Title of Invention

IMPROVED INHIBITORS FOR THE SOLUBLE EPOXIDE HYDROLASE

Abstract Inhibitors of the soluble epoxide hydrolase (sEH) are provided that incorporate multiple pharmacophores and are useful in the treatment of diseases.
Full Text WO 2006/045119 PCT/US2005/038282
IMPROVED INHIBITORS FOR THE SOLUBLE EPOXIDE
HYDROLASE
CROSS-REFERENCE TO RELATED APPLICATION
[0001] This application claims the benefit of U.S. Patent Application No. 60/651,487, filed
October 20, 2004, the content of which is incorporated herein by reference.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER
FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] The U.S. Government has certain rights to the invention pursuant to contract
ES02710 awarded by the National Institutes of Health.
REFERENCE TO A "SEQUENCE LISTING," A TABLE, OR A COMPUTER
PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK.
[0003] NOT APPLICABLE
BACKGROUND OF THE INVENTION
[0004] Epoxide hydrolases (EHs, EC 3.3.2.3) catalyze the hydrolysis of epoxides or arene
oxides to their corresponding diols by the addition of water (see, Oesch, F., et al.,
Xenobiotica 1973, 3, 305-340). Some EHs play an important role in the metabolism of a
variety of compounds including hormones, chemotherapeutic drugs, carcinogens,
environmental pollutants, mycotoxins, and other harmful foreign compounds.
[0005] There are two well-studied EHs, microsomal epoxide hydrolase (mEH) and soluble
epoxide hydrolase (sEH). These en2ymes are very distantly related, have different
subcellular localization, and have different but partially overlapping substrate selectivities.
The soluble and microsomal EH forms are known to complement each other in degrading
some plant natural products (see, Hammock, B.D., et al., COMPREHENSIVE
TOXICOLOGY. Oxford: Pergamon Press 1977,283-305 and Fretland, A.J., et al., Chem.
Biol. Intereract 2000,129, 41-59).
[0006] The major role of the sEH is in the metabolism of lipid epoxides including the
metabolism of arachidonic acid (see, Zeldin, D.C., et al., J. Biol. Chem. 1993, 268, 6402-

WO 2006/045119 PCT/US2005/038282
2
6407), linoleic (see, Moghaddam, M.F., et al, Nat. Med. 1991,3, 562-567) acid, some of
which are endogenous chemical mediators (see, Carroll, M.A., et al., Thorax 2000,55, S13-
16). Epoxides of arachidonic acid (epoxyeicosatrienoic acids or EETs) and other lipid
epoxides and diols are known effectors of blood pressure (see, Capdevila, J.H., et al., J. Lipid.
Res. 2000, 41,163-181), and modulators of vascular permeability (see, Oltman, C.L., et al.,
Circ Res. 1998, 83, 932-939). The vasodilatory properties of EETs are associated with an
increased open-state probability of calcium-activated potassium channels leading to
hyperpolarization of the vascular smooth muscle (see Fisslthaler, B., et al., Nature 1999,401,
493-497). Hydrolysis of the arachidonate epoxides by sEH diminishes this activity (see,
Capdevila, J.H., et al., J. Lipid. Res. 2000, 41,163-181). sEH hydrolysis of EETs also
regulates their incorporation into coronary endothelial phospholipids, suggesting a regulation
of endothelial function by sEH (see, Weintraub, N.L., et al., Am. J. Physiol. 1992, 277,
H2098-2108), It has recently been shown that treatment of spontaneous hypertensive rats
(SHRs) with selective sEH inhibitors significantly reduces their blood pressure (see, Yu, Z.,
et al., Circ. Res. 2000, 87, 992-998). In addition, male knockout sEH mice have significantly
lower blood pressure than wild-type mice (see Sinal, C.J., et al., J. Biol. Chem. 2000,275,
40504-405010), further supporting the role of sEH in blood pressure regulation.
[0007] The EETs have also demonstrated anti-inflammatory properties in endothelial cells
(see, Node, K., et al., Science 1999, 285,1276-1279 and Campbell, W.B. Trends Pharmacol.
Sci. 2000, 21,125-127). In contrast, diols derived from epoxy-linoleate (leukotoxin) perturb
membrane permeability and calcium homeostasis (see, Moghaddam, M.F., et al., Nat. Med.
1997, 3, 562-567), which results in inflammation that is modulated by nitric oxide synthase
and endothelin-1 (see, Ishizaki, T.? et al., Am. J. Physiol. 1995,269, L65-70 and Ishizaki, T.,
et al., J. Appl. Physiol. 1995, 79, 1106-1611). Micromolar concentrations of leukotoxin
reported in association with inflammation and hypoxia (see, Dudda, A., et al., Chem. Phys.
Lipids 1996, 82, 39-51), depress mitochondrial respiration in vitro (see, Sakai, T., et al., Am.
J. Physiol. 1995, 269, L3 26-3 31), and cause mammalian cardiopulmonary toxicity in vivo
(see, Ishizaki, T., et al., Am. J. Physiol. 1995, 269, L65-70; Fukushima, A., et al., Cardiovasc.
Res. 1988, 22, 213-218; and Ishizaki, T., et al., Am. J. Physiol. 1995, 268, L123-128).
Leukotoxin toxicity presents symptoms suggestive of multiple organ failure and acute
respiratory distress syndrome (ARDS) (see, Ozawa, T. et al., Am. Rev. Respir. Dis. 1988,
137, 535-540). In both cellular and organismal models, leukotoxin-mediated toxicity is
dependent upon epoxide hydrolysis (see, Moghaddam, M.F., et al., Nat. Med. 1997, 3, 562-

