scholarly journals Microrna 29b Mediates Immune Evasion of Natural Killer Cells in Acute Myeloid Leukemia

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 207-207
Author(s):  
Steven Scoville ◽  
Bethany Mundy-Bosse ◽  
Michael Zhang ◽  
Li Chen ◽  
Ryan Sanderson ◽  
...  

Abstract Natural killer (NK) cells are innate effector cells that can spontaneously recognize and kill cancer cells. While important in inducing long-term disease free survival (DFS) in the setting of killer immunoglobulin-like receptor (KIR) mismatch for acute myeloid leukemia (AML), harnessing NK cells to kill autologous or self AML blasts to extend DFS has had no success. In this study, we uncover one potential mechanism by which AML blasts can evade NK cell cytotoxicity. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that has now been shown to be expressed in immature human NK cells. Furthermore, activation of AHR in these immature cells suppresses human NK cell maturation and function. We and others have shown that human AML blasts secrete ligands that can activate AHR, leading us to hypothesize that AML may evade autologous NK cell cytotoxicity in part by inhibiting NK cell maturation. Expression of microRNA (miR)-29b has previously been shown to suppress the expression of transcription factors Tbx21 (TBET) and Eomesodermin (EOMES), both of which are critical for terminal NK cell differentiation and function. Here we show that AHR is able to directly regulate the expression of miR-29b and thus may serve as the link between AML and immune evasion of NK cells. We first identified putative AHR binding sites within the proximal promoter of miR-29b, suggesting that AHR may directly regulate miR-29b expression. To test this, we transfected the AHR responsive HepG2 human cell line with a miR-29b promoter driven luciferase reporter and measured luciferase activity after treatment with the known AHR agonist, FICZ, compared to vehicle control. We discovered that cells treated with 6-formylindolol[3,2-b]carbazole (FICZ) had increased luciferase activity compared to cells treated with vehicle control (P<0.05). This effect was subsequently shown to be directly mediated by AHR, as mutation of the putative AHR binding site as well as siRNA targeting of AHR mRNA both significantly diminished the induced luciferase activity (P<0.01). We then established the importance of miR-29b in human NK cell development by transducing immature NK cells, characterized as Lin(-)CD117(+)CD94(-), with a miR-29b knockdown virus and testing for the ability of these cells to become mature [i.e., Lin(-)CD117(-)CD94(+)] NK cells, after two weeks in IL-15 and FICZ. Indeed, in early experiments, knockdown of miR-29b resulted in increased percentages of mature NK cells compared to cells transduced with control virus (16.8% compared to 3.6%, respectively), despite being cultured with an AHR agonist. Finally, utilizing a translational in vivo murine AML model developed by our laboratory that recapitulates human AML, we have found that AML blasts harvested from these leukemic mice release an AHR agonist (P < 0.01), similar to previous reports that have described AHR ligands being produced by human AML blasts. In addition,NK cells isolated from these mice have increased levels of miR-29b when leukemic, compared to NK cells from wild type control littermates, consistent with our hypothesis that AHR regulates miR-29b expression. Thus, we propose that AHR, when activated by AML-derived ligands, upregulates miR-29b in NK cells to ultimately suppress the primary regulators of NK cell maturation and function, resulting in immune evasion (See Figure). Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2574-2574
Author(s):  
Mi roslaw J Szczepanski ◽  
Malgorzata Czystowska ◽  
Marta E Szajnik ◽  
Magis Mandapathil ◽  
Benedict Hilldorfer ◽  
...  

