scholarly journals Dual Targeting of Aurora Kinases with AMG 900 Exhibits Potent Preclinical Activity Against Acute Myeloid Leukemia with Distinct Post-Mitotic Outcomes

2018 ◽  
Vol 17 (12) ◽  
pp. 2575-2585 ◽  
Author(s):  
Marc Payton ◽  
Hung-Kam Cheung ◽  
Maria Stefania S. Ninniri ◽  
Christian Marinaccio ◽  
William C. Wayne ◽  
...  
2020 ◽  
Vol 4 (7) ◽  
pp. 1478-1491 ◽  
Author(s):  
Andrew S. Moore ◽  
Amir Faisal ◽  
Grace W. Y. Mak ◽  
Farideh Miraki-Moud ◽  
Vassilios Bavetsias ◽  
...  

Abstract Internal tandem duplication of FLT3 (FLT3-ITD) is one of the most common somatic mutations in acute myeloid leukemia (AML); it causes constitutive activation of FLT3 kinase and is associated with high relapse rates and poor survival. Small-molecule inhibition of FLT3 represents an attractive therapeutic strategy for this subtype of AML, although resistance from secondary FLT3 tyrosine kinase domain (FLT3-TKD) mutations is an emerging clinical problem. CCT241736 is an orally bioavailable, selective, and potent dual inhibitor of FLT3 and Aurora kinases. FLT3-ITD+ cells with secondary FLT3-TKD mutations have high in vitro relative resistance to the FLT3 inhibitors quizartinib and sorafenib, but not to CCT241736. The mechanism of action of CCT241736 results in significant in vivo efficacy, with inhibition of tumor growth observed in efficacy studies in FLT3-ITD and FLT3-ITD-TKD human tumor xenograft models. The efficacy of CCT241736 was also confirmed in primary samples from AML patients, including those with quizartinib-resistant disease, which induces apoptosis through inhibition of both FLT3 and Aurora kinases. The unique combination of CCT241736 properties based on robust potency, dual selectivity, and significant in vivo activity indicate that CCT241736 is a bona fide clinical drug candidate for FLT3-ITD and TKD AML patients with resistance to current drugs.


2008 ◽  
Vol 7 (5) ◽  
pp. 1140-1149 ◽  
Author(s):  
Anna Morena D'Alise ◽  
Giovanni Amabile ◽  
Mariangela Iovino ◽  
Francesco Paolo Di Giorgio ◽  
Marta Bartiromo ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1276-1276
Author(s):  
Mario Scarpa ◽  
Prerna Singh ◽  
Shivani Kapoor ◽  
Jonelle K. Lee ◽  
Sandrine Niyongere ◽  
...  

