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
. 2017 Feb 2;129(5):609-618.
doi: 10.1182/blood-2016-08-735365. Epub 2016 Dec 1.

An anti-CD3/anti-CLL-1 bispecific antibody for the treatment of acute myeloid leukemia

Affiliations

An anti-CD3/anti-CLL-1 bispecific antibody for the treatment of acute myeloid leukemia

Steven R Leong et al. Blood. .

Abstract

Acute myeloid leukemia (AML) is a major unmet medical need. Most patients have poor long-term survival, and treatment has not significantly changed in 40 years. Recently, bispecific antibodies that redirect the cytotoxic activity of effector T cells by binding to CD3, the signaling component of the T-cell receptor, and a tumor target have shown clinical activity. Notably, blinatumomab is approved to treat relapsed/refractory acute lymphoid leukemia. Here we describe the design, discovery, pharmacologic activity, pharmacokinetics, and safety of a CD3 T cell-dependent bispecific (TDB) full-length human IgG1 therapeutic antibody targeting CLL-1 that could potentially be used in humans to treat AML. CLL-1 is prevalent in AML and, unlike other targets such as CD33 and CD123, is not expressed on hematopoietic stem cells providing potential hematopoietic recovery. We selected a high-affinity monkey cross-reactive anti-CLL-1 arm and tested several anti-CD3 arms that varied in affinity, and determined that the high-affinity CD3 arms were up to 100-fold more potent in vitro. However, in mouse models, the efficacy differences were less pronounced, probably because of prolonged exposure to TDB found with lower-affinity CD3 TDBs. In monkeys, assessment of safety and target cell depletion by the high- and low-affinity TDBs revealed that only the low-affinity CD3/CLL1 TDB was well tolerated and able to deplete target cells. Our data suggest that an appropriately engineered CLL-1 TDB could be effective in the treatment of AML.