Abstract Interleukin-15 (IL-15) has been demonstrated to play a critical role in the regulation of natural killer (NK) cells. IL-15 induces the differentiation of NK cells from hematopoietic progenitors, stimulates the expansion of peripheral NK cells, and supports their survival. We investigated the role of IL-15 as a homeostatic regulator of NK cells in 29 patients diagnosed with acute myeloid leukemia (AML) and the potential role of IL-15 in enhancing the anti-tumor activity of NK cells in AML patients. The percentage of circulating NK cells was lower (p&lt;0.0001) in the AML patients (6%± 0.7, range 1–17%) compared to the NK cells of healthy donors (12%± 1, range 9–17 %). At diagnosis the mean level of IL-15 in patient plasma was 1.9 pg/ml (range 0.03–8.9) and increased (p &lt;0.02) to 5.2 pg/ml (range 0.06–13.4) after the completion of induction chemotherapy, when the NK levels had been reduced to zero cells/microliter. The mean level of IL-15 subsequently decreased to pre-treatment levels in the AML patients who achieved complete remission (mean 1.6 pg/ml, range 0.4–2.3). To assess effects of IL-15 on the NK cytotoxicity, we sorted NK cells from PBMC obtained from AML patients prior to treatment (at diagnosis) and cultured them in the presence of IL-15. Following IL-15 stimulation, a significant increase in NK-cell cytotoxicity against K562 targets and the patients’ autologous leukemic blasts was observed (p&lt;0.05) as was up-regulation in expression of the activating natural cytotoxicity receptors, NKp30 and NKp46 and the C-type lectin receptors NKG2D and NKG2C (p&lt;0.02–0.001). Addition of blocking antibodies to the activating receptors reduced NK-cell cytotoxicity. We determined that IL-15, a homeostatic NK-cell cytokine, increases after severe depletion of NK cells following intensive chemotherapy and this leads to increased NK-cell lytic activity in AML patients. These data suggest that modulation of IL-15 levels in AML could be therapeutically beneficial as IL-15 enhances NK-cell recovery following intensive chemotherapy and increases NK-cell anti-tumor activity.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 754-754 ◽  
Author(s):  
Bethany Mundy-Bosse ◽  
McConnell Kathleen ◽  
Charlene Mao ◽  
Elshafa Ahmed ◽  
Li Chen ◽  
...  

Abstract Acute myeloid leukemia (AML) is a devastating disease primarily affecting adults. Immune evasion is a major mechanism of AML persistence, and represents a barrier for long-term clinical success. Natural killer (NK) cells are a key component of the innate immune system and hold promise as a tool for effective anti-leukemia therapy. However, to date, the clinical success of NK therapy has been disappointing, indicating additional immune evasion strategies may affect the ability of NK cells to function in patients. We hypothesized that AML may evade the innate immune system by inhibiting NK cell maturation. To evaluate NK function and maturation during AML progression, our lab utilized a novel knock-in model of AML that expresses both Flt3-ITD and Mll-PTD mutations (PTD/ITD), and develops AML with 100% penetrance and recapitulates human disease. For these studies, both primary PTD/ITD mice and transplanted AML blasts were used. For transplant studies, AML or wild-type (WT) control cells (CD45.2+) were transplanted into irradiated C57BL/6 (CD45.1+) naïve recipients. Normal (CD45.1+) NK cells were defined as NK1.1+/CD3- by flow cytometry, and expression of receptors was determined within this population. NK cells in leukemic mice exhibited a reduction in the activating receptors Ly49D and Ly49H in the spleen, blood, LN, and bone marrow. There was also an increase in the inhibitory receptor Ly49C/I and NKG2C/A/E (both p<0.05). However, these alterations were not consistent with all activating and inhibitory receptors, as CD69 was elevated, while NKp46 expression was decreased (both p<0.05). NK function was determined by ELISA measurement of soluble interferon-gamma (IFN-g) production. While there was no change in IFN-g production at baseline, there was surprisingly a significant elevation in IFN-g detected upon stimulation with interleukin-12 and interleukin-18 in the NK cells isolated from leukemic mice as compared to WT control mice (p=0.0004). NK maturation was then evaluated by examining CD11b and CD27 protein expression on NK1.1+/CD3- cells in the spleen, bone marrow, blood, and LN. CD11b+/CD27+ (DP) NK cells were reduced in the spleen as well as bone marrow, blood and LN (p<0.02 for all organs). AML splenocytes had a corresponding increase in NK cells negative for both proteins (DN) when compared to WT control splenocytes (p=0.0004). Previous studies indicate the DP population of NK cells have the highest cytotoxic and cytokine producing functions, while DN NK cells are functionally immature. These alterations were more distinct as AML burden increased in the mice (p=0.0004). Interestingly, there was no significant difference in the most mature CD11b+/CD27- NK population in the spleen, but it there was a significant increase in the CD11b+/CD27- population in the blood (p=0.002). Additional analysis of two transcription factors known to regulate NK development, T-bet and Eomes, was also performed. A reduction in expression of both transcription factors was seen in total NK cells of leukemic mice, as well as NK subsets in the spleen (p<0.02). These data suggest that NK maturation is altered in the presence of AML, and that this alteration becomes more pronounced as leukemic burden increases. Studies evaluating the mechanistic link between AML and NK maturation are ongoing. Future studies will evaluate the efficacy of combinatorial therapies addressing both this imbalance in maturation as well as enhancing recognition to improve NK killing of AML cells. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 134 (2) ◽  
pp. 261-271 ◽  
Author(s):  
Zhiding Wang ◽  
Yang Xiao ◽  
Wei Guan ◽  
Mengzhen Wang ◽  
Jinghong Chen ◽  
...  

