Aurora Kinase Inhibition: A New Light in the Sky? - Journal of

Jun 25, 2015 - Copyright © 2015 American Chemical Society. *E-mail: [email protected]. Telephone: +44(0)2087224051. Cite this:J. Med. Chem. 58, ...
0 downloads 3 Views 371KB Size
Viewpoint pubs.acs.org/jmc

Aurora Kinase Inhibition: A New Light in the Sky? Spiros Linardopoulos†,‡ and Julian Blagg*,† †

Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, U.K. Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London SW3 6JB, U.K.



ABSTRACT: The quest for potent and selective small molecule inhibitors of the Aurora kinases has been long and resource intensive with multiple agents progressed to the clinic. To definitively explore the potential for clinical efficacy at well-tolerated dosing schedules requires a well-characterized, selective inhibitor with pharmacokinetic properties, flexible dosing regimen, and suite of target engagement biomarkers suitable for clinical use. AMG900 is a promising opportunity to definitively test the clinical benefit of dual Aurora kinase A and B inhibition.

P

some aspects of the AMG900 biological profile including in vivo efficacy of AMG900 alone and in combination with taxanes and epothilones in multidrug-resistant cell lines and human tumor xenografts representative of triple negative breast cancer.8−10 Of particular interest in the article is the rationale for selection of AMG900 as a preclinical development candidate, nicely illustrating the complexities often encountered by drug discovery teams in translating in vitro pharmacology and SAR to in vivo studies. In order to differentiate AMG900 from other small molecule Aurora kinase inhibitors and to definitively test the clinical benefit and mechanism-based toxicity of Aurora kinase inhibition, the Amgen team set out to maintain the selectivity of leads 1 and 2 (Figure 1) as well as their potency in MDRoverexpressing cell lines in order to maximize the potential for translation into cell-based assays and to minimize the potential for clinical resistance through transporter-mediated efflux. The team also set out to discover compounds with potency, solubility, and an oral pharmacokinetic profile consistent with low predicted human dose sufficient to support flexibility of dosing regimen in the clinic in the event that, for example, sustained target inhibition should be necessary to completely target unsynchronized cell populations in their continual passage through mitosis. In designing and testing compounds to address these aims, the authors illustrate some notable medicinal chemistry design themes. First is a lack of correlation between compound potencies observed in in vitro kinase biochemical assays versus specific cell-based pharmacodynamic (PD) biomarker assays of Aurora kinase A or B inhibition, consistent with the notion that in vitro biochemical assays using recombinant enzyme preparations may fail to capture the cell-based kinase conformation that is presented to small molecule inhibitors in the cellular context. Thus, the medicinal chemistry optimization and compound selection described by the Amgen team in this article is driven by cell-based PD assays that correlate with cellbased antiproliferative activity. Second, the incorporation of polarity and/or basic centers into small molecule inhibitors in order to improve solubility and reduce in vitro metabolic

rotein kinases Aurora A and B are among the most studied in the kinome largely because of their pivotal role in mitosis.1 Defects in the function and amplification or overexpression of these kinases are associated with tumorigenesis.2 Aurora A plays an important role in centrosome maturation, spindle assembly, meiotic maturation, and metaphase I spindle orientation,1 while Aurora B is a member of the chromosomal passenger complex (CPC), critical for chromosome condensation, chromosome orientation on the mitotic spindle and the spindle-assembly checkpoint, as well as the final stages of cytokinesis.1 The role of Aurora kinases A and B in mitosis, coupled with evidence that amplification or overexpression of these kinases and other functions such as stabilization of N-MYC by Aurora A drives tumorigenesis,3 has driven a long history of medicinal chemistry design in search of small molecule Aurora kinase inhibitors. Many of the first wave of Aurora kinase inhibitors to reach clinical evaluation lacked one or more of the key features now regarded as essential for clinical hypothesis testing of molecularly targeted agents, namely, pharmacological selectivity, pharmacokinetic profile, flexibility of dosing regimen, and the potential for clinical assessment of pharmacodynamic biomarkers to correlate clinical outcome and toxicity with target engagement.4,5 In addition, the optimal genetic context for clinical evaluation of Aurora kinase inhibitors has evolved in parallel with medicinal chemistry design.6 Although our understanding of the role of Aurora kinases in cancer has improved significantly and several pan-Aurora kinase or subtype-specific inhibitors have been discovered, only a small number have moved forward in clinical trials, the majority of which focus on hematopoietic cancers. A possible explanation for limited clinical responses in the treatment of solid tumors is the likely need for sustained exposures through a number of cell cycles (for Aurora B inhibitors) or time in mitosis (for Aurora A inhibitors) to elicit a maximum therapeutic response. At such sustained exposures, mechanism-based toxicity may become dose-limiting. The Featured Article in this issue of the Journal of Medicinal Chemistry describes the evolution of a series of phthalazinebased Aurora kinase inhibitors from advanced leads to the selection of AMG900 as a preclinical development candidate.7 Genesis of the hit series has been previously described, as have © XXXX American Chemical Society

