Although adoptive transfer of NK cells with IL-2 can induce complete remissions in 30-50% of patients with refractory AML, efficacy is limited by IL-2 mediated induction of Tregs and by lack of antigen specificity. Thus we generated a 161533 trispecific killer engager (TriKE) molecule containing an anti-CD16 scFv to engage NK cells, an anti-CD33 scFv to engage myeloid targets (including myelodysplastic syndrome [MDS]), and a modified IL-15 linker. We have previously shown that this molecule is superior to a 1633 bispecific killer engager (BiKE) in killing of AML targets and that it promotes enhanced survival and in vivo expansion of NK cells. We questioned the mechanism for the increased potency of the 161533 TriKE and if the TriKE could activate the dysfunctional and suppressed NK cells found in patients with MDS. Cryopreserved mononuclear cells (obtained from the NMDP Sample Repository) collected from 8 patients with advanced MDS were tested to investigate how the TriKE might enhance functionality in this setting. Previously we reported that MDS patients have significantly decreased frequencies of NK cells due to increased CD33+ myeloid derived suppressor cells. Our 161533 TriKE enhanced the function of NK cells derived from MDS patients against acute promyelocytic CD33+ leukemia HL-60 tumor targets (Figure 1A), when compared to 1633 BiKE, in a flow cytometry assay measuring NK cell degranulation (% CD107a: 41.8±3.8 vs. 30.3±3.2, p=0.004), and inflammatory cytokine production (% IFNg: 40.7±5.0 vs. 30.0±4.9, p=0.009; % TNFa: 36.9±5.5 vs. 28.4±4.8, p=0.009). State of the art microchip-based live cell imaging was then employed to evaluate NK cell function and contact-to-target dynamics (Figure 1B). Briefly, resting NK cells and HL-60 target cells were stained with distinct dyes and then co-incubated in the presence of BiKE or TriKE in microwells at 37°C and 5% CO2. Cells were then imaged for 12 hours using a Zeiss 880 microscope and analyzed by Matlab. In contrast to those incubated with BiKE, NK cells cultured in the presence of TriKE had augmented cytotoxicity (37%±6% vs. 59%±6%, p=0.02) and killed their targets remarkably faster (time to first target kill = 148±30 min vs. 75±26 min, p< 0.0001). In addition, NK cell serial killers were more common in the presence of TriKE compared to BiKE (number of killed targets ≥3: 18%±7% vs. 9%±1%, p=0.04). Having shown the robust killing dynamics of TriKE primed NK cells, we next designed an in vivo dose escalation study to evaluate HL-60 tumor control in our xenogeneic mouse model. NSG (NOD scid gamma) mice were conditioned with a sub-lethal dose of radiation (275 cGy) and engrafted with 750,000 HL-60luc cells, which allow for tracking of tumor growth using bioluminescent imaging (BLI). Mice were infused with 1 million fresh healthy donor human NK cells and treated daily with recombinant IL-15 (5 ug/injection), 20-200 ug of 161533 TriKE, or left untreated. Higher doses of TriKE provided better tumor control (6.3x109±2.9x109 [200 ug 1615133] vs. 1.6 x1010±2.7x109 [20 ug 161533] p/sec/cm2/sr) demonstrating dose responsiveness at days 14 and 21 This was not due to increased proliferation of effectors (NK cell numbers) induced by the IL-15 segment of the TriKE as the absolute PBNK cell counts in mice were not different across the TriKE concentrations. However, the day 14 and 21 absolute PBNK counts were higher in all of the TriKE groups when compared to the IL-15 treated mice, including in the 20 ug 161533 TriKE group which best matched the 5 ug IL-15 dose. These data indicated that the TriKE molecule mediates superior NK cell expansion or maintenance in vivo. Taken together the in vivo data suggests that 161533 TriKE not only mediates tumor control by inducing NK cell proliferation and survival in a methodology that exceeds signals provided by IL-15 alone, but also increases tumor killing through enhanced killing kinetics. In summary, we have shown that the 161533 TriKE can rescue dysfunctional NK cells suppressed in patients with MDS and can mediate potent in vivo tumor killing presumably through better NK cell maintenance and enhanced killing kinetics This is of translational importance and demonstrates that the cancer induced immune suppression is reversible by the activating signals induced by the TriKE molecule. The 161533 TriKE represents a promising modality to maximizing NK cell based immunotherapies against MDS and AML and will be in phase I clinical testing the first half of 2017.

Disclosures

Cooley:Fate Therapeutics: Research Funding. Vallera:Oxis Biotech: Consultancy, Membership on an entity's Board of Directors or advisory committees. Miller:Fate Therapeutics: Consultancy, Research Funding; Oxis Biotech: Consultancy, Other: SAB.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution