scholarly journals Antileukemic effects of the novel, mutant FLT3 inhibitor NVP-AST487: effects on PKC412-sensitive and -resistant FLT3-expressing cells

Blood ◽  
2008 ◽  
Vol 112 (13) ◽  
pp. 5161-5170 ◽  
Author(s):  
Ellen Weisberg ◽  
Johannes Roesel ◽  
Guido Bold ◽  
Pascal Furet ◽  
Jingrui Jiang ◽  
...  

Abstract An attractive target for therapeutic intervention is constitutively activated, mutant FLT3, which is expressed in a subpopulation of patients with acute myelocyic leukemia (AML) and is generally a poor prognostic indicator in patients under the age of 65 years. PKC412 is one of several mutant FLT3 inhibitors that is undergoing clinical testing, and which is currently in late-stage clinical trials. However, the discovery of drug-resistant leukemic blast cells in PKC412-treated patients with AML has prompted the search for novel, structurally diverse FLT3 inhibitors that could be alternatively used to override drug resistance. Here, we report the potent and selective antiproliferative effects of the novel mutant FLT3 inhibitor NVP-AST487 on primary patient cells and cell lines expressing FLT3-ITD or FLT3 kinase domain point mutants. NVP-AST487, which selectively targets mutant FLT3 protein kinase activity, is also shown to override PKC412 resistance in vitro, and has significant antileukemic activity in an in vivo model of FLT3-ITD+ leukemia. Finally, the combination of NVP-AST487 with standard chemotherapeutic agents leads to enhanced inhibition of proliferation of mutant FLT3-expressing cells. Thus, we present a novel class of FLT3 inhibitors that displays high selectivity and potency toward FLT3 as a molecular target, and which could potentially be used to override drug resistance in AML.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1823-1823
Author(s):  
Monica Pallis ◽  
Francis Burrows ◽  
Nigel H. Russell

Abstract Abstract 1823 In clinical trials, FLT3 inhibitors are reported to kill circulating AML blasts, but the bone marrow is protected. We have previously reported that niche-like conditions (fibronectin and a cytokine cocktail) significantly reduced the in vitro toxicity of the FLT3 inhibitor AG1296 to AML cells. Moreover, the toxicity of AG1296 to the chemoresistant leukaemic CD34+CD38-CD123+ subset was completely abolished under niche-like conditions. The novel multi-kinase inhibitor TG02 has selectivity against cell cycle and transcriptional CDKs and JAK2 as well as FLT3. TG02 has efficacy in in vivo models and induces apoptosis in primary AML cells. We have now evaluated its in vitro toxicity under niche-like conditions in bulk AML cells and in the CD34+CD38-CD123+ subset. In a cohort of six FLT3-ITD and five FLT3-wildtype samples, 100nM TG02 induced decreases of 30% in bulk cells and 32% in CD34+CD38-CD123+ cells, whereas AG1296 (5μM) induced a median 21% decrease in bulk cells under niche-like conditions but a 0% decrease in CD34+CD38-CD123+ cells. Lestaurtinib, sorafenib and sunitinib were used as comparators (all at 100 nM) and induced, respectively, 13%, 4% and 13% decrease in bulk cells and 10%, 0% and 8% decrease in CD34+CD38-CD123+ cells. FLT3 wildtype as well as ITD samples were targeted. In order to establish the molecular pathways involved in niche-mediated chemoresistance and its reversal, we treated primary AML samples with TG02 or AG1296 for 3 hours in the presence and absence of niche proteins; we measured activating phosphorylations of STAT3 (tyr705), STAT5 (tyr694), ERK1/2 (thr202/tyr404) and AKT(ser473). Basal levels of activating phosphorylations were generally higher in the bulk cells than the CD34+CD38-CD123+ cells, possibly reflecting the increased quiescence of the latter subset. STAT3, STAT5 and ERK1/2 phosphorylation were reduced by TG02 to a slightly greater degree than by AG1296 in bulk cells. However, in CD34+CD38-CD123+ cells this contrast was enhanced, such that AG1296 was ineffective, whereas TG02 was at least as effective as in bulk cells. Niche-like conditions induced an increase in phosphorylation of STAT5, but not of the other proteins tested. TG02 reduced this to basal levels in both bulk cells and CD34+CD38-CD123+ cells. AG1296 partially blocked niche-induced STAT5 phosphorylation in bulk cells, but not in CD34+CD38-CD123+ cells. It had no effect on ERK signalling. AKT phosphorylation was not informative. In conclusion, TGO2 is more cytotoxic than comparatively selective FLT3 inhibitors towards CD34+CD38-CD123+ AML cells as well as bulk cells under niche conditions and the toxicity is associated with downregulation of STAT3, STAT5 and ERK activation. Disclosures: Pallis: Tragara Pharmaceuticals: Research Funding. Burrows:Tragara Pharmaceuticals: Employment.


