Abstract

Monopolar spindle 1 (Mps1) has been shown to function as the key kinase that activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we report the structure and functional characterization of two novel selective Mps1 inhibitors, BAY 1161909 and BAY 1217389, derived from structurally distinct chemical classes. BAY 1161909 and BAY 1217389 inhibited Mps1 kinase activity with IC50 values below 10 nmol/L while showing an excellent selectivity profile. In cellular mechanistic assays, both Mps1 inhibitors abrogated nocodazole-induced SAC activity and induced premature exit from mitosis ("mitotic breakthrough"), resulting in multinuclearity and tumor cell death. Both compounds efficiently inhibited tumor cell proliferation in vitro (IC50 nmol/L range). In vivo, BAY 1161909 and BAY 1217389 achieved moderate efficacy in monotherapy in tumor xenograft studies. However, in line with its unique mode of action, when combined with paclitaxel, low doses of Mps1 inhibitor reduced paclitaxel-induced mitotic arrest by the weakening of SAC activity. As a result, combination therapy strongly improved efficacy over paclitaxel or Mps1 inhibitor monotreatment at the respective MTDs in a broad range of xenograft models, including those showing acquired or intrinsic paclitaxel resistance. Both Mps1 inhibitors showed good tolerability without adding toxicity to paclitaxel monotherapy. These preclinical findings validate the innovative concept of SAC abrogation for cancer therapy and justify clinical proof-of-concept studies evaluating the Mps1 inhibitors BAY 1161909 and BAY 1217389 in combination with antimitotic cancer drugs to enhance their efficacy and potentially overcome resistance. Mol Cancer Ther; 15(4); 583-92. ©2016 AACR.

Highlights

  • Cell-cycle deregulation represents one of the classical hallmarks of cancer, and cell-cycle arrest is the predominant mode of action of antimitotic cancer drugs

  • Monotherapy of Monopolar spindle 1 (Mps1) inhibitor BAY 1217389 dosed upon 4 mg/kg (80% of efficacious dose in monotherapy) led to 80% reduction of basal phosphorylated KNL1 (pKNL1); 2 mg/kg

  • Antitubulin–based chemotherapy is used as standard of care in many cancer patients across a variety of tumor types, such as breast, lung, or ovarian cancers

Read more

Summary

Introduction

Cell-cycle deregulation represents one of the classical hallmarks of cancer, and cell-cycle arrest is the predominant mode of action of antimitotic cancer drugs (e.g., taxanes and vinca alkaloids). Targeted disruption of mitotic cell-cycle checkpoints offers a novel approach to cancer treatment: driving tumor cells into cell division despite DNA damage or unattached/misattached chromosomes, resulting in a lethal degree of DNA damage or aneuploidy. Monopolar spindle 1 (Mps1), known as TTK, is a serine threonine kinase, which ensures proper biorientation of sister chromatids on the mitotic spindle by the activation of the spindle assembly checkpoint (SAC). The SAC controls attachments between microtubules (MT) and kinetochores (KT) during prometaphase and blocks transition to anaphase until all chromosomes are correctly tensed, attached, and bioriented at the metaphase plate [1]. Bayer Pharma AG, Drug Discovery, Berlin, Germany. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/)

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call