Substituted phenyl groups improve the pharmacokinetic profile and anti-inflammatory effect of urea-based soluble epoxide hydrolase inhibitors in murine models

https://doi.org/10.1016/j.ejps.2012.12.013Get rights and content

Abstract

Soluble epoxide hydrolase inhibitors (sEHIs) are anti-inflammatory, analgesic, anti-hypertensive, cardio- and renal-protective in multiple animal models. However, the earlier adamantyl-containing urea-based inhibitors are rapidly metabolized. Therefore, new potent inhibitors with the adamantyl group replaced by a substituted phenyl group were synthesized to presumptively offer better pharmacokinetic (PK) properties. Here we describe the improved PK profile of these inhibitors and the anti-inflammatory effect of the most promising one in a murine model. The PK profiles of inhibitors were determined following p.o. administration and serial bleeding in mice. The anti-inflammatory effect of 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl)urea (TPPU), the most promising inhibitor among the five sEHIs tested, was investigated in a lipopolysaccharide (LPS)-challenged murine model. The earlier broadly-used adamantyl-containing sEHI, trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB), was used for comparison. Compared with the earlier adamantyl-containing urea-based inhibitors, substituted phenyl-containing urea-based inhibitors afford more favorable PK properties, such as higher Cmaxs, larger AUCs and longer t1/2s, which, as expected, show more stable metabolic stability. Moreover, oral administration of TPPU dramatically reversed the shifts caused by LPS-challenge in plasma levels of inflammatory cytokines, epoxides and corresponding diols, which is more potent than t-AUCB. The substituted phenyl-containing sEHIs are more metabolically stable than those with adamantyl group, resulting in more potent efficacy in vivo. This indicates a new strategy for development of sEHIs for further study toward clinical trials.

Introduction

Soluble epoxide hydrolase (sEH) is a ubiquitous enzyme that catalyzes the conversion of epoxides into the corresponding vicinal diols by the addition of water (Hammock et al., 1980, Morisseau and Hammock, 2005, Newman et al., 2005). For example, sEH catalyzes the metabolism of anti-inflammatory and vasodilatory epoxyeicosatrienoic acids (EETs) into the more polar and pro-inflammatory dihydroxyeicosatrieneoic acid (DHETs) (Chacos et al., 1983, Morisseau and Hammock, 2005, Newman et al., 2005). Pharmacological inhibition of sEH by sEH inhibitors (sEHIs) has been demonstrated to be an effective approach to reduce infammation, pain, and hypertension among others (Ingraham et al., 2011, Qiu et al., 2011). Significant progress has been made in recent years in the development of the amide-, urea- and carbamate-based compounds as potent sEHIs (Morisseau et al., 1999). N,N′-Dicyclohexylurea (DCU) was the first sEHI tested in spontaneously hypertensive rats (SHRs), and it was effective in lowering blood pressure (Yu et al., 2000). Another sEHI 1-cyclohexyl-3-dodecyl urea (CDU) was reported not only to attenuate human vascular smooth muscle cell proliferation dose-dependently in vitro (Davis et al., 2002), but also to be anti-hypertensive and renal protective in a rodent model of angiotensin II-induced hypertension (Imig et al., 2002, Zhao et al., 2004). However, these inhibitors have high melting points and poor solubility in either water or oil, which limits their pharmacological use. Therefore a new series of N,N′-substituted urea was then designed to desirably improve the water solubility and melting point from the early inhibitors (Kim et al., 2004, Morisseau et al., 2002). Among such series, 1-adamantanyl-3-{5-[2-(2-ethoxyethoxy)ethoxy]pentyl}urea (AEPU), N-adamantyl-N′-dodecylurea (ADU), 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA) and its butyl ester (AUDA-BE) have been widely tested in animal models for anti-inflammation (Schmelzer et al., 2005, Smith et al., 2005), analgesic (Inceoglu et al., 2006, Schmelzer et al., 2006), anti-hypertension (Imig et al., 2005, Loch et al., 2007, Revermann et al., 2009), renal-protection (Huang et al., 2007, Parrish et al., 2009), and cardio-protection (Dorrance et al., 2005, Xu et al., 2006). These compounds have improved water solubility and melting points in comparison to the earlier sEHIs such as DCU and CDU. However, they were rapidly metabolized and excreted due to the facile oxidation of their flexible alkyl chain, which limits their use in vivo. Therefore, conformationally-restricted sEHIs such as 1-(1-acetypiperidin-4-yl)-3-adamantanylurea (APAU) and trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB) were synthesized based on the hypothesis that conformationally-restricted compounds will eliminate beta oxidation, resulting in improved metabolic stability (Hwang et al., 2007, Jones et al., 2006). This hypothesis was supported by subsequent pharmacokinetics (PK) studies in murine and canine models (Liu et al., 2009, Tsai et al., 2010).

