scholarly journals 852 Trifunctional NKp46/CD16a-NK cell engager targeting CD123 overcomes acute myeloid leukemia resistance to ADCC

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.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3307-3307
Author(s):  
Jeffrey J. Bednarski ◽  
Clare Zimmerman ◽  
Amanda F Cashen ◽  
Sweta Desai ◽  
Mark Foster ◽  
...  

Acute myeloid leukemia (AML) accounts for 18% of pediatric leukemias. For high-risk AML, standard treatment includes multi-agent chemotherapy followed allogeneic hematopoietic cell transplantation (HCT). Despite a high remission rate, 50% of patients relapse and have a poor prognosis with < 20% of patients surviving more than 3 years. Salvage chemotherapy alone or combined with donor lymphocyte infusions (DLI) have little curative potential, and new treatment strategies are needed for relapsed-refractory AML. Previous studies have shown that natural killer (NK) cells can be stimulated ex vivo with IL-12/15/18 to generate a memory-like phenotype with enhanced anti-leukemia effect. In adults with relapsed-refractory AML, adoptive transfer of MHC-haploidentical cytokine-induced memory-like (CIML or ML) NK cells induced remission in 54% of patients (PMID27655849). The infused donor ML NK cells expand in vivo but are rapidly eliminated following recovery of recipient T cells, providing a window of therapeutic activity of 2-3 weeks. We sought to test the safety and efficacy of ML NK cells for treatment of pediatric/young adult patients with post-HCT relapsed AML. We hypothesized that ML NK cells derived from the HCT donor would be well-tolerated, exhibit anti-leukemia activity, and expand with prolonged persistence following transfer into pediatric AML patients. Here, we report the results of the first pediatric patient treated on a phase I clinical trial using ML NK cell therapy for relapsed AML after allogeneic HCT (NCT03068819). Briefly, patients are treated with FLAG (fludarabine, cytarabine and granulocyte colony stimulating factor) salvage chemotherapy to reduce the bulk of AML and provide lymphodepletion for ML NK cell expansion. Two weeks after chemotherapy, a non-mobilized leukapheresis product is collected from the original HCT donor and processed into a T cell-based DLI and ML NK cells. The T cell DLI (1 x 106 T cells/kg) is immediately infused, and the ML NK cells are generated by stimulation with IL-12/15/18 ex vivo for 12-16 hours and then infused (10x106/kg). An 18-month-old male with infant AML had relapse of his leukemia 3 months after MHC-haploidentical HCT. Treatment with chemotherapy, including mitoxantrone and daunorubicin-cytarabine liposome, failed to induce remission. At the time of enrollment on the phase I trial, he had AML blasts in his bone marrow (Table 1). He was treated with FLAG chemotherapy followed by infusion of DLI and ML NK cells from the original haploidentical HCT donor. Assessment at 30 days, 3 months and 6 months post NK cell infusion showed complete remission with no evidence of leukemia and full donor engraftment. Remarkably, donor-derived ML NK cells expanded to 77% of donor lymphocytes on day 28 and still comprised 24% percent of lymphocytes at 6 months post infusion (Figure 1A-C). The expanded donor NK cell phenotype was consistent with ML NK cells (e.g., NKG2A+KIR+) utilizing CyTOF multidimensional analysis previously confirmed to identify ML NK cells (Figure 1D). The ML NK cells were functional as demonstrated by leukemia-triggered IFN-γ production immediately ex vivo from day 7-28 samples (Figure 1E-F). The patient's clinical course was complicated by mild gastrointestinal graft-versus-host disease that resolved with low-dose steroids and tociluzimab. These early results demonstrate proof-of-principle that adoptive transfer of donor-derived ML NK cells in combination with DLI is feasible and offers a novel immunotherapy option for patients with relapsed AML after HCT. Moreover, in this T and NK cell compatible immune environment post-HCT, donor ML NK cells expand and persist robustly in vivo for > 6 months without exogenous cytokine support and have potent anti-leukemic activity. Thus, ML NK cells are a cellular therapy platform to treat AML that has relapsed after allogeneic HCT. Disclosures Cashen: Celgene: Other: Speaker's Bureau; Seattle Genetics: Other: Speaker's Bureau; Novartis: Other: Speaker's Bureau. Fehniger:Horizon Pharma PLC: Other: Consultancy (Spouse); Cyto-Sen Therapeutics: Consultancy.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3090-3090 ◽  
Author(s):  
Folashade Otegbeye ◽  
Nathan Mackowski ◽  
Evelyn Ojo ◽  
Marcos De Lima ◽  
David N. Wald

