Preclinical assessment of synergistic efficacy of MELK and CDK inhibitors in adrenocortical cancer

J Exp Clin Cancer Res. 2022 Sep 23;41(1):282. doi: 10.1186/s13046-022-02464-5.

Abstract

Background: Adrenocortical cancer (ACC) is a rare and aggressive cancer with dismal 5-year survival due to a lack of effective treatments. We aimed to identify a new effective combination of drugs and investigated their synergistic efficacy in ACC preclinical models.

Methods: A quantitative high-throughput drug screening of 4,991 compounds was performed on two ACC cell lines, SW13 and NCI-H295R, based on antiproliferative effect and caspase-3/7 activity. The top candidate drugs were pairwise combined to identify the most potent combinations. The synergistic efficacy of the selected inhibitors was tested on tumorigenic phenotypes, such as cell proliferation, migration, invasion, spheroid formation, and clonogenicity, with appropriate mechanistic validation by cell cycle and apoptotic assays and protein expression of the involved molecules. We tested the efficacy of the drug combination in mice with luciferase-tagged human ACC xenografts. To study the mRNA expression of target molecules in ACC and their clinical correlations, we analyzed the Gene Expression Omnibus and The Cancer Genome Atlas.

Results: We chose the maternal embryonic leucine zipper kinase (MELK) inhibitor (OTS167) and cyclin-dependent kinase (CDK) inhibitor (RGB-286638) because of their potent synergy from the pairwise drug combination matrices derived from the top 30 single drugs. Multiple publicly available databases demonstrated overexpression of MELK, CDK1/2, and partnering cyclins mRNA in ACC, which were independently associated with mortality and other adverse clinical features. The drug combination demonstrated a synergistic antiproliferative effect on ACC cells. Compared to the single-agent treatment groups, the combination treatment increased G2/M arrest, caspase-dependent apoptosis, reduced cyclins A2, B1, B2, and E2 expression, and decreased cell migration and invasion with reduced vimentin. Moreover, the combination effectively decreased Foxhead Box M1, Axin2, glycogen synthase kinase 3-beta, and β-catenin. A reduction in p-stathmin from the combination treatment destabilized microtubule assembly by tubulin depolymerization. The drug combination treatment in mice with human ACC xenografts resulted in a significantly lower tumor burden than those treated with single-agents and vehicle control groups.

Conclusions: Our preclinical study revealed a novel synergistic combination of OTS167 and RGB-286638 in ACC that effectively targets multiple molecules associated with ACC aggressiveness. A phase Ib/II clinical trial in patients with advanced ACC is therefore warranted.

Keywords: Adrenocortical cancer; Cyclin-dependent kinase; Maternal embryonic leucine zipper kinase; Quantitative high-throughput screening; Targeted therapy; β-catenin.

MeSH terms

  • Adrenal Cortex Neoplasms* / drug therapy
  • Adrenal Cortex Neoplasms* / genetics
  • Adrenal Cortex Neoplasms* / metabolism
  • Adrenocortical Carcinoma* / drug therapy
  • Adrenocortical Carcinoma* / genetics
  • Adrenocortical Carcinoma* / metabolism
  • Animals
  • Apoptosis
  • Caspase 3
  • Cell Line, Tumor
  • Cell Proliferation
  • Cyclin-Dependent Kinases
  • Cyclins
  • G2 Phase Cell Cycle Checkpoints
  • Glycogen Synthase Kinase 3 / pharmacology
  • Glycogen Synthase Kinase 3 / therapeutic use
  • Humans
  • Mice
  • Protein Serine-Threonine Kinases
  • Pyrazoles
  • RNA, Messenger
  • Stathmin
  • Tubulin
  • Urea / analogs & derivatives
  • Vimentin
  • beta Catenin

Substances

  • Cyclins
  • Pyrazoles
  • RGB 286638
  • RNA, Messenger
  • Stathmin
  • Tubulin
  • Vimentin
  • beta Catenin
  • Urea
  • MELK protein, human
  • Protein Serine-Threonine Kinases
  • Cyclin-Dependent Kinases
  • Glycogen Synthase Kinase 3
  • Caspase 3