Novel Activities of Select NSAID R-Enantiomers against Rac1 and Cdc42 GTPases

PLoS One. 2015 Nov 11;10(11):e0142182. doi: 10.1371/journal.pone.0142182. eCollection 2015.

Abstract

Rho family GTPases (including Rac, Rho and Cdc42) collectively control cell proliferation, adhesion and migration and are of interest as functional therapeutic targets in numerous epithelial cancers. Based on high throughput screening of the Prestwick Chemical Library® and cheminformatics we identified the R-enantiomers of two approved drugs (naproxen and ketorolac) as inhibitors of Rac1 and Cdc42. The corresponding S-enantiomers are considered the active component in racemic drug formulations, acting as non-steroidal anti-inflammatory drugs (NSAIDs) with selective activity against cyclooxygenases. Here, we show that the S-enantiomers of naproxen and ketorolac are inactive against the GTPases. Additionally, more than twenty other NSAIDs lacked inhibitory action against the GTPases, establishing the selectivity of the two identified NSAIDs. R-naproxen was first identified as a lead compound and tested in parallel with its S-enantiomer and the non-chiral 6-methoxy-naphthalene acetic acid (active metabolite of nabumetone, another NSAID) as a structural series. Cheminformatics-based substructure analyses-using the rotationally constrained carboxylate in R-naproxen-led to identification of racemic [R/S] ketorolac as a suitable FDA-approved candidate. Cell based measurement of GTPase activity (in animal and human cell lines) demonstrated that the R-enantiomers specifically inhibit epidermal growth factor stimulated Rac1 and Cdc42 activation. The GTPase inhibitory effects of the R-enantiomers in cells largely mimic those of established Rac1 (NSC23766) and Cdc42 (CID2950007/ML141) specific inhibitors. Docking predicts that rotational constraints position the carboxylate moieties of the R-enantiomers to preferentially coordinate the magnesium ion, thereby destabilizing nucleotide binding to Rac1 and Cdc42. The S-enantiomers can be docked but are less favorably positioned in proximity to the magnesium. R-naproxen and R-ketorolac have potential for rapid translation and efficacy in the treatment of several epithelial cancer types on account of established human toxicity profiles and novel activities against Rho-family GTPases.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Anti-Inflammatory Agents, Non-Steroidal / chemistry
  • Anti-Inflammatory Agents, Non-Steroidal / metabolism
  • Anti-Inflammatory Agents, Non-Steroidal / pharmacology*
  • Cell Line, Tumor
  • Cell Movement / drug effects
  • Cell Survival / drug effects
  • HeLa Cells
  • Humans
  • Immunoblotting
  • Ketorolac / chemistry
  • Ketorolac / metabolism
  • Ketorolac / pharmacology*
  • Mice
  • Microscopy, Confocal
  • Molecular Docking Simulation
  • Molecular Structure
  • NIH 3T3 Cells
  • Naproxen / chemistry
  • Naproxen / metabolism
  • Naproxen / pharmacology*
  • Protein Binding
  • Protein Structure, Tertiary
  • Stereoisomerism
  • cdc42 GTP-Binding Protein / antagonists & inhibitors*
  • cdc42 GTP-Binding Protein / chemistry
  • cdc42 GTP-Binding Protein / metabolism
  • rac1 GTP-Binding Protein / antagonists & inhibitors*
  • rac1 GTP-Binding Protein / chemistry
  • rac1 GTP-Binding Protein / metabolism

Substances

  • Anti-Inflammatory Agents, Non-Steroidal
  • Naproxen
  • cdc42 GTP-Binding Protein
  • rac1 GTP-Binding Protein
  • Ketorolac