A Novel Aurora-a Kinase Inhibitor MLN8237 Induces Cytotoxicity and Cell Cycle Arrest in Experimental Multiple Myeloma Models.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1719-1719 ◽  
Author(s):  
Gullu Gorgun ◽  
Elisabetta Calabrese ◽  
Mala Mani ◽  
Teru Hideshima ◽  
Hiroshi Ikeda ◽  
...  

Abstract Multiple myeloma (MM) is an incurable bone marrow derived plasma cell malignancy. Despite significant improvements in treating patients suffering from this disease, MM remains uniformly fatal owing to intrinsic or acquired drug resistance. Thus, additional modalities for treating MM are required. In this study, we examined the anti-tumor activity of MLN8237, a small molecule Aurora-A kinase inhibitor, in experimental models of MM. Aurora-A is a mitotic kinase that localizes to centrosomes and the proximal mitotic spindle and functions in mitotic spindle formation and in regulating chromatid congression and segregation. Aurora-A gene amplification and protein overexpression is a common event in many cancers, and has been experimentally linked to genetic instability and tumorigenesis. In MM, increased Aurora-A gene expression has previously been correlated with centrosome amplification and a worsened disease prognosis. Thus, inhibition of Aurora A in MM may prove to be therapeutically beneficial. Here we show that Aurora-A protein is highly expressed in eight distinct MM cell lines. The affect of Aurora-A inhibition in these cell lines was examined in cytotoxicity (MTT viability) and proliferation (3[H]thymidine incorporation) assays by treating with MLN8237 (0.25 mM −32 mM) for 24, 48 and 72h. Although there was no significant inhibition of cell viability and proliferation at 24h, a marked effect occurred 48 and 72h after compound addition at concentrations as low as 0.25 mM. Interestingly, the melphalan resistant line (LR5) and Doxorubucin resistant line (Dox40) were among the least sensitive to MLN8237 induced cell cytotoxicity. The affect of MLN8237 on peripheral blood mononuclear cells (PBMCs) from healthy donors was also examined at the same concentrations and exposure time used for the MM cell lines. In healthy PBMCs, MLN8237 did not induce cytotoxicity as measured by the MTT assay, but there was a significant inhibition of proliferation at 48 and 72h as measured by the 3[H]thymidine incorporation assay at concentrations above 4uM. To delineate the mechanisms of cytotoxicity and growth inhibitory activity of MLN8237, apoptotic markers and cell cycle profiles were examined in the MM cell lines. Fluorescence conjugated-Annexin V and propidium iodide (PI) co-staining of MM cell lines after culturing in the presence or absence of MLN8237 at 1 mM (IC50) for 24, 48 and 72h demonstrated that MLN8237 induces apoptosis in these lines. This finding was corroborated by demonstrating increased capase-9 expression by Western blot analysis. Cell cycle analysis by flow cytometry demonstrated that MLN8237 results in an accumulation of tetraploid cells, presumably by abrogating G2/M progression. These results suggest that MLN8237 represents a possible novel agent for treating MM patients. Additional studies are ongoing to assess the anti-tumor effects of MLN8237 alone and in combination with other therapeutic agents in xenograft models of MM.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3830-3830
Author(s):  
Gullu Gorgun ◽  
Elisabetta Calabrese ◽  
Teru Hideshima ◽  
Jeffrey Ecsedy ◽  
Giada Bianchi ◽  
...  

