scholarly journals MDM2 inhibitor APG-115 exerts potent antitumor activity and synergizes with standard-of-care agents in preclinical acute myeloid leukemia models

2021 ◽  
Vol 7 (1) ◽  
Author(s):  
Douglas D. Fang ◽  
Qiuqiong Tang ◽  
Yanhui Kong ◽  
Tao Rong ◽  
Qixin Wang ◽  
...  

AbstractAcute myeloid leukemia (AML) is a clinically and genetically heterogeneous clonal disease associated with unmet medical needs. Paralleling the pathology of other cancers, AML tumorigenesis and propagation can be ascribed to dysregulated cellular processes, including apoptosis. This function and others are regulated by tumor suppressor P53, which plays a pivotal role in leukemogenesis. Opposing P53-mediated activities is the mouse double minute 2 homolog (MDM2), which promotes P53 degradation. Because the TP53 mutation rate is low, and MDM2 frequently overexpressed, in patients with leukemia, targeting the MDM2-P53 axis to restore P53 function has emerged as an attractive AML treatment strategy. APG-115 is a potent MDM2 inhibitor under clinical development for patients with solid tumors. In cellular cultures and animal models of AML, we demonstrate that APG-115 exerted substantial antileukemic activity, as either a single agent or when combined with standard-of-care (SOC) hypomethylating agents azacitidine (AZA) and decitabine (DAC), or the DNA-damaging agent cytarabine (Ara-C). By activating the P53/P21 pathway, APG-115 exhibited potent antiproliferative and apoptogenic activities, and induced cell cycle arrest, in TP53 wild-type AML lines. In vivo, APG-115 significantly reduced tumor burden and prolonged survival. Combinations of APG-115 with SOC treatments elicited synergistic antileukemic activity. To explain these effects, we propose that APG-115 and SOC agents augment AML cell killing by complementarily activating the P53/P21 pathway and upregulating DNA damage. These findings and the emerging mechanism of action afford a sound scientific rationale to evaluate APG-115 (with or without SOC therapies) in patients with AML.

2020 ◽  
Vol 38 (36) ◽  
pp. 4260-4273
Author(s):  
Olga Salamero ◽  
Pau Montesinos ◽  
Christophe Willekens ◽  
José Antonio Pérez-Simón ◽  
Arnaud Pigneux ◽  
...  

PURPOSE Iadademstat is a novel, highly potent, and selective inhibitor of LSD1 (KDM1A), with preclinical in vitro and in vivo antileukemic activity. This study aimed to determine safety and tolerability of iadademstat as monotherapy in patients with relapsed/refractory acute myeloid leukemia (R/R AML). METHODS This phase I, nonrandomized, open-label, dose-escalation (DE), and extension-cohort (EC) trial included patients with R/R AML and evaluated the safety, pharmacokinetics (PK), pharmacodynamics (PD), and preliminary antileukemic activity of this orally bioavailable first-in-class lysine-specific demethylase 1 inhibitor. RESULTS Twenty-seven patients were treated with iadademstat on days 1 to 5 (5-220 µg/m2/d) of each week in 28-day cycles in a DE phase that resulted in a recommended dose of 140 µg/m2/d of iadademstat as a single agent. This dose was chosen to treat all patients (n = 14) in an EC enriched with patients with MLL/KMT2A-rearranged AML. Most adverse events (AEs) were as expected in R/R AML and included myelosuppression and nonhematologic AEs, such as infections, asthenia, mucositis, and diarrhea. PK data demonstrated a dose-dependent increase in plasma exposure, and PD data confirmed a potent time- and exposure-dependent induction of differentiation biomarkers. Reductions in blood and bone marrow blast percentages were observed, together with induction of blast cell differentiation, in particular, in patients with MLL translocations. One complete remission with incomplete count recovery was observed in the DE arm. CONCLUSION Iadademstat exhibits a good safety profile together with signs of clinical and biologic activity as a single agent in patients with R/R AML. A phase II trial of iadademstat in combination with azacitidine is ongoing (EudraCT No.: 2018-000482-36).