WO 2006/045119 PCT/US2005/038282
3
567; Morisseau, C., et al., Proc. Natl. Acad. Sci USA 1999, 96, 8849-8854; and Zheng, J., et
al., Am. J. Respir. CellMol. Biol. 2001, 25,434-438), suggesting a role for sEH in the
regulation of inflammation and vascular permeability. The bioactivity of these epoxy-fatty
acids suggests that inhibition of vicnal-dihydroxy-lipid biosynthesis may have therapeutic
value, making sEH a promising pharmacological target.
[0008] Recently, 1,3-disubstituted ureas, carbamates, and amides have been reported as
new potent and stable inhibitors of sEH (Figure 1). See, U.S. Patent No. 6,150,415.
Compounds 192 and 686 are representative structures for this type of inhibitors (Figure 1).
These compounds are competitive tight-binding inhibitors with nanomolar KJ values that
interact stoichiometrically with purified recombinant sEH (see, Morisseau, C, et al., Proc.
Natl. Acad. Sci. USA 1999, 96, 8849-8854). Based on the X-ray crystal structure, the urea
inhibitors were shown to establish hydrogen bonds and to form salt bridges between the urea
function of the inhibitor and residues of the sEH active site, mimicking features encountered
in the reaction coordinate of epoxide ring opening by this enzyme (see, Argiriadi, M.A., et
al., Proc. Natl. Acad. Sci. USA 1999, 96,10637-10642 and Argiriadi, M.A., et al., J. Biol.
Chem. 2000, 275,15265-15270). These inhibitors efficiently reduced epoxide hydrolysis in
several in vitro and in vivo models (see, Yu, Z., et al., Circ. Res. 2000, 87, 992-998;
Morisseau, C, et al., Proc. Natl. Acad. Sci. USA 1999, 96, 8849-8854; and Newman, J.W., et
al., Environ. Health Perspect. 2001, 109, 61-66). Despite the high activity associated with
these inhibitors, there exists a need for compounds possessing similar or increased activities,
with improved solubility and pharmacokinetic properties to facilitate formulation and
delivery.
[0009] Surprisingly, the present invention provides such compounds along with methods
for their use and compositions that contain them.
BRIEF SUMMARY OF THE INVENTION
[0010] In one aspect, the present invention provides a method for inhibiting a soluble
epoxide hydrolase, comprising contacting the soluble epoxide hydrolase with an inhibiting
amount of a compound having a formula selected from the group consisting of:


WO 2006/045119 PCT/US2005/038282
4
and their pharmaceutically acceptable salts, wherein the symbol; R1 is a member selected
from the group consisting of substituted or unsubstituted alkyi, substituted or unsubstituted
heteroalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted
cycloalkylheteroalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted
arylheteroalkyl, substituted or unsubstituted C5-C12 cycloalkyl, substituted or unsubstituted
aryl, substituted or unsubstituted heteroaryl and combinations thereof, wherein said
cycloalkyl portions are monocyclic or polycyclic; P1 is a primary pharmacophore selected
from the group consisting of -OC(O)O-, -OC(O)CH2-, CH2C(O)O-, -OqO)-, -C(O)O-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-NHC(O)NH-,
-OC(O)NH-, -NHC(O)O-,-NHC(S)NH-, -NHC(S)CH2, CH2C(S)NH-, -SC(O)CH2-, -
CH2C(O)S-, -SC(NH)CH2-; - CH2C(NH)S-, -NON-, -CH2C(O)NH-, -NHC(O)CH2-;
-C(O)NH-, -NHC(O)-,

