

AT1413 bTCE was tested for inducing T-cell–mediated lysis of CD43s-expressing SH-2 cells in an in vitro Calcein AM retention assay (30). Using PBMCs as effector cells at an E:T ratio of 10:1, concentration-dependent target cell lysis was observed with EC50 values of 30–60 pmol/L (Fig. 2A; Table 1). Neither the control bTCE, nor UCHT1 scFv or AT1413-Fc0 induced target cell death. Incubation of TCRα−/− Jurkat cells with AT1413 bTCE and PBMC did not induce target cell lysis, assuring target cell specificity (Fig. 2A). At decreased E:T ratios of as low as 0.5:1, AT1413 bTCE–induced SH-2 target cell lysis was retained, albeit with gradually reduced efficacies (Supplementary Fig. S5).

Figure 2.

In vitro cytotoxicity of AT1413 bTCE against AML but not endothelial cells. Redirected T-cell–mediated lysis monitored by viable target cell count in Calcein AM retention assay with PBMCs as effector cells (unless otherwise indicated). A, SH-2 AML and Jurkat TCRα−/− target cell lines treated with AT1413 bTCE or control drugs. B, A panel of selected AML cell lines: THP-1, Molm13, HL-60, and Kasumi3, the CML cell line BV173 and primary AML blasts (donor BL-123) treated with AT1413 bTCE or AT1002 bTCE control. C, SH-2 cells incubated with full or T-cell–depleted PBMCs as effector cells and treated with AT1413 bTCE. D, Endothelial cells (HAEC and HUVEC) treated with AT1413 bTCE or AT1002 bTCE control.

Figure 2.

In vitro cytotoxicity of AT1413 bTCE against AML but not endothelial cells. Redirected T-cell–mediated lysis monitored by viable target cell count in Calcein AM retention assay with PBMCs as effector cells (unless otherwise indicated). A, SH-2 AML and Jurkat TCRα−/− target cell lines treated with AT1413 bTCE or control drugs. B, A panel of selected AML cell lines: THP-1, Molm13, HL-60, and Kasumi3, the CML cell line BV173 and primary AML blasts (donor BL-123) treated with AT1413 bTCE or AT1002 bTCE control. C, SH-2 cells incubated with full or T-cell–depleted PBMCs as effector cells and treated with AT1413 bTCE. D, Endothelial cells (HAEC and HUVEC) treated with AT1413 bTCE or AT1002 bTCE control.

Close modal
Table 1.

Summary of in vitro potencies and efficacies of AT1413 bTCE–mediated target cell lysis for different AML target cell lines incubated with PBMCs (E:T ratio 10:1)

AML cell lineEC50 (pmol/L)Max. kill (%)
SH-2 31–56 45–92 
THP-1 252 78 
Molm13 45–90 33–73 
BV173 39–85 20–53 
HL-60 75–187 26–50 
Kasumi3 25 29 
AML cell lineEC50 (pmol/L)Max. kill (%)
SH-2 31–56 45–92 
THP-1 252 78 
Molm13 45–90 33–73 
BV173 39–85 20–53 
HL-60 75–187 26–50 
Kasumi3 25 29 

AT1413 bTCE was also tested on other cell lines and primary AML blast cells. AT1413 binds the AML cell lines THP-1, Molm13, HL-60, and Kasumi3, as well as the B-cell precursor leukemia cell line BV173, although at reduced levels compared with SH-2 cells (8). Notably, AT1413 bTCE demonstrated in vitro cytotoxicity also toward these cell lines, with EC50 values in the range of 40–250 pmol/L (Fig. 2B; Table 1). Activity of AT1413 bTCE toward primary AML blast cells was tested on 14 samples of PB or BM from patients with various AML subtypes (Fig. 2B; Supplementary Table S1). AT1413 bTCE induced lysis in all 14 tested AML blast samples; blasts were lysed with a wide range in efficacy (maximal lysis: 9%–92%). No correlations between the extend of maximal target cell lysis and AML subtype or AT1413 binding were observed in samples, where corresponding data were available (Supplementary Fig. S6A). Consistently, no significant differences in AT1413 binding were found in a set of 52 blast samples of various AML subtypes (Supplementary Fig. S6B).

To confirm that AT1413 bTCE–induced lysis is mediated by T-cell cytotoxicity, Calcein AM retention assay was performed with T-cell–depleted PBMCs as effector cells, which abolished target cell lysis (Fig. 2C).

