<<

Bioorganic & Medicinal Letters 22 (2012) 5889–5892

Contents lists available at SciVerse ScienceDirect

Bioorganic & Medicinal Chemistry Letters

journal homepage: www.elsevier.com/locate/bmcl

Biologically active derivatives as potent inhibitors of the soluble hydrolase

In-Hae Kim , Kosuke Nishi , Takeo Kasagami à, Christophe Morisseau, Jun-Yan Liu, Hsing-Ju Tsai, ⇑ Bruce D. Hammock

Department of Entomology and UC Davis Comprehensive Cancer Center, University of California, One Shields Avenue, Davis, CA 95616, USA article info abstract

Article history: Substituted with a carboxylic ester as a secondary pharmacophore are potent soluble epoxide Received 15 May 2012 hydrolase (sEH) inhibitors. Although the ester imparts better physical properties, such com- Revised 18 July 2012 pounds are quickly metabolized to the corresponding less potent . Toward producing biologically Accepted 23 July 2012 active ester compounds, a series of were prepared and evaluated for potency on the human Available online 2 August 2012 , stability in human microsomes, and physical properties. Modifications around the ester function enhanced in vitro metabolic stability of the ester inhibitors up to 32-fold without a decrease Keywords: in inhibition potency. Further, several compounds had improved physical properties. sEH Ó 2012 Elsevier Ltd. All rights reserved. sEH inhibitors Substituted –ester derivatives Metabolic stability

Epoxyeicosatrienoic acids (EETs), which are metabolites of ara- sEHI chidonic acid by cytochrome P450 epoxygenases, have important O O 1–6 7–11 roles in the regulation of hypertension, inflammation, and OH sEH OH other cardiovascular related diseases.12,13 Recently a role in modu- 14 lating pain was also reported. However, of EETs (1) O HO OH to their corresponding hydrated products (2) by soluble epoxide 14,15-EET 14,15-DHET hydrolase (sEH) generally reduces these biological activities 1 2 (Fig. 1).1 Both in vitro and in vivo studies have demonstrated that the anti-hypertensive and cardio protective effects mediated by Figure 1. Metabolism of biologically active EETs (1; e.g., 14,15-EET) to the less the EETs are reversely dependent on the extent of sEH active DHETs (2; e.g., 14,15-DHET) by sEH. The sEH converts a variety of lipid of the EETs.2–4,6–8,13,15 Thus, maintaining the in vivo concentration to their corresponding diols. The sEHIs act by blocking this enzyme, increasing the in vivo concentrations of EETs which have direct actions on of EETs through sEH inhibition is a promising therapeutic pathway hypertension, inflammation, and pain. EET, epoxyeicosatrienoic acid; DHET, to treat cardiovascular and other diseases. Urea, , and carba- dihydroxyeicosatrienoic acid; sEHI, soluble epoxide hydrolase inhibitor. mate compounds substituted with hydrophobic groups are very potent and stable inhibitors of sEH with significant biological activ- ities in both in vitro and in vivo models.3,4,16 However, poor phys- one of the urea-substitution is effective in increasing in ical properties of the early compounds, such as low and either or organic solvents, and also in improving in vivo high melting points, likely result in limited in vivo availability.17 availability while maintaining the inhibition potency on the target The addition of a polar on specific positions of enzyme.18–21 Among a variety of polar functional groups explored previously, ester functionalities located on the fifth atom from the urea function were found to be among the most suitable polar Abbreviations: sEH, soluble epoxide hydrolase; EET, epoxyeicosatrienoic acid; EDCI, 1-[3-(dimethylamino)propyl]-3-ethylcarbodiimide; DMAP, 4-dimethyl- groups for making potent inhibitors with improved physical prop- 18 . erties. However, as expected, the ester functionality is rapidly ⇑ Corresponding author. Tel.: +1 530 752 7519; fax: +1 530 752 1537. hydrolyzed both in vitro and in vivo producing the corresponding E-mail address: [email protected] (B.D. Hammock). short chain acid metabolite that has dramatically reduced inhibi-

Present address: Department of Applied Bioscience, Faculty of Agriculture, Ehime tion potency on the human sEH.20 Although rapid enzymatic University, 3-5-7 Tarumi, Matsuyama, Ehime 790-8566, Japan. à Present address: Drug Development Service Segment, Mitsubishi Chemical hydrolysis of the ester compound is not favorable for high in vivo Medience Corporation, 14-1 Sunayama Kamisu-shi, Ibaraki 314-0255, Japan. efficacy, it offers some advantage. First, moieties of ester