Abstract Introduction: A crucial component of the innate immune response system, natural killer (NK) cells are uniquely competent to mediate anti-myeloid leukemia responses. NKG2D is an activating receptor on the surface of NK cells that engages stress ligands MICA and MICB, typically upregulated on myeloid leukemia cells. Adoptive transfer of NK cells is a promising treatment strategy for AML. Strategies to optimize the anti-leukemia effect of NK cell adoptive transfer are an area of active research. These include attempts to enhance NK cell activity and to maintain the activation status and proliferation of the NK cells in vivo. Traditionally, IL-2 has been used to maintain the in vivo proliferation of adoptively transferred NK cells, but it leads to unwanted proliferation of regulatory T cells and suboptimal NK cell proliferation. IL-15 may be superior to IL-2, without the effects on T regulatory cells. The IL-15 superagonist, ALT-803 exhibits >25 fold enhancement in biological activity as compared to IL-15. ALT-803 is a fusion protein of an IL-15 mutant and the IL-15Rα/Fc complex that has recently entered clinical trials as a direct immunomodulatory agent in cancer clinical trials We hypothesized ALT-803 would augment the activity and/or proliferation of adoptively transferred NK cells in vitro and in a mouse model system.. Methods: Human NK cells were isolated from healthy donor peripheral blood and were expanded over a 21-day period in co-culture with irradiated K562 cells genetically modified to express membrane-bound IL-21. (Somanchi et al. 2011 JoVE 48. doi: 10.3791/2540) The NK cells were expanded with IL-2 (50mU/mL) and/or ALT-803 (200ng/mL). On Day 21, NK cells were examined for cytotoxicity against AML cells as well as by flow cytometry for expression of known activating receptors. An NSG murine xenograft model of human AML was developed to test the in vivo function of NK cells expanded above. Briefly, NSG mice (n=5 per group) were non-lethally irradiated and each injected IV with 5 x106 OCI-AML3 leukemic cells. Two days later, each mouse received weekly NK cell infusions for 2 weeks. Mice that received NK cells expanded with IL2 got cytokine support with IL-2 (75kU IP three times a week). Mice infused with ALT-803 expanded cells (alone or in combination with IL2) received ALT-803 (0.2mg/kg IV weekly). One control group received OCI cells but were infused weekly only with 2% FBS vehicle, no NK cells. Leukemic burden in each mouse was assessed by flow cytometry of bone marrow aspirates on day 28 following start of NK cell infusions). This time point was chosen as the control mice appeared moribund. Results: ALT-803 did not have any differential effect on the proliferation of the NK cells ex vivo as compared to IL-2. However, the presence of ALT-803 either alone or in combination with IL-2 resulted in a significant increase (30% increase, p<0.0001) in the cytotoxic activity of the NK cells against leukemia cells as compared with IL-2 alone in vitro (figure 1). In addition, the percentages of NK cells that express the activating receptor NKG2D as well as CD16 were significantly higher (p<0.001 for both) after ALT-803 exposure (figure 1). Finally, in the murine xenograft AML model, ALT-803 expanded NK cells, which were also supported in vivo with ALT-803, resulted in an 8-fold reduction in disease burden in the bone marrow (p<0.0001). Importantly the efficacy of NK cells in the ALT-803 injected mice was significantly higher (3-fold, p= 0.0447) than IL-2 treated mice (figure 2). Discussion: Our results suggest that the presence of ALT-803 during ex-vivo expansion of NK cells results in increased activation and cytotoxicity against AML cells. In addition our results using a murine model of human AML show that the use of ALT-803 in combination with adoptively transferred NK cells provides a significant anti-leukemic benefit as compared to IL-2. Future studies to test larger panels of leukemia cells as well as other cancer cell lines are currently in progress. It is hoped that this work will lead to an improvement in the efficacy of adoptively transferred NK cells for AML patients due to an improvement in survival and activity of the NK cells. Disclosures Wald: Invenio Therapeutics: Equity Ownership.


Blood ◽  
2011 ◽  
Vol 118 (12) ◽  
pp. 3273-3279 ◽  
Author(s):  
Antonio Curti ◽  
Loredana Ruggeri ◽  
Alessandra D'Addio ◽  
Andrea Bontadini ◽  
Elisa Dan ◽  
...  