Previously, AT1413 was shown to bind normal endothelial cells, but no triggering of ADCC nor of CDC against endothelial cells was observed (8). Because AT1413 bTCE has a higher cytotoxic potency than AT1413, we investigated whether AT1413 bTCE triggers T cells to lyse endothelial cells isolated from human aorta (HAEC) and human umbilical vein (HUVEC). Although AT1413 binds to these endothelial cells at IgG concentrations of ≥6.7 nmol/L, neither HAEC nor HUVEC were lysed by PBMCs in the presence of AT1413 bTCE (Fig. 2D).

Expression of T-cell activation markers by AT1413 bTCE requires CD43s+ target cells

To confirm that AT1413 bTCE activates T cells only in the presence of CD43s-expressing target cells, upregulation of the T-cell activation markers CD69 and CD25 was monitored. In the presence of SH-2 cells, CD69 expression was observed only on T cells incubated with AT1413 bTCE, but not with the control AT1002 bTCE, AT1413-Fc0, or UCHT1 scFv alone (Fig. 3A). In the absence of target cells, AT1413 bTCE did not induce CD69 expression. CD69 expression on CD4- and CD8-positive T cells from PBMCs of four different donors was compared (Supplementary Fig. S7A). While PBMC donors showed variations in the maximal percentage of CD69- CD4-positive and CD8-positive T cells, potency was similar for all donors.

Figure 3.

On T cells, activation markers are upregulated by AT1413 bTCE only in the presence of target cells. A and B, Percentages of CD69-positive CD8-positive T cells after incubation with AT1413 bTCE, AT1002 bTCE, and anti-CD3/anti-CD28 containing beads, AT1413-Fc0 or UCHT1 scFv (A) and CD25-positive CD4- and CD8-positive T cells after incubation with AT1413 bTCE or AT1002 bTCE in the presence or absence of SH-2 target cells (B). C and D, Percentages of CD69- and CD25-positive (C) and CD107a-positive CD4- and CD8-positive T cells (D) within different T-cell subsets according to CD45RO and CCR7 gating (naïve, CM, E, and EM) after incubation with AT1413 bTCE or AT1002 bTCE control. Dot plots, overlay of activation marker expressing T cells (colored dots) with all T cells (density plot) of the same subset. Statistical analysis, two-way ANOVA (C and D); *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant.

Figure 3.

On T cells, activation markers are upregulated by AT1413 bTCE only in the presence of target cells. A and B, Percentages of CD69-positive CD8-positive T cells after incubation with AT1413 bTCE, AT1002 bTCE, and anti-CD3/anti-CD28 containing beads, AT1413-Fc0 or UCHT1 scFv (A) and CD25-positive CD4- and CD8-positive T cells after incubation with AT1413 bTCE or AT1002 bTCE in the presence or absence of SH-2 target cells (B). C and D, Percentages of CD69- and CD25-positive (C) and CD107a-positive CD4- and CD8-positive T cells (D) within different T-cell subsets according to CD45RO and CCR7 gating (naïve, CM, E, and EM) after incubation with AT1413 bTCE or AT1002 bTCE control. Dot plots, overlay of activation marker expressing T cells (colored dots) with all T cells (density plot) of the same subset. Statistical analysis, two-way ANOVA (C and D); *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant.

Close modal

CD25 expression was also detected on both CD4- and CD8-positive T cells within PBMCs, but percentages of CD25-positive T cells were lower than those of CD69-positive T cells (Fig. 3B). In addition, the EC50 values for CD25 expression (140–170 nmol/L for CD4-positive T cells and 170–200 pmol/L for CD8-positive T cells) were higher than those for CD69 expression. Importantly, no activation markers were induced in PBMC in the absence of target cells, which implies that monocytes despite expression of low levels of CD43s do not activate T cells in the presence of AT1413 bTCE.

Using previously stimulated, resting CD8-positive T cells, higher CD25-expression levels were observed with AT1413 bTCE, whereas cells treated with the control bTCE remained CD25-negative. In addition to SH-2, other AML cell lines, such as THP-1 and HL-60, induced CD25 expression when incubated with AT1413 bTCE in this setting (Supplementary Fig. S7B).

Expression of CD69 and CD25 was compared for naïve, effector (E), central memory (CM), and effector memory (EM) T-cell subsets (Fig. 3C). Both activation markers were significantly upregulated on CD4 and CD8 T cells across most subsets in AT1413 bTCE compared with AT1002 bTCE–treated samples. However, activation was more pronounced in some subsets compared with others. Of CD8 T cells, primarily those of CM or EM phenotype expressed CD69 and CD25. On CD4 T cells, CD25 expression showed a similar pattern, although less pronounced, whereas CD69 expression was also high on naïve CD4 T cells.