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 581-581
Author(s):  
Patrick Griffin ◽  
Steffan T Nawrocki ◽  
Takashi Satou ◽  
Claudia M Espitia ◽  
Kevin R. Kelly ◽  
...  

Abstract Abstract 581 The long-term prognosis for the majority of patients diagnosed with acute myeloid leukemia (AML) is very poor due, in part, to pre-existing myelodysplasia, multidrug resistance, and co-existing morbidities that limit therapeutic options. Novel strategies are essential in order to improve clinical outcomes. TAK-901 is an investigational small molecule kinase inhibitor that is currently being evaluated in Phase I trials. In preclinical studies, TAK-901 has demonstrated significant effects against a number of kinases with important roles in cancer including the Aurora kinases, which are key regulators of mitosis and whose overexpression in cancer promotes genetic instability, malignant pathogenesis, and drug resistance. We hypothesized that simultaneously targeting the activity of the Auroras and other oncogenic kinases with TAK-901 would disrupt AML pathogenesis. In order to test our hypothesis, we investigated the efficacy and pharmacodynamic activity of TAK-901 human AML cell lines, primary AML specimens, and an orthotopic bioluminescent disseminated mouse model of AML. TAK-901 potently diminished the viability of a panel of 8 AML cell lines as well as primary cells obtained from patients with AML. Acute exposure to TAK-901 ablated clonogenic survival, triggered the accumulation of polyploid cells, and induced apoptosis. The cytostatic and cytotoxic effects of TAK-901 were associated with significantly increased expression of the cyclin-dependent kinase inhibitor p27, growth arrest and DNA-damage-inducible 45a (GADD45a), and the BH3-only pro-apoptotic protein PUMA. Chromatin immunoprecipitation (ChIP) assays revealed that the elevation in the expression of these genes caused by administration of TAK-901 was due to increased FOXO3a transcriptional activity. The in vivo anti-leukemic activity of TAK-901 was investigated in a disseminated xenograft mouse model of AML established by intravenous injection of luciferase-expressing MV4-11 cells. IVIS Xenogen imaging was utilized to monitor disease burden throughout the study. In this mouse model, administration of TAK-901 was very well-tolerated and significantly more effective than the standard of care drug cytarabine with respect to suppressing disease progression and prolonging overall survival. Analysis of specimens collected from mice demonstrated that TAK-901 inhibited the homing of AML cells to the bone marrow microenvironment and induced AML cell apoptosis in vivo. Our collective findings indicate that TAK-901 is a novel multi-targeted kinase inhibitor that has significant preclinical activity in AML models and warrants further investigation. Disclosures: Satou: Takeda Pharmaceuticals: Employment. Hasegawa:Takeda Pharmaceuticals: Employment. Romanelli:Millennium Pharmaceuticals: Employment. de Jong:Takeda San Diego: Employment. Carew:Millennium Pharmaceuticals: Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2236-2236 ◽  
Author(s):  
Dirkje W Hanekamp ◽  
Megan K Johnson ◽  
Scott Portwood ◽  
Joshua Acklin ◽  
Eunice S. Wang