In several biological studies both the cis- and trans-isomers of AUCB demonstrated anti-inflammatory (Liu et al., 2009, Manhiani et al., 2009, Rose et al., 2010), anti-hypertensive (Revermann et al., 2009), cardio-protective (Ai et al., 2009, Chaudhary et al., 2010, Li et al., 2009) and renal-protective effects (Manhiani et al., 2009). However, the adamantyl group has been shown in pharmacology to be metabolically unstable (Lamoureux and Artavia, 2010). For example, the adamantyl group at t-AUCB gave an attractive PK profile for once a day oral dosing in dogs, but in other species it required higher and more frequent doses for efficacy. Interestingly, we found in previous studies that TPAU (containing 4-trifluoro-methoxy-phenyl group) is more metabolically stable than APAU (containing adamantyl group) (Liu et al., 2009, Tsai et al., 2010), suggesting a possibility that replacement of an adamantyl group by a substituted phenyl group may improve the metabolic stability of urea-based sEHIs. To test this hypothesis, a series of substituted phenyl urea-based compounds were synthesized. The PK profiles of five such inhibitors that afford favorable IC50s in vitro (Table 1) were then tested in a murine model at four different doses with single oral administration. Here we present the PK profiles of these compounds and the anti-inflammatory effect of 1-(4-trifluoro-methoxy-phenyl)-3-(1-propionylpiperidin-4-yl)urea (TPPU), the most promising compound among the five tested compounds in murine models.

Section snippets

Materials

Methanol, acetonitrile, and ethyl acetate were purchased from Fisher Scientific (Pittsburgh, PA). Acetic acid, polyethylene glycol (average molecular weight: 400, PEG400) and LPS (Escherichia coli serotype 0111:B4) were purchased from Sigma–Aldrich (St. Louis, NJ). EDTA(K3) was purchased from Tyco Health Group LP (Mansfield, MA). Water (>18.0 MΩ) was purified by a NANO pure system (Barnstead, Newton, MA). All the sEHIs used in this study were synthesized in this laboratory, and their structures

In vitro inhibitory potency of five inhibitors against human and murine sEHs

The structure and in vitro inhibitory activity of five urea-based sEH inhibitors containing substituted phenyl groups and two urea-based sEH inhibitors containing an adamantyl group are presented in Table 1. In regard to the in vitro potency against human sEH, substituted phenyl-containing compounds give lower IC50 values by the fluorescent assay than those by radioactive assay. Tsai et al. cautioned earlier that for some potent compounds, particular piperidine derivatives, the fluorescent

Discussion

The adamantane group was used in some previous studies from this laboratory because of its ease of detection on positive mode LC/MS/MS. This high sensitivity allowed rapid PK screening of analogs to optimize another side of the molecules for potency. However, adamantane group is rapidly hydroxylated in the presence of some cytochrome P450 enzymes. Previous PK studies in murine and canine models showed that TPAU, a substituted phenyl group-containing sEHI, is more metabolically stable than APAU,

Conclusion

In summary, this study shows that the substituted phenyl group-containing sEH inhibitor TPPU is better than the adamantane-containing t-AUCB in terms of PK properties and in vivo potency. This statement is supported by the improvement in the efficacy of TPPU over t-AUCB in attenuation of the LPS-challenged inflammation and the reverse in the shifts of epoxides, diols and epoxide/diol ratios as well as the suppression of the inflammation-associated cytokines. TPPU appears to be a better