Cell surface expression of CD107a throughout the incubation with bTCE and target cells was also monitored for different T-cell subsets (Fig. 3D). Consistent with their primary cytotoxic function, mainly CD8 T cells stained CD107a positive. CD107a-positive CD8 T cells spread across the CM, E, and EM phenotypes, whereas naïve CD8 T cells, with low levels of preformed cytotoxic granules remained widely CD107a negative as expected.

AT1413 bTCE induces cytokine production and T-cell proliferation only in presence of CD43s+ tumor cells

Next, we studied the ability of AT1413 bTCE to induce cytokine (IFNγ) production, and to stimulate T-cell proliferation. IFNγ production was only observed when previously stimulated T cells were used as effector cells and was dependent on AT1413 bTCE and the presence of CD43s-expressing target cells (Fig. 4A). With the control bTCE or in the absence of target cells, no IFNγ was detected. With SH-2 as target cells, IFNγ production was observed at lower AT1413 bTCE concentrations than for THP-1 and HL-60 cells, consistent with AT1413 binding (Fig. 4B). When coincubated with SH-2 cells and AT1413 bTCE over 5 days, T cells proliferated in a concentration-dependent manner whereas control bTCE–treated T cells remained unaffected (Fig. 4C).

Figure 4.

AT1413 bTCE induces IFNγ production and T-cell proliferation. A and B, IFNγ concentration determined by ELISA in supernatants of cultured CD8-positive T cells incubated for 48 hours with AT1413 bTCE or AT1002 bTCE and with or without SH-2 target cells (A) or with different AML target cell lines (B). C, Proliferation of CellTrace CFSE–stained cultured CD8-positive T cells incubated with SH-2 cells and varying concentrations of AT1413 bTCE or 10 nmol/L AT1002 bTCE.

Figure 4.

AT1413 bTCE induces IFNγ production and T-cell proliferation. A and B, IFNγ concentration determined by ELISA in supernatants of cultured CD8-positive T cells incubated for 48 hours with AT1413 bTCE or AT1002 bTCE and with or without SH-2 target cells (A) or with different AML target cell lines (B). C, Proliferation of CellTrace CFSE–stained cultured CD8-positive T cells incubated with SH-2 cells and varying concentrations of AT1413 bTCE or 10 nmol/L AT1002 bTCE.

Close modal

In conclusion, our analyses of T-cell marker expression, cytokine production, and T-cell proliferation induced by AT1413 bTCE indicate that, in the presence of CD43s-positive target cells, AT1413 bTCE is capable of activating T cells and engaging them to kill CD43s-expressing AML cells.

AT1413 bTCE induces strong tumor growth inhibition of AML cell line SH-2 in vivo

To test the in vivo efficacy of AT1413 bTCE we used a xenograft NSG mouse model, inoculated with human luciferase/ZsGreen-labeled SH-2 cells and engrafted with human PBMC. Mice were treated biweekly with AT1413 bTCE or control (AT1002) bTCE with 2 mg/kg i.v. Treatment with AT1413 bTCE for 15 days resulted in tumor growth inhibition of 99% (P < 0.001) compared with AT1002 bTCE–treated mice, demonstrating that AT1413 bTCE is efficacious against SH-2 cells in vivo (Fig. 5A). During the course of the experiment, no obvious signs of discomfort or weight loss were observed.

Figure 5.

In vivo efficacy of AT1413 bTCE in PBMC coinjected NSG and HIS-NSG mice. A, Tumor growth in PBMC coinjected mice treated with AT1413 bTCE or AT1002 bTCE for individual mice (left) or as average (right). B, Tumor growth in HIS-NSG mice treated with AT1413 bTCE or AT1002 bTCE as average. Arrows, bTCE injection; suns, BLI. C, Percentages of human stem cells (CD34+), granulocytes (CD66b+), and monocytes (CD14+) in bone marrow or selected organs of HIS-NSG mice at sacrifice. Statistical analysis, two-way repeated measures ANOVA (A); mixed-effects analysis (REML; B); two-way ANOVA (C); **, P < 0.01; ***, P < 0.001; ns, not significant.

Figure 5.

In vivo efficacy of AT1413 bTCE in PBMC coinjected NSG and HIS-NSG mice. A, Tumor growth in PBMC coinjected mice treated with AT1413 bTCE or AT1002 bTCE for individual mice (left) or as average (right). B, Tumor growth in HIS-NSG mice treated with AT1413 bTCE or AT1002 bTCE as average. Arrows, bTCE injection; suns, BLI. C, Percentages of human stem cells (CD34+), granulocytes (CD66b+), and monocytes (CD14+) in bone marrow or selected organs of HIS-NSG mice at sacrifice. Statistical analysis, two-way repeated measures ANOVA (A); mixed-effects analysis (REML; B); two-way ANOVA (C); **, P < 0.01; ***, P < 0.001; ns, not significant.

