Anti-Myeloma Activity of the Small-Molecule Aurora Kinase Inhibitor VE465.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3468-3468 ◽  
Author(s):  
Joseph M. Negri ◽  
Douglas W. McMillin ◽  
Nicholas Mitsiades ◽  
Teru Hideshima ◽  
Dharminder Chauhan ◽  
...  

Abstract Multiple Myeloma (MM) remains an incurable plasma cell neoplasia, despite recent additions in the therapeutic arsenal for its management. Aurora kinases play integral roles in the orchestration of chromosomes and cytoskeletal mobility during the process of cell division. Aurora kinase activity has been implicated in several tumor types, including ovarian, colon, and prostate cancers. To determine whether inhibition of Aurora kinase activity could attenuate myeloma cell survival, we performed studies of the Aurora kinase inhibitor VE465 (Vertex Pharmaceuticals / Merck & Co., Inc.). VE465 inhibits all 3 Aurora isoforms (Aur A, B and C) with approximate Ki values of 1, 26, and 8.7 nM respectively. MTT colormetric survival assays (72–96hrs exposure) showed that VE465 is active against a wide panel of human MM cell lines: 26 of 38 MM cell lines had IC50 values at or < 100 nM, which are significantly lower than IC50 values for normal hematopoietic cells, e.g. unstimulated or PHA-stimatuled PBMCs. Importantly, VE465 was active in vitro against MM cell lines and/or primary MM tumor cells resistant to various anti-MM therapeutics, including dexamethasone, alkylating agents, anthracyclines, the proteasome inhibitor bortezomib, and/or immunomodulatory thalidomide derivatives (IMiDs). Moreover, VE465 maintained its activity despite the presence of protective bone marrow-derived cytokines (e.g. IL-6). PI cell cycle analyses showed that VE465 causes (even within 8 hrs of treatment) caused pronounced G2 arrest, followed by significant shift of MM cells to sub-G1 gate, consistent with cell death. Immunoblotting analyses confirmed that VE465 treatment induces cleavage of PARP, as well as cleavage of caspases-8 and -9, without significant changes in the expression levels of several key molecular effectors (e.g. Mcl-1, Bax, p53, hsp70, hsp90, hs27) which have been previously implicated in the mechanism of anti-MM activity of diverse other therapeutics. Screening of VE465-based combination regimens with other anti-MM agents showed additive effects of VE465 with the histone deacetylase inhibitor Vorinostat (SAHA) (Merck & Co., Inc). Ongoing studies in our Center are addressing the identification of specific molecular markers correlating with the degree of sensitivity of MM cells to VE465. Our in vitro evidence for induction of MM cell death and therapeutic window for the anti-MM effect of VE465, its ability to overcome protective effect of BM-derived cytokines, and the clearly distinct pattern of molecular sequelae of VE465 compared to several other agents in our current anti-MM therapeutic armamentarium, all suggest that Aurora kinase inhibition represents an intriguing novel targeted treatment strategy in MM. Importantly, these studies, particularly the identification of a sizeable subset of MM cell lines with higher sensitivity to VE465 than normal cells, provide the framework for in vivo VE465 studies in progress, alone and in combination with other anti-MM agents, to inform the design of potential clinical trials of this class of agents for MM.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 649-649 ◽  
Author(s):  
Douglas W. McMillin ◽  
Joseph M. Negri ◽  
Jake Delmore ◽  
Melissa Ooi ◽  
Patrick Hayden ◽  
...  