Abstract Acute myeloid leukemia (AML) is a malignant disorder of hemopoietic stem cells. AML can escape immunosurveillance of natural killer (NK) by gene mutation, fusions and epigenetic modification. The mechanism of AML immune evasion is not clearly understood. Here we show that CD48 high expression is a favorable prognosis factor that is down-regulated in AML patients, which can help AML evade from NK cell recognition and killing. Furthermore, we demonstrate that CD48 expression is regulated by methylation and that a hypomethylating agent can increase the CD48 expression, which increases the NK cells killing in vitro. Finally, we show that CD48 high expression can reverse the AML immune evasion and activate NK cells function in vivo. The present study suggests that a combination the hypomethylating agent and NK cell infusion could be a new strategy to cure AML.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2678-2678
Author(s):  
Miroslaw J Szczepanski ◽  
Marta E Szajnik ◽  
Malgorzata Czystowska ◽  
Magis Mandapathil ◽  
Ann Welsh ◽  
...  

Abstract Abstract 2678 Poster Board II-654 Natural killer (NK) cell cytotoxicity in patients with acute myeloid leukemia (AML) is significantly decreased relative to that in normal controls (NC). However, the mechanisms responsible for low NK cell activity in AML are not known. We considered the possibility that tumor-cell-derived microvesicles (MV) mediate suppression of NK cells. MV originate from the endosomal compartment of activated normal and neoplastic cells. Evidence suggests that tumor-derived MV exert detrimental effects on cells of the immune system and may play a role in tumor progression. To determine their contribution to immune suppression in AML, MV were isolated from sera of patients newly diagnosed with AML prior to any treatment and used to evaluate MV-mediated NK cell suppression. The protein content of MV isolated using exclusion chromatography and ultracentrifugation from sera of 19 AML patients was significantly higher than that of MV isolated from sera of 25 NC (75μg±12/mL vs 1.2μg±0.4/mL, p<0.001 ). MV from AML patients were positive for membrane-associated TGFb-1 and FasL in Western blots, whereas no TGFb-1 or FasL was detected in MV from NC. For functional assays, NK cells sorted from peripheral blood of NC were cultured with MV isolated from sera of the AML patients. A significant decrease in NK cell cytotoxicity was observed after co-incubation with MV (2412 LU before vs 1640 LU after, p<0.002). Concomitantly, a decrease in the expression of the NK cell activating receptor, NKG2D, was observed (57% before vs 38% after, p<0.001). The addition of TGFb1-neutralizing antibody abrogated the effects of MV on the NK cell cytotoxicity and receptor expression. The increased levels in sera of AML patients of MV mediating potent NK cell suppression is likely to compromise anti-tumor immune responses. Therefore, modulation of the levels and functions of MV might provide new immunotherapeutic approaches in AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2298-2298
Author(s):  
Miroslaw J. Szczepanski ◽  
Malgorzata Czystowska ◽  
Marta Szajnik ◽  
Ann Welsh ◽  
Kenneth A. Foon ◽  
...  