Blood ◽  
1996 ◽  
Vol 87 (7) ◽  
pp. 2723-2731 ◽  
Author(s):  
P Schwarzenberger ◽  
S Spence ◽  
N Lohrey ◽  
T Kmiecik ◽  
DL Longo ◽  
...  

To develop a rapid preclinical in vivo model to study gene transfer into human hematopoietic progenitor cells, MO-7e cells (CD-34+, c-kit+) were infected with multidrug resistance (MDR1)-containing retroviruses and then transplanted into nonobese diabetic severe combined immunodeficient mice (NOD SCID). MO-7e cells infected with a retrovirus encoding the human MDR1 cDNA showed integration, transcription, and expression of the transfered MDR1 gene. This resulted in a 20-fold increase in the resistance of MO-7e cells to paclitaxel in vitro. The expression of the MDR1 gene product was stable over a 6-month period in vitro without selection in colchicine. MO-7e and MDR1-infected MO-7e cells were transplanted into NOD SCID mice to determine whether MDR1 could confer drug resistance in vivo. A sensitive polymerase chain reaction method specific for human sequences was developed to quantitate the level of human cell engraftment in NOD SCID bone marrow (BM) cells. The percentage of human DNA in BM cells from MO-7e- transplanted mice was 10.9% and decreased to 0.7% in mice treated with paclitaxel. The percentage of human DNA in infected-MO-7e transplanted mice was 7.6% and that level was unchanged in mice treated with paclitaxel. These results show that expression of the MDR1 gene in human hematopoietic progenitor cells can confer functional drug resistance in an in vivo model.


2016 ◽  
Vol 113 (43) ◽  
pp. E6669-E6678 ◽  
Author(s):  
Mark A. Gregory ◽  
Angelo D’Alessandro ◽  
Francesca Alvarez-Calderon ◽  
Jihye Kim ◽  
Travis Nemkov ◽  
...  

Activating mutations in FMS-like tyrosine kinase 3 (FLT3) are common in acute myeloid leukemia (AML) and drive leukemic cell growth and survival. Although FLT3 inhibitors have shown considerable promise for the treatment of AML, they ultimately fail to achieve long-term remissions as monotherapy. To identify genetic targets that can sensitize AML cells to killing by FLT3 inhibitors, we performed a genome-wide RNA interference (RNAi)-based screen that identified ATM (ataxia telangiectasia mutated) as being synthetic lethal with FLT3 inhibitor therapy. We found that inactivating ATM or its downstream effector glucose 6-phosphate dehydrogenase (G6PD) sensitizes AML cells to FLT3 inhibitor induced apoptosis. Examination of the cellular metabolome showed that FLT3 inhibition by itself causes profound alterations in central carbon metabolism, resulting in impaired production of the antioxidant factor glutathione, which was further impaired by ATM or G6PD inactivation. Moreover, FLT3 inhibition elicited severe mitochondrial oxidative stress that is causative in apoptosis and is exacerbated by ATM/G6PD inhibition. The use of an agent that intensifies mitochondrial oxidative stress in combination with a FLT3 inhibitor augmented elimination of AML cells in vitro and in vivo, revealing a therapeutic strategy for the improved treatment of FLT3 mutated AML.


2022 ◽  
Vol 11 ◽  
Author(s):  
Yajun Wang ◽  
Lan Yao ◽  
Yao Teng ◽  
Hua Yin ◽  
Qiuling Wu

