Targeting the 5-Lipoxygenase as a Novel Principle of Stem Cell Therapy In Acute Myeloid Leukemia

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1846-1846
Author(s):  
Jessica Roos ◽  
Astrid Fischer ◽  
Dieter Steinhilber ◽  
Hubert Serve ◽  
Oliver Ottmann ◽  
...  

Abstract Abstract 1846 Chromosomal translocations such as t(15;17), t(8,21) or t(6;9) lead to the formation of chimeric genes encoding the PML/RAR, AML-1/ETO or DEK/CAN fusion proteins (FP). These FP are able to induce and to maintain acute myeloid leukemia (AML) by both blocking terminal differentiation of early hematopoietic progenitors and increasing the self renewal potential of the leukemic stem cells (LSC). LSCs are potential therapeutic targets and it is of great importance to elucidate which signaling pathways control their development and maintenance. Recently it has been shown that the presence of the 5-Lipoxygenase activity (5-LO) is indispensable for the induction and the maintenance of the BCR/ABL induced CML-like disease in mice. Its depletion or inhibition impairs the LSCs in the CML-like disease. 5-LO is the key enzyme in the biosynthesis pathway of leukotrienes, a group of proinflammatory lipid mediators derived from arachidonic acid. Furthermore we have shown that Sulindac sulfide, a dual Cycloxygenase/5 –LO inhibitor, was able, at 5-LO inhibitory concentrations, to interfere with the stem cell capacity of PML/RAR-positive LSC. It also overcame the differentiation block in PML/RAR-positive HSC. To disclose whether a “leukemic stem cell therapy” in AML is feasible if based on selectively targeting the 5-LO, we used two different selective 5-LO inhibitors, Zileuton and CJ-13,610, in a PML/RAR- and DEK/CAN-positive leukemia model. Zileuton, an anti-asthmatic drug, is a reversible inhibitor of 5-LO activity which leads to the inhibition of leukotrienes (LTB4, LTC4, LTD4, and LTE4) formation. CJ-13,610 is novel non redox, non iron chelating 5-LO inhibitor. As stem cell models we used Sca-1+/lin-murine HSC retrovirally transduced either with PML/RAR or DEK/CAN. Here we report that both Zileuton and CJ -13,610 at clinically feasible concentrations of 0.3 – 3μM interfered with the aberrant replating efficiency of PML/RAR and DEK/CAN expressing HSCs; ii.) inhibited the short-term stem cell (ST-HSC) capacity of PMR/RAR- and DEK/CAN-positive HSCs as assessed by colony forming unit-spleen day 12 assays in lethally irradiated recipient mice; and iii.) reduced the frequency of long-term HSC in a long term competitive repopulation stem cell assays. The effects of both compounds were not due related to the induction of apoptosis. Interestingly, on normal control HSC both Zileuton and CJ-13,610 exhibited a “paradox” effect by increasing ST-HSC as well as LT-HSC capacity. Our here presented data establish the inhibition of 5-LO by selective inhibitors as a feasible approach of molecular stem cell therapy in AML. Furthermore it strongly suggest an important role of leukotrienes for the maintenance of leukemic stem cells. The exact mechanisms by which the inhibition of 5-LO interferes with the LSC have still to be disclosed. Disclosures: Off Label Use: The use of anti-inflammatory drugs such as Zileuton and CJ-13610 as novel approach for stem cell treatment in AML is discussed.

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A465-A465
Author(s):  
Catherine Sabatos-Peyton ◽  
Tyler Longmire ◽  
Lisa Baker ◽  
Nidhi Patel ◽  
Anne-Sophie Wavreille ◽  
...  