Abstract Thirteen patients with acute myeloid leukemia, 5 with active disease, 2 in molecular relapse, and 6 in morphologic complete remission (CR; median age, 62 years; range, 53-73 years) received highly purified CD56+CD3− natural killer (NK) cells from haploidentical killer immunoglobulin-like receptor–ligand mismatched donors after fludarabine/cyclophosphamide immunosuppressive chemotherapy, followed by IL-2. The median number of infused NK cells was 2.74 × 106/Kg. T cells were < 105/Kg. No NK cell–related toxicity, including GVHD, was observed. One of the 5 patients with active disease achieved transient CR, whereas 4 of 5 patients had no clinical benefit. Both patients in molecular relapse achieved CR that lasted for 9 and 4 months, respectively. Three of 6 patients in CR are disease free after 34, 32, and 18 months. After infusion, donor NK cells were found in the peripheral blood of all evaluable patients (peak value on day 10). They were also detected in BM in some cases. Donor-versus-recipient alloreactive NK cells were shown in vivo by the detection of donor-derived NK clones that killed recipient's targets. Adoptively transferred NK cells were alloreactive against recipient's cells, including leukemia. In conclusion, infusion of purified NK cells is feasible in elderly patients with high-risk acute myeloid leukemia. This trial was registered at www.clinicaltrial.gov as NCT00799799.


2021 ◽  
Vol 11 ◽  
Author(s):  
Yang Xiao ◽  
Jinghong Chen ◽  
Jia Wang ◽  
Wei Guan ◽  
Mengzhen Wang ◽  
...  

Acute myeloid leukemia (AML), a malignant disorder of hemopoietic stem cells. AML can escape immunosurveillance of natural killer (NK) by gene mutation, fusions, and epigenetic modification, while the mechanism is not clearly understood. Here we show that the expression of Intercellular adhesion molecule‐1 (ICAM‐1, CD54) is silenced in AML cells. Decitabine could upregulate ICAM-1 expression, which contributes to the NK-AML cell conjugates and helps NK cells kill AML cells. We also show that ICAM-1 high expression can reverse the AML immune evasion and activate NK cells function in vivo. This study suggests that a combination of the hypomethylating agent and NK cell infusion could be a new strategy to cure AML.


2021 ◽  
Vol 20 (1) ◽  
Author(s):  
Yu-Jun Dai ◽  
Si-Yuan He ◽  
Fang Hu ◽  
Xue-Ping Li ◽  
Jian-Ming Zhang ◽  
...  

AbstractAcute myeloid leukemia (AML) is still incurable due to its heterogeneity and complexity of tumor microenvironment. It is imperative therefore to understand the molecular pathogenesis of AML and identify leukemia-associated biomarkers to formulate effective treatment strategies. Here, we systematically analyzed the clinical characters and natural killer (NK) cells portion in seventy newly-diagnosis (ND) AML patients. We found that the proportion of NK cells in the bone marrow of ND-AML patients could predict the prognosis of patients by analyzing the types and expression abundance of NK related ligands in tumor cells. Furthermore, MCL1 inhibitor but not BCL2 inhibitor combined with NK cell-based immunotherapy could effectively improve the therapeutic efficiency via inhibiting proliferation and inducing apoptosis of AML primary cells as well as cell lines in vitro. There results provide valuable insights that could help for exploring new therapeutic strategies for leukemia treatment.


2021 ◽  
Vol 12 ◽  
Author(s):  
Berna Bou-Tayeh ◽  
Vladimir Laletin ◽  
Nassim Salem ◽  
Sylvaine Just-Landi ◽  
Joanna Fares ◽  
...  

Natural Killer (NK) cells are potent anti-leukemic immune effectors. However, they display multiple defects in acute myeloid leukemia (AML) patients leading to reduced anti-tumor potential. Our limited understanding of the mechanisms underlying these defects hampers the development of strategies to restore NK cell potential. Here, we have used a mouse model of AML to gain insight into these mechanisms. We found that leukemia progression resulted in NK cell maturation defects and functional alterations. Next, we assessed NK cell cytokine signaling governing their behavior. We showed that NK cells from leukemic mice exhibit constitutive IL-15/mTOR signaling and type I IFN signaling. However, these cells failed to respond to IL-15 stimulation in vitro as illustrated by reduced activation of the mTOR pathway. Moreover, our data suggest that mTOR-mediated metabolic responses were reduced in NK cells from AML-bearing mice. Noteworthy, the reduction of mTOR-mediated activation of NK cells during AML development partially rescued NK cell metabolic and functional defects. Altogether, our data strongly suggest that NK cells from leukemic mice are metabolically and functionally exhausted as a result of a chronic cytokine activation, at least partially IL-15/mTOR signaling. NK cells from AML patients also displayed reduced IL-2/15Rβ expression and showed cues of reduced metabolic response to IL-15 stimulation in vitro, suggesting that a similar mechanism might occur in AML patients. Our study pinpoints the dysregulation of cytokine stimulation pathways as a new mechanism leading to NK cell defects in AML.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1041-1041
Author(s):  
Julie Gertner-Dardenne ◽  
Eloise Perrot ◽  
Thomas Prebet ◽  
Aude Charbonnier ◽  
Helene Sicard ◽  
...  