0960-894X/$ - see front matter Ó 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.bmcl.2012.07.074 5890 I.-H. Kim et al. / Bioorg. Med. Chem. Lett. 22 (2012) 5889–5892

(A) HO O O O d

O O OH a O O N N N N O H H H H 3 O O 9

a (b, c)

O O OR1 O N N N N H H H H O R OCN 4: R= H, R1= n-Butyl 7 5: R= Methyl, R1= n-Butyl 6: R= Ethyl, R1= n-Butyl 8: R= H, R1= (2-Ethoxyethoxy)methyl

(B)

O O a OH OR1 N N N N H H H H 10 O O R 11: R= H, R1= n-Butyl 12: R= Ethyl, R1= n-Butyl 13: R= Phenyl, R1= (2-Ethoxyethoxy)methyl

Scheme 1. Syntheses of 4-(3-adamantan-1-yl-ureido)-butyric acid or -cyclohexanecarboxylic acid derivatives. Reagents and conditions: (a) EDCI, DMAP, a corresponding alcohol (2-hexanol for compound 5, 3-heptanol for compounds 6 and 12, and diethylene glycol monoethyl for compounds 8 and 13) in MC or 1-bromopentane for compounds 4 and 11 in DMF, rt; (b) pentanal, TMSCN, ZnI, chloroform, rt/3 N HCl; (c) coupling of compound 3 with the product of reaction; (b) by using EDCE/DMAP in MC, rt; (d) 2-ethoxyethanol, NaH, DMF, rt.