Abstract Abstract 3830 Poster Board III-766 Multiple myeloma (MM) is an incurable bone marrow derived plasma cell malignancy. Despite significant improvements in treating patients suffering from this disease, MM remains uniformly fatal due to intrinsic or acquired drug resistance. Thus, additional modalities for treating MM are required. Targeting cell cycle progression proteins provides such a novel treatment strategy. Here we assess the in vivo and in vitro anti-MM activity of MLN8237, a small molecule Aurora A kinase (AURKA) inhibitor. AURKA is a mitotic kinase that localizes to centrosomes and the proximal mitotic spindle, where it functions in mitotic spindle formation and in regulating chromatid congression and segregation. In MM, increased AURKA gene expression has been correlated with centrosome amplification and a worse prognosis; thus, inhibition of AURKA in MM may prove to be therapeutically beneficial. Here we show that AURKA protein is highly expressed in eight MM cell lines and primary patient MM cells. The affect of AURKA inhibition was examined using cytotoxicity (MTT viability) and proliferation (3[H]thymidine incorporation) assays after treatment of these cell lines and primary cells with MLN8237 (0.0001 μM – 4 μM) for 24, 48 and 72h Although there was no significant inhibition of cell viability and proliferation at 24h, a marked effect on both viability and proliferation occurred after 48 and 72h treatment at concentrations as low as 0.01 μM. Moreover, MLN8237 inhibits cell growth and proliferation of primary MM cells and cell lines even in the presence of bone marrow stromal cells (BMSCs) or cytokines IL-6 and IGF1. Similar experiments revealed that MLN8237 did not induce cytotoxicity in normal peripheral blood mononuclear cells (PBMCs) as measured by MTT assay, but did inhibit proliferation at 48 and 72h, as measured by the 3[H]thymidine incorporation assay. To delineate the mechanisms of cytotoxicity and growth inhibitory activity of MLN8237, apoptotic markers and cell cycle profiles were examined in both MM cell lines and primary MM cells. Annexin V and propidium iodide staining of MM cell lines cultured in the presence or absence of MLN8237 (1 μM) for 24, 48 and 72h demonstrated apoptosis, which was further confirmed by increased cleavage of PARP, capase-9, and caspase-3 by immunoblotting. In addition, MLN8237 upregulated p53-phospho (Ser 15) and tumor suppressor genes p21 and p27. Cell cycle analysis demonstrated that MLN8237 treatment induces an accumulation of tetraploid cells by abrogating G2/M progression. We next determined whether combining MLN8237 with conventional (melphalan, doxorubucin, dexamethasone) and other novel (VELCADE®) therapeutic agents elicited synergistic/additive anti-MM activity by isobologram analysis using CalcuSyn software. Combining MLN8237 with melphalan, dexamethasone, or VELCADE® induces synergistic/additive anti-MM activity against MM cell lines in vitro (p≤0.05, CI<1). To confirm in vivo anti-MM effects of MLN8237, MM.1S cells were injected s.c. into g-irradiated CB-17 SCID mice (n=40, 10 mice EA group). When tumors were measurable (>100 mm3), mice were treated with daily oral doses of vehicle alone or 7.5mg/kg, 15mg/kg, 30mg/kg MLN8237 for 21 days. Overall survival (defined as time between initiation of treatment and sacrifice or death) was compared in vehicle versus- MLN8237- treated mice by Kaplan-Meier method. Tumor burden was significantly reduced (p=0.02) and overall survival was significantly increased (p=0.02, log-rank test) in animals treated with 30mg/kg MLN8237. In vivo anti-MM effects of MLN8237 were further validated by performing TUNEL apoptosis-cell death assay in tumor tissues excised from control or treated animals. Importantly, a significant dose-related increase in apoptotic cells was observed in tumors from animals that received MLN8237 versus controls. These results suggest that MLN8237 represents a promising novel targeted therapy in MM. Disclosures: Ecsedy: Millennium Pharmaceutical: Employment. Munshi:Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees. Richardson:Celgene: Membership on an entity's Board of Directors or advisory committees; Millennium: Membership on an entity's Board of Directors or advisory committees. Anderson:Millennium: Research Funding; Novartis: Research Funding; Celgene: Research Funding.


Blood ◽  
2010 ◽  
Vol 115 (25) ◽  
pp. 5202-5213 ◽  
Author(s):  
Güllü Görgün ◽  
Elisabetta Calabrese ◽  
Teru Hideshima ◽  
Jeffrey Ecsedy ◽  
Giulia Perrone ◽  
...  

Abstract Aurora-A is a mitotic kinase that regulates mitotic spindle formation and segregation. In multiple myeloma (MM), high Aurora-A gene expression has been correlated with centrosome amplification and proliferation; thus, inhibition of Aurora-A in MM may prove to be therapeutically beneficial. Here we assess the in vitro and in vivo anti-MM activity of MLN8237, a small-molecule Aurora-A kinase inhibitor. Treatment of cultured MM cells with MLN8237 results in mitotic spindle abnormalities, mitotic accumulation, as well as inhibition of cell proliferation through apoptosis and senescence. In addition, MLN8237 up-regulates p53 and tumor suppressor genes p21 and p27. Combining MLN8237 with dexamethasone, doxorubicin, or bortezomib induces synergistic/additive anti-MM activity in vitro. In vivo anti-MM activity of MLN8237 was confirmed using a xenograft-murine model of human-MM. Tumor burden was significantly reduced (P = .007) and overall survival was significantly increased (P < .005) in animals treated with 30 mg/kg MLN8237 for 21 days. Induction of apoptosis and cell death by MLN8237 were confirmed in tumor cells excised from treated animals by TdT-mediated dUTP nick end labeling assay. MLN8237 is currently in phase 1 and phase 2 clinical trials in patients with advanced malignancies, and our preclinical results suggest that MLN8237 may be a promising novel targeted therapy in MM.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1551-1551
Author(s):  
Steven I. Park ◽  
Carolina P. Lin ◽  
Dirk P. Dittmer ◽  
Steven P. Angus ◽  
Greg G. Wang ◽  
...  