Close modal

Next, AT1413 bTCE was tested in a HIS-mouse xenograft model, to better mimic its effect on human immune cell subsets. Successfully engrafted female HIS-NSG mice were inoculated with luciferase/ZsGreen-labeled SH-2 cells. AT1413 bTCE or control bTCE treatment (biweekly, 2 mg/kg i.v.) was initiated 23 days after AML inoculation. In this model, after 20 days of treatment, BLI revealed a tumor growth inhibition of 89% (P < 0.01) in AT1413 bTCE–treated mice compared with the control bTCE group (Fig. 5B).

To detect whether AT1413 bTCE had an effect on the engrafted human hematopoietic cells in vivo, populations of human hematopoietic stem cells, granulocytes, and monocytes were analyzed at sacrifice (after 20 days of treatment with AT1413 bTCE). In BM, the percentages of neither human hematopoietic stem cells (CD34+), nor monocytes (CD14+), nor granulocytes (CD66b+) were significantly reduced by AT1413 bTCE (Fig. 5C). In addition, liver, PB, and spleen monocytes were present at comparable percentages within the human compartment compared with the control group. To further investigate a potentially cytotoxic effect of AT1413 bTCE toward human monocytes or granulocytes, whole blood from healthy donors was incubated with AT1413 bTCE in the presence or absence of SH-2 cells in an in vitro assay. While up to 29% of SH-2 cells were lysed in this setup at 1 nmol/L AT1413 bTCE, cytotoxicity toward monocytes and granulocytes remained at a background level and was hardly affected by SH-2 cell lysis (Supplementary Fig. S8).

Taken together, AT1413 bTCE revealed strong tumor growth inhibition against SH-2 AML cells in vivo. In a HIS-NSG mouse model, AT1413 bTCE did not show a depletion of the engrafted human myeloid cells.

AT1413 bTCE reduces tumor burden in a patient-derived xenograft AML model

To assess its efficacy in vivo against primary AML blast cells, AT1413 bTCE was tested in a patient-derived xenograft (PDX) mouse model. hGM-CSF/hIL-3 NOG mice were engrafted with patient-derived (BL059), T-cell–depleted AML blast samples. Greater than or equal to 1% CD45+CD33+ cells were observed on day 49 after engraftment. Treatment was initiated 4 days later and CD45+CD33+ cells were monitored on day 59.

On day 59, CD45+CD33+ cells were reduced in AT1413 bTCE–treated mice by a median of 89% compared with day 49 (Fig. 6A). In the same period, CD45+CD33+ cells further increased by a median of 57% in the AT1002 bTCE–treated mice. At sacrifice, the percentages of human CD33+ cells within human CD45+ cells were assessed in PB, BM, liver, and spleen (Fig. 6B). In all three organs and PB, CD33+ cells were significantly reduced in AT1413 bTCE compared with AT1002 bTCE–treated mice.

Figure 6.

In vivo efficacy of AT1413 bTCE against primary AML blast samples. A, Human CD33+ cells in the blood of PDX mice on day 59 as percentage of initial number before start of treatment (day 49) in the same animal. B, Percentages of human CD33+ cells within human CD45+ cells in BM, liver, PB, and spleen at sacrifice. Statistical analysis, Mann–Whitney test (A); **, P < 0.01; error bars, median with 95 % confidence interval; two-way ANOVA (B), significant at P < 0.05 for treatment allocation.

Figure 6.

In vivo efficacy of AT1413 bTCE against primary AML blast samples. A, Human CD33+ cells in the blood of PDX mice on day 59 as percentage of initial number before start of treatment (day 49) in the same animal. B, Percentages of human CD33+ cells within human CD45+ cells in BM, liver, PB, and spleen at sacrifice. Statistical analysis, Mann–Whitney test (A); **, P < 0.01; error bars, median with 95 % confidence interval; two-way ANOVA (B), significant at P < 0.05 for treatment allocation.