PubMed Disclaimer

Figures

Figure 1.
Figure 1.
AML tumor cell growth suppression is dependent on affinity to CD3ε and effects of CLL1/CD3H TDB on AML BMMNCs. (A) Dose-dependent survival curves determined by Prism-6 using a nonlinear regression with sigmoidal dose response where X is the log [ ]. Percent survival is calculated by dividing the treatment live events by its untreated control replicate live events, and multiplying by 100. HL60 was performed in a separate experiment from the other cell lines (data not shown for CLL1/CD3H). (B) EOL-1 dose-dependent survival and percent of CD8+CD69+CD25+ effector cells. One of 3 experiments, each using a different blood donor. Similar results were observed for other AML cell lines. (C-D) CD8+ T cells were purified from peripheral blood of a healthy human donor (ALLCells, Alameda, CA) for donor 1 and a Genentech donor for donor 2 by Ficoll density gradient centrifugation and a human CD8+ T cell isolation kit from Miltenyi (130-096-495). Patient AML bone marrow was obtained from the Stanford Cancer Center and purified by Ficoll centrifugation. The AML blasts were preincubated for ∼6 hours with hIgG (EU = 0.07/mg) to reduce nonspecific binding to FcγRs. The E:T ratio was 3:1 (150 000 CD8 T cells to 50 000 blasts). Medium used for the assay was RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum, 2 mM glutamine, Pen-Strep, 1X cytokine bullet (IL-3, IL-6, SCF, Flt3; StemCell Technologies, Vancouver, Canada), 0.1 μg/mL GM-CSF and 0.1 μg/mL G-CSF. The test articles were NT/CD3H and CLL1/CD3H diluted serially in threefold steps. The assay was set up in triplicate with either a 20-hour (donor 2; blue lines) or 40-hour (donor 1; red lines) exposure to test article. (C) Dot plot shows SSC/CD45 profile of AML patient donor 1 and donor 2. Histogram shows positive staining of CD11b/CD34+ AML blasts for human CD33 (red) and CLL-1 (blue) compared with isotype control antibody. (D) Allogeneic CD8+ effector T cells in the presence of threefold serially diluted CLL1/CD3H shows concentration-dependent killing of AML blasts with an EC50 ∼0.45 ng/mL for donor 1 compared with the NT/CD3H TDB. Dotted line (NT/CD3H) and solid line (CLL1/CD3H). Donor 1 phenotype was consistent with AML LSCs—CD45+/CD34+/CD33+/CLL-1+/CD38 (99%) with <1% CD38+. Donor 2 phenotype was consistent with AML progenitors—CD45+/CD34+/CD33+/CLL-1+/CD38+ (∼97%).
Figure 2.
Figure 2.
Depletion of human CLL-1+ 2xBAC-Tg mouse cells from blood and bone marrow after single IV dose administration of 0.5 mg/kg CLL-1 TDB. (A) CLL1/CD3VH, (B) CLL1/CD3H, and (C) CLL1/CD3L. Percent of hCLL-1+ cells were derived from the average naïve untreated mice hCLL-1+ cells; blood (n = 39), bone marrow and spleen (n = 4). Upregulation of CD69 on CD8+ and CD4+ T cells by (D) CLL1/CD3VH, (E) CLL1/CD3H, and (F) CLL1/CD3L. Time points are terminal bleeds. The mean and standard deviation are represented by horizontal and vertical lines, respectively.
Figure 3.
Figure 3.
PK profiles for CLL-1/CD3 TDBs in target-deficient and target-expressing mice. Animals were administered a single IV dose of 0.5 mg/kg of either nontargeting (NT) TDBs or CLL-1–specific TDBs over a period of 14 to 21 days. (A) Target-deficient SCID.Beige mice PK values, (B) target-expressing 2xBACT-Tg (human CLL-1 and human CD3ε) PK values.
Figure 4.
Figure 4.
Cytokine levels in cynomolgus monkeys receiving CLL1/CD3H (HA) and CLL1/CD3L (LA). (A) G-CSF, (B) tumor necrosis factor-α (TNFα), (C) IL-2, and (D) IL-6. Cytokine release typically occurred within 2 to 6 hours of administration of either molecule and was markedly increased in those animals that received 0.5 mg/kg CLL1/CD3H compared with those that received CLL1/CD3L. All animals that required unscheduled euthanasia had comparatively elevated cytokine levels compared with those that survived to scheduled necropsy. The one animal that received 0.5 mg/kg CLL1/CD3L and required unscheduled necropsy had perimortem increases in G-CSF, IL-6, and other innate cytokines such as MCP-1 (data not shown).
Figure 5.
Figure 5.
Clinical pathology and peripheral blood flow cytometry from cynomolgus monkeys receiving CLL1/CD3L TDB. Target cell depletion of neutrophils (A) and monocytes (B) is similar between both dose groups, with nadirs between day 4 and 8 and 2 and 8 post dose, respectively. Rebound neutrophilia between days 10 and 15 is marked and greater in the 0.2 mg/kg dose group. Note, the mild decrease in neutrophil count on day 2 in the vehicle group is likely secondary to repeat blood sampling. Reduction of lymphocytes (C) occurred in 2 waves (days 2-8) concurrent to target cell depletion, elevation of acute phase proteins such as C-reactive protein (D), and increased activation of CD8+ cytotoxic T cells as measured by CD69 (E) and CD25 (F) expression. The immediate increase in percentage of CD69+ CD8+ T cells was greater (peak 71.3%-80.7% vs 25.2%-57.0%), and the percentage of CD25+ T cells was elevated for longer (return to baseline D15-D22 vs D8-D15) in the 0.5 mg/kg group.
Figure 6.
Figure 6.
Myeloid depletion in the bone marrow of cynomolgus monkeys receiving 0.2 mg/kg CLL1-CD3L. Myeloid cell number, assessed by either CD11b reactivity (A) or CLL-1 reactivity (B), were markedly attenuated on day 8 (nadir of peripheral neutrophils) compared with day 22 (peripheral neutrophil recovery). Horizontal and vertical lines represent mean and standard deviation, respectively. (C) Bone marrow histology at day 8 revealed marked depletion of late-stage myeloid cells with a relative increase in early myeloid progenitors (arrows), suggesting that hematopoietic stem cells were not affected at this dose. (D) By day 22, late-stage myeloid cells were recovered and minimally increased in relative abundance (asterisks). Images of FFPE 5-µm sections stained with hematoxylin and eosin were acquired on an Olympus BX53 (Olympus, Burlingame, CA) camera, objective UPlanSApo 40x/0.95 at 20-23°C, and photographed with an Infinity 2 camera and analyzed with Infinity Analyze software (Lumenera, Ottawa, Ontario, Canada).

References

    1. Freireich EJ, Wiernik PH, Steensma DP. The leukemias: a half-century of discovery. J Clin Oncol. 2014;32(31):3463-3469. - PubMed
    1. Wander SA, Levis MJ, Fathi AT. The evolving role of FLT3 inhibitors in acute myeloid leukemia: quizartinib and beyond. Ther Adv Hematol. 2014;5(3):65-77. - PMC - PubMed
    1. Topp MS, Gökbuget N, Zugmaier G, et al. . Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood. 2012;120(26):5185-5187. - PubMed
    1. Topp MS, Kufer P, Gökbuget N, et al. . Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol. 2011;29(18):2493-2498. - PubMed
    1. Kung Sutherland MS, Walter RB, Jeffrey SC, et al. . SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. Blood. 2013;122(8):1455-1463. - PubMed

MeSH terms