As an important member of the Argonaute protein family, PIWI-like protein 1 (PIWIL1) plays a key role in tumor cell viability. However, the exact function of PIWIL1 in multiple myeloma (MM) and the underlying mechanism remain unclear. Here, we revealed that PIWIL1 was highly expressed in myeloma cell lines and newly diagnosed MM patients, and that its expression was notably higher in refractory/relapsed MM patients. PIWIL1 promoted the proliferation of MM cells and conferred resistance to chemotherapeutic agents both in vitro and in vivo. More importantly, PIWIL1 enhanced the formation of autophagosomes, especially mitophagosomes, by disrupting mitochondrial calcium signaling and modulating mitophagy-related canonical PINK1/Parkin pathway protein components. Mitophagy/autophagy inhibitors overcome PIWIL1-induced chemoresistance. In addition, PIWIL1 overexpression increased the proportion of side population (SP) cells and upregulated the expression of the stem cell-associated genes Nanog, OCT4, and SOX2, while its inhibition resulted in opposite effects. Taken together, our findings demonstrated that PIWIL1 induced drug resistance by activating mitophagy and regulating the MM stem cell population. PIWIL1 depletion significantly overcame drug resistance and could be used as a novel therapeutic target for reversing resistance in MM patients.


Blood ◽  
2010 ◽  
Vol 116 (24) ◽  
pp. 5256-5267 ◽  
Author(s):  
Lina Happo ◽  
Mark S. Cragg ◽  
Belinda Phipson ◽  
Jon M. Haga ◽  
Elisa S. Jansen ◽  
...  

Abstract DNA-damaging chemotherapy is the backbone of cancer treatment, although it is not clear how such treatments kill tumor cells. In nontransformed lymphoid cells, the combined loss of 2 proapoptotic p53 target genes, Puma and Noxa, induces as much resistance to DNA damage as loss of p53 itself. In Eμ-Myc lymphomas, however, lack of both Puma and Noxa resulted in no greater drug resistance than lack of Puma alone. A third B-cell lymphoma-2 homology domain (BH)3-only gene, Bim, although not a direct p53 target, was up-regulated in Eμ-Myc lymphomas incurring DNA damage, and knockdown of Bim levels markedly increased the drug resistance of Eμ-Myc/Puma−/−Noxa−/− lymphomas both in vitro and in vivo. Remarkably, c-MYC–driven lymphoma cell lines from Noxa−/−Puma−/−Bim−/− mice were as resistant as those lacking p53. Thus, the combinatorial action of Puma, Noxa, and Bim is critical for optimal apoptotic responses of lymphoma cells to 2 commonly used DNA-damaging chemotherapeutic agents, identifying Bim as an additional biomarker for treatment outcome in the clinic.


Blood ◽  
2011 ◽  
Vol 117 (12) ◽  
pp. 3286-3293 ◽  
Author(s):  
Takashi Sato ◽  
Xiaochuan Yang ◽  
Steven Knapper ◽  
Paul White ◽  
B. Douglas Smith ◽  
...  

AbstractWe examined in vivo FLT3 inhibition in acute myeloid leukemia patients treated with chemotherapy followed by the FLT3 inhibitor lestaurtinib, comparing newly diagnosed acute myeloid leukemia patients with relapsed patients. Because we noted that in vivo FLT3 inhibition by lestaurtinib was less effective in the relapsed patients compared with the newly diagnosed patients, we investigated whether plasma FLT3 ligand (FL) levels could influence the efficacy of FLT3 inhibition in these patients. After intensive chemotherapy, FL levels rose to a mean of 488 pg/mL on day 15 of induction therapy for newly diagnosed patients, whereas they rose to a mean of 1148 pg/mL in the relapsed patients. FL levels rose even higher with successive courses of chemotherapy, to a mean of 3251 pg/mL after the fourth course. In vitro, exogenous FL at concentrations similar to those observed in patients mitigated FLT3 inhibition and cytotoxicity for each of 5 different FLT3 inhibitors (lestaurtinib, midostaurin, sorafenib, KW-2449, and AC220). The dramatic increase in FL level after chemotherapy represents a possible obstacle to inhibiting FLT3 in this clinical setting. These findings could have important implications regarding the design and outcome of trials of FLT3 inhibitors and furthermore suggest a rationale for targeting FL as a therapeutic strategy.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e14551-e14551
Author(s):  
P. C. Cheng ◽  
J. Crane ◽  
T. Hunter