Close modal

With AT1413 bTCE we have generated a T-cell–engaging antibody against CD43s as a potential new drug for the treatment of AML. Previously we had demonstrated that AT1413 induced ADCC in vitro (5). By converting AT1413 into a bTCE, we expected to increase its cytotoxic potency. Indeed, AT1413 bTCE induced SH-2 target cell death with EC50 values of 30–60 pmol/L, whereas naked AT1413 antibody induced ADCC with EC50 values only in the low nanomolar range (8). In addition, we demonstrated AT1413 bTCE to induce lysis of other AML cell lines, as well as patient-derived primary AML blast cells. Consistently, we observed more efficient tumor growth inhibition in vivo using a lower dose of AT1413 bTCE (2 mg/kg) compared with the naked AT1413 antibody (15 mg/kg). Further, AT1413 bTCE also inhibited growth of patient-derived primary AML blasts in an in vivo PDX model. Despite the potency increase, AT1413 bTCE did not mediate lysis of monocytes or granulocytes, which expressed some CD43s, in an in vitro cytotoxicity assay in whole blood. Consistently, human monocytes were not depleted by AT1413 bTCE in the in vivo HIS-NSG mouse model. We conclude therefore that AT1413 bTCE has a very strong tumor reducing activity without significantly impacting the normal myeloid cells.

Although the final reason for this difference remains elusive, we expect that the lower level of CD43s expression protects normal myeloid cells from T-cell–mediated lysis or at least reduces their susceptibility. To directly compare lysis of primary AML blasts and normal myeloid cells from the same donor, whole-blood Calcein AM retention assays could be conducted on fresh AML patient samples, which contain similar amounts of AML blasts compared with the normal myeloid populations. In addition to the CD43s expression level, protection of normal myeloid cells against T-cell–mediated lysis may explain the difference. For instance, naïve blood monocytes have been described to be able to suppress T-cell function (32), which is partially dependent on nitric oxide synthase (NOS). If this is applicable, AT1413 bTCE–mediated T-cell cytotoxicity against this monocyte population should be enhanced by the addition of a NOS inhibitor. Finally, we speculate that the target, CD43s, may be exposed differently on AML cells compared with normal myeloid cells hampering efficient formation of a cytolytic synapse.

Compared with other bTCE molecules, features of our bTCE-format are, its relatively high molecular weight and its bivalent binding to the tumor antigen. Unlike smaller bTCE-formats such as the bispecific T-cell engager or the dual-affinity retargeting protein formats, our bTCE-format is expected to have a longer in vivo half-life, as it is not prone to renal clearance (33–35). Further advantages of our bTCE-format are the perpetuation of integrity and stability of the full-length parent IgG, as well as its bivalency for tumor cell binding. Particularly if the T-cell–engaging antibody binds a glycopeptide epitope, as in case of AT1413 bTCE, antibodies often have lower affinities as when targeting peptide epitopes (36). Thus, the avidity effect of bivalent target binding gives a valuable addition to its overall functional affinity.

Our bTCE-format contains two CD3ϵ-binding domains. Knowing that CD3ϵ-targeting antibodies such as UCHT1 and OKT3 are mitogenic toward T cells and observing that this effect is not fully abrogated by using an antibody with abolished FcγR interaction, one of our initial concerns was whether bivalent binding of AT1413 bTCE to CD3ϵ could cause CD43s-independent T-cell activation (37). However, AT1413 bTCE neither induced lysis of CD43s-negative TCRα−/− Jurkat cells nor activation of T cells in the absence of CD43s-positive target cells. Likewise, the AT1002 control bTCE had no effect on either target cell death or T-cell activation. Hence, triggering of T-cell activation and cytotoxicity by this bTCE-format is dependent on target binding, which is consistent with previously described T-cell–engaging antibody formats with bivalent CD3ϵ binding (19, 38).

AT1413 bTCE is the first bTCE generated through chemo-enzymatic linkage of a CD3ϵ-targeting scFv to an IgG by combining sortase-catalyzed transpeptidation and click chemistry. This bTCE-format provides a platform for simple conversion of any recombinantly expressed antibody into a T-cell–engaging format by adding just the short six amino acid sortase-recognition motif (LPETGG) to its heavy chain C-termini. Thus, extensive antibody engineering, which is often required to prepare an antibody-fusion product, or other bispecific formats (e.g., knob-into-hole) may be bypassed. This allows for quick screening of panels of antibody candidates for use as a bTCE. Alternative to T-cell–engaging antibodies, bispecific antibodies with other modalities (e.g., cytokines) or antibody–drug conjugates are readily accessible, by coupling a functional group of interest to the antibody C-termini via the presented sortase + click procedure (39).

To summarize, with AT1413 bTCE we present a bTCE against CD43s, a novel target overexpressed in AML. Efficacy of AT1413 bTCE toward AML cell lines and toward primary AML blasts in vitro and in vivo validates both the antibody and the target for T-cell engagement, and confirms the therapeutic potential of AT1413 in a T-cell–engaging format.