Introduction fms-like tyrosine like kinase 3 internal tandem duplication (FLT3-ITD), present in acute myeloid leukemia (AML) cells of 30% of patients, results in constitutive and aberrant FLT3 signaling and, clinically, short disease-free survival. Efficacy of FLT3 inhibitors is limited and transient, but may be enhanced by dual targeting of FLT3-ITD signaling pathways. The tumor suppressor protein phosphatase 2A (PP2A) is inhibited in cells with FLT3-ITD. The oncogenic serine/threonine kinase Pim-1 is transcriptionally upregulated and also stabilized by PP2A inhibition in cells with FLT3-ITD. Pim-1 contributes directly to FLT3-ITD proliferative and anti-apoptotic effects, and also phosphorylates and stabilizes FLT3-ITD in a positive feedback loop. Moreover FLT3-ITD, PP2A and Pim-1 all regulate the transcription factor c-Myc. PP2A-activating drugs enhance efficacy of FLT3 inhibitors. We sought to identify mechanisms underlying the efficacy of this combination. Methods Ba/F3-ITD and MV4-11 cells, with FLT3-ITD, and blasts from patients with AML with FLT3-ITD were cultured with the FLT3 inhibitors gilteritinib (15 nM) or quizartinib (1 nM) and/or the PP2A-activating drugs FTY720 (2-4 µM) or DT-061 (10 µM), or with DMSO control. Pim-1, c-Myc, p-AKT (S473 and T308) and AKT protein expression was measured by immunoblotting, along with p-STAT5 (Y694), STAT5, p-PP2A (Y307) and PP2A expression. To study post-translational regulation, cells were cultured with cycloheximide (100 µg/mL) with and without the proteasome inhibitor MG-132 (20 µM). Ubiquitinated c-Myc was measured by co-immunoprecipitation and immunoblotting with c-Myc and ubiquitin antibodies. Ba/F3-ITD cells were stably transfected with estrogen receptor (ER)-c-Myc, kinase-dead Pim-1 or myristoylated AKT plasmids or corresponding empty vectors. Apoptosis was detected by Annexin V and propidium iodine staining, measured by flow cytometry. Cells were also cultured with the pan-Pim kinase inhibitor AZD1208 (1 µM), the Myc inhibitor 10058-F4 (100 µM) or the pan-AKT inhibitor MK-2206 (5 µM). Results Concurrent treatment of Ba/F3-ITD and MV4-11 cells and primary AML cells with FLT3-ITD with a FLT3 inhibitor (gilteritinib or quizartinib) and a PP2A-activating drug (FTY720 or DT-061) decreased growth and increased apoptosis induction, relative to treatment with single drugs. Concurrent FLT3 inhibitor and PP2A-activating drug treatment decreased expression of both Pim-1 and c-Myc protein. Concurrent treatment decreased Pim-1 half-life from 15 to 5 minutes, and c-Myc half-life from 30 to 5 minutes, while half-lives were restored by concurrent treatment with the proteasome inhibitor MG-132. Concurrent treatment was also shown to increase c-Myc ubiquitination. Effects of concurrent treatment on Pim-1 and c-Myc were independent, as transfection with kinase-dead Pim-1 or treatment with Pim inhibitor AZD1208 did not alter c-Myc downregulation, and c-Myc overexpression or treatment with Myc inhibitor 10058-F4 did not alter Pim-1 downregulation. Concurrent treatment with FLT3 inhibitor and PP2A-activating drug did not alter expression of c-Myc deubiquitinases, but rapidly decreased AKT S473 and T308 phosphorylation. FLT3 inhibitor and PP2A activator co-treatment did not induce downregulation or increased turnover of Pim-1 and c-Myc protein or apoptosis in cells with constitutive AKT activation caused by transfection of myristoylated AKT. Moreover, AKT inhibition downregulated Pim-1 and c-Myc protein expression, decreased Pim-1 and c-Myc protein half-lives from 15 to 5 minutes and 30 to 10 minutes, respectively, and induced apoptosis of cells with FLT3-ITD, replicating the effects of FLT3 inhibitor and PP2A activator co-treatment. Conclusion PP2A activators enhance the efficacy of FLT3 inhibitors in AML cells with FLT3-ITD through AKT inactivation-dependent increased Pim-1 and c-Myc proteasomal degradation, which is a novel mechanism. The data support further preclinical and clinical testing of this dual targeting approach to treatment of AML with FLT3-ITD. Disclosures Baer: Takeda: Research Funding; Incyte: Research Funding; Kite: Research Funding; Forma: Research Funding; AI Therapeutics: Research Funding; Abbvie: Research Funding; Astellas: Research Funding.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 119-119 ◽  
Author(s):  
Amir T. Fathi ◽  
Seth A. Wander ◽  
Traci M. Blonquist ◽  
Karen K. Ballen ◽  
Eyal C. Attar ◽  
...  