Abstract Abstract 1041 Poster Board I-63 BACKGROUNd: Compelling evidences have demonstrated the role of the immune system in the control of acute myeloid leukemia (AML). So far, T cells and natural killer (NK) cells are the major immune effectors shown to be involved in AML control. The graft-versus-leukemia (GVL) effect following allogenic stem cell transplantation as well as donor lymphocyte infusions indicate that T lymphocytes can control and eliminate AML cells. Leukemia-specific antigenic peptides have been characterized (proteinase-3 and Wilms tumor 1 protein) and serve as targets for peptide-based vaccine trials in AML. Allogenic NK cells have anti-leukemic activity as shown by killer cell inhibitory receptor (KIR)-mismatched haplo-identical stem cell transplantation. Less is known regarding the role of gd T cells in the control of AML. Recently the reconstitution of Vd1 T lymphocytes post transplantation has been shown to correlate with a better prognosis. In the present study, we have analyzed gd T cells in patients with AML and in a mouse model of human AML and focused on (Vg9) Vd2 T cells, the main subset of circulating gd T cells with anti-neoplastic activity. Human Vg9Vd2 T lymphocytes can be activated by nonpeptidic antigens such as the mevalonate pathway-derived isopentenyl pyrophosphate or synthetic phosphoantigen such as bromohydrin pyrophosphate (BrHPP). This population may be suitable for the adoptive immunotherapy of acute myeloid leukemia (AML). However little is known about the frequency, the function and the mechanisms underlying Vg9Vd2 T-cell recognition of AML. We have focused this study on AMoL which are targets of NK and ab T cells. OBJECTIVE OF THE STUDY to describe Vg9Vd2 T cells in patients with AML and investigate their ability to induce an effective cytotoxic response against autologous AML blast in vitro and in vivo. EXPERIMENTAL PROCEDURe: We compared the phenotype and the absolute circulating Vg9Vd2 T cell levels in the blood and the bone marrow (BM) in 12 patients with AMoL (FAB AML-M4 and -M5) and 12 healthy volunteers (HV) using multi parametric flow cytometry. All patients and volunteers gave written informed consent. Vg9Vd2 T cells of AML patient were expanded ex vivo using BrHPP or Zoledronic acid plus IL2. The functions of expanded Vg9Vd2 T cells were assessed in vitro by their cytotoxicity against leukemic blasts (CD107a staining, 51Cr assay) and in vivo in immunodeficient mice transplanted with human AML cell line (U937). In these experiments, the ability of adoptively transferred Vg9Vd2 T cells to migrate into BM and improve mice survival was assessed after i.v. infusion of U937 cells into healthy female NOD-SCID, common _-chain knockout mice (NOG mice). Mice were then treated twice i.v. with 40.106 Vg9Vd2 T cells. RESULTs: Vg9Vd2 T lymphocytes are present in the blood as well as BM of AMoL patients at a lower frequency as compared to HV (median 2.07/μl vs 34/μL respectively P<0.001). Vg9Vd2 T lymphocytes from AML patients are endowed with in vitro proliferation in response to BrHPP or Zoledronic acid plus IL2 but lower than HV (fold increase median 33 versus 69, P=0.051). Expanded Vg9Vd2 express activation markers (CD69 and CCR5) and exhibit an effector/memory phenotype (CD45RA- CD27-). Their lytic potential toward autologous AML blast was equivalent to those of HV by 51Cr experiments and CD107a staining and involves the perforin-granzyme pathway. Their activity depends on both TCRVd2 and DNAX accessory molecule-1 (DNAM-1) as demonstrated by antibody blockade. In vivo data show that, upon sacrifice, Vg9Vd2 were detected in BM, spleen and blood of mice. Preliminary Kaplan-Meier analysis of pooled cohorts of Vg9Vd2-treated and untreated mice reveals that mice receiving Vg9Vd2 T cells displayed superior survival compared with untreated controls (P=0.0047). CONCLUSIOn: Altogether, our data indicate that Vg9Vd2 T cells are decreased in AML patients and have a more limited expansion potential. However, they are able to kill autologous AML blast upon stimulation in a TCRVd2 as well as the DNAM-1 receptor dependent manner. These results provide a rationale for the clinical evaluation of adoptive transfer of ex vivo expanded allogenic Vg9Vd2T cells or direct activation of Vg9Vd2T cells with IL2 + phosphoantigens in patients with AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1120-1120
Author(s):  
Andreas T Björklund ◽  
Mattias Carlsten ◽  
Marie Schaffer ◽  
Lisa Liu ◽  
Sarah A. Cooley ◽  
...  