M.A. Gillissen has ownership interest (including stock, patents, etc.) in AIMM Therapeutics. A.Q. Bakker is a senior scientist at AIMM Therapeutics. H. Spits reports receiving commercial research grant from, has ownership interest (including stock, patents, etc.) in AIMM Therapeutics, and is a consultant/advisory board member for GSK. K. Wagner has ownership interest (including stock, patents, etc.) in AIMM Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: G. de Jong, A.Q. Bakker, M.D. Hazenberg, H. Spits, K. Wagner

Development of methodology: L. Bartels, M.A. Gillissen, E. Yasuda, C. Bru, P.M. van Helden, J. Villaudy, K. Wagner

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Bartels, G. de Jong, E. Yasuda, V. Kattler, C. Bru, C. Fatmawati, S.E. van Hal-van Veen, M.G. Cercel, J. Villaudy, M.D. Hazenberg, K. Wagner

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L. Bartels, E. Yasuda, C. Fatmawati, J. Villaudy, K. Wagner

Writing, review, and/or revision of the manuscript: L. Bartels, G. de Jong, G. Moiset, P.M. van Helden, J. Villaudy, M.D. Hazenberg, H. Spits, K. Wagner

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): V. Kattler, C. Fatmawati, G. Moiset, A.Q. Bakker, K. Wagner

Study supervision: A.Q. Bakker, P.M. van Helden, M.D. Hazenberg, H. Spits, K. Wagner