Abstract Context: Akt plays a pivotal role in the survival and proliferation of multiple myeloma (MM) cells and functions as a central link between upstream signaling pathways, such as growth factor receptors (e.g. IL-6/IL-6R, IGF/IGF-1R) and the kinase PI-3K with their downstream signaling effectors, such as the multifunctional mTOR-p70S6K cascade. The pivotal role of Akt in these proliferative/anti-apoptotic cascades for MM has provided the impetus for development of small molecule inhibitors against the kinase activity of Akt. We show that the dual Akt/p70S6K kinase inhibitor EXEL-6075 has potent anti-MM activity in preclinical models. Methods/Results: We tested a panel of 15 human MM cell lines for in vitro response to EXEL-6075 using MTT colorimetric survival assays, which showed that the majority of MM cells responded to EXEL-6075 with IC50 values <0.5 μM, including a subset of 6/15 MM cell lines with IC50 values in the range of 0.1 μM. Importantly, <16 hrs of exposure to 0.5 μM of EXEL-6075 was sufficient to commit these EXEL-6075-sensitive MM cell lines to cell death. Peripheral blood mononuclear cells (PBMCs) remain insensitive to the drug up to 5 μM, consistent with a 1-log therapeutic window compared to the aforementioned sensitive MM cell lines. Other non-malignant tissue, such as bone marrow stromal cells (BMSCs) and immortalized human hepatocyte cells were even less sensitive to EXEL-6075 (IC50>5 μM), further underscoring its selectivity towards neoplastic cells. EXEL-6075-responsive cells included MM cells resistant to diverse conventional and novel agents, such as dexamethasone, bortezomib, and immunomodulatory thalidomide derivatives, suggesting that dual Akt/p70S6K inhibition may overcome mechanisms of constitutive resistance to other currently available therapeutics. EXEL-6075 was also able to overcome the protective effects of IGF-1 and IL-6 on MM.1S cells. In addition, there was modest, if any, protection conferred to MM cells by BMSCs at low drug doses. Interestingly, when we compared the in vitro activity of EXEL-6075 against cell lines from MM vs. other neoplasias (including 11 epithelial cancer cell lines which also exhibited dose-dependent response to this kinase inhibitor), EXEL-6075 was significantly more active against MM cells, as evidenced by the distribution of IC50 values and AUCs of the dose-response curves for each group of cell lines (p=0.028, t-test). Ongoing studies are utilizing gene expression profiles of MM cells highly- vs. moderately-responsive to EXEL-6075, in order to identify putative markers of sensitivity vs. resistance to this kinase inhibitor Conclusion: Targeting the Akt-mTOR-p70S6K pathway is an attractive strategy for the treatment of MM due to the central role Akt plays in proliferation and survival of MM. Inhibition of Akt using the small molecule inhibitor EXEL-6075 resulted in sub-μM killing of MM cell lines with > 1-log differential activity against the non-neoplastic tissues tested in our study. This remains an interesting strategy for the treatment of MM. Future in vivo studies will be needed to confirm these interesting in vitro results.


Blood ◽  
2010 ◽  
Vol 116 (9) ◽  
pp. 1498-1505 ◽  
Author(s):  
Jürgen den Hollander ◽  
Sara Rimpi ◽  
Joanne R. Doherty ◽  
Martina Rudelius ◽  
Andreas Buck ◽  
...  

Myc oncoproteins promote continuous cell growth, in part by controlling the transcription of key cell cycle regulators. Here, we report that c-Myc regulates the expression of Aurora A and B kinases (Aurka and Aurkb), and that Aurka and Aurkb transcripts and protein levels are highly elevated in Myc-driven B-cell lymphomas in both mice and humans. The induction of Aurka by Myc is transcriptional and is directly mediated via E-boxes, whereas Aurkb is regulated indirectly. Blocking Aurka/b kinase activity with a selective Aurora kinase inhibitor triggers transient mitotic arrest, polyploidization, and apoptosis of Myc-induced lymphomas. These phenotypes are selectively bypassed by a kinase inhibitor-resistant Aurkb mutant, demonstrating that Aurkb is the primary therapeutic target in the context of Myc. Importantly, apoptosis provoked by Aurk inhibition was p53 independent, suggesting that Aurka/Aurkb inhibitors will show efficacy in treating primary or relapsed malignancies having Myc involvement and/or loss of p53 function.