Abstract Introduction: We here report from an ongoing phase I/II study of HLA-haploidentical NK cell therapy to patients with high-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) not eligible for standard therapies. The preparative regimen consisted of intermediate doses of Cyklophosphamide (Cy), Fludarabin (Flu) and titrated doses of total lymphoid irradiation (TLI). The trial design excluded systemic IL-2 treatment to avoid expansion of regulatory T cells and to test if in vivo expansion could be obtained without IL-2 support. Patients:The first 12 patients were treated with Cy/Flu and an escalating dose of TLI (2 Gy and 4 Gy), followed by infusion of short-term IL-2 activated (16 hours) NK cells. Three patients received daily cyclosporine A after the conditioning. Three had relapsed, chemotherapy-refractory, primary AML, seven had secondary relapsed or refractory MDS-AML and two had high risk MDS with fibrosis. Results: The treatment was well tolerated and no severe non-infectious toxicity could be observed in the patients. The endpoint of expansion (>100 donor NK cells/ul at day 14) was not reached, but six patients had positive microchimerism, NK cells of donor origin detectable by RT-PCR at day 7-14, that thereafter became undetectable within 7-14 days. Four of these six patients achieved complete remission (CR) whereafter they become eligible for and could proceed to allogeneic stem cell transplantation. None of the patients with negative microchimerism obtained CR. Four patients died from progressive disease and three patients, with minor response and progressive disease, died in infections within three months of therapy. Discussion: Although the long-term efficacy needs to be evaluated, the results suggest that a combined regimen with mild conditioning followed by NK cell therapy may induce remission in patients with chemo-refractory disease and provide a bridge to allogeneic stem cell transplantation. Notably, clinical responses were observed after only a minimal in vivo NK cell expansion and were independent on KIR-ligand mismatch. Disclosures Blomberg: VECURA: Employment. Hellström-Lindberg:Celgene: Research Funding.


Viruses ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 1365
Author(s):  
Michael A. Morgan ◽  
Arnold Kloos ◽  
Daniela Lenz ◽  
Nadine Kattre ◽  
Juliette Nowak ◽  
...  

Anti-cancer activity can be improved by engineering immune cells to express chimeric antigen receptors (CARs) that recognize tumor-associated antigens. Retroviral vector gene transfer strategies allow stable and durable transgene expression. Here, we used alpharetroviral vectors to modify NK-92 cells, a natural killer cell line, with a third-generation CAR designed to target the IL-3 receptor subunit alpha (CD123), which is strongly expressed on the surface of acute myeloid leukemia (AML) cells. Alpharetroviral vectors also contained a transgene cassette to allow constitutive expression of human IL-15 for increased NK cell persistence in vivo. The anti-AML activity of CAR-NK-92 cells was tested via in vitro cytotoxicity assays with the CD123+ AML cell line KG-1a and in vivo in a patient-derived xenotransplantation CD123+ AML model. Unmodified NK-92 cells or NK-92 cells modified with a truncated version of the CAR that lacked the signaling domain served as controls. Alpharetroviral vector-modified NK-92 cells stably expressed the transgenes and secreted IL-15. Anti-CD123-CAR-NK-92 cells exhibited enhanced anti-AML activity in vitro and in vivo as compared to control NK-92 cells. Our data (1) shows the importance of IL-15 expression for in vivo persistence of NK-92 cells, (2) supports continued investigation of anti-CD123-CAR-NK cells to target AML, and (3) points towards potential strategies to further improve CAR-NK anti-AML activity.


Sign in / Sign up

Export Citation Format

Share Document