Received: June 15, 2015

A

DOI: 10.1021/acs.jmedchem.5b00918 J. Med. Chem. XXXX, XXX, XXX−XXX

Journal of Medicinal Chemistry

Viewpoint

Figure 1. Evolution of AMG900 from compounds 1 and 2.

duration of exposure and also of Aurora B target inhibition that are required for in vivo efficacy in human tumor xenograft animal experiments. These predictions were evaluated in in vivo efficacy studies in HCT116 human tumor xenograft models to demonstrate that intermittent dosing schedules can, in the context of this solid tumor animal model, deliver equivalent efficacy to continual daily dosing. These findings auger well for the exploration of clinical dosing regimens in order to maximize efficacy at well tolerated dosing schedules, either as a single agent or in combination with other therapies. Interestingly, despite relatively high lipophilicity (log P = 4.4), AMG900 exhibits excellent kinase selectivity; furthermore, its low aqueous solubility (1 μg/mL in PBS at pH 7) and relatively high clearance in in vitro microsomal metabolism assays do not preclude acceptable oral pharmacokinetic properties, presumably because of masking of high plasma free clearance by extensive plasma protein binding. The in vitro biochemical potency and exquisite cell-based potency of AMG900 in both PD and antiproliferative assays likely overcome the low free fraction available to engage with its biochemical targets and contribute to an acceptable dose-prediction to man. In summary, AMG900 represents a promising addition to the armory of Aurora kinase inhibitors for clinical evaluation. Its selectivity profile, coupled with flexibility of dosing regimen in the clinic should, with clinical translation of the described suite of PD biomarkers, allow definitive testing of the clinical hypothesis that dual inhibition of Aurora A and B can lead to clinical benefit at a well-tolerated dosing regimen either as a single agent or in combination.

turnover frequently leads to increased transporter-mediated efflux as demonstrated by the comparison between compounds 3 and 4 (Figure 1); in this article, evaluation of phosphohistone H3 (p-HH3), a distal PD biomarker of Aurora B inhibition, in both a parental and an MDR-overexpressing cell line proved important for compound prioritization. Third is the complexities inherent in interpreting how small changes in physicochemical properties impact the fraction unbound in in vitro cell-based and in vivo assays. Interpretation of these changes requires a pharmacological audit trail of in vitro cellbased and in vivo PD biomarkers of compound action and an understanding of free fraction in each context.11,12 Fourth is the importance of investigating both time- and concentrationdependent effects in cell-based assays. In the case of AMG900, evidence is provided to demonstrate slow equilibration to maximal cell-based inhibition of the distal PD biomarker pHH3 consistent with in vitro determination of slow binding kinetics by surface plasmon resonance. However, AMG900 does not exhibit long residence time in cell-based assays, again suggesting that in vitro biochemical enzyme assays using recombinant protein preparations may fail to represent the true physiological context of kinase enzymes in intact cell-based assays and raising the possibility that the observed slow onset of effect in cell-based assays is driven by high nonspecific binding/ low free fraction of AMG900. AMG900 is an extremely potent inhibitor of Aurora kinases A and B (IC50(AurA/TPX2) = 0.0054 ± 0.0024 μM; IC50(AurB) = 0.0036 ± 0.0034 μM) with excellent translation to cell-based activity (inhibition of p-AurA in the HCT116 human colorectal cancer cell line IC50 = 0.004 ±