P2 is a secondary pharmacophore selected from the group consisting of -NH-, -OC(O)O-
,-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -NHC(NH)NH-, -NHC(NH)CH2-,
-CH2C(NH)NH-, -NHC(O)KB-, -OC(O)NH-, -NHC(O)O-, -C(O)NH-, -NHC(O)-; -
NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, -

WO 2006/045119 PCT/US2005/038282
5
CH2C(NH)S-, -N=C=N-,

P3 is a tertiary pharmacophore selected from the group consisting of C2-C6 alkenyl, C2-C6
alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, -O(CH2CH2O)q-R2, - OR2,
-C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4a1kyl-C(O)OR2, -C(O)R2, -C(O)OR2
and carboxylic acid analogs, wherein R2 is a member selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl; L1 is a first linker selected from the group
consisting of substituted or unsubstituted C1-C6 alkylene, substituted or unsubstituted C3-C6
cycloalkylene, substituted or unsubstituted arylene and substituted or unsubstituted
heteroarylene; L2 is a second linker selected from the group consisting of substituted or
unsubstituted C1-C12 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, substituted
or unsubstituted arylene, substituted or unsubstituted heteroarylene; an amino acid, a
dipeptide and a dipeptide analog; and combinations thereof; or is selected from the group
consisting of H and CH3 when m is 0. In the above formulae, the subscripts n and m are each
independently 0 or 1, and at least one of n or m is 1, and the subscript q is 0 to 6. When n is
0, then L1 and L2 are combined. When m is 0, then L2 can also be H.

WO 2006/045119 PCT/US2005/038282
6
[0011] Turning next to the linking groups, the symbol L1 represents a first linker that is a
substituted or unsubstituted C1-C6 alkylene, C3-C6-cycloalkylene, or an arylene or
heteroarylene group; the symbol L2 represents a second linker selected from substituted or
unsubstituted C1-C12 alkylene, substituted or unsubstituted arylene, an amino acid, a
dipeptide, a dipeptide analog, and combinations thereof; or is H when m is 0.
[0012] In a related aspect, the present invention provides methods of treating diseases
modulated by soluble epoxide hydrolases, the method comprising administering to a subject
in need of such treatment an effective amount of a compound having a formula selected from
formula (I), above.
[0013] In other aspects, the present invention provides methods of reducing renal
deterioration in a subject, the method comprising administering to the subject an effective
amount of a compound of formula (I), above.
[0014] In a related aspect, the present invention provides methods method for inhibiting
progression of nephropathy in a subject, the method comprising administering to the subject
an effective amount of a compound of formula (I), above.
[0015] In another aspect, the present invention provides for reducing blood pressure in a
subject, the method comprising administering to the subject an effective amount of a
compound of formula (I), above.
[0016] In a related aspect, the present invention provides methods of inhibiting the
proliferation of vascular smooth muscle cells in a subject, the method comprising
administering to the subject an effective amount of a compound of formula (I), above.
[0017] In another aspect, the present invention provides methods of inhibiting the
progression of an obstructive pulmonary disease, an interstitial lung disease, or asthma in a
subject, the method comprising administering to the subject an effective amount of a
compound of formula (T), above. The obstructive pulmonary disease can be, for example,
chronic obstructive pulmonary disease ("COPD"), emphysema, or chronic bronchitis. The
interstitial lung disease can be, for example, idiopathic pulmonary fibrosis, or one associated
with occupational exposure to a dust
[0018] In yet another aspect, the present invention provides compounds having a formula
(I) above, as well as pharmaceutical compositions containing one or more of the subject
compounds.