groups can be rapidly altered to explore binding properties of the prepared by the reaction of 2-ethoxyethanol with 2-phenyloxirane, right hand side of the inhibitors such as those shown in Scheme 1. was coupled with compound 3 in the above same method as that Second, the introduction of a secondary pharmacophore roughly used for the preparation of compound 7 to afford compound 9 in 7–8 Å from the urea or amide carbonyl of the primary pharmaco- 48% yield. Scheme 1(B) shows syntheses of derivatives with a phore often improves solubility while retaining high po- cyclohexyl linker (10–13) between the urea and ester pharmaco- tency.18,21–23 Finally, to date the urea inhibitors of the sEH phores.21 Compound 10, which was prepared from the coupling appear highly selective with low toxicity. The epoxy-lipids stabi- lized by these inhibitors have numerous biological activities. Although the activities of the epoxy-lipids are so far found to be Table 1 beneficial, it may be important to direct the activities to individual Inhibition of human sEH by 4-(3-adamantan-1-yl-ureido)-butyric acid derivatives tissues. Administering short acting compounds may be also impor- O tant for the treatment of pulmonary inflammation by inhalation. O N N R Thus for these latter two approaches it is important to develop es- H H ters of varying metabolic stability with increasing stability for clas- O sical systemic administration and decreasing stability for short 24 a term or local effects. In the present study, we explored structural Compound R Human sEH IC50 (nM) modifications of urea–ester inhibitors toward altering the enzy- 3 H 684 matic stability of the ester function in an in vitro metabolic system 4 10 while maintaining sEH inhibition potency and appropriate physical 5 4 properties. 18,21 Compound 3, which was prepared as previously reported, 6 4 was coupled with a corresponding alcohol by using 1-[3-(dimeth- ylamino)propyl]-3-ethylcarbodiimide (EDCI) in the presence of 4- 7 12 dimethylaminopyridine (DMAP) in dichloromethane to afford CN O compounds 5, 6, and 8 or was alkylated with 1-bromopentane in 8 O 67 the presence of K CO in DMF to afford compound 4 in a range of O 2 3 O 64–95% yield (Scheme 1(A)). For the synthesis of cyanoalkyl ester 9 4 (7), 1-pentanal was first reacted with trimethylsilyl cyanide in the presence of zinc iodide in chloroform at 0 °C, followed by a hydrolysis with an aqueous of 3 N HCl (80%). The resulting Human sEH (0.96 nM) was incubated with inhibitors for 5 min in 25 mM Bis– /HCl buffer (200 lL; pH 7.0) at 30 °C before fluorescent substrate introduction cyanohydrin intermediate was then coupled with compound 3 by ([S] = 5 lM). Results are averages of three separate measurements. The fluorescent using EDCI and DMAP in dichloromethane to provide 7 in 70% assay as performed here has a standard error between 10% and 20%, suggesting that yield. In addition, ethoxyethanol with a , which was differences of 2-fold or greater are significant. I.-H. Kim et al. / Bioorg. Med. Chem. Lett. 22 (2012) 5889–5892 5891 of 1-adamantyl with 3-aminocyclohexanoic acid, was was synthesized as seen in compound 10 in Table 2, alkylated or coupled with a corresponding bromide or alcohol in further reduced inhibition was exhibited. This indicates that a the same reaction condition as that used for the preparation of short chain acid attached to the urea function like urea–cyclo- compounds 4–9 in Scheme 1(A) to give target compounds 11–13 hexylcarboxylic acid is a poor inhibitor and substitution of the acid in 65–95% yield.21 could also result in potent compounds. When cyclohexylcarboxylic Five derivatives (5–9) were prepared to first explore the modi- acid esters were prepared as seen in Table 2, as expected, a dra- fication effect of the alcohol moiety of the ester function of com- matic improvement in inhibition on the target human enzyme pound 4 on inhibition potency. As shown in Table 1, the was observed in compound substituted with a (11). incorporation of a (5) on the alpha of the alco- The incorporation of an (12) on the alpha carbon of hol moiety showed a 2.5-fold improvement in inhibition potency the alcohol moiety of the pentyl ester 11 provided a highly potent compared to compound 4. An ethyl group (6) substituted on the al- inhibitor, as well. A diethyleneglycol group with a phenyl also pha carbon of compound 4 exhibited a similar enhancement in po- made a potent inhibitor (13), although it was less potent than com- tency, indicating that an substituent on the alpha position is pounds 11 or 12. The results in Table 2 imply that alkylation of the effective for improving potency. On the other hand, the introduc- acid function of urea–cyclohexylcarboxylic acid (10) is effective for tion of a cyano group (7) induced a 3-fold decrease in inhibition producing highly potent inhibitors like butyrate derivatives in potency in comparison with that of methyl (5) or ethyl (6) groups. Table 1. This suggests that hydrophobic are better than a polar In general, liphophilicity of compounds causes limited solubility group to be incorporated on the alpha carbon of the alcohol moiety in water, which probably affects in vivo efficacy of com- of the ester function for enhanced inhibition potency. In a previous pounds.17,21,25 In addition, the stability of the crystals of com- report, we demonstrated that diethylene glycol groups attached to pounds, indicated by their high melting points, leads to a general hydrophobic urea inhibitors are useful for improving water solubil- lack of solubility even in organic solvents. These poor physical ity and bioavailability without a loss in inhibition potency.21 Two properties result in undesirable pharmacokinetic properties and derivatives with a diethylene glycol group (8 and 9) were prepared difficulty in compound formulation in either an aqueous or oil to investigate whether the polar glycol group affects inhi- . We examined physical properties of the above potent deriv- bition potency of urea–ester compounds. As illustrated by com- atives in Tables 1 and 2. As seen in Table 3, butyric acid (3) and the pound 8, attachment of a diethylene glycol resulted in a 7- to 17- corresponding ester derivatives (4 and 6) had very low solubility in fold reduction in inhibition when compared to those of non-substi- water, while dramatic improvement in solubility was observed in tuted (4) and alkyl substituted compounds (5 and 6). However, diethylene glycol compounds (8 and 9). Similar increase in solubil- interestingly, a big improvement in inhibition was obtained by ity was obtained in cyclohexylcarboxylic acid ester with a diethyl- incorporating a phenyl group (9) on the alpha carbon of the alcohol ene glycol group (13). The diethylene glycol derivatives (8, 9, and moiety of compound 8, which was as potent as that of the alkyl 13) also possessed much lower melting points in comparison with derivatives (5 and 6). This further indicates that a hydrophobic the acids (3 and 10) or other ester compounds (4, 6, and 12). This substituent on the alpha carbon of the ester function is highly result indicates that hydrophilic alcohol moiety of the ester func- effective to lead to potent urea–butyrate compounds as inhibitors tion of compounds 9 and 13 is highly beneficial for improving of the human sEH. As described, the corresponding acid compound physical properties while producing potent inhibition. In order to (3) of butyrate derivatives in Table 1 was poor as an inhibitor of the see in vitro metabolism of the urea-ester compounds we used hu- human enzyme. In addition, when a corresponding cyclohexyl- man liver microsomes, which contain various kinds of such as esterases, amidases, and cytochrome P450s playing a pri- mary role in drug metabolism. When compounds 3 and 10 were