Abstract Background: Acute myeloid leukemia (AML) is an aggressive hematological malignancy occurring primarily in older adults. Despite high remission rates following upfront therapy, the disease eventually recurs in most patients, and overall cure rates remain only 20-30%. Preclinical studies have recently demonstrated that the marrow microenvironment in acute leukemic hosts to be intrinsically hypoxic, with AML progression associated with further hypoxia. Moreover, human AML cells and primary AML colonies cultured under hypoxia are markedly less sensitive to cytarabine chemotherapy than normoxic cells. We hypothesized that AML cells may respond to hypoxic stress and mediate chemoresistance in part by invoking autophagy, a highly regulated catabolic process by which cells evade apoptosis by degrading damaged cellular components. To test our hypothesis, we investigated the effects of two known autophagy inhibitors (bafilomycin A1 (Baf) and chloroquine (CQ)) on the sensitivity of human AML cells to various therapeutic agents under differing oxygen levels. Methods: We treated HEL (FLT-3 wildtype) and MV4-11 (FLT-3 ITD mutant) AML cells with autophagy inhibitors (Baf and CQ) alone and in combination with a chemotherapeutic drug (cytarabine (AraC), doxorubicin (Dox), decitabine (Dac)) or a tyrosine kinase inhibitor (sorafenib, SFN) under normoxic (21% O2) or hypoxic (1% O2) conditions. Apoptosis /cell death and proliferation were measured by flow cytometry for Annexin-PI and MTT assays, respectively. Autophagy was assessed by flow cytometry using Cyto-ID Green Dye (Enzo Life Sciences), fluorescent microscropy for acridine orange dye accumulation, and western blot analysis. Results: Autophagy in human ALL and AML cell lines was significantly increased following 24-72 hours of hypoxia (1% O2) as compared with normoxia and was a relatively late response to prolonged low oxygen levels (> 24 hours). Treatment with cytotoxic agents (AraC or Dox) or hypomethylating agent (Dac) resulted in a dose-dependent increases in the number of autophagic vesicles in AML cells consistent with autophagy induction. Low-doses of Baf which selectively inhibits the vacuolar H+ ATPase to prevent lysosomal acidification, and CQ, which blocks lysosome-autophagosome fusion by raising the pH of lysosomes and endosomes, both resulted in buildup of autophagic vesicles by flow cytometry consistent with inhibition of autophagic flux in human AML cells. Combination treatment with an autophagy inhibitor (Baf, CQ) and cytotoxic chemotherapy (AraC, Dox) significantly enhanced apoptosis and cell death over single agent therapy. Treatment with Baf combined with hypomethylating therapy (Dac) synergistically improved the anti-leukemic effects as compared with monotherapy (CI 0.09-0.31)(see Figure). The addition of Baf also improved cell death induced by sorafenib (SFN) on FLT-3 ITD mutant human AML cells (MV4;11) (CI 0.36-0.9) (see Figure). Single agent Baf or CQ treatment resulted in significantly higher levels of apoptosis and cell death in AML cells under hypoxia. The anti-tumor activity of almost all combination regimens was consistently improved under hypoxic versus normoxic culture conditions. In vivo CQ treatment (25-50 mg/kg i.p. daily) in preclinical human AML xenograft models significantly inhibited systemic leukemia progression as a single agent. Further experiments investigating the in vivo effects of CQ combined with other chemotherapeutic agents in preclinical AML xenograft models are ongoing. Conclusions: Our data suggest that human AML cells preferentially induce autophagy to promote survival under chronic hypoxia and following cytotoxic, hypomethylating, and FLT-3 tyrosine kinase inhibitor therapy. Strategies targeting autophagy therefore may have the potential to improve therapeutic responses and overcome chemoresistance of AML cells within the hypoxic bone marrow microenvironment. Figure 1 Figure 1. Figure 2 Figure 2. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5154-5154
Author(s):  
Armelle Goubard ◽  
Martine Humbert ◽  
Colin Mansfield ◽  
Olivier Hermine ◽  
Patrice Dubreuil ◽  
...  