Cancers ◽  
2020 ◽  
Vol 12 (3) ◽  
pp. 583 ◽  
Author(s):  
Jean Camille Mattei ◽  
Corinne Bouvier-Labit ◽  
Doriane Barets ◽  
Nicolas Macagno ◽  
Mathieu Chocry ◽  
...  

Soft tissue sarcoma (STS) are rare and aggressive tumours. Their classification includes numerous histological subtypes of frequent poor prognosis. Liposarcomas (LPS) are the most frequent type among them, and the aggressiveness and deep localization of dedifferentiated LPS are linked to high levels of recurrence. Current treatments available today lead to five-year overall survival has remained stuck around 60–70% for the past three decades. Here, we highlight a correlation between Aurora kinasa A (AURKA) and AURKB mRNA overexpression and a low metastasis-free survival. AURKA and AURKB expression analysis at genomic and protein level on a 9-STS cell lines panel highlighted STS heterogeneity, especially in LPS subtype. AURKA and AURKB inhibition by RNAi and drug targeting with AMG 900, a pan Aurora Kinase inhibitor, in four LPS cell lines reduces cell survival and clonogenic proliferation, inducing apoptosis and polyploidy. When combined with doxorubicin, the standard treatment in STS, aurora kinases inhibitor can be considered as an enhancer of standard treatment or as an independent drug. Kinome analysis suggested its effect was linked to the inhibition of the MAP-kinase pathway, with differential drug resistance profiles depending on molecular characteristics of the tumor. Aurora Kinase inhibition by AMG 900 could be a promising therapy in STS.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e14105-e14105
Author(s):  
Janaki Parameswaran ◽  
Jong Woo Lee ◽  
Teresa Sandoval-Schaefer ◽  
Jaseok P. Koo ◽  
Barbara Burtness

e14105 Background: Aurora Kinase A (AURKA) is overexpressed in HNSCC, and correlates with poor prognosis. It has been identified as a potential therapeutic target, yet the response rate for the AURKA inhibitor MLN8237 is only 9% in treatment refractory HNSCC. We hypothesized that although AURKA inhibitors lead to defective spindle assembly, they may also reduce mitotic entry, undermining cytotoxic effect. We predicted that adding a WEE1 inhibitor to an AURKA inhibitor would mitigate this effect and enhance cell death Methods: Cell viability assays were performed on FaDu ( p53 mut.), Detroit562 ( p53 mut.), and UNC7 ( p53 WT) HPV negative HNSCC cell lines treated with AZD1775 (AZD), MLN8237 (MLN), and combination of AZD+MLN. Oncosphere formation assays were used to confirm findings of cell death, and western blot analysis and confocal microscopy were used to investigate mechanism of synergy. The above drugs were also given at varying doses via oral gavage to FaDu xenografted nude mice. Results: There was clear synergy of AZD and MLN in-vitro. Combination Indices were determined by the Chou-Talalay method: FaDu 0.4, Detroit562 0.5, and UNC7 0.6 (synergy = < 0.8). Oncopshere assays showed inability of AZD+MLN treated cells to re-differentiate. FaDu cells treated with MLN had increased p-CDK1 and reduced phospho-histone H3 (pHH3), suggesting reduced mitotic entry. AZD+MLN treated cells had reduced p-CDK1 and increased pHH3, similar to AZD treated cells; they also had spindle disarray with poor chromatin organization on confocal microscopy indicating mitotic catastrophe. In mice, the combination of AZD+MLN inhibited tumor growth, with no apparent toxicity. Mice treated with either drug alone had tumor volumes over 1000mm3 and were sacrificed at day 21; those treated with AZD 90mg/kg and MLN 30mg/kg had tumors volumes around 300mm3 on day 28. Conclusions: AZD and MLN synergistically enhance cell death in HNSCC cell lines and significantly inhibit tumor growth in mouse xenograft models. The ability of AZD to overcome intrinsic resistance to MLN may underlie mechanism of synergy. We recommend further investigation of AURKA and WEE1 inhibition in other cancers with high AURKA expression, and in patients with HNSCC.