Table 2 Inhibition of human sEH by 3-(3-adamantan-1-yl-ureido)-cyclohexylcarboxylic acid derivatives Table 3 Physical properties and in vitro metabolic stability of 4-(3-adamantan-1-yl-ureido)- Compound Structure Human butyric acid or -cyclohexylcarboxylic acid derivatives sEH a b a Compound Solubility in water Mp (°C) Stability IC50 (nM) (lg/mL) (% remaining) O 3 7 165 97 OH 10 N N 1160 4 2 114 <1 H H 6 3724 O 8 878 Oil 10 O 9 89 Oil 14 O 10 NDc >250 95 11 N N 2 H H 12 1 124 10 O 13 41 oil 32

O a An excess of the test compound was added to a vial containing phos- O phate buffer, 0.1 M pH 7.4 (1 mL), and a suspension of the mixture was equilibrated 12 N N 3 H H during 1 h of sonication and 24 h of shaking, followed by centrifugation. The water O supernatant was analyzed by LC–MS/MS. A regression curve for each compound O was obtained from five standard stock by using LC–MS/MS. Then, the 19 O O absolute amount of each compound was calculated. N N O b Human liver microsomal (0.125 mg) was incubated with a solution of 13 H H 9 O test compound under a NADPH generating system. Incubation mixture was shaken at 37 °C for 60 min. The absolute quantity of parent compounds remaining after the incubation was analyzed by LC–MS/MS and was converted to a percentage. The a Human sEH (0.96 nM) was incubated with inhibitors for 5 min in 25 mM Bis– corresponding acid metabolites (3 and 10) of ester compounds were analyzed in all Tris/HCl buffer (200 lL; pH 7.0) at 30 °C before fluorescent substrate introduction determinations (data not shown). The results given are averages of triplicate ([S] = 5 lM). Results are averages of three separate measurements. The fluorescent independent analyses. c assay as performed here has a standard error between 10% and 20%, suggesting that Not-determined because the crystal of compound 10 was not dissolved in water differences of 2-fold or greater are significant. which seems to be due to the high (>250 °C). 5892 I.-H. Kim et al. / Bioorg. Med. Chem. Lett. 22 (2012) 5889–5892 tested for microsomal incubation, >95% of the parent acids were Acknowledgments determined. This implies that the adamantylurea–acid structure of compounds 3 and 10 are stable under the incubation conditions This work was supported in part by NIEHS Grant R01 ES02710, and the human liver microsomal incubation is useful to investigate NIEHS Center for Environmental Health Sciences P30 ES05707, relative in vitro metabolic stability of urea–ester compounds. As NIH/NIEHS Superfund Basic Research Program P42 ES04699, NIH/ expected, pentyl ester of butyric acid (4) was all metabolized to NHLBI R01 HL59699-06A1, and UCDMC Translational Technology the corresponding butyric acid (3) in the incubation. On the other Research Grant. B.D.H. is a George and Judy Marcus Senior Fellow hand, a little increased stability was shown in compound 6 with an of the American Asthma Foundation. ethyl branch on the alpha carbon of the ester function. Further, diethylene glycol compounds (8 and 9) had >2.5-fold improved Supplementary data metabolic stability compared to compound 6, suggesting that a po- lar alcohol moiety of the ester function is more effective for Supplementary data associated with this article can be found, in improving the ester stability over a hydrophobic alkyl group. Inter- the online version, at http://dx.doi.org/10.1016/j.bmcl.2012.07. estingly, cyclohexylcarboxylic acid ester derivatives (12 and 13) 074. exhibited much higher metabolic stability than the corresponding butyric acid esters (6 