Compound AB8939 is a structurally novel, small chemical molecule, synthesized tubulin inhibitor that can circumvent two of the major resistance mechanisms in acute myeloid leukemia (AML), namely P-glycoprotein (Pgp) and myeloperoxidase (MPO) mediated resistance, thereby conferring an important advantage over traditional tubulin inhibitors. A series of ex vivo and in vivo studies provide proof-of-concept that AB8939 has broad anti-proliferative activity across the breath of acute myeloid leukemia (AML) subtypes, i.e. M0 through M7 of the French-American-British AML classification. Acute myeloid leukemia blasts were isolated from patients' peripheral blood and/or bone marrow samples, collected either at the time of diagnosis or following relapse, and also from patient derived xenograft (PDX) models. After purification, mononuclear cells were treated for 48 hours with various concentrations of AB8939 or cytarabine (Ara-C) and analyzed in a cell proliferation/viability assay. AB8939 produced a strong anti-proliferative action against blasts isolated from AML patients with a majority of IC50 values ranging from 1.4 nM to 1.0 µM. Two-thirds of AML patients had nanomolar sensitivity to AB8939 (IC50 ≤ 500 nM), while 44% where very sensitive (IC50 ≤ 100 nM) and 11% were highly sensitive (IC50 ≤ 10 nM). The potential of AB8939 to overcome Ara-C-resistance was also evident with 66% of Ara-C-resistant blasts (i.e. IC50 >5 µM) being sensitive to AB8939. Notably, AB8939 demonstrated activity across the entire spectrum of AML subtypes, according to the French-American-British (FAB) AML classification, with an IC50 of < 50 nM in M0, M1, M4, M5 and M6 subtypes, corresponding to over 90% of the AML patient population. A slightly lower sensitivity was observed for the M3 subtype (IC50 = 1.25 µM). Additionally, potent AB8939 activity was also seen in Ara-C-insensitive biphenotypic and mixed-phenotype acute leukemia samples. All FAB categories other than M7 were tested in the abovementioned ex vivo assessment. Acute megakaryocytic leukemia (FAB AML subtype M7) is a rare form of adult AML, accounting for only 1% of cases, and is associated with resistance to standard treatment and poor prognosis. The effect of AB8939 in this subtype was assessed in vivo using an AMKL26 model, an NSG mouse model based on cells isolated from a patient with an aggressive acute megakaryocytic leukemia presenting the ETO2-GLIS2 fusion oncogene. Following post graft detection of blasts, single agent AB8939 was administered intravenously at a dose of 2 mg/kg for three consecutive days per week (3 ON / 4 OFF) for 2 weeks and then at 5 mg/kg for three consecutive days per week (3 ON / 4 OFF) for 1 week. At the end of the 3-week treatment period, blast detection in bone marrow was performed via bioluminescence imaging with comparison to vehicle-treated controls. As seen in the figure below, single agent AB8939 showed strong anti-leukemic activity in this AMKL26 mouse model as evidenced by the near eradication of blasts. No blasts could be detected in 6 out of 8 mice treated with AB8939. At the described dosing schedule, AB8939 was well-tolerated with no toxicity-related deaths and no impact on animal body weight or behavior. These findings provide preclinical proof of concept for the development of AB8939 as a next-generation tubulin inhibitor for AML, in particular for poor-prognosis AML subsets and relapse/refractory AML; i.e. patients that currently have very limited therapeutic options and represent the highest unmet medical need. Disclosures Goubard: AB Science: Employment. Humbert:AB Science: Employment. Mansfield:AB Science: Employment, Patents & Royalties. Hermine:AB Science: Membership on an entity's Board of Directors or advisory committees. AB8939 Study Group:AB Science: Consultancy, Employment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4026-4026
Author(s):  
Nathaniel C. Doro ◽  
Deepika Lal ◽  
Peter King ◽  
Eddy Freyne ◽  
Tim Perera ◽  
...  