Abstract The aurora kinases play an essential role in regulating mitosis and cell division. Over-expression of aurora kinases A and B has been associated with poor-risk features in acute myeloid leukemia (AML). Preclinical studies suggest that small molecule aurora kinase inhibitors have activity in AML cell lines. Alisertib (MLN8237), a novel Aurora-A kinase inhibitor, has been generally well tolerated in early clinical trials of hematological malignancies, including AML. This study sought to evaluate tolerability of alisertib combined with standard “7+3” induction chemotherapy for AML. In this phase I, 3+3 cohort dose-escalation study, alisertib was administered with standard 7+3 induction for newly diagnosed AML. Those with acute promyelocytic leukemia or with AML and core-binding factor alterations were excluded. All patients received 7+3 induction (continuous infusion cytarabine 200mg/m2 x 7 days and intravenous idarubicin 12mg/m2 x 3 days), after which on day 8, alisertib was administered twice daily (BID) for 7 days. Dose escalation occurred via three cohorts (10mg BID, 20mg BID, 30mg BID). All underwent a mid-induction bone marrow biopsy, following the course of alisertib, to assess for residual disease, which if present, was to be treated with 5+2 re-induction (cytarabine 200mg/m2 x 5 days, idarubicin 12mg/m2 x 2 days) without alisertib. Following induction, up to four cycles of consolidation were allowed, using cytarabine (3g/m2 BID days 1,3,5 for those aged <60 and 2g/m2 daily days 1-5 for those aged ≥60) followed by alisertib, according to dose cohort, on days 6-12. After consolidation, alisertib maintenance was initiated, at cohort dose level, for days 1-7 of 21 day cycles, to be continued for 12 months or until disease progression. Those eligible for stem cell transplant (SCT), following induction and/or consolidation courses, were removed from study for this purpose. Currently, 14 patients are enrolled on study (n=3, 10mg BID; n=7, 20mg BID; n=4, 30mg BID). The median age was 62 (range 43-75); 9 (64%) were male, and all but 3 were Caucasian. One patient (7%) had therapy-related AML and six (43%) had underlying myelodysplasia. FLT3 mutations were detected in 3 (21%), NPM1 mutations in 2 (14%), CEBPA mutations in 2 (14%), and IDH1/IDH2 mutations in 4 patients (28.4%). One patient in cohort 2 died of sepsis, deemed unrelated to study drug, prior to completion of the toxicity assessment period and was replaced. Five patients have gone on to SCT. All patients received induction, 7 patients have undergone first consolidation, 3 a second consolidation, 2 a third consolidation, 1 a fourth consolidation, and 1 patient has started maintenance therapy. Alisertib has been well tolerated. All patients have experienced expected grade 4 toxicities of anemia, thrombocytopenia, and febrile neutropenia seen during the induction phase of treatment. At initial dose of 10mg BID, no dose-limited toxicities (DLTs) were encountered. In the 20mg BID cohort, one DLT was encountered, of delayed thrombocytopenia (G4 at day 40). The final cohort, at 30mg BID, is currently accruing (4 of 6 enrolled). No other DLTs have been detected, and other toxicities on study will be presented. The maximum tolerated dose (MTD) will be established upon completion of this cohort. To date, 11 of 12 (92%) evaluable patients have achieved remission following induction (8 cases of CR and 3 of CRp). During the course of the study, one patient has experienced relapse, and none have died. Thirteen of 14 evaluable patients had an ablated mid-induction marrow biopsy, with the remaining patient showing 6% blasts in a 20% cellular marrow at mid-induction. None of the patients have undergone re-induction with 5+2 at mid-induction. We anticipate that the study will be fully enrolled and closed to further accrual by the time of presentation. Alisertib, a novel aurora A kinase inhibitor, appears to be well tolerated in conjunction with standard induction chemotherapy in newly diagnosed AML. Currently, 13 of 14 treated patients have demonstrated marrow ablation at the mid-induction point of therapy, and none have required re-induction with 5+2. Eleven of 12 evaluable patients have achieved remission following induction. These results suggest that alisertib is well-tolerated and may hold promise as a therapeutic agent in AML, when combined with induction chemotherapy. Ongoing efforts seek to define the MTD and establish appropriate dosing for phase II studies. Disclosures Fathi: Millennium: Research Funding; Seattle Genetics: Advisory Board, Advisory Board Other; Agios: Advisory Board, Advisory Board Other. Attar:Agios: Employment.


Oncotarget ◽  
2015 ◽  
Vol 6 (10) ◽  
pp. 8062-8070 ◽  
Author(s):  
Marco Colamonici ◽  
Gavin Blyth ◽  
Diana Saleiro ◽  
Amy Szilard ◽  
Meghan Bliss-Moreau ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document