e14551 Background: FLT3 receptor tyrosine kinase activating mutations contribute to leukemogenesis and poor prognosis in approximately 30% of acute myeloid leukemia (AML). An internal tandem duplication (ITD) mutation in the juxtamembrane domain of FLT3 results in loss of autoinhibition that leads to constitutive, ligand-independent activation of the receptor, with subsequent activation of multiple downstream signaling pathways, including RAS/MAPK and STAT5. Early phase clinical trials of FLT3 inhibitors show a lack of durability of responses, suggesting combination therapy may work better. Methods: In the course of conducting a synthetic lethality screen with a FLT3 inhibitor on the Ba/F3 murine cell line stably expressing human FLT3 or FLT3-ITD, we identified bortezomib, a proteasome inhibitor, as having potent activity against FLT3-ITD cells. The effects of drugs on proliferation, apoptosis, and phosphosignaling were quantified in Ba/F3 cells and in the HL60 (WT FLT3) and MV4–11 (FLT3-ITD) human cell lines, using an MTS- based colorimetric assay, caspase 3 and 7 activity assays, and immunoblotting, respectively. Results: A highly potent and specific FLT3 inhibitor was shown to have an IC50 of 1.9 nM, inhibiting proliferation, inducing apoptosis, and abrogating downstream STAT5 and ERK phosphorylation in FLT3-ITD cells. The IC50 of bortezomib for FLT3-ITD cells was 10 nM as compared to an IC50 of 40 nM for WT FLT3 cells. Surprisingly, bortezomib abrogated tyrosine phosphorylation of FLT3, STAT5, and ERK within 60 min of adding drug. When the FLT3 inhibitor and bortezomib were used at IC25 concentrations, a more pronounced inhibition of cell proliferation was observed when they were used in combination than with either alone. Conclusions: Bortezomib preferentially kills FLT3- ITD cells, showing a four-fold more potent inhibition of cell proliferation, induces apoptosis, and abrogates activation of FLT3 and its downstream effector pathways. The combination of proteasome inhibition with FLT3 inhibition results in enhanced cytotoxicity. The mechanism by which bortezomib affects the FLT3-ITD signaling axis and the possible synergy with FLT3 inhibitors remains to be elucidated. No significant financial relationships to disclose.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 28-28
Author(s):  
Ky-Youb Nam ◽  
Jeejin Im ◽  
June H-J H-J Han ◽  
Kyu-Tae Kim ◽  
Jeong-Hyeok Yoon ◽  
...  

Background: FMS-like tyrosine kinase 3 (FLT3), a member of the class III receptor tyrosine kinase family, plays a pivotal role in regulating cell growth and differentiation of hematopoietic cells. FLT3 is mutated in approximately 30% of AML patients either by internal tandem duplication (ITD) within the juxtamembrane portion or by point-mutations in the kinase domain (TKD). Thus, FLT3 inhibitors including quizartinib (AC220), midostaurin (PKC412) and gilteritinib (ASP2215) have been developed and undergone clinical testing for the treatment of AML. However, patients frequently relapse due to intrinsic and extrinsic resistance to these FLT3 inhibitors in spite of the initial clinical efficacy. We have developed a potent third-generation FLT3 inhibitor, PHI-101, capable of overcoming some of these mechanisms of resistance. Methods: Biochemical kinase assays for PHI-101 have been performed on 9 different FLT3 mutants and wild type FLT3. Cellular potencies of PHI-101 have also been assessed using various patient-derived AML cells as well as MV4-11, MOLM14 and BaF3 cell lines transformed with human FLT3 mutants including single mutations [FLT3(ITD), FLT3(D835Y)], double or triple mutations [FLT3(ITD/D835Y), FLT3(ITD/F691L), FLT3(ITD/F691L/D835Y)]. In order to evaluate in vivo efficacies of oral administration of PHI-101, xenograft mouse models and in vivo bioluminescence imaging have been utilized. Results: PHI-101 possessed excellent enzymatic potencies against FLT3 potential resistant mutants such as ITD/D835V and the gatekeeper ITD/F691L mutation, as well as against FLT3 single activating mutants that include ITD, D835V, D835H, and D835Y. PHI-101 inhibited the phosphorylation of FLT3 and downstream STAT5 and ERK1/2 more effectively than quizartinib and gilteritinib at equivalent doses. Moreover, oral administration of PHI-101 induced tumor regression in the xenograft mouse models developed by injection of BaF3 cells transformed with FLT3-ITD, FLT3-TKD or FLT3-ITD/TKD mutants in a dose-dependent fashion with no appreciable toxicities. In luciferase-bearing blood circulating mouse models with FLT3-double mutants (ITD/D835Y and ITD/F691L) or triple mutants (ITD/D835Y/F691L), oral administration (30 mpk, QD) of PHI-101 diminished more than 89% of bioluminescent intensity and reduced leukemic burden. PHI-101 also showed increased efficacy in extending the lifespan of xenograft mice compared to quizartinib. PHI-101 strongly suppressed proliferation and induced apoptosis in primary AML samples harboring FLT3/ITD and FLT3/TKD mutations with little effect on wild-type FLT3 samples. Conclusion: PHI-101, an orally bioavailable novel small molecule, is a potent third-generation FLT3 inhibitor able to overcome resistance to several resistance mutations based on in vitro and in vivo experiments. PHI-101 possesses excellent in vitro and in vivo activities against not only FLT3 single activating mutations (ITD or TKD mutants) but also FLT3 double (ITD/D835Y or ITD/F691L) and triple (ITD/D835Y/F691L) resistant mutations with no pronounced toxicities. Preclinical evaluation of PHI-101 showed clear evidence of antileukemic activity and improved efficacy in both in vitro and in vivo models. PHI-101 is currently under investigation in first-in-human clinical trials with relapsed or refractory AML patients. Disclosures Nam: Pharos I&BT Co., Ltd.: Current equity holder in private company. Im:Pharos I&BT Co., Ltd.: Current Employment. Han:Pharos I&BT Co., Ltd.: Current equity holder in private company. Kim:Pharos I&BT Co., Ltd.: Current Employment. Yoon:Pharos I&BT Co., Ltd.: Current equity holder in private company. Cho:Pharos I&BT Co., Ltd.: Research Funding. Choi:Pharos I&BT Co., Ltd.: Research Funding. Young:Pharos I&BT Co., Ltd.: Research Funding. Nguyen:Pharos I&BT Co., Ltd.: Research Funding. Zhu:Pharos I&BT Co., Ltd.: Research Funding. Li:Pharos I&BT Co., Ltd.: Research Funding. Small:Pharos I&BT Co., Ltd.: Consultancy, Research Funding. Sim:Pharos I&BT Co., Ltd.: Research Funding.