BackgroundTIM-3 is expressed on leukemic stem cells (LSCs) and blasts in AML,1 2 and TIM-3 expression on MDS blasts correlates with disease progression.3 Functional evidence for TIM-3 in AML was established with an anti-TIM-3 antibody which inhibited engraftment and development of human AML in immuno-deficient murine hosts.1 TIM-3 promotes an autocrine stimulatory loop via the TIM-3/Galectin-9 interaction, supporting LSC self-renewal.4 In addition to its cell-autonomous role on LSCs/blasts, TIM-3 also has a critical role in immune system regulation, in adaptive (CD4+ and CD8+ T effector cells, regulatory T cells) and innate (macrophages, dendritic cells, NK cells) immune responses.5 MBG453 is a high-affinity, humanized anti-TIM-3 IgG4 antibody (Ab) (stabilized hinge, S228P), which blocks the binding of TIM-3 to phosphatidylserine (PtdSer). Recent results from a multi-center, open label phase Ib dose-escalation study (NCT03066648) in patients with high-risk MDS and no prior hypomethylating agent therapy evaluating MBG453 in combination with decitabine demonstrated encouraging preliminary efficacy with an overall response rate of 58%,6 and MBG453 combined with azacitidine also showed encouraging response rates.7 Preclinical experiments were undertaken to define the mechanism of action of the hypomethylating agent and anti-TIM-3 combination.MethodsTHP-1 cells (a human monocytic AML cell line) were pre-treated with decitabine and co-cultured with anti-CD3 activated healthy human donor peripheral blood mononuclear cells (PBMCs) in an Incucyte-based assay to measure cell killing. The ability of MBG453 to mediate antibody-dependent cellular phagocytosis (ADCP) was measured by determining the phagocytic uptake of an engineered TIM-3-overexpressing Raji cell line in the presence of MBG453 by phorbol 12-myristate 13-acetate (PMA)-activated THP-1 cells. Patient-derived AML xenograft studies were undertaken in immune-deficient murine hosts to evaluate the combination of decitabine and MBG453.ResultsMBG453 was determined to partially block the TIM-3/Galectin-9 interaction in a plate-based MSD (Meso Scale Discovery) assay, supported by a crystal structure of human TIM-3.8 Pre-treatment of THP-1 cells with decitabine enhanced sensitivity to immune-mediated killing in the presence of MBG453. MBG453 was determined to mediate modest ADCP, relative to controls. MBG453 did not enhance the anti-leukemic activity of decitabine in patient-derived xenograft studies in immuno-deficient hosts.ConclusionsTaken together, these results support both direct anti-leukemic effects and immune-mediated modulation by MBG453. Further studies are ongoing to determine: (1) whether MBG453 can mediate physiologically relevant ADCP of TIM-3-expressing leukemic cells; and (2) the potential of MBG453 to impact the autocrine feedback loop of TIM-3/Galectin-9.Ethics ApprovalThe human tissue used in these studies was under the Novartis Institutes of BioMedical Research Ethics Board IRB, Approval Number 201252867.ReferencesKikushige Y, Shima T, Takayanagi S, et al. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell 2010;7(6):708–717.Jan M, Chao MP, Cha AC, et al. Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker. Proc Natl Acad Sci USA 2011; 108(12): 5009–5014.Asayama T, Tamura H, Ishibashi M, et al. Functional expression of Tim-3 on blasts and clinical impact of its ligand galectin-9 in myelodysplastic syndromes. Oncotarget 2017;8(51): 88904–88917.Kikushige Y, Miyamoto T, Yuda J, et al. A TIM-3/Gal-9 autocrine stimulatory loop drives self-renewal of human myeloid leukemia stem cells and leukemic progression. Cell Stem Cell 2015; 17(3):341–352.Acharya N, Sabatos-Peyton C, Anderson AC. Tim-3 finds its place in the cancer immunotherapy landscape. J Immunother Cancer 2020; 8(1):e000911.Borate U, Esteve J, Porkka K, et al. Phase Ib Study of the Anti-TIM-3 Antibody MBG453 in combination with decitabine in patients with high-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Blood 2019;134 (Supplement_1):570.Borate U, Esteve J, Porkka K, et al. Abstract S185: Anti-TIM-3 antibody MBG453 in combination with hypomethylating agents (HMAs) in patients (pts) with high-risk myelodysplastic syndrome (HR-MDS) and acute myeloid leukemia (AML): a Phase 1 study. EHA 2020.Sabatos-Peyton C. MBG453: A high affinity, ligand-blocking anti-TIM-3 monoclonal Ab. AACR 2016.