Acknowledgements

This work was supported in part by the National Institute of Environmental Health Sciences (NIEHS) ES02710, and NIEHS Superfund P42 ES04699 to BDH. The work is also supported in part by NIH/NHLBI (R01 HL075274 and HL085844) and the VA Merit Review Grant to NC. BDH is a George and Judy Marcus Senior Fellow of the American Asthma Foundation. H.Q. was supported by American Heart Association Western Affiliates postdoctoral fellowship award.

References (40)

  • H.J. Tsai et al.

    Pharmacokinetic screening of soluble epoxide hydrolase inhibitors in dogs

    Eur. J. Pharm. Sci.

    (2010)
  • D. Ai et al.

    Soluble epoxide hydrolase plays an essential role in angiotensin II-induced cardiac hypertrophy

    Proc. Natl. Acad Sci. USA

    (2009)
  • K.R. Chaudhary et al.

    Inhibition of soluble epoxide hydrolase by trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid is protective against ischemia-reperfusion injury

    J. Cardiovasc. Pharm.

    (2010)
  • B.B. Davis et al.

    Inhibitors of soluble epoxide hydrolase attenuate vascular smooth muscle cell proliferation

    Proc. Natl. Acad Sci. USA

    (2002)
  • 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. Pharm.

    (2005)
  • R. Faggioni et al.

    LPS-induced anorexia in leptin-deficient (ob/ob) and leptin receptor-deficient (db/db) mice

    Am. J. Physiol.-Reg. I

    (1997)
  • H. Huang et al.

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

    Am. J. Physiol.-Renal.

    (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)
  • J.D. Imig et al.

    An orally active epoxide hydrolase inhibitor lowers blood pressure and provides renal protection in salt-sensitive hypertension

    Hypertension

    (2005)
  • Cited by (61)

    • Anandamide and other N-acylethanolamines: A class of signaling lipids with therapeutic opportunities

      2023, Progress in Lipid Research
      Citation Excerpt :

      Recently, attention has focused on sEH inhibition in the CNS, where EETs exert neuroprotective and anti-inflammatory effects. Development towards sEH inhibitors has afforded orally bioavailable compounds such as 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl)urea (TPPU), a subnanomolar inhibitor with low to moderate blood-brain barrier penetration (Fig. 5) [319]. Highlighting the significance of lipid epoxides in the CNS, sEH inhibition by TPPU decreased reperfusion injury after focal cerebral ischemia in rats, significantly reduced infarction, the inflammatory response and improved sensorimotor function [320,321].

    • 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) Urea Exerts Neuro-Protective Effects Against Ischemic Injury via Suppressing JNK/p38 MAPK-Mediated Mitochondrial Apoptosis Pathway

      2021, Journal of Stroke and Cerebrovascular Diseases
      Citation Excerpt :

      Inhibitors of sEH, including 12-(3-adamantan-1-yl-ureido) dodecanoic acid (AUDA), 1-trifluoromethoxyphenyl-3-(1-acetylpiperidin-4-yl) urea (TPAU), trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB), have also been shown to reduce infarct volume and peri-infarct cell death, attenuate the neuroinflammatory response following MCAO in mice and rats,26,27,28 and maintain mitochondrial function following cellular stress in myocardial ischemic model.16 TPPU, as a newer generation sEH inhibitor, offers greater metabolic stability, possessed longer circulatory half-life and higher potency than many of other sEH inhibitors in stroke models,17,18 and may protect against ischemic brain injury in rat by maintaining the integrity of blood-brain barrier and decreasing the number of apoptotic cells.19 However, the mitochondrial protection and mechanism of sEH inhibitors after cerebral ischemia remain to be determined.

    • Soluble Epoxide Hydrolase Hepatic Deficiency Ameliorates Alcohol-Associated Liver Disease

      2021, Cellular and Molecular Gastroenterology and Hepatology
    View all citing articles on Scopus
    View full text