Abstract JNJ-26483327 is a novel oral Pan Her/Src/VEGFR-3 inhibitor which has previously been shown in preclinical models to cross the blood-brain barrier and to reach high levels in brain, solid tumor, and bone marrow sites. JNJ-26483327 is not an active substrate for P-glycoprotein pumps and has been well tolerated to date in an ongoing phase I trial. VEGF-C signaling through the VEGFR-3 (FLT-4) receptor has been shown to promote growth of acute myeloid leukemia (AML) cells and to mediate resistance to multiple chemotherapy drugs in vitro. Anti-VEGFR-3 antibody therapy decreased angiogenesis, increased hypoxia and necrosis, and reduced lymph node metastases in solid tumor xenografts. To date, however, VEGF-C/VEGFR-3 inhibition has not been actively been investigated for treatment of hematological malignancies. We hypothesized that JNJ- 26483327 treatment of VEGFR-3 expressing systemic AML would limit tumor growth and lymphatic spread via VEGF-C/VEGFR-3 mechanisms. An initial dose-finding pilot experiment was performed using SCID mice engrafted via tail vein with ten million HELluc cells, human acute myeloid leukemia cells with known expression of VEGF-A/C and VEGFR-2/3 and stably transfected with luciferase constructs to facilitate small animal imaging. Mice were treated with PBS, vehicle (200 mL by mouth twice daily), low dose JNJ-26483327 (75 mg/kg by mouth twice daily, total 150 mg/kg/day) and high dose JNJ- 26483327 (125 mg/kg by mouth daily, total 250 mg/kg/day) for 10 consecutive days. We found that low dose JNJ-26483327 therapy significantly improved the median survival of HELluc systemic xenografts by 46% (26 days longer than vehicle-treated controls) (p&lt;0.05). Although high dose JNJ-26483327 prolonged median survival over vehicletreated controls, the difference was not statistically significant. Moreover, although JNJ- 26483327 improved survival, HELluc leukemia burden (as measured by bioluminescent imaging) was not significantly reduced or eradicated as compared to control, consistent with cytostatic but not cytotoxic anti-tumor effects. VEGFR-3 signaling has also been shown to mediate leukemia cell proliferation, survival, and resistance to chemotherapy. Based on preclinical and clinical data demonstrating improved anti-tumor activity of VEGF inhibitors when combined with chemotherapy, we hypothesized that combining JNJ-26483327 with chemotherapy used in conventional AML therapy may result in additive synergistic anti-tumor effects. To determine if JNJ-26483327 inhibition enhanced the effects of cytotoxic chemotherapy, systemic HELluc tumor bearing mice were treated with low dose JNJ-26483327 (150 mg/kg/day for 10 days) and a single maximally tolerated dose of doxorubicin (1.5mg/kg). Both single agent doxorubicin and single agent JNJ-26483327 treatment resulted in significant reduction of HELluc tumor burden. However, no significant decrease in leukemia burden was observed after combination JNJ-26483327+doxorubicin treatment when compared to single agent groups. Lastly we postulated that combination therapies of JNJ-26483327 with other anti-VEGF therapies directed at inhibition of VEGF-A, VEGFR-1, or VEGFR-2 would result in inhibition of all known VEGFR signaling pathways and result in improved anti-leukemic effects of JNJ-26483327 therapy. Systemic HELluc bearing mice were treated with PBS, vehicle, low dose JNJ-26483327, an anti-hVEGF-A antibody BV (bevacizumab, Genentech) or combination JNJ-26483327+ BV. Results showed that single agent low dose JNJ- 26483327 or single agent BV significantly reduced HELluc tumor burden up until day 20. Combination JNJ-26483327+BV treatment, however, did not result in additive/synergistic anti-leukemic effects as compared to single agent therapy and may in fact have resulted in possible antagonistic effects. Conclusions: Single agent JNJ-26483327 therapy prolongs survival of mice engrafted with VEGFR-3+ HEL AML cells. Limitations of the above studies include the short duration of JNJ-26483327 administration (10 days only) and lack of synergistic effects of JNJ-26483327 when combined with doxorubicin and anti-hVEGF-A antibody therapy. Future studies will address the effects of long-term JNJ-26483327 administration on AML stem cell growth using NOD/SCID mouse models engrafted with patient samples and combination JNJ-26483327+ cytarabine/anthracycline chemotherapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3302-3302
Author(s):  
Timothy Pardee ◽  