Abstract Background: The proto-oncogene Myc is a key regulator of cell growth and survival, and aberrant Myc expression plays a significant role in various tumors, including non-Hodgkin lymphoma (NHL). Myc-associated lymphoma is clinically aggressive, more resistant to standard therapies, and associated with a significantly higher rate of mortality. Novel treatment paradigms are needed to improve survival of patients with Myc-associated NHL. Expression of Aurora Kinase (Aurk) has been associated with Myc, and Aurk is thought to be essential for the maintenance of Myc-driven lymphoma. Aurk is required for assembly of the mitotic spindle and plays key roles in cell proliferation. Amplification and overexpression of Aurk have been observed in various human tumors, including lymphoma, and are frequently associated with tumor progression as well as resistance to chemotherapy. Inhibition of Aurk may overcome resistance to chemotherapy and improve clinical outcomes in patients with Myc-overexpressing lymphoma. Methods: Cytotoxicity assays using MTS and trypan blue were used to compare levels of drug sensitivity in lymphoma cell lines resistant or sensitive to a conventional chemotherapeutic drug cyclophosphamide. Apoptosis and cell cycle assays were performed using Annexin V and Propidium Iodide staining. The Multiplexed Inhibitor Beads and quantitative Mass Spectrometry (MIB/MS) assays were used to profile kinome changes in response to Aurk inhibition. Murine xenograft models were used to assess the efficacy and tolerability of single vs. combined therapy. Results: Two Myc-overexpressing cell lines were identified as resistant (Raji) or sensitive (Ramos) to cyclophosphamide, with IC50 of ~ 400 µM and ~ 250 µM, respectively. Raji cells were characterized by increased expression of multidrug resistant protein 1 (MDR1) and mutated p53. There were no significant differences in baseline Aurk or Myc expressions between Raji and Ramos cells. Both cell lines were sensitive to alisertib, an aurora A kinase inhibitor, with maximum cytotoxicity achieved at ~ 100 nM. Combined treatment with alisertib and cyclophosphamide induced more significant cell growth inhibition as compared to treatment with the single agent alone. The combination index (CI) values were less than 1, indicating that alisertib was synergistic to cyclophosphamide in terms of inhibitory effect on tumor cell viability. Alisertib induced apoptosis and pronounced cell cycle arrest, resulting in polyploidy, in Raji cells. Alisertib had little to no effect on Myc, p53, or the total aurora A kinase protein expression in Raji cells although p-Histone-3-Ser10, a downstream target of Aurk, and p-Src levels were significantly decreased at 24 hours of treatment in vitro. Nocodazole-treated cells had reduced p-Aurk level and increased p-Rb as well as increased Mdm2 when treated with alisertib for 24 hours. Athymic nude mice bearing Ramos or Raji lymphoma xenografts were treated with cyclophosphamide, alisertib, or the combination. As expected, all mice bearing Ramos xenograft had complete tumor regression by day 35 of treatment while all mice bearing Raji xenograft had rapid disease progression with median survival of ~ 35 days when treated with cyclophosphamide alone. In contrast, when treated with the combination of cyclophosphamide and alisertib, all mice bearing Raji xenograft had complete regression of tumor by day 35 and had significant improvement in survival (median survival not reached by day 100) compared to the single agent control (p=0.022). Lastly, kinome analysis of Raji xenograft tumors treated with alisertib showed suppression of various kinases involved in Aurk, Src, and PI3K pathways. Western blot of the Raji tumors treated with a prolonged course (25 days) of alisertib showed significant decrease in p-Src and p53 protein levels. Conclusion: Our data demonstrates that alisertib induces synthetic lethality and overcomes chemoresistance in Myc-overexpressing tumors even in the presence of MDR1 overexpression and p53 mutation. The synergistic effect was largely independent of depletion of cytoplasmic level of Myc. Alisertib, when combined with a conventional chemotherapy drug, induced apoptosis and cell cycle arrest of Myc-overexpressing tumor cells in vitro and showed promising anti-tumor activity in mice bearing chemoresistant Myc-overexpressing lymphoma. Disclosures Park: Janssen: Other: travel; Seattle Genetics: Research Funding; Teva: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2848-2848
Author(s):  
Enrico Derenzini ◽  
Daniela Buglio ◽  
Hiroshi Katayama ◽  
Yuan Ji ◽  
Subrata Sen ◽  
...  

