Pharmacokinetic screening of soluble epoxide hydrolase inhibitors in dogs

https://doi.org/10.1016/j.ejps.2010.03.018Get rights and content

Abstract

Epoxyeicosatrienoic acids that have anti-hypertensive and anti-inflammatory properties are mainly metabolized by soluble epoxide hydrolase (sEH, EC 3.3.2.3). Therefore, sEH has emerged as a therapeutic target for treating various cardiovascular diseases and inflammatory pain. N,N′-Disubstituted ureas are potent sEH inhibitors in vitro. However, in vivo usage of early sEH inhibitors has been limited by their low bioavailability and poor physiochemical properties. Therefore, a group of highly potent compounds with more drug-like physiochemical properties were evaluated by monitoring their plasma profiles in dogs treated orally with sEH inhibitors. Urea compounds with an adamantyl or a 4-trifluoromethoxyphenyl group on one side and a piperidyl or a cyclohexyl ether group on the other side of the urea function showed pharmacokinetic profiles with high plasma concentrations and long half lives. In particular, the inhibitor trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB) not only is very potent with good physiochemical properties, but also shows high oral bioavailability for doses ranging from 0.01 to 1 mg/kg. This compound is also very potent against the sEH of several mammals, suggesting that t-AUCB will be an excellent tool to evaluate the biology of sEH in multiple animal models. Such compounds may also be a valuable lead for the development of veterinary therapeutics.

Introduction

Epoxyeicosatrienoic acids (EETs) are endogenous signalling molecules produced by cytochrome P450 from arachidonic acid (Spector, 2008). The EETs act as vasodilators by activating Ca2+-activated K+ channels in vascular smooth muscle cells (Fleming, 2007). EETs also possess anti-inflammatory properties in endothelial cells (Node et al., 1999). EETs and related epoxy fatty acids are mainly metabolized by the soluble epoxide hydrolase (sEH; EC 3.3.2.3) (Liu et al., 2005, Newman et al., 2005), which hydrolyzes the EETs into dihydroxyeicosatrienoic acids (DHETs), resulting in partial or complete loss of their initial biological activities. Inhibition of sEH stabilizes endogenous EETs and thus reduces hypertension in multiple rodent models (Imig et al., 2002, Imig et al., 2005, Jung et al., 2005, Chiamvimonvat et al., 2007, Huang et al., 2007, Loch et al., 2007). The inhibition of sEH also reduces the production of nitric oxide, cytokines and pro-inflammatory lipid mediators, in addition to significantly improving survival in lipopolysaccharide (LPS)-induced systemic inflammation in mice (Liu et al., 2005, Schmelzer et al., 2005, Smith et al., 2005). The sEH is also a potential target for the treatment of ischemic stroke (Dorrance et al., 2005, Zhang et al., 2007a) and cardiac hypertrophy (Xu et al., 2006). Furthermore, sEH inhibition reduces pain in several rodent models (Inceoglu et al., 2006). Thus, sEH is an emerging target for pharmacological treatment of cardiovascular diseases (Monti et al., 2008), inflammation, and possibly other diseases (Gauthier et al., 2007, Harris et al., 2008).

N,N′-Disubstituted ureas are potent sEHIs (sEH inhibitors) with Kis in the low nanomolar range (Morisseau et al., 1999b). Early urea-based inhibitors, such as 12-(3-adamantane-1-yl-ureido)-dodecanoic acid (AUDA), 12-(3-adamantane-1-yl-ureido)-dodecanoic acid n-butyl ester (AUDA-nBE) and 1-adamantan-1-yl-3-{5-[2-(2-ethoxyethoxy)ethoxy]pentyl}urea (AEPU) (Imig et al., 2002, Kim et al., 2004), were used to investigate the biology associated with the inhibition of sEH in animal models (Dorrance et al., 2005, Imig et al., 2005, Jung et al., 2005, Schmelzer et al., 2005, Sellers et al., 2005, Smith et al., 2005, Inceoglu et al., 2006, Xu et al., 2006, Ai et al., 2007, Huang et al., 2007, Loch et al., 2007, Zhang et al., 2007a, Li et al., 2008, Olearczyk et al., 2009, Parrish et al., 2009). However, their in vivo usage is limited due to poor physiochemical properties, rapid metabolism and/or poor bioavailability. Tedious formation such as the use of nanocrystals from cryomilling with careful selection of salts and solvents are needed to formulate such materials for efficient delivery in a small capsule (Ghosh et al., 2008). As an alternative to such formulation approaches, a medicinal chemistry approach was taken to develop new potent inhibitors that are more water soluble and more metabolically stable by methodically modifying their structures (Zhao et al., 2004, Hwang et al., 2006, Jones et al., 2006, Li et al., 2006, Morisseau et al., 2006, Hwang et al., 2007, Kim et al., 2007a, Kim et al., 2007b, Kasagami et al., 2009, Shen et al., 2009).