Abstract Natural killer (NK) cells lyse malignant cells without prior antigen-specific priming and play a critical role in the innate immune response. A balance of signals from activating and inhibiting receptors expressed on each NK cell controls its activity. The growth, differentiation and survival of NK cells have been found to be dependent on interleukin-15 (IL-15). Using multicolor flow cytometry we investigated the receptor repertoire and also measured the NK cell activity in twenty three patients with newly diagnosed acute myeloid leukemia (AML) prior to any treatment. Further, we investigated the ex-vivo effect of IL-15 on the NK cell repertoire and NK cell cytotoxicity. The percentage of circulating NK cells was lower (p&lt;0.0001) in the AML patients (6%± 0.7, range 1–17%) compared to the NK cells of healthy donors (12%± 1, range 9–17%). The expression of the activating natural cytotoxicity (NCR) receptors NKp30 and NKp46 and the C-type lectin receptors NKG2D and NKG2C was significantly decreased in the AML patients compared to the NK cells of healthy donors: NKp30 24 vs 51% p&lt;0.0001, NKp46 32 vs 73% p&lt;0.0001, NKG2D 43 vs 83% p&lt;0.0001, NKG2C 17 vs 28% p&lt;0.03. In addition, the receptor expression (mean fluorescence intensity, MFI) was also significantly lower in AML patients compared to healthy donors. No significant differences in the expression of the NCR NKp44 and the NK- cell inhibitory receptors were observed. Furthermore, the NK cytotoxicity in the AML patients at diagnosis was significantly lower (p&lt;0.0003) compared to the NK cytotoxicity of healthy donors (4 vs 75 LU). When NK cells obtained from AML patients were cultured with IL-15, significant increases in the expression of the NK- cell activating receptors (Table 1) were observed. The upregulation of the activating receptors was associated with a concomitant significant increase (p&lt;0.001) of the NK cell cytotoxicity (4 vs 70 LU). The data suggest that IL-15, a homeostatic NK cell cytokine, can upregulate the expression of activating receptors and concomitantly increase the NK lytic activity. The use of IL-15 as a platform for NK- based therapies for AML patients should be considered in the future. Table 1. The effect of IL-15 on the receptor expression in AML patients NK cell Activating Receptors NK cells in AML pts at diagnosis NK cells in AML pts after IL-15 stimulation p value % positive, MFI % positive, MFI NKp30 24, 2.6 84, 6.0 0.001 NKp44 4, 2.4 71, 6.7 0.001 NKp46 32, 2.6 83, 7 0.002 NKG2C 17, 2.2 58, 6.9 0.005 NKG2D 43, 2.5 87, 7.8 0.01


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2591-2591
Author(s):  
Kate Stringaris ◽  
Takuye Sekine ◽  
Ahmad Khoder ◽  
Abdullah Alsuliman ◽  
Pavlu Jiri ◽  
...  

Abstract Abstract 2591 Despite favorable initial responses to induction chemotherapy, most patients with acute myeloid leukemia (AML) will relapse. We hypothesized that just as cancers evade immunosurveillance by suppressing the immune response (“immunoediting”), remission and relapse in AML may be determined by similar immune interactions. After stem cell transplantation natural killer (NK) cells exert powerful allogeneic graft vs. leukemia effects. To explore immunosurveillance by autologous NK cells and immunoediting by AML blasts, we prospectively analyzed NK surface phenotype and function in AML patients at presentation and following remission induction. Using multi-color flow cytometry, we analyzed the surface expression of natural cytotoxicity receptors (NCRs), killer immunoglobulin receptors (KIRs) and C-type lectins directly ex-vivo in 32 consecutive patients at presentation and following complete remission (CR) in 12 patients for whom remission samples were available. Results were compared with 15 healthy controls. NK effector function in AML was measured against K562 leukemia targets and autologous AML blasts by CD107a degranulation and interferon-gamma (IFNγ) and TNF-alpha (TNFα) production. NK cells from 32 patients with AML at diagnosis had an abnormal phenotype compared to controls, with downregulation of the activatory receptor NKp46 (MFI 187± 15 vs. 266± 24, p=0.007) and upregulation of the inhibitory receptor NKG2A (mean 45%± 4.2 vs. 32%± 2.7, p=0.046). Moreover, AML-NK cells were defective in their effector function with significantly reduced CD107a degranulation (5% vs. 11%, p=0.0002), TNFα (1% vs. 3%, p=0.008) and IFNγ production (1% vs. 5%, p=<0.0001). In the 12 patients who achieved remission following induction chemotherapy NKp46 expression normalized from an MFI 122 at presentation to 242 (p=0.01); however, NKG2A expression continued to increase (45% vs. 61%, p=0.008). At remission AML-NK cells displayed CD107a degranulation levels comparable to that of healthy donor NK cells (9% vs. 11%, p=0.4). In contrast, TNFα and IFNγ production only partially normalized (2% vs. 3%, p=0.3) and (3% vs. 5%, p=0.04), respectively. AML patients with NKG2A overexpression (> median of 32.6%) at diagnosis were significantly less likely to achieve CR post chemotherapy compared to those with lower NKG2A expression (78% vs. 32% p= 0.041). Furthermore, patients who failed to respond to induction chemotherapy had significantly reduced NK effector function at diagnosis compared to normal controls (mean TNFα production 1% cf. 5% p=0.019). We then sought for evidence of immunoediting by AML on NK cell phenotype and function. Co-incubation of healthy donor NK cells with primary AML blasts for 24 hours resulted in significant reduction in TNFα production (p=0.02), IFNγ production (p=0.01) and a trend to reduced CD107a degranulation (p=0.07) against K562 leukemia targets. Our results indicate that AML blasts can produce long-lasting changes in NK cell subsets and impair their effector function and favouring immune escape from NK cell control. This work justifies larger prospective analyses to relate NK-based prognostic factors to classical predictive factors for remission and relapse, and should guide the development of NK cell based immunotherapy to improve outcome in AML. Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 320
Author(s):  
Matthew R. Lordo ◽  
Steven D. Scoville ◽  
Akul Goel ◽  
Jianhua Yu ◽  
Aharon G. Freud ◽  
...  