Abstract Abstract 2848 Hodgkin Lymphoma (HL) cell proliferation and survival is sustained by a complex network of cytokine signaling, involving the Hodgkin and Reed-Sternberg cells and tumor microenvironment. Following cytokine stimulation, JAK-STAT activation promotes the transcription of target genes involved in proliferation, survival, and immune escape. Programmed Death-ligands 1 and 2 (PD-L1 and PD-L2) and the Th2 chemokine TARC are immune-modulators involved in immune evasion, respectively through inhibition of effector T cell function (PD-L1, PD-L2) and attraction and homing of Th2 cells (TARC). Aurora kinases are frequently overexpressed in human cancers and play essential functions in chromosome alignment and cytokinesis. The role of Aurora kinases in Hodgkin lymphomagenesis is not defined yet. In this study we report the activity profile of the JAK2 inhibitor AZD1480 in HL cell lines (HD-LM2, L-428, KM-H2, L-540). To assess the effect of AZD1480 on cell proliferation, cells were incubated with increasing concentrations of AZD1480 (from 0.1 to 10 μM) for 24, 48 and 72 hours (hrs). A significant growth inhibition was evident after 72 hrs of incubation, specially using the high doses of AZD1480 (5μM). The L-540 cell line showed the highest sensitivity, with a decrease in cell viability close to 50% following incubation with AZD1480 1μM. Inhibition of STAT3, STAT5 and STAT6 phosphorylation in the L-540, L-428 and HD-LM2 cell lines was observed with concentrations equal to 0.1 μM or higher. Using Annexin V- propidium iodide staining, we found that AZD1480 induced cell death by apoptosis in a dose dependent manner after 72 hrs of incubation when a high concentration (5μM) of the drug was used. Lower concentrations of AZD1480 (1μM) promoted a statistically significant increase in cell death only in the L-540 and to a lesser extent in the L-428 cell line. Consistent with this data, also caspase 9, 3 and PARP cleavage was observed in all the cell lines exposed to AZD1480 5 μM. AZD1480 5μM promoted a marked increase in the G2/M fraction in all the cell lines as soon as 24 hrs after incubation, especially in the HD-LM2 and L-428 cell lines. Treatment with lower doses (1μM) did not affect significantly the cell cycle. Since AZD1480 was also reported to inhibit Aurora A kinase at nanomolar concentrations in enzymatic assays, we assessed if the significant increase in the G2/M fraction was related to the inhibition of the Aurora A kinase. We evaluated the levels of autophosphorylation on Thr-288 by western blotting. Cells were pretreated with Nocodazole 400 ng/ml for 18 hrs in order to achieve a mitotic block, and then exposed to AZD1480 (1-5μM) and/or the proteasome inhibitor MG132 (20μM) (in order to prevent the potential overriding of the Nocodazole induced mitotic block), for 3 hours. A dose-dependent inhibition of Aurora A was detected in all the cell lines, with a complete abrogation when higher doses of AZD1480 were used (5μM). These findings are consistent with the analysis of the cell cycle fractions, showing dose-dependent changes of the cell cycle at 24 hrs following incubation with AZD1480. AZD1480 also decreased the secretion of key cytokines involved autocrine and paracrine survival loops and immune escape. Following incubation with AZD1480 1μM for 72 hrs cell culture supernatants were analyzed by ELISA: decreased levels of IL-6, IL-13, TARC, and IL-21 were observed in HD-LM2, L-428 and L-540 cells. Moreover we assessed the expression of PD-L1 and PD-L2 by flow cytometry and observed significant downregulation in the PD-L1/PD-L2 overexpressing cell lines (L-540 and HD-LM2). These data suggest that AZD1480 has a pleiotropic mechanism of action in HL by targeting the JAK-STAT and the Aurora kinase pathway, and by altering the pattern of cytokine and chemokine secretion and the expression of factors involved in immune escape. Our study provides the rationale for further clinical investigation of AZD1480 in HL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 248-248
Author(s):  
Dirk Hose ◽  
Thierry Rème ◽  
Tobias Meißner ◽  
Jérôme Moreaux ◽  
Anja Seckinger ◽  
...  