Evan Gomes ◽  
Jamie Jennings-Gee ◽  
David L. Caudell ◽  
William Gmeiner

Abstract Abstract 3302 Acute Myeloid Leukemia (AML) is an aggressive myeloid malignancy that leads to marrow failure and death. This disease affects approximately 12,000 people per year in the United States, causing 9,000 deaths. Despite decades of research, therapy remains essentially unchanged and outcomes are poor. In patients over the age of 60 less then 10% of patients survive 5 years from diagnosis. There is a desperate need for the identification of new active agents with favorable toxicity profiles. The novel polymeric fluoropyrimidine (FP) FdUMP[10] is an oligodeoxynucleotide pro-drug of the thymidylate synthase (TS)-inhibitory FP metabolite 5-fluoro-2'-deoxyuridine-5`-O-monophosphate (FdUMP). The observation that this compound was highly active against several leukemia lines in the NCI 60 cell line screen prompted us to evaluate its activity in several preclinical models of AML. In vitro, FdUMP[10] exhibited remarkable activity against 3 human acute leukemia cell lines, HL60, Jurkat and THP-1, with IC50 values of 3.378 nM (95% CI 2.984 to 3.825), 5.438 nM (4.609 to 6.417) and 4.093 nM (3.413 to 4.907) respectively. We next tested its efficacy against a more genetically defined murine model of AML driven by expression of MLL-ENL. FdUMP[10] exhibited even greater activity against all murine lines tested. The IC50 values of FdUMP[10] against two MLL-ENL driven murine AML cell lines were 214 pM (95%CI 178.9 to 255.9) and 292.3 pM (251.8 to 339.4). The IC50 values observed for FdUMP[10] for all the murine lines tested were lower than both Ara-C (30-40 nM) and doxorubicin (2-4 nM). We then determined the cytotoxic mechanism for FdUMP[10] in vitro. Upon treatment with FdUMP[10] both the human and murine cell lines undergo extensive apoptosis as indicated by Annexin V and propidium iodide staining. Treated cells developed γH2AX foci, rapid and complete TS inhibition and display trapped Topoisomerase I (Topo I) cleavage complexes. FdUMP[10]-mediated induction of apoptosis was p53 independent as murine AML cells that had p53 knocked down by RNAi demonstrated resistance to both Ara-C and doxorubicin, but not to FdUMP[10]. We next tested the efficacy of FdUMP[10] in vivo. The MLL-ENL driven murine AML model results in blasts that can be transplanted into sublethally irradiated, immunocompetent, syngeneic recipients. The recipients develop a fatal and therapy-resistant AML. Lines were generated that expressed a luciferase reporter. Animals were imaged 6–7 days after injection of the leukemias to ensure engraftment and then began treatment with either the combination of Ara-C plus doxorubicin, single-agent FdUMP[10], or observation. Studies were performed using 2 doses of FdUMP[10] at 150 or 300 mg/kg injected on days 1 and 3 and compared to animals treated with 100 mg/kg Ara-C and 3mg/kg doxorubicin injected on days 1 through 5. Both treatments resulted in a statistically significant survival advantage over observation. A preliminary toxicology study compared FdUMP[10], 150 mg/kg daily, to 5-fluorouracil (5 FU), 150 mg/kg daily, or the combination of Ara-C at 100 mg/kg plus doxorubicin at 3 mg/kg daily. All groups were treated for 3, 4 or 5 days. On day 6 animals were sacrificed and organs harvested, sectioned, and stained. Slides were then reviewed by a veterinary pathologist. Tissues most affected were the small intestine, colon, and the bone marrow. The 5FU-treated animals had severe villous blunting and fusion with crypt necrosis in both large and small intestine. In contrast, FdUMP[10]-treated animals had only mild crypt epithelial apoptosis with mitoses. The 5 FU and Ara-C plus doxorubicin groups had a severe pan-cytopenia in the marrow compared to FdUMP[10] treated animals that showed only minimal to mild apoptosis. These data support the assertion that FdUMP[10] has lower toxicity then either Ara-C plus doxorubicin or identically dosed 5 FU. In summary FdUMP[10] exhibited remarkable activity against AML cells in vitro and in vivo. Additionally, FdUMP[10] had decreased toxicity compared to treatment with either single agent 5 FU or combination treatment with Ara-C plus doxorubicin. Disclosures: Gmeiner: Salzburg Therapeutics: Equity Ownership.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3726-3726
Author(s):  
Daniel Olive ◽  
Audrey Benyamine ◽  
Aude Le Roy ◽  
Rémy Castellano ◽  
Julie Gertner-Dardenne ◽  
...  