Other (in vivo and in vitro experiments): C. Bru

The authors thank the patients for participating in this study and the Trial Office of the Amsterdam UMC, location AMC, Department of Hematology for the provision of the AML samples. We would also like to thank Andrea Solar and Wouter Pos for their initial contribution to the work on bispecific antibodies at AIMM Therapeutics and Sophie Levie for her assistance in testing AT1413 binding to AML blast samples. This study was financially supported by the Netherland's Organization for Scientific Research (NWO), the Dutch Cancer Society (KWF), and Amsterdam UMC, location AMC. Zwaartekracht grant by the Netherland's Organization for Scientific Research (NWO) to H. Spits (ICI00004 to L. Bartels). Intramural AMC PhD Scholarship (to M. A. Gillissen). KWF Kankerbestrijding to H. Spits and M.D. Hazenberg (CA301006 to G. de Jong and G. Moiset). NWO ZonMW VIDI to M.D. Hazenberg (grant no. 91715362).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Alibhai
SMH
,
Leach
M
,
Minden
MD
,
Brandwein
J
. 
Outcomes and quality of care in acute myeloid leukemia over 40 years
.
Cancer
2009
;
115
:
2903
11
.
2.
Almeida
AM
,
Ramos
F.
Acute myeloid leukemia in the older adults
.
Leuk Res Reports
2016
;
6
:
1
7
.
3.
Howlader
N
,
Noone
A
,
Krapcho
M
,
Miller
D
,
Bishop
K
,
Kosary
C
, et al
SEER cancer statistics review, 1975–2014
.
NCI
.
Bethesda, MD
; 
2017
.
Available from
: https://seer.cancer.gov/archive/csr/1975_2014/.
4.
Dombret
H
,
Gardin
C.
An update of current treatments for adult acute myeloid leukemia
.
Blood
2016
;
127
:
53
62
.
5.
Krug
U
,
Büchner
T
,
Berdel
WE
,
Müller-Tidow
C
. 
The treatment of elderly patients with acute myeloid leukemia
.
Dtsch Arztebl Int
2011
;
108
:
863
70
.
6.
Estey
E
,
de Lima
M
,
Tibes
R
,
Pierce
S
,
Kantarjian
H
,
Champlin
R
, et al
Prospective feasibility analysis of reduced-intensity conditioning (RIC) regimens for hematopoietic stem cell transplantation (HSCT) in elderly patients with acute myeloid leukemia (AML) and high-risk myelodysplastic syndrome (MDS)
.
Blood
2007
;
109
:
1395
400
.
7.
Gillissen
MA
,
De Jong
G
,
Levie
SE
,
Yasuda
E
,
Bakker
AQ
,
Evers
LM
, et al
AML relapse after rituximab treatment for GvHD: crucial role for B cells in GvL responses
.
Bone Marrow Transplant
2016
;
51
:
1245
8
.
8.
Gillissen
MA
,
Jong
G De
,
Kedde
M
,
Yasuda
E
,
Levie
SE
,
Moiset
G
, et al
Patient-derived antibody recognizes a unique CD43 epitope expressed on all AML and has antileukemia activity in mice
.
Blood Adv
2017
;
1
:
1551
64
.
9.
Gillissen
MA
,
Kedde
M
,
Jong
G de
,
Moiset
G
,
Yasuda
E
,
Levie
SE
, et al
AML-specific cytotoxic antibodies in patients with durable graft versus leukemia responses
.
Blood
2018
;
131
:
131
43
.
10.
Wu
CJ
,
Yang
XF
,
McLaughlin
S
,
Neuberg
D
,
Canning
C
,
Stein
B
, et al
Detection of a potent humoral response associated with immune-induced remission of chronic myelogenous leukemia
.
J Clin Invest
2000
;
106
:
705
14
.
11.
Kwakkenbos
MJ
,
Diehl
SA
,
Yasuda
E
,
Bakker
AQ
,
van Geelen
CMM
,
Lukens
MV
, et al
Generation of stable monoclonal antibody–producing B cell receptor–positive human memory B cells by genetic programming
.
Nat Med
2010
;
16
:
123
8
.
12.
Kwakkenbos
MJ
,
van Helden
PM
,
Beaumont
T
,
Spits
H
. 
Stable long-term cultures of self-renewing B cells and their applications
.
Immunol Rev
2016
;
270
:
65
77
.
13.
Strohl
WR
. 
Current progress in innovative engineered antibodies
.
Protein Cell
2018
;
9
:
86
120
.
14.
Aigner
M
,
Feulner
J
,
Schaffer
S
,
Kischel
R
,
Kufer
P
,
Schneider
K
, et al
T lymphocytes can be effectively recruited for ex vivo and in vivo lysis of AML blasts by a novel CD33/CD3-bispecific BiTE antibody construct
.
Leukemia
2013
;
27
:
1107
15
.
15.
Kantarjian
H
,
Stein
A
,
Gökbuget
N
,
Fielding
AK
,
Schuh
AC
,
Ribera
J-M
, et al
Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia
.
N Engl J Med
2017
;
376
:
836
47
.
16.
Bluemel
C
,
Hausmann
S
,
Fluhr
P
,
Sriskandarajah
M
,
Stallcup
WB
,
Baeuerle
PA
, et al
Epitope distance to the target cell membrane and antigen size determine the potency of T cell-mediated lysis by BiTE antibodies specific for a large melanoma surface antigen
.
Cancer Immunol Immunother
2010
;
59
:
1197
209
.
17.
Cyster
JG
,
Shotton
DM
,
Williams
AF
. 
The dimensions of the T lymphocyte glycoprotein leukosialin and identification of linear protein epitopes that can be modified by glycosylation
.
EMBO J
1991
;
10
:
893
902
.
18.
Kuo
SR
,
Wong
L
,
Liu
JS
. 
Engineering a CD123xCD3 bispecific scFv immunofusion for the treatment of leukemia and elimination of leukemia stem cells