and 9). Comparing both compounds with an References and notes ethyl branch on the alpha carbon (6 and 12) a 2.5-fold increase in stability was observed in the cyclohexane derivative (12). Approx- 1. Capdevila, J. H.; Falck, J. R.; Harris, R. C. J. Lipid Res. 2000, 41, 163. imately a 2-fold improved stability was also shown in compound 2. Yu, Z.; Xu, F.; Huse, L. M.; Morisseau, C.; Draper, A. J.; Newman, J. W.; Parker, C.; Graham, L.; Engler, M. M.; Hammock, B. D.; Zeldin, D. C.; Kroetz, D. L. Circ. Res. 13 compared to that of the corresponding ethyleneglycol butyric 2000, 87, 992. acid ester (9), indicating that sterically rigid cyclohexane linker 3. Imig, J. D.; Zhao, X.; Capdevila, J. H.; Morisseau, C.; Hammock, B. D. Hypertension present between the urea primary pharmacophore and the ester 2002, 39, 690. 4. Zhao, X.; Yamamoto, T.; Newman, J. W.; Kim, I.-H.; Watanabe, T.; Hammock, B. secondary pharmacophore is highly effective for increasing the es- D.; Stewart, J.; Pollock, J. S.; Pollock, D. M.; Imig, J. D. J. Am. Soc. Nephrol. 2004, ter stability. The results in Table 3 showed that solubility and sta- 15, 1244. bility of urea–ester compounds are varied up to 390-fold and 32- 5. Jung, O.; Brandes, R. P.; Kim, I. H.; Schweda, F.; Schmidt, R.; Hammock, B. D.; fold, respectively, through incorporation of a substituent and/or Busse, R.; Fleming, I. Hypertension 2005, 45, 759. 6. Imig, J. D.; Zhao, X.; Zaharis, C. Z.; Olearczyk, J. J.; Pollock, D. M.; Newman, J. W.; modification in the linker structure. Kim, I. H.; Watanabe, T.; Hammock, B. D. Hypertension 2006, 46, 975. In conclusion, we synthesized a series of urea-ester compounds 7. Smith, K. R.; Pinkerton, K. E.; Watanabe, T.; Pedersen, T. L.; Ma, S. J.; Hammock, as inhibitors for the human soluble epoxide hydrolase (sEH) to pro- B. D. Proc. Natl. Acad. Sci. U.S.A. 2005, 102, 2186. 8. Schmelzer, K. R.; Kubala, L.; Newman, J. W.; Kim, I. H.; Eiserich, J. P.; Hammock, duce compounds of varying metabolic stability by improving the B. D. Proc. Natl. Acad. Sci. U.S.A. 2005, 102, 9772. biodegradable ester function while possessing potent inhibition 9. Node, K.; Huo, Y.; Ruan, X.; Yang, B.; Spiecker, M.; Ley, K.; Zeldin, D. C.; Liao, J. K. and improved physical properties. The SAR study showed that Science 1999, 285, 1276. 10. Campbell, W. B. Trends Pharmacol. Sci. 2000, 21, 125. the incorporation of an alkyl group on the alpha carbon of the alco- 11. Yang, B.; Graham, L.; Dikalov, S.; Mason, R. P.; Falck, J. R.; Liao, J. K.; Zeldin, D. C. hol moiety of the ester increased inhibition potency on the sEH, Mol. Pharmacol. 2001, 60, 310. while a polar group in the alcohol moiety resulted in a big decrease 12. Node, K.; Ruan, X.; Dai, J.; Yang, S.; Graham, L.; Zeldin, D. C.; Liao, J. K. J. Biol. Chem. 2001, 276, 15983. in potency. However, interestingly, a substituted polar group with 13. Xu, D.; Li, N.; He, Y.; Timofeyev, V.; Lu, L.; Tsai, H.-J.; Kim, I. H.; Tuteja, D.; a hydrophobic function made potent inhibitors (9 and 13). More- Mateo, R. K. P.; Singapuri, A.; Davis, B. B.; Low, R.; Hammock, B. D.; over, the potent polar compounds (9 and 13) exhibited approxi- Chiamvimonvat, N. Proc. Natl. Acad. Sci. U.S.A. 2006, 103, 18733. 14. Inceoglu, B.; Wagner, K.; Schebb, N. H.; Morisseau, C.; Jinks, S. L.; Ulu, A.; mately a 20- to 45-fold improved water solubility and low Hegedus, C.; Rose, T.; Brosnan, R.; Hammock, B. D. Proc. Natl. Acad. Sci. U.S.A. melting points when compared to those of alkyl substituted inhib- 2011, 108, 5093. itors (4 or 6). Because we found that adamantylureas with a short 15. Moghaddam, M. F.; Grant, D. F.; Cheek, J. M.; Greene, J. F.; Williamson, K. C.; chain acid (3 and 10 in Table 3) are not metabolized in incubation Hammock, B. D. Nat. Med. 1997, 3, 562. 