Abstract As they can kill Acute Myeloid Leukemia (AML) blasts in vitro and in vivo, Vg9Vd2T cells are key players in the design of new strategies of immunotherapy. AminoBisphonates (NBP) can enhance their activation in vitro and in vivo. Their combination with low-dose IL2 has shown promising results in 2 patients with AML who underwent partial remission. NBP treatment of blasts inhibits the Mevalonate pathway. The subsequent accumulation of Isopentenyl Diphosphate sensitize AML blasts to Vg9Vd2T cells killing but some AML cell lines blasts are resistant to this TCR mediated-lysis. Butyrophilin 3 A1 (BTN3A1) has been shown to be involved in IPP recognition and Vg9Vd2 T cells activation. Agonist monoclonal antibodies (mAb) recognizing the 3 isoforms of BTN3, can trigger BTN3 on tumor cell lines and sensitize them to Vg9Vd2 T cells lysis. We show that primary AML blasts from patient at diagnosis are heterogeneously killed by allogenic-IL-2-NBP-expanded Vg9Vd2 T. Some are resistant to this lysis and/or poorly sensitized by NBP. BTN3 molecules are highly expressed by blasts of AML cell lines and primary AML samples. We show that treatment of primary AML blasts with agonist anti-BTN3 mAb can overcome the resistance to Vg9Vd2 cells lysis in vitro. We assess this effect in vivo, showing that the addition of agonist anti-BTN3 mAb to Vg9Vd2 cells infusion decreased the tumor burden and increased the survival of NOG mice xenografted with luciferase-transduced U937 cell line. We confirm this effect in a model of mice xenografted with primary AML blasts, showing that treatment with anti-BTN3 mAb added to Vg9Vd2 cells infusion can decrease the number of blastic cells in the spleen, bone marrow and the blood, without requiring additional cytokine infusion. This drastic effect on sensitization of primary AML blasts to Vg9Vd2T cells killing could be of great interest especially in cases of refractory or relapsing AML. Disclosures No relevant conflicts of interest to declare.


Leukemia ◽  
2021 ◽  
Author(s):  
Uwe Platzbecker ◽  
Anne Sophie Kubasch ◽  
Collin Homer-Bouthiette ◽  
Thomas Prebet

AbstractMyelodysplastic syndromes (MDS) represent a heterogeneous group of myeloid neoplasms that are characterized by ineffective hematopoiesis, variable cytopenias, and a risk of progression to acute myeloid leukemia. Most patients with MDS are affected by anemia and anemia-related symptoms, which negatively impact their quality of life. While many patients with MDS have lower-risk disease and are managed by existing treatments, there currently is no clear standard of care for many patients. For patients with higher-risk disease, the treatment priority is changing the natural history of the disease by delaying disease progression to acute myeloid leukemia and improving overall survival. However, existing treatments for MDS are generally not curative and many patients experience relapse or resistance to first-line treatment. Thus, there remains an unmet need for new, more effective but tolerable strategies to manage MDS. Recent advances in molecular diagnostics have improved our understanding of the pathogenesis of MDS, and it is becoming clear that the diverse nature of genetic abnormalities that drive MDS demands a complex and personalized treatment approach. This review will discuss some of the challenges related to the current MDS treatment landscape, as well as new approaches currently in development.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 853-853
Author(s):  
Shruti Bhatt ◽  
Vineeth Kumar Murali ◽  
Holly Zhu ◽  
Sophia Adamia ◽  
Amanda L Christie ◽  
...  

