1. Academic Validation
  2. Protein kinase Cδ is a therapeutic target in malignant melanoma with NRAS mutation

Protein kinase Cδ is a therapeutic target in malignant melanoma with NRAS mutation

  • ACS Chem Biol. 2014 Apr 18;9(4):1003-14. doi: 10.1021/cb400837t.
Asami Takashima 1 Brandon English Zhihong Chen Juxiang Cao Rutao Cui Robert M Williams Douglas V Faller
Affiliations

Affiliation

  • 1 Cancer Center, ‡Departments of Medicine, Biochemistry, Pediatrics, Microbiology, Pathology and Laboratory Medicine, and §Department of Dermatology, Boston University School of Medicine , 72 E Concord Street, Boston, Massachusetts 02118, United States.
Abstract

NRAS is the second most frequently mutated gene in melanoma. Previous reports have demonstrated the sensitivity of Cancer cell lines carrying KRAS mutations to Apoptosis initiated by inhibition of protein kinase Cδ (PKCδ). Here, we report that PKCδ inhibition is cytotoxic in melanomas with primary NRAS mutations. Novel small-molecule inhibitors of PKCδ were designed as chimeric hybrids of two naturally occurring PKCδ inhibitors, staurosporine and rottlerin. The specific hypothesis interrogated and validated is that combining two domains of two naturally occurring PKCδ inhibitors into a chimeric or hybrid structure retains biochemical and biological activity and improves PKCδ isozyme selectivity. We have devised a potentially general synthetic protocol to make these chimeric species using Molander trifluorborate coupling chemistry. Inhibition of PKCδ, by siRNA or small molecule inhibitors, suppressed the growth of multiple melanoma cell lines carrying NRAS mutations, mediated via caspase-dependent Apoptosis. Following PKCδ inhibition, the stress-responsive JNK pathway was activated, leading to the activation of H2AX. Consistent with recent reports on the apoptotic role of phospho-H2AX, knockdown of H2AX prior to PKCδ inhibition mitigated the induction of caspase-dependent Apoptosis. Furthermore, PKCδ inhibition effectively induced cytotoxicity in BRaf mutant melanoma cell lines that had evolved resistance to a BRaf Inhibitor, suggesting the potential clinical application of targeting PKCδ in patients who have relapsed following treatment with BRaf inhibitors. Taken together, the present work demonstrates that inhibition of PKCδ by novel small molecule inhibitors causes caspase-dependent Apoptosis mediated via the JNK-H2AX pathway in melanomas with NRAS mutations or BRaf Inhibitor resistance.

Figures
Products