2021 ◽  
Vol 11 ◽  
Author(s):  
Noortje van Gils ◽  
Fedor Denkers ◽  
Linda Smit

Standard induction chemotherapy, consisting of an anthracycline and cytarabine, has been the first-line therapy for many years to treat acute myeloid leukemia (AML). Although this treatment induces complete remissions in the majority of patients, many face a relapse (adaptive resistance) or have refractory disease (primary resistance). Moreover, older patients are often unfit for cytotoxic-based treatment. AML relapse is due to the survival of therapy-resistant leukemia cells (minimal residual disease, MRD). Leukemia cells with stem cell features, named leukemic stem cells (LSCs), residing within MRD are thought to be at the origin of relapse initiation. It is increasingly recognized that leukemia “persisters” are caused by intra-leukemic heterogeneity and non-genetic factors leading to plasticity in therapy response. The BCL2 inhibitor venetoclax, combined with hypomethylating agents or low dose cytarabine, represents an important new therapy especially for older AML patients. However, often there is also a small population of AML cells refractory to venetoclax treatment. As AML MRD reflects the sum of therapy resistance mechanisms, the different faces of treatment “persisters” and LSCs might be exploited to reach an optimal therapy response and prevent the initiation of relapse. Here, we describe the different epigenetic, transcriptional, and metabolic states of therapy sensitive and resistant AML (stem) cell populations and LSCs, how these cell states are influenced by the microenvironment and affect treatment outcome of AML. Moreover, we discuss potential strategies to target dynamic treatment resistance and LSCs.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Qingya Cui ◽  
Chongsheng Qian ◽  
Nan Xu ◽  
Liqing Kang ◽  
Haiping Dai ◽  
...  

AbstractAllogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative treatment for acute myeloid leukemia (AML). However, most patients experience relapse after allo-HSCT, with a poor prognosis, and treatment options are limited. The lack of an ideal targetable antigen is a major obstacle for treating patients with relapsed AML. CD38 is known to be expressed on most AML and myeloma cells, and its lack of expression on hematopoietic stem cells (HSCs) renders it a potential therapeutic target for relapsed AML. To investigate the clinical therapeutic efficacy and safety of CD38-targeted chimeric antigen receptor T (CAR-T-38) cells, we enrolled 6 AML patients who experienced relapse post-allo-HSCT (clinicaltrials.gov: NCT04351022). Prior to CAR-T-38 treatment, the blasts in the bone marrow of these patients exhibited a median of 95% (92–99%) CD38 positivity. Four weeks after the initial infusion of CAR-T-38 cells, four of six (66.7%) patients achieved complete remission (CR) or CR with incomplete count recovery (CRi); the median CR or CRi time was 191 (range 117–261) days. The cumulative relapse rate at 6 months was 50%. The median overall survival (OS) and leukemia-free survival (LFS) times were 7.9 and 6.4 months, respectively. One case relapsed 117 days after the first CAR-T-38 cell infusion, with remission achieved after the second CAR-T-38 cell infusion. All six patients experienced clinically manageable side effects. In addition, multiparameter flow cytometry (FCM) revealed that CAR-T-38 cells eliminated CD38 positive blasts without off-target effects on monocytes and lymphocytes. Although this prospective study has a limited number of cases and a relatively short follow-up time, our preliminary data highlight the clinical utility and safety of CAR-T-38 cell therapy in treating relapsed AML post-allo-HSCT.


Sign in / Sign up

Export Citation Format

Share Document