Abstract Identification of genetic heterogeneity in acute myeloid leukemia (AML) has provided a unique opportunity for the greater individualization of therapy. However the implementation of new therapies has lagged far behind the ability to initially recognize operationally important genetic lesions. Until we have further bridged this gap between the identification of genetic lesions and the resultant knowledge of effective therapies, alternative strategies for rapidly identifying candidate therapies can become an important tool for precision medicine. Since most agents, regardless of whether "cytotoxic" or "targeted" ultimately function by activating the mitochondrial apoptotic pathway, we hypothesized that a tool that measures mitochondrial sensitivity may serve as a broadly predictable biomarker. We developed a dynamic BH3 profiling (DBP) technique that measures early death signaling within 8-16 hours after exposure to drugs. Increased cell death signaling is reflected by increased mitochondrial sensitivity (i.e. increased priming) to standardized BH3 peptides mimicking pro-apoptotic proteins. To develop a personalized therapy for AML using DBP, we utilized 20 independent patient derived xenograft (PDX) models, established from de novo, primary refractory or relapsed (R/R) patients (available at http://www.PRoXe.org). Human myeloblasts from spleen and bone marrow of xenotransplanted NSG mice were exposed to 30 targeted and 3 standard of care drugs to determine mitochondrial responses. Unsupervised hierarchical clustering of ex-vivo DBP measurements across AML PDXs revealed several distinct clusters. Majority of targeted agents with an ability to induce priming in selective PDXs were enriched within a cluster, including kinase inhibitors, epigenetic modifiers, SMAC mimetic and chemotherapy drugs. In contrast, a discrete subcluster of drugs showed sensitivity across majority of PDXs, including BH3 mimetics, CDK9 inhibitors and HDAC inhibitors. Drugs with identical mechanism of action showed similar priming patterns across PDXs. Of note, 3 non-myeloid PDXs clustered distinctly from AML, an indication of differential priming responses owing to their cells of origin. AML PDXs developed from treatment naïve patients clustered adjacently and showed greater priming responses to a large number of drugs as opposed to PDXs from R/R patients that formed a discrete cluster. These data reveal that mitochondrial priming can stratify AML PDXs according to its predicted sensitivity to targeted agents. Next, we validated ability of DBP to predict in-vivo responses of single agent birinapant (SMAC mimetic), JQ-1 (BRD-4 inhibitor), quizartinib (FLT-3 inhibitor), and venetoclax (BCL-2 inhibitor) across 6 AML PDX models, prioritized based on their greatest range of priming responses. We found that birinapant was most efficacious nonetheless as expected from ex-vivo DBP studies, responses varied between different PDX models. Myeloblasts of those PDXs that showed the greatest drug-induced changes in apoptotic priming were indeed the PDXs with the highest in-vivo responses. When we compared the ability of DBP to identify sensitive PDXs with additional precision medicine tools such as genomics, we found that DBP was able to accurately predict quizartinib activity in PDXs expressing WT FLT-3, which would have been predicted to be unresponsive based on genomic analysis. Collectively, priming responses obtained from ex-vivo DBP was able to rank different PDX models according to their sensitivities to targeted agents (AUC of ROC curve 0.8731, p&lt;0.005). To investigate if DBP can predict combination therapies in relapsed settings, we first developed resistant models of single agents and then repeated DBP. Myeloblasts from relapsing clone showed reduced overall mitochondrial priming and lacked acquisition of a new chemical dependency compared to initial clone. This suggests that targeting of pre-existing dependencies might be more crucial than therapy induced dependency for AML. In summary, our findings highlight that mitochondria-based measurements could identifying individualized therapy for a heterogeneous population and may serve as a as a powerful biomarker to identify the best responders to patient therapies. Disclosures Letai: AbbVie, AstraZeneca, Novartis: Consultancy, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document