.
Protein Eng Des Sel
2012
;
25
:
561
9
.
19.
Xu
H
,
Cheng
M
,
Guo
H
,
Chen
Y
,
Huse
M
,
Cheung N-K
V
. 
Retargeting T Cells to GD2 pentasaccharide on human tumors using bispecific humanized antibody
.
Cancer Immunol Res
2014
;
3
:
266
77
.
20.
Durben
M
,
Schmiedel
D
,
Hofmann
M
,
Vogt
F
,
Nübling
T
,
Pyz
E
, et al
Characterization of a bispecific FLT3 X CD3 antibody in an improved, recombinant format for the treatment of leukemia
.
Mol Ther
2015
;
23
:
648
55
.
21.
Harwood
SL
,
Alvarez-Cienfuegos
A
,
Nuñez-Prado
N
,
Compte
M
,
Hernández-Pérez
S
,
Merino
N
, et al
ATTACK, a novel bispecific T cell-recruiting antibody with trivalent EGFR binding and monovalent CD3 binding for cancer immunotherapy
.
Oncoimmunology
2018
;
7
:
e1377874
.
22.
Beverley
PC
,
Callard
RE.
Distinctive functional characteristics of human “T” lymphocytes defined by E rosetting or a monoclonal anti‐T cell antibody
.
Eur J Immunol
1981
;
11
:
329
34
.
23.
Witte
MD
,
Cragnolini
JJ
,
Dougan
SK
,
Yoder
NC
,
Popp
MW
,
Ploegh
HL
. 
Preparation of unnatural N-to-N and C-to-C protein fusions
.
Proc Natl Acad Sci U S A
2012
;
109
:
11993
8
.
24.
Wagner
K
,
Kwakkenbos
MJ
,
Claassen
YB
,
Maijoor
K
,
Böhne
M
,
Van Der Sluijs
KF
, et al
Bispecific antibody generated with sortase and click chemistry has broad antiinfluenza virus activity
.
Proc Natl Acad Sci U S A
2014
;
111
:
16820
5
.
25.
Bartels
L
,
Ploegh
HL
,
Spits
H
,
Wagner
K
. 
Preparation of bispecific antibody-protein adducts by site-specific chemo-enzymatic conjugation
.
Methods
2019
;
154
:
93
101
.
26.
Horton
HM
,
Bernett
MJ
,
Pong
E
,
Peipp
M
,
Karki
S
,
Chu
SY
, et al
Potent in vitro and in vivo activity of an Fc-engineered anti-CD19 monoclonal antibody against lymphoma and leukemia
Cancer Res
2008
;
68
:
8049
57
.
27.
Antos
JM
,
Chew
GL
,
Guimaraes
CP
,
Yoder
NC
,
Grotenbreg
GM
,
Popp
MWL
, et al
Site-specific N- and C-terminal labeling of a single polypeptide using sortases of different specificity
.
J Am Chem Soc
2009
;
131
:
10800
1
.
28.
Arnett
KL
,
Harrison
SC
,
Wiley
DC
. 
Crystal structure of a human CD3-epsilon/delta dimer in complex with a UCHT1 single-chain antibody fragment
.
Proc Natl Acad Sci U S A
2004
;
101
:
16268
73
.
29.
Yssel
H
,
De Vries
JE
,
Koken
M
,
Van Blitterswijk
W
,
Spits
H
,
Vries
JE De
, et al
Serum-free medium for generation and propagation of functional human cytotoxic and helper T cell clones
.
J Immunol Methods
1984
;
72
:
219
27
.
30.
Gillissen
MA
,
Yasuda
E
,
de Jong
G
,
Levie
SE
,
Go
D
,
Spits
H
, et al
The modified FACS calcein AM retention assay: a high throughput flow cytometer based method to measure cytotoxicity
.
J Immunol Methods
2016
;
434
:
16
23
.
31.
Blackman
ML
,
Royzen
M
,
Fox
JM
. 
Tetrazine ligation: fast bioconjugation based on inverse-electron-demand Diels-Alder reactivity
.
J Am Chem Soc
2008
;
130
:
13518
9
.
32.
Slaney
CY
,
Toker
A
,
La Flamme
A
,
Bäckström
BT
,
Harper
JL
. 
Naïve blood monocytes suppress T-cell function. A possible mechanism for protection from autoimmunity
.
Immunol Cell Biol
2011
;
89
:
7
13
.
33.
Nagorsen
D
,
Kufer
P
,
Baeuerle
PA
,
Bargou
R
. 
Blinatumomab: a historical perspective
.
Pharmacol Ther
2012
;
136
:
334
42
.
34.
Bargou
R
,
Leo
E
,
Zugmaier
G
,
Klinger
M
,
Goebeler
M
,
Knop
S
, et al
Tumor regression in cancer patients by very low doses of a T cell-engaging antibody
.
Science
2008
;
321
:
974
7
.
35.
Chichili
GR
,
Huang
L
,
Li
H
,
Burke
S
,
He
L
,
Tang
Q
, et al
A CD3xCD123 bispecific DART for redirecting host T cells to myelogenous leukemia: preclinical activity and safety in nonhuman primates
.
Sci Transl Med
2015
;
7
:
289ra82
.
36.
Brooks
CL
,
Schietinger
A
,
Borisova
SN
,
Kufer
P
,
Okon
M
,
Hirama
T
, et al
Antibody recognition of a unique tumor-specific glycopeptide antigen
.
Proc Natl Acad Sci U S A
2010
;
107
:
10056
61
.
37.
Hoffmann
P
,
Hofmeister
R
,
Brischwein
K
,
Brandl
C
,
Crommer
S
,
Bargou
R
, et al
Serial killing of tumor cells by cytotoxic T cells redirected with a CD19-/CD3-bispecific single-chain antibody construct
.
Int J Cancer
2005
;
115
:
98
104
.
38.
Reusch
U
,
Duell
J
,
Ellwanger
K
,
Herbrecht
C
,
Knackmuss
SHJ
,
Fucek
I
, et al
A tetravalent bispecific TandAb (CD19/CD3), AFM11, efficiently recruits T cells for the potent lysis of CD19+ tumor cells
.
MAbs
2015
;
7
:
584
604
.
39.
Witte
MD
,
Theile
CS
,
Wu
T
,
Guimaraes
CP
,
Blom
AEM
,
Ploegh
HL
. 
Production of unnaturally linked chimeric proteins using a combination of sortase-catalyzed transpeptidation and click chemistry
.
Nat Protoc
2013
;
8
:
1808
19
.