A novel screening approach comparing kinase activity of small molecule inhibitors with similar molecular structures and distinct biologic effects in triple-negative breast cancer to identify targetable signaling pathways

Supplemental Digital Content is available in the text. Breast cancer affects women globally; the majority of breast cancer-related mortalities are due to metastasis. Acquisition of a mesenchymal phenotype has been implicated in the progression of breast cancer cells to an invasive, metastatic state. Triple-negative breast cancer (TNBC) subtypes have high rates of metastases, recurrence, and have poorer prognoses compared to other breast cancer types, partially due to lack of commonly targeted receptors. Kinases have diverse and pivotal functions in metastasis in TNBC, and discovery of new kinase targets for TNBC is warranted. We previously used a screening approach to identify intermediate-synthesis nonpotent, nonselective small-molecule inhibitors from the Published Kinase Inhibitor Set that reversed the mesenchymal phenotype in TNBC cells. Two of these inhibitors (GSK346294A and GSK448459A) are structurally similar, but have unique kinase activity profiles and exhibited differential biologic effects on TNBC cells, specifically on epithelial-to-mesenchymal transition (EMT). Here, we further interrogate these effects and compare activity of these inhibitors on transwell migration, gene (qRT-PCR) and protein (western blot) expressions, and cancer stem cell-like behavior. We incorporated translational patient-derived xenograft models in these studies, and we focused on the lead inhibitor hit, GSK346294A, to demonstrate the utility of our comparative analysis as a screening modality to identify novel kinase targets and signaling pathways to pursue in TNBC. This study introduces a new method for discovering novel kinase targets that reverse the EMT phenotype; this screening approach can be applied to all cancer types and is not limited to breast cancer.

[1]  S. Spencer,et al.  Ki67 is a Graded Rather than a Binary Marker of Proliferation versus Quiescence. , 2018, Cell reports.

[2]  Cathy H. Wu,et al.  Integrative annotation and knowledge discovery of kinase post-translational modifications and cancer-associated mutations through federated protein ontologies and resources , 2018, Scientific Reports.

[3]  Jennifer Abbasí Personal Genomics and Cryptocurrency Team Up. , 2018, JAMA.

[4]  E. Martin,et al.  Panobinostat suppresses the mesenchymal phenotype in a novel claudin-low triple negative patient-derived breast cancer model , 2018, Oncoscience.

[5]  Andrea Sottoriva,et al.  Patient-derived organoids model treatment response of metastatic gastrointestinal cancers , 2018, Science.

[6]  H. Rupasinghe,et al.  Kinase-targeted cancer therapies: progress, challenges and future directions , 2018, Molecular Cancer.

[7]  E. Martin,et al.  A novel patient-derived xenograft model for claudin-low triple-negative breast cancer , 2018, Breast Cancer Research and Treatment.

[8]  C. Sotiriou,et al.  Unravelling triple-negative breast cancer molecular heterogeneity using an integrative multiomic analysis , 2018, Annals of oncology : official journal of the European Society for Medical Oncology.

[9]  A. Jemal,et al.  Cancer statistics, 2018 , 2018, CA: a cancer journal for clinicians.

[10]  M. Redondo,et al.  Protein Kinase Targets in Breast Cancer , 2017, International journal of molecular sciences.

[11]  D. Drewry,et al.  Novel application of the published kinase inhibitor set to identify therapeutic targets and pathways in triple negative breast cancer subtypes , 2017, PloS one.

[12]  P. Lønning,et al.  Genomic Evolution of Breast Cancer Metastasis and Relapse , 2017, Cancer cell.

[13]  C. Kelly,et al.  Therapeutic Advances and New Directions for Triple-Negative Breast Cancer , 2017, Breast Care.

[14]  Rajarshi Guha,et al.  Pharos: Collating protein information to shed light on the druggable genome , 2016, Nucleic Acids Res..

[15]  Ashish Ranjan Sharma,et al.  PLK-1: Angel or devil for cell cycle progression. , 2016, Biochimica et biophysica acta.