Abstract BACKGROUND. At the time of diagnosis, myeloma cells are characterized by a low proliferation rate that increases in relapse. Presence of proliferation correlates with adverse prognosis. At the same time, myeloma cells harbor a high median number of chromosomal aberrations, often associated with genetic instability. Cellular proliferation and genetic instability in turn have been associated with Aurora-kinase expression in several cancer entities, including multiple myeloma. PATIENTS AND METHODS. Expression of Aurora-A, -B and -C was assessed using Affymetrix DNA-microarrays in 784 samples including two independent sets of 233 and 345 CD138-purified myeloma cells from previously untreated patients. Chromosomal aberrations were assessed by comprehensive iFISH using a set of probes for the chromosomal regions 1q21, 6q21, 8p21, 9q34, 11q23, 11q13, 13q14.3, 14q32, 15q22, 17p13, 19q13, 22q11, as well as the translocations t(4;14)(p16.3;q32.3) and t(11;14) (q13;q32.3). Proliferation of primary myeloma cells (n=67) was determined by propidium iodine staining. The effect of the clinical Aurora-kinase inhibitor VX680 on proliferation of 20 human myeloma cell lines and survival of 5 primary myeloma cell-samples was tested. RESULTS. We found Aurora-A and -B to be expressed at varying frequencies in primary myeloma cells of different patient-cohorts, including 23% for Aurora A in our first cohort of patients treated with high dose therapy (see figure shown below). Aurora-C expression was found in testis-samples only. Myeloma cell-samples with detectable Aurora-A expression show a significantly higher proliferation rate, whereas the number of chromosomal aberrations (aneuploidy) is not higher compared to myeloma-cells with absent Aurora-A expression. The same holds true for subclonal aberrations (i.e. genetic instability). The Aurora-kinase inhibitor VX680 induces apoptosis in all myeloma cell lines and primary myeloma cell-samples tested. Presence of Aurora-A expression delineates significantly inferior event-free and overall survival in two independent cohorts of patients undergoing high-dose chemotherapy and autologous stem cell transplantation. This observation is independent of conventional prognostic factors, i.e. serum-ß2-microglobulin or ISS-stage. CONCLUSION. Aurora-kinase inhibitors (including VX680 tested here) are very active on myeloma cell lines as well as primary myeloma cells and represent a promising weapon in the therapeutic arsenal against multiple myeloma. Gene expression profiling allows the assessment of Aurora-kinase expression and thus in turn a tailoring of treatment to patients expressing Aurora-A associated with adverse prognosis. Figure Figure


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3495-3495 ◽  
Author(s):  
Garrett O’Sullivan ◽  
Xavier Leleu ◽  
Xiaoying Jia ◽  
Evdoxia Hatjiharisi ◽  
Hai Ngo ◽  
...  