Blood ◽  
2004 ◽  
Vol 104 (4) ◽  
pp. 1145-1150 ◽  
Author(s):  
Mark Levis ◽  
Rosalyn Pham ◽  
B. Douglas Smith ◽  
Donald Small

AbstractPatients with acute myeloid leukemia (AML) harboring internal tandem duplication mutations of the FLT3 receptor (FLT3/ITD mutations) have a poor prognosis compared to patients lacking such mutations. Incorporation of FLT3 inhibitors into existing chemotherapeutic regimens has the potential to improve clinical outcomes in this high-risk group of patients. CEP-701, an indolocarbazole-derived selective FLT3 inhibitor, potently induces apoptosis in FLT3/ITD-expressing cell lines and primary leukemic blasts. We conducted a series of in vitro cytotoxicity experiments combining CEP-701 with chemotherapy using the FLT3/ITD-expressing cell lines MV4-11 and BaF3/ITD as well as a primary blast sample from a patient with AML harboring a FLT3/ITD mutation. CEP-701 induced cytotoxicity in a synergistic fashion with cytarabine, daunorubicin, mitoxantrone, or etoposide if used simultaneously or immediately following exposure to the chemotherapeutic agent. In contrast, the combination of pretreatment with CEP-701 followed by chemotherapy was generally antagonistic, particularly with the more cell cycle-dependent agents such as cytarabine. This effect appears to be due to CEP-701 causing cell cycle arrest. We conclude that in FLT3/ITD-expressing leukemia cells, CEP-701 is synergistic with standard AML chemotherapeutic agents, but only if used simultaneously with or immediately following the chemotherapy. These results should be considered when designing trials combining chemotherapy with each of the FLT3 inhibitors currently in clinical development. (Blood. 2004; 104:1145-1150)


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4049-4049
Author(s):  
Herman O Sintim ◽  
M. Javad Aman ◽  
Frederick Holtsberg ◽  
Ashkan Emadi ◽  
Rena G Lapidus