16. Newman, J. W.; Denton, D. L.; Morisseau, C.; Koger, C. S.; Wheelock, C. E.; with human liver microsomal enzymes, this incubation was used Hinton, D. E.; Hammock, B. D. Environ. Health Perspect. 2001, 109, 61. to investigate relative ester bond stability of urea–ester com- 17. Morisseau, C.; Goodrow, M. H.; Newman, J. W.; Wheelock, C. E.; Dowdy, D. L.; pounds. Among the modified urea–ester derivatives, the highest Hammock, B. D. Biochem. Pharmacol. 2002, 63, 1599. 18. Kim, I. H.; Morisseau, C.; Watanabe, T.; Hammock, B. D. J. Med. Chem. 2004, 47, improvement (32-fold) in stability was obtained from a cyclohexyl 2110. analogue with a polar substituent (13). Approximately a 2-fold 19. Kim, I. H.; Heirtzler, F. R.; Morisseau, C.; Nishi, K.; Tsai, H. J.; Hammock, B. D. J. lower stability was observed when the cyclohexyl linker of com- Med. Chem. 2005, 48, 3621. 20. Kim, I. H.; Nishi, K.; Tsai, H.-J.; Bradford, T.; Koda, Y.; Watanabe, T.; Morisseau, pound 13 was replaced by an alkyl chain linker (9). In compounds C.; Blanchfield, J.; Toth, I.; Hammock, B. D. Bioorg. Med. Chem. 2007, 15, 312. with a cyclohexyl linker (12) or polar group (8), a 3-fold decreased 21. (a) Kim, I. H.; Tsai, H.-J.; Nishi, K.; Kasagami, T.; Morisseau, C.; Hammock, B. D. J. stability was shown compared to that of 13. A dramatic decrease in Med. Chem. 2007, 50, 5217; (b) Kasagami, T.; Kim, I. H.; Tsai, H.-J.; Nishi, K.; Hammock, B. D.; Morisseau, C. Bioorg. Med. Chem. Lett. 2009, 19, 1784. stability was exhibited in butyrate derivatives with an alkyl substi- 22. Kim, I. H.; Park, Y. K.; Hammock, B. D.; Nishi, K. J. Med. Chem. 2011, 54, 1752. tution (4 and 6). The overall structural modifications altered the 23. (a) Anandan, S.-K.; Gless, R. D. Bioorg. Med. Chem. Lett. 2010, 20, 2740; (b) Shen, relative metabolism of the ester inhibitors up to 32-fold without H. C.; Ding, F.-X.; Wang, S.; Xu, S.; Chen, H.-S.; Tong, X.; Tong, V.; Mitra, K.; Kumar, S.; Zhang, X.; Chen, Y.; Zhou, G.; Pai, L.-Y.; Alonso-Galicia, M.; Chen, X.; a decrease in inhibition potency. Further, several compounds had Zhang, B.; Tata, J. R.; Berger, J. P.; Colletti, S. Bioorg. Med. Chem. Lett. 2009, 19, improved physical properties. These findings will facilitate devel- 3398. opment of biologically active urea–ester compounds as potent 24. Richard, B. S. The of Drug Design and Drug Action, 2nd ed.; inhibitors of the sEH. The ester compounds described in this study Elsevier Academic Press, 2004. pp 471–473. 25. Hwang, S. H.; Tsai, H.-J.; Liu, J.-Y.; Morisseau, C.; Hammock, B. D. J. Med. Chem. could be particularly applicable for administration directly to in- 2007, 50, 3825. flamed tissue by inhalation for chronic obstructive pulmonary dis- 26. Pillarisetti, S.; Khanna, I. Inflamm. Allergy Drug Targets 2012, 11, 143. order (COPD) or asthma. Alternatively they could be given by slow 27. Gross, G. J.; Gauthier, K. M.; Moore, J.; Falck, J. R.; Hammock, B. D.; Campbell, 26 W. B.; Nithipatikom, K. Am. J. Physiol. Heart Circ. Physiol. 2008, 294, H2838. release formulations to address inflammatory bowel diseases. 28. Shen, H.; Hammock, B. D. J. Med. Chem. 2012, 55, 1789. sEH inhibitors have been shown to be active on a variety of cardio- vascular, inflammatory, and pain related disorders in rodents.27,28