While simple, rapid and efficient methods have been developed to estimate the inhibitory potency of new compounds (Jones et al., 2005, Wolf et al., 2006) as well as their solubility and other physiochemical properties, it is more difficult and costly to evaluate their pharmacokinetic properties. It is believed that compounds with favorable pharmacokinetics are more likely to be efficacious and safe (Dingemanse and Appel-Dingemanse, 2007). We previously developed a rapid pharmacokinetic screening method using cassette dosing and measuring compounds with minuscule serial bleedings in mice (Watanabe et al., 2006). This method was efficient in classifying compounds on their relative bioavailability. However, because of the small size of the animal used, it is difficult to extrapolate to larger animals or humans. Although we routinely monitor blood levels with 5 μl of blood for these compounds (Watanabe et al., 2006), the small volume of blood in a mouse limits our ability to monitor multiple blood biomarkers and, in particular, oxylipins which are promising indicators of the efficacy of sEHIs in vivo. Herein we report the use of dogs (black Labrador retrievers) for rapid pharmacokinetics screen of sEHIs. Due to the relatively large body size of the dogs (∼20 kg each), this method allows for easier administration of drugs and collection of plasma with larger sample size. Moreover, dogs are much closer phylogenetically and in size to humans than rodents, making extrapolation to humans more reliable (Tibbitts, 2003). Finally, there is a need for novel anti-inflammatory and pain therapies in dogs and cats (Lascelles et al., 2005). For example, the non-steroid anti-inflammatory drugs (NSAIDs) are potent canine analgesic and anti-inflammatory drugs, but their use is limited by a variety of adverse effects such as causing gastrointestinal disorders. Thus, the canine data obtained herein will not only be valuable in refining the structure of sEHI for clinical use in humans and for use in canine models of human diseases, but also these data could lead to the development of novel veterinary therapeutics either used alone or as synergists with existing drugs such as NSAIDs (Schmelzer et al., 2006).

Section snippets

Chemicals

All sEHIs tested were synthesized according to previously reported methods (Kim et al., 2004, Kim et al., 2005, Kim et al., 2007a, Kim et al., 2007b, Hwang et al., 2006, Jones et al., 2006, Morisseau et al., 2006, Hwang et al., 2007, Kasagami et al., 2009). References for the synthesis and properties are given in Table S1. Details for the preparation of new compounds (17, 18, 25, 26, 27, 29, 30, 31, and 32) are described in supplementary material. The purity and structural assignment of all

The effects of structure of sEH inhibitors on potency and oral availability

The general structure of sEH urea inhibitors screened in this study is given in Table 1. They are composed of non-polar groups (R1), usually a urea central pharmacophore, linker moieties (L), varied polar functional (P) groups, which are putative secondary pharmacophores, and R2 groups. The inhibitors are divided into four groups based on the nature of the linker moiety L attached to the 3 position on the urea as follows (Table 1): a flexible alkyl; a piperidyl; a cyclohexyl ether; and a phenyl

Discussion

In this paper, sEHIs were screened for their pharmacokinetic profiles in dogs and these data used to select sEHIs for further study. Using a cassette of three compounds administered in a single dose to a dog allowed relatively fast screening, but this approach may result in some errors so caution must be exercised with fine interpretation of the data. First, drug–drug interactions may affect the prediction of pharmacokinetic profiles for each individual compound. To minimize such interactions (

Conclusion

The pharmacokinetic profiles of sEHIs were successfully evaluated and ranked by cassette dosing in dogs with LC–MS/MS. For three compounds cassette dosing was found to be predictive of the pharmacokinetic profiles obtained when the compounds were doses individually. Based on the results, the inhibitors with cycloheptyl or 4-trifluoromethoxyphenyl groups on the 1 position of the urea and with a piperidyl or cyclohexyl ether linker have shown the best PK profiles. APAU and t-AUCB were found to

Acknowledgments

We thank Dr. Robert Gunther, Linda Talken and staff of the UC Davis animal resource center for helping with treating the dogs and their husbandry. We also thank Pharsight for providing the pharmacokinetic software WinNonlin 5.0. Hsing-Ju Tsai was supported in part by the UC Davis-Howard Hughes Medical Institute (UC Davis-HHMI) training program. Jun Yang was supported by the Elizabeth Nash Memorial fellowship from the Cystic Fibrosis Foundation Inc. Bruce D. Hammock is a George and Judy Marcus

References (62)

  • C.A. Lipinski et al.

    Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings

    Adv. Drug Deliv. Rev.

    (2001)
  • A. Luria et al.

    Compensatory mechanism for homeostatic blood pressure regulation in Ephx2 gene-disrupted mice

    J. Biol. Chem.

    (2007)
  • P. Manitpisitkul et al.

    Whatever happened to cassette-dosing pharmacokinetics?

    Drug Discov. Today

    (2004)
  • C. Morisseau et al.

    Peptidyl-urea based inhibitors of soluble epoxide hydrolases

    Bioorg. Med. Chem. Lett.

    (2006)
  • J.W. Newman et al.

    Epoxide hydrolases: their roles and interactions with lipid metabolism

    Prog. Lipid Res.

    (2005)
  • M. Taketani et al.

    Carboxylesterase in the liver and small intestine of experimental animals and human

    Life Sci.

    (2007)
  • T. Watanabe et al.

    High-throughput pharmacokinetic method: Cassette dosing in mice associated with minuscule serial bleedings and LC/MS/MS analysis

    Anal. Chim. Acta

    (2006)
  • R.N. Wixtrom et al.

    Affinity purification of cytosolic epoxide hydrolase using derivatized epoxy-activated Sepharose gels

    Anal. Biochem.

    (1988)
  • N.M. Wolf et al.

    Development of a high-throughput screen for soluble epoxide hydrolase inhibition

    Anal. Biochem.

    (2006)
  • D. Ai et al.

    Angiotensin II up-regulates soluble epoxide hydrolase in vascular endothelium in vitro and in vivo

    Proc. Natl. Acad. Sci. U.S.A.

    (2007)
  • N. Chiamvimonvat et al.

    The soluble epoxide hydrolase as a pharmaceutical target for hypertension

    J. Cardiovasc. Pharmacol.

    (2007)
  • J. Dingemanse et al.

    Integrated pharmacokinetics and pharmacodynamics in drug development

    Clin. Pharmacokinet.

    (2007)
  • A.M. Dorrance et al.

    An epoxide hydrolase inhibitor, 12-(3-adamantan-1-yl-ureido)dodecanoic acid (AUDA), reduces ischemic cerebral infarct size in stroke-prone spontaneously hypertensive rats

    J. Cardiovasc. Pharmacol.

    (2005)
  • O.L. Frick et al.

    Allergen immunotherapy with heat-killed Listeria monocytogenes alleviates peanut and food-induced anaphylaxis in dogs

    Allergy

    (2005)
  • K.M. Gauthier et al.

    Roles of epoxyeicosatrienoic acids in vascular regulation and cardiac preconditioning

    J. Cardiovasc. Pharmacol.

    (2007)
  • S. Ghosh et al.

    Oral delivery of 1,3-dicyclohexylurea nanosuspension enhances exposure and lowers blood pressure in hypertensive rats

    Basic Clin. Pharmacol. Toxicol.

    (2008)
  • G.A. Gomez et al.

    Human soluble epoxide hydrolase: structural basis of inhibition by 4-(3-cyclohexylureido)-carboxylic acids

    Protein Sci.

    (2006)
  • T.R. Harris et al.

    The potential of soluble epoxide hydrolase inhibition in the treatment of cardiac hypertrophy

    Congest Heart Fail.

    (2008)
  • H. Huang et al.

    Increasing or stabilizing renal epoxyeicosatrienoic acid production attenuates abnormal renal function and hypertension in obese rats

    Am. J. Physiol. Renal Physiol.

    (2007)
  • S.H. Hwang et al.

    Orally bioavailable potent soluble epoxide hydrolase inhibitors

    J. Med. Chem.

    (2007)
  • J.D. Imig et al.

    Soluble epoxide hydrolase inhibition lowers arterial blood pressure in angiotensin II hypertension

    Hypertension

    (2002)
  • Cited by (74)

    View all citing articles on Scopus
    View full text