Over the past 50 years, few therapeutic advances have been made in treating acute myeloid leukemia (AML), an aggressive form of blood cancer, despite vast improvements in our ability to classify the disease. Emerging evidence suggests the immune system is important in controlling AML progression and in determining prognosis. Natural killer (NK) cells are important cytotoxic effector cells of the innate lymphoid cell (ILC) family that have been shown to have potent anti-leukemic functions. Recent studies are now revealing impairment or dysregulation of other ILCs in various types of cancers, including AML, which limits the effectiveness of NK cells in controlling cancer progression. NK cell development and function are inhibited in AML patients, which results in worse clinical outcomes; however, the specific roles of other ILC populations in AML are just now beginning to be unraveled. In this review, we summarize what is known about the role of ILC populations in AML.


2021 ◽  
Vol 12 ◽  
Author(s):  
Laura Kiekens ◽  
Wouter Van Loocke ◽  
Sylvie Taveirne ◽  
Sigrid Wahlen ◽  
Eva Persyn ◽  
...  

T-bet and Eomes are transcription factors that are known to be important in maturation and function of murine natural killer (NK) cells. Reduced T-BET and EOMES expression results in dysfunctional NK cells and failure to control tumor growth. In contrast to mice, the current knowledge on the role of T-BET and EOMES in human NK cells is rudimentary. Here, we ectopically expressed either T-BET or EOMES in human hematopoietic progenitor cells. Combined transcriptome, chromatin accessibility and protein expression analyses revealed that T-BET or EOMES epigenetically represses hematopoietic stem cell quiescence and non-NK lineage differentiation genes, while activating an NK cell-specific transcriptome and thereby drastically accelerating NK cell differentiation. In this model, the effects of T-BET and EOMES are largely overlapping, yet EOMES shows a superior role in early NK cell maturation and induces faster NK receptor and enhanced CD16 expression. T-BET particularly controls transcription of terminal maturation markers and epigenetically controls strong induction of KIR expression. Finally, NK cells generated upon T-BET or EOMES overexpression display improved functionality, including increased IFN-γ production and killing, and especially EOMES overexpression NK cells have enhanced antibody-dependent cellular cytotoxicity. Our findings reveal novel insights on the regulatory role of T-BET and EOMES in human NK cell maturation and function, which is essential to further understand human NK cell biology and to optimize adoptive NK cell therapies.


Haematologica ◽  
2020 ◽  
pp. 0-0
Author(s):  
Jessica Li ◽  
Sarah Whelan ◽  
Maya F. Kotturi ◽  
Deborah Meyran ◽  
Criselle D’Souza ◽  
...  