Abstract Introduction Presence of FLT3-ITD gene mutations is a poor prognostic factor in acute myeloid leukemia (AML). Midostaurin, a multikinase inhibitor, is approved for treatment of patients with newly diagnosed FLT3 mutant AML, in combination with cytarabine and daunorubicin standard chemotherapy. Midostaurin is not indicated as a single-agent induction therapy for AML treatment. Newer FLT3 inhibitors such as Quizartinib, Crenolanib and Gilteritinib have shown promising single agent activity in clinical trials involving patients with relapsed or refractory FLT3-mutated AML. Unfortunately, initial responses to FLT3 inhibitors are not durable, and leukemia progresses in virtually all patients. While several mechanisms of resistance to FLT3 inhibitors have been proposed, occurrence of new tyrosine kinase domain (TKD) mutations are among the most frequent and important mechanisms of resistance to current FLT3 inhibitors, making the development of novel FLT3 inhibitors imperative. F691, D835, and N676 are the most common mutations occurring in the kinase domain of the FLT3 protein; these confer resistance to currently available FLT3 inhibitors. We have synthesized a novel and selective FLT3 inhibitor, KRX-101 (a 4-substituted aminoisoquinoline), that has shown a superior anti-AML activity compared to available FLT3 inhibitors in vitro and in vivo. Importantly, KRX-101 possesses favorable pharmaceutical properties and has potent activity against the D835 and F691 mutations that arise during treatment with Quizartinib and Gilteritinib. Methods Using commercially available starting materials, the aminoisoquinoline compound was synthesized via a Sonogashira reaction. For enzymatic activity, KRX-101 and other FLT3 inhibitors were evaluated in protein based assays targeting FLT3-ITD including those with D835Y and F691L mutations (Reaction Biology, Malvern, PA; Eurofins DiscoveryX, Fremont, CA). In anti-proliferative assays, AML cell lines were exposed to KRX-101 and other FLT3 inhibitors for 72h in a 96-well plate. Assays were terminated with an MTT-like agent. IC50s were determined by GraphPad Prism. In pharmacokinetic assays, KRX-101 was administered to female rats (n=6) intravenously (IV, single dose, 5 mg/kg) or by oral gavage (single dose, 50 mg/kg). Blood was drawn at specific time points and plasma was isolated. The plasma concentrations of KRX-101 were determined by liquid chromatography mass spectrometry at Metabolite Profiling Facility, Bindley Bioscience Center, Purdue University. For in vivo efficacy studies, 1x106 MV4-11 cells expressing firefly luciferase were injected IV into female NSG mice. Three days later, mice were sorted into groups so that baseline luminescence (ie, disease burden) was equivalent and dosing started. Mice were imaged weekly. Results KRX-101 was synthesized with overall yield of 54% and has been scaled up for pharmaceutical development. KRX-101 in enzymatic activity assays inhibited FLT3-ITD at 2.3 nM (IC50) and FLT3-D835Y at 1.8 nM (IC50). In in vitro anti-proliferative assays, KRX-101 demonstrated robust activity in all tested AML cell lines harboring FLT3 mutations ranging from 0.07 to 7 nM (Table 1). KRX-101 showed similar or superior anti-AML activity in vitro compared to other FLT3 inhibitors. KRX-101 was readily orally bioavailable in rats. Twenty four hours after oral gavage, the plasma concentration of KRX-101 was >10 µg/mL (>18 µM); 1000 fold higher than in vitro IC50s suggesting that three times weekly dosing is reasonable. In a head to head comparative study, KRX-101 appeared to be superior to Gilteritinib in a FLT3-ITD AML orthotopic model (Fig. 1A) by inducing undetectable disease at Day 22. Additionally, two mice whose disease progressed on Gilteritinib responded to KRX-101 (Fig. 1B). In another study, animals with clearly detectable FLT3-ITD AML were treated with aminoisoquinoline for 44 days. In the absence of detectable AML at day 44, treatment was discontinued. Mice were then monitored until day 175; 4 of 5 mice (80%) had no measurable disease. In all studies, KRX-101 dosed daily or thrice weekly was tolerated well. Conclusion KRX-101 is a novel agent with promising activity in FLT3 inhibitor-resistant AML. Testing in other FLT3 inhibitor-resistant animal models with various tyrosine kinase domain mutations is ongoing. Investigational New Drug (IND) enabling studies are underway. The Phase I clinical trial is planned. . Disclosures Sintim: KinaRx, LLC: Other: Founder and Scientific Advisor. Aman:KinaRx, LLC: Other: Founder. Holtsberg:KinaRx, LLC: Other: Founder. Emadi:NewLink Genetics: Research Funding. Lapidus:KinaRx, LLC: Other: Founder and Scientific Advisor.


Sign in / Sign up

Export Citation Format

Share Document