[16]  Jaebong Kim,et al.  PLK-1 Targeted Inhibitors and Their Potential against Tumorigenesis , 2015, BioMed research international.

[17]  Xin-hua Liang,et al.  Links between cancer stem cells and epithelial–mesenchymal transition , 2015, OncoTargets and therapy.

[18]  Nicolas Stransky,et al.  Targeting cancer with kinase inhibitors. , 2015, The Journal of clinical investigation.

[19]  Andrew L. Kung,et al.  Examining the utility of patient-derived xenograft mouse models , 2015, Nature Reviews Cancer.

[20]  Wong Cheng Lee,et al.  Multivariate biophysical markers predictive of mesenchymal stromal cell multipotency , 2014, Proceedings of the National Academy of Sciences.

[21]  Judy Lucas,et al.  Advances in patient-derived tumor xenografts: from target identification to predicting clinical response rates in oncology. , 2014, Biochemical pharmacology.

[22]  Vandana G Abramson,et al.  Molecular Heterogeneity of Triple-Negative Breast Cancer , 2014, Current Breast Cancer Reports.

[23]  Philip M. Kim,et al.  A systematic approach to identify novel cancer drug targets using machine learning, inhibitor design and high-throughput screening , 2014, Genome Medicine.

[24]  V. Findlay,et al.  Epithelial to mesenchymal transition and the cancer stem cell phenotype: Insights from cancer biology with therapeutic implications for colorectal cancer , 2014, Cancer Gene Therapy.

[25]  G. Johnson,et al.  Implications of Mesenchymal Cells in Cancer Stem Cell Populations: Relevance to EMT , 2014, Current Pathobiology Reports.

[26]  A. A. Moustafa Epithelial-mesenchymal transition and its regulators are major targets of triple-negative breast cancer , 2013 .

[27]  A. Al Moustafa Epithelial-mesenchymal transition and its regulators are major targets of triple-negative breast cancer , 2013, Cell adhesion & migration.

[28]  Haixu Tang,et al.  A new method for stranded whole transcriptome RNA-seq. , 2013, Methods.

[29]  J. R. Peterson,et al.  The Human Kinome and Kinase Inhibition , 2013, Current protocols in pharmacology.

[30]  Dario R Alessi,et al.  Kinase drug discovery--what's next in the field? , 2013, ACS chemical biology.

[31]  J. Crown,et al.  Emerging targeted therapies in triple-negative breast cancer. , 2012, Annals of oncology : official journal of the European Society for Medical Oncology.

[32]  A. Shuttleworth,et al.  Inhibition of Platelet-Derived Growth Factor Receptor Signaling Regulates Oct4 and Nanog Expression, Cell Shape, and Mesenchymal Stem Cell Potency , 2012, Stem cells.

[33]  Theonie Anastassiadis,et al.  Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity , 2011, Nature biotechnology.

[34]  A. Ridley Life at the Leading Edge , 2011, Cell.

[35]  L. Rhodes,et al.  Effects of human mesenchymal stem cells on ER-positive human breast carcinoma cells mediated through ER-SDF-1/CXCR4 crosstalk , 2010, Molecular Cancer.

[36]  Jorge S Reis-Filho,et al.  Triple-negative breast cancer. , 2010, The New England journal of medicine.

[37]  J. Jackson,et al.  Design of potent thiophene inhibitors of polo-like kinase 1 with improved solubility and reduced protein binding. , 2009, Bioorganic & medicinal chemistry letters.

[38]  Erik Sahai,et al.  The actin cytoskeleton in cancer cell motility , 2009, Clinical & Experimental Metastasis.

[39]  J. Condeelis,et al.  Regulation of the actin cytoskeleton in cancer cell migration and invasion. , 2007, Biochimica et biophysica acta.

[40]  J. Peters,et al.  The Small-Molecule Inhibitor BI 2536 Reveals Novel Insights into Mitotic Roles of Polo-like Kinase 1 , 2007, Current Biology.

[41]  Johannes Gerdes,et al.  The Ki‐67 protein: From the known and the unknown , 2000, Journal of cellular physiology.