Abstract Background: The PI3K and NFk-B/proteasome pathways are major regulators of survival in Multiple Myeloma (MM). Previous studies have demonstrated clinical efficacy of bortezomib in MM; however, not all patients responded to this agent. mTOR inhibitors have demonstrated significant in vitro and in vivo activity in MM, specifically clinical trials with the mTOR inhibitor CCI-779 (Wyeth) in MM. Therefore, we examined whether inhibition of the PI3K pathway by mTOR and proteasome inhibitors may lead to synergistic activity in MM. Methods: MM cell lines (MM.1S, RPMI, U266, OPM2) were treated with rapamycin 1–5nM (Sigma Aldrich), bortezomib 2.5–10nM (Millenium, MA), or the combination. Cytotoxicity was measured by the MTT assay at 48 hrs; DNA synthesis was measured using thymidine uptake assay; apoptosis was studied using Apo2.7 by flow cytometry, and cell cycle regulation was determined using flow cytometry. To determine whether these agents can overcome the growth advantage conferred by bone marrow stromal cells (BMSCs), we co-cultured cell lines with stromal cells. Normal peripheral blood mononuclear cells (PBMCs) were obtained from healthy volunteers. Determination of the additive or synergistic effect of the combination was calculated using the CalcuSyn software (Biosoft, MO) based on the Chou-Talalay method, with synergistic activity determined as a combination index (CI) of <1.0. Results: Rapamycin induced dose-dependent cytotoxicity from 0.1nM to 1nM, with an IC50 of 5nM in MM.1S and OPM2. Interestingly, higher doses did not induce further cytotoxicity, confirming that low doses of rapamycin are as effective as higher doses. RPMI and U266 MM cell lines were less sensitive to rapamycin, with 5nM inducing 40% and 20% decrease in survival, respectively. Bortezomib induced significant inhibition of survival in all MM cell lines with an IC50 of 2.5nM, as previously reported. The combination of agents induced significant inhibition of proliferation as compared to each agent alone, specifically with the combination of 5nM rapamycin with 5nM of bortezomib. In the DNA synthesis assay, the combination of bortezomib and rapamycin was significantly cytotoxic compared to each agent alone, specifically at the dose of 5nM rapamycin and bortezomib 2.5nM. The combination of rapamycin 1 to 5 nM and bortezomib 5 to 10 nM were synergistic with a CI index less than 1.0, as in RPMI (CI=0.4) and U266 (CI=0.2) cell lines. The combination of rapamycin and bortezomib at serial concentrations did not trigger cytotoxicity in PBMCs from normal volunteers, indicating significant cytotoxicity in malignant cells, with lack of toxicity in normal PBMCs and suggesting a therapeutic index. The combination of bortezomib and rapamycin demonstrated a significant inhibitory effect on the growth of MM cell lines even in coculture with stromal cells. Cell cycle analysis demonstrated G1 arrest at 24 and 48 hrs in MM.1S cells. Similar results were obtained using primary CD138+ myeloma cells from patients. Conclusion: The combination of rapamycin and bortezomib resulted in synergistic in vitro cytotoxicity in MM cells. These results provide the framework for clinical trials evaluating the combination of CCI-779 and bortezomib in MM.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2087-2087
Author(s):  
Kevin R. Kelly ◽  
Ronan T. Swords ◽  
Devalingam Mahalingam ◽  
Steffan T. Nawrocki ◽  
Ernest Medina ◽  
...  

Abstract Abstract 2087 Poster Board II-64 Acute myeloid leukemia (AML) most frequently affects the elderly, many of whom are unable to tolerate intensive chemotherapy. Therefore, improvements in clinical outcomes for patients with AML largely depend upon the development of novel targeted therapies. Aurora A is a serine/threonine kinase that plays a key role in mitosis by regulating the G2-M transition, centrosome separation, spindle assembly and chromosome segregation. It is overexpressed in AML and has been implicated in genetic instability, disease progression and drug resistance. MLN8237 is a novel orally available Aurora A inhibitor that has entered Phase I and Phase II trials for cancer therapy. Based on the critical role of Aurora A in cell cycle regulation and its intrinsic overexpression in AML cells, we hypothesized that MLN8237 would possess significant antileukemic activity. We tested our hypothesis in established human AML cell lines, primary AML patient specimens, and a mouse model of AML. MLN8237 potently inhibited the in vitro growth and survival of all AML cell lines and primary human AML cells from patients with newly diagnosed AML and patients with relapsed, chemotherapy resistant disease. Mechanistic studies showed that MLN8237 reduced the activity of Aurora A kinase as evidenced by reduced phosphorylation of Aurora A at Thr288. MLN8237 treatment also disrupted normal cell cycle kinetics and induced apoptosis in a dose- and time-dependant manner characterized by the accumulation of G2/M and aneuploid cells prior to the onset of apoptosis. Exposure to MLN8237 led to activation of FOXO3a followed by a dose-dependent increase in the expression of its transcriptional target Bim. These events appear to be important mediators of the pro-apoptotic effects of this agent. We next investigated the ability of MLN8237 to increase the efficacy of cytarabine in AML. Treatment with the combination of MLN8237 and cytarabine resulted in significantly greater apoptosis and more effective inhibition of survival than treatment with either agent alone in AML cell lines and primary patient specimens. MLN8237 and cytarabine cooperated to enhance mitochondrial-mediated apoptosis as evidenced by increased processing of caspases-9 and -3 to active forms. Daily oral administration of MLN8237 to immunodeficient mice bearing AML xenografts was well tolerated, effectively reduced tumor growth and led to the disruption of mitotic spindles and centrosome amplification. Moreover, MLN8237 significantly enhanced the activity of cytarabine to achieve tumor regression. Our data demonstrates that MLN8237 has a multifaceted mechanism of action comprised of pro-apoptotic and growth inhibitory effects. Our collective findings indicate that the combination of MLN8237 and cytarabine represents a novel, very promising therapeutic strategy for AML. A clinical study investigating the safety and efficacy of this combination in patients with refractory AML is planned. Disclosures: Ecsedy: Millennium Pharmaceuticals: Employment.