This study explored the novel immune checkpoint poliovirus receptor-related immunoglobulin domain-containing (PVRIG) in acute myeloid leukemia (AML). We showed that AML patient blasts consistently expressed the PVRIG ligand (poliovirus receptor-related 2, PVRL2). Furthermore, PVRIG blockade significantly enhanced NK cell killing of PVRL2+, poliovirus receptor (PVR)lo AML cell lines, and significantly increased NK cell activation and degranulation in the context of patient primary AML blasts. However, in AML patient bone marrow, NK cell PVRIG expression levels were not increased. To understand how PVRIG blockade might potentially be exploited therapeutically, we investigated the biology of PVRIG and revealed that NK cell activation resulted in reduced PVRIG expression on the cell surface. This occurred whether NK cells were activated by tumour cell recognition, cytokines (IL-2 and IL-12) or activating receptor stimulation (CD16 and NKp46). PVRIG was present at higher levels in the cytoplasm than on the cell surface, particularly on CD56bright NK cells, which further increased cytoplasmic PVRIG levels following IL-2 and IL-12 activation. PVRIG was continually transported to the cell surface via the endoplasmic reticulum (ER) and Golgi in both unstimulated and activated NK cells. Taken together, our findings suggest that anti- PVRIG blocking antibody functions by binding to surface-bound PVRIG, which undergoes rapid turnover in both unstimulated and activated NK cells. We conclude that the PVRIGPVRL2 immune checkpoint axis can feasibly be targeted with PVRIG blocking antibody for NK-mediated immunotherapy of PVRL2+ AML.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A893-A893
Author(s):  
Laurent Gauthier ◽  
Angela Virone-Oddos ◽  
Angela Virone-Oddos ◽  
Jochen Beninga ◽  
Benjamin Rossi ◽  
...  

BackgroundThere is a clear need for targeted therapies to treat acute myeloid leukemia (AML), the most common acute leukemia in adults. CD123 (IL-3 receptor alpha chain) is an attractive target for AML treatment.1 However, cytotoxic antibody targeting CD123 proved insufficiently effective in a combination setting in phase II/III clinical trials.2 T-cell engagers targeting CD123 displayed some clinical efficacy but were often associated with cytokine release syndrome and neurotoxicity.3 Interest in the use of NK cells for therapeutic interventions has increased in recent years, as a potential safer alternative to T cells. Several NK-cell activating receptors, such as CD16a, NKG2D, and the natural cytotoxicity receptors NKp30 and NKp46, can be targeted to induce antitumor immunity. We previously reported the development of trifunctional NK-cell engagers (NKCEs) targeting a tumor antigen on cancer cells and co-engaging NKp46 and CD16a on NK cells.4MethodsWe report here the design, characterization and preclinical development of a novel trifunctional NK cell engager (NKCE) targeting CD123 on AML cells and engaging the activating receptors NKp46 and CD16a on NK cells. The CD123 NKCE therapeutic molecule was engineered with humanized antibodies targeting NKp464 and CD123.5 We compared CD123-NKCE and a cytotoxic ADCC-enhanced antibody (Ab) targeting CD123, in terms of antitumor activity in vitro, ex vivo and in vivo. Pharmacokinetic, pharmacodynamic and safety profile of CD123-NKCE were evaluated in non-human primate (NHP) studies.ResultsThe expression of the high affinity Fc gamma receptor CD64 on patient-derived AML cells inhibited the ADCC of the Ab targeting CD123 in vitro and ex vivo, but not the antitumor activity of CD123-NKCE. CD123-NKCE had potent antitumor activity against primary AML blasts and AML cell lines, promoted strong NK-cell activation and induced cytokine secretion only in the presence of AML target cells. Its antitumor activity in mouse model was greater than that of the comparator antibody. Moreover, CD123-NKCE had strong and prolonged pharmacodynamic effects in NHP when used at very low doses, was well-tolerated up to high 3 mg/kg dose and triggered only minor cytokine release.ConclusionsThe data for activity, safety, pharmacokinetics, and pharmacodynamics provided here demonstrate the superiority of CD123-NKCE over comparator cytotoxic antibody, in terms of antitumor activity in vitro, ex vivo, in vivo, and its favorable safety profile, as compared to T-cell therapies. These results constitute proof-of-principle for the efficacy of CD123-NKCE for controlling AML tumors in vivo, and provide consistent support for their clinical development.ReferencesEhninger A, Kramer M, Rollig C, et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J 2014;4:e218.Montesinos P, Gail J Roboz GJ, et al. Safety and efficacy of talacotuzumab plus decitabine or decitabine alone in patients with acute myeloid leukemia not eligible for chemotherapy: results from a multicenter, randomized, phase 2/3 study. Leukemia 2021;35(1):62–74.Uy GL, Aldoss I, Foster MC, et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021;137(6):751–762.Gauthier L, Morel A, Anceriz N, et al. Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell 2019;177(7):1701–13.Jin L, Lee EM, Ramshaw HS, et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell 2009;5:31–42.


Sign in / Sign up

Export Citation Format

Share Document