1. Academic Validation
  2. Further exploration of the structure-activity relationship of dual soluble epoxide hydrolase/fatty acid amide hydrolase inhibitors

Further exploration of the structure-activity relationship of dual soluble epoxide hydrolase/fatty acid amide hydrolase inhibitors

  • Bioorg Med Chem. 2021 Dec 1;51:116507. doi: 10.1016/j.bmc.2021.116507.
Stephanie Wilt 1 Sean Kodani 2 Leah Valencia 1 Paula K Hudson 1 Stephanie Sanchez 3 Taylor Quintana 3 Christophe Morisseau 2 Bruce D Hammock 2 Ram Kandasamy 4 Stevan Pecic 5
Affiliations

Affiliations

  • 1 Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States.
  • 2 Department of Entomology and Nematology, and UCD Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, United States.
  • 3 Department of Psychology, California State University, East Bay, 25800 Carlos Bee Blvd. Science S229, Hayward, CA 94542, United States.
  • 4 Department of Psychology, California State University, East Bay, 25800 Carlos Bee Blvd. Science S229, Hayward, CA 94542, United States. Electronic address: [email protected].
  • 5 Department of Chemistry & Biochemistry, California State University, Fullerton, 800 N. State College, Fullerton, CA 92834, United States. Electronic address: [email protected].
Abstract

Fatty acid amide hydrolase (FAAH) is a membrane protein that hydrolyzes endocannabinoids, and its inhibition produces analgesic and anti-inflammatory effects. The soluble Epoxide Hydrolase (sEH) hydrolyzes epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatetraenoic acids. EETs have anti-inflammatory and inflammation resolving properties, thus inhibition of sEH consequently reduces inflammation. Concurrent inhibition of both enzymes may represent a novel approach in the treatment of chronic pain. Drugs with multiple targets can provide a superior therapeutic effect and a decrease in side effects compared to ligands with single targets. Previously, microwave-assisted methodologies were employed to synthesize libraries of benzothiazole analogs from which high affinity dual inhibitors (e.g. 3, sEH IC50 = 9.6 nM; FAAH IC50 = 7 nM) were identified. Here, our structure-activity relationship studies revealed that the 4-phenylthiazole moiety is well tolerated by both enzymes, producing excellent inhibition potencies in the low nanomolar range (e.g. 6o, sEH IC50 = 2.5 nM; FAAH IC50 = 9.8 nM). Docking experiments show that the new class of dual inhibitors bind within the catalytic sites of both enzymes. Prediction of several pharmacokinetic/pharmacodynamic properties suggest that these new dual inhibitors are good candidates for further in vivo evaluation. Finally, dual inhibitor 3 was tested in the Formalin Test, a rat model of acute inflammatory pain. The data indicate that 3 produces antinociception against the inflammatory phase of the Formalin Test in vivo and is metabolically stable following intraperitoneal administration in male rats. Further, antinociception produced by 3 is comparable to that of ketoprofen, a traditional nonsteroidal anti-inflammatory drug. The results presented here will help toward the long-term goal of developing novel non-opioid therapeutics for pain management.

Keywords

4-Phenylthiazole moiety; ADMET predictions; Docking experiments; Enzyme inhibition; Formalin test; Microwave-assisted synthesis; Polypharmacology; Structure-Activity Relationship study.

Figures
Products