2010 ◽  
Vol 8 (5) ◽  
pp. 72
Author(s):  
L. Vidarsdottir ◽  
A.M. Halldorsdottir ◽  
G. Steingrimsdottir ◽  
S.K. Bodvarsdottir ◽  
H.M. Ogmundsdottir ◽  
...  

Author(s):  
Tuğçe Balcı Okcanoğlu ◽  
Çağla Kayabaşı ◽  
Cumhur Gündüz

Long non-coding RNAs (lncRNAs) are involved in a range of biological processes, such as cellular differentiation, migration, apoptosis, invasion, proliferation, and transcriptional regulation. The aberrant expression of lncRNAs plays a significant role in several cancer types. Aurora kinases are increasingly expressed in various malignancies; accordingly, the inhibition of these enzymes may represent a novel approach for the treatment of various cancers. CCT137690, an Aurora kinase inhibitor, displays an anti-proliferative activity in human cancer cell lines. The aim of the present study was to investigate the anti-proliferative and cytotoxic effects of CCT137690 on estrogen receptor (ER)-positive human breast cancer cell line (MCF-7) and ER-negative human breast cancer cell line (MDA-MB-231). In addition, this study was targeted toward determining the changes induced in lncRNA expression levels following the initiation of Aurora kinase inhibitor treatment. The cytotoxic effects of CCT137690 were determined by means of the xCELLigence system. Furthermore, the anti-proliferative role of CCT137690 in breast cancer was investigated by checking the changes in lncRNA expression profiles using quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The half-maximal inhibitory concentrations (IC50) of CCT137690 were determined as 4.5 μM (MCF-7) and 7.27 μM (MDA-MB-231). Several oncogenic lncRNAs (e.g., PRINS, HOXA1AS, and NCRMS) were downregulated in both ER-negative and ER-positive cell lines. On the other hand, tumor suppressor lncRNAs (e.g., DGCR5 and IGF2AS) were upregulated in the ER-positive cell line. After CCT137690 treatment, HOXA11AS and PCAT-14 lncRNAs were downregulated in the ER-positive cell lines. In addition, MER11C, SCA8, BC200, HOTAIR, PCAT-1, UCA1, SOX2OT, and HULC lncRNAs were downregulated in the ER-negative cell lines. The results of the present study indicated that Aurora kinase inhibitor CCT137690 could be a potential anti-cancer agent for breast cancer treatment.


2020 ◽  
Author(s):  
Khairul Ansari ◽  
Arunoday Bhan ◽  
Mike Chen ◽  
Rahul Jandial

Abstract Leptomeningeal carcinomatosis (LC), when tumor cells spread to leptomeninges surrounding the brain and spinal cord. HER2+ breast cancer is the most common origin of LC. HER2+ LC remains incurable, with few treatment options and response rates of <20%. One major limitation in development of HER2+ LC therapies has been lack of clinically relevant HER2+ LC primary cell-lines and animal models. To address this, we generated cell lines and patient-derived xenograft models using nodular HER2+ LC. This led to identification of granulocyte-macrophage colony-stimulating factor (GM-CSF) as an oncogenic autocrine driver of HER2+ LC. We observed that oligodendrocyte progenitor cells (OPCs) inhibit growth of HER2+ LC in vitro and in vivo. Furthermore, OPC-derived factor TPP1 degrades GM-CSF, decreasing GM-CSF signaling and suppressing HER2+ LC growth. Lastly, we determined that synergistic inactivation of GM-CSF signaling via the intrathecal delivery neutralizing anti-GM-CSF antibodies and a pan-Aurora kinase inhibitor (CCT137690) antagonizes development of HER2+ LC in vivo.


Sign in / Sign up

Export Citation Format

Share Document