2012 ◽  
Vol 198 (4) ◽  
pp. 591-605 ◽  
Author(s):  
Amy B. Foraker ◽  
Stéphane M. Camus ◽  
Timothy M. Evans ◽  
Sophia R. Majeed ◽  
Chih-Ying Chen ◽  
...  

Clathrin depletion by ribonucleic acid interference (RNAi) impairs mitotic spindle stability and cytokinesis. Depletion of several clathrin-associated proteins affects centrosome integrity, suggesting a further cell cycle function for clathrin. In this paper, we report that RNAi depletion of CHC17 (clathrin heavy chain 17) clathrin, but not the CHC22 clathrin isoform, induced centrosome amplification and multipolar spindles. To stage clathrin function within the cell cycle, a cell line expressing SNAP-tagged clathrin light chains was generated. Acute clathrin inactivation by chemical dimerization of the SNAP-tag during S phase caused reduction of both clathrin and ch-TOG (colonic, hepatic tumor overexpressed gene) at metaphase centrosomes, which became fragmented. This was phenocopied by treatment with Aurora A kinase inhibitor, suggesting a centrosomal role for the Aurora A–dependent complex of clathrin, ch-TOG, and TACC3 (transforming acidic coiled-coil protein 3). Clathrin inactivation in S phase also reduced total cellular levels of ch-TOG by metaphase. Live-cell imaging showed dynamic clathrin recruitment during centrosome maturation. Therefore, we propose that clathrin promotes centrosome maturation by stabilizing the microtubule-binding protein ch-TOG, defining a novel role for the clathrin–ch-TOG–TACC3 complex.


2021 ◽  
Vol 14 (7) ◽  
pp. 682
Author(s):  
Jianling Bi ◽  
Garima Dixit ◽  
Yuping Zhang ◽  
Eric J. Devor ◽  
Haley A. Losh ◽  
...  

Angiogenesis plays a crucial role in tumor development and metastasis. Both bevacizumab and cediranib have demonstrated activity as single anti-angiogenic agents in endometrial cancer, though subsequent studies of bevacizumab combined with chemotherapy failed to improve outcomes compared to chemotherapy alone. Our objective was to compare the efficacy of cediranib and bevacizumab in endometrial cancer models. The cellular effects of bevacizumab and cediranib were examined in endometrial cancer cell lines using extracellular signal-related kinase (ERK) phosphorylation, ligand shedding, cell viability, and cell cycle progression as readouts. Cellular viability was also tested in eight patient-derived organoid models of endometrial cancer. Finally, we performed a phosphoproteomic array of 875 phosphoproteins to define the signaling changes related to bevacizumab versus cediranib. Cediranib but not bevacizumab blocked ligand-mediated ERK activation in endometrial cancer cells. In both cell lines and patient-derived organoids, neither bevacizumab nor cediranib alone had a notable effect on cell viability. Cediranib but not bevacizumab promoted marked cell death when combined with chemotherapy. Cell cycle analysis demonstrated an accumulation in mitosis after treatment with cediranib + chemotherapy, consistent with the abrogation of the G2/M checkpoint and subsequent mitotic catastrophe. Molecular analysis of key controllers of the G2/M cell cycle checkpoint confirmed its abrogation. Phosphoproteomic analysis revealed that bevacizumab and cediranib had both similar and unique effects on cell signaling that underlie their shared versus individual actions as anti-angiogenic agents. An anti-angiogenic tyrosine kinase inhibitor such as cediranib has the potential to be superior to bevacizumab in combination with chemotherapy.


Sign in / Sign up

Export Citation Format

Share Document