WO 2006/045119 PCT/US2005/038282
7
BRIEF DESCRIPTION OF THE DRAWINGS
[0019] Figure 1 provides structures of known sEH inhibitors having only a primary
pharmacophore: l-adamantyl-3-cyclohexylurea (192), l-adamantyl-3-dodecylurea (686).
[0020] Figure 2 provides a structural diagram defining the sEH inhibitors primary,
secondary, and tertiary pharmacophores. The nomenclature used refers to the three
pharmacophores and two substituents (R and R' groups). The secondary and tertiary
pharmacophores located in the R' area are illustrated linearly from the primary
pharmacophore. The secondary pharmacophore generally consists of a polar carbonyl group
or a polar ether group. When the secondary pharmacophore is a carbonyl group, it is located
about 7.5 ± 1 A from the carbonyl of the primary pharmacophore, with either side of the
carbonyl (X and Y) being a CH2, O or NH. When the secondary pharmacophore is a ether
group it is preferably located about 1 carbon unit further from the carbonyl of the primary
pharmacophore. The tertiary pharmacophore is also a polar group located approximately 11
carbon units (17 ± 1 Å) from the carbonyl of the primary pharmacophore with the Z group as
an OH, or a substituted amine or alcohol or a heterocyclic or acyclic structure mimicing the
terminal ester or acid.
[0021] Figure 3 provides a hydrophobicity map of the mouse sEH substrate binding pocket
co-crystalyzed with the inhibitor l-cyclohexyl-3-dodecyl urea. A shading gradient indicates
degrees of hydrophobicity. A series of hydrophilic residues were observed on the "top" side
of the channel, while the "bottom" of the channel was very hydrophobic, with the exception
of the catalytic aspartate (Asp333). This structural analysis indicated that a number of
potential hydrogen bonding sites are observed in the substrate binding pocket of the soluble
epoxide hydrolase, primarily located on the surface opposite Asp333 (the catalytic nucleophile
which reacts with the substrate or binds to the primary pharmacophores).
[0022] Figure 4 provides mammalian soluble epoxide hydrolase protein sequence
alignments (residue 1-340).
[0023] Figure 5 provides mammalian soluble epoxide hydrolase protein sequence
alignments (residue 341-554).
[0024] Figure 6 is a graph illustrating the metabolic stabilities of l-adamantyl-3-dodecyl
urea (686) and 1-cyclohexyl- 3-dodecyl urea (297) in rat hepatic microsomes. Microsomes

WO 2006/045119 PCT/US2005/038282
8
were incubated with 1 μM 686 or 297 in the presence of an NADPH generating system. Data
are expressed as mean ± SD of triplicate experiments.
[0025] Figure 7 is a graph illustrating.the metabolic stabilities of 686 and 687 in rat hepatic
microsomes as described above.
[0026] Figure 8 is a series of graphs illustrating the metabolic conversion of 1 -adamantyl-
3-dodecyl urea (686) in microsomal preparations from rat, mouse, and human hepatic tissues.
The metabolites identified are the omega hydroxyl (686-M1), the omega aldehyde (686-M2),
the omega acid (687), and a mixture of monohydroxy adamantyl omega hydroxylated
compounds (686-M3). These structures are shown in Table 13.
[0027] Figure 9 provides a mass spectrum showing collision induced dissociation of a
dominant urinary metabolite of l-adamantyl-3-dodecyl urea (686) and the 3-dodecanoic acid
analog (687) suggesting that these compounds can ultimately enter beta-oxidation to produce
chain shortened inhibitors.
[0028] Figure 10 is a graph illustrating the blood concentration vs. time profiles of 687
after oral administration of 5 mg/kg of either 687 or 800 to mice. The ester compound delays
the time to achieve the maximum circulating dose, and increases the maximum circulating
concentration of 687 observed. This translates into a longer half-life for the inhibitor.
[0029] Figure 11 provides a structural evaluation of conserved hydrogen bond donors in
the sEH substrate binding pocket with linear distances to the primary pharmacophore noted
and further illustrating the effect of functional group distances on interactions with the
mammalian soluble epoxide hydrolases.
[0030] Figure 12 is a graph illustrating the relative substrate turnover/relative inhibitor
potency as a function of terminal carboxyl distance to either substrate epoxide of inhibitor 3-
position nitrogen.
[0031] Figure 13 is a bar graph showing the levels of urinary octadecanoids (A) and
urinary eicosanoids (B) in rats treated with angiotensin II in the presence of absence of 687.
[0032] Figure 14 is a graph showing blood concentration vs. time profiles of 950 after
single oral administration of 0.1 to 1.0 mg/kg of 950 to 70 kg rats. The presence of the
polyether secondary pharmacophore increases the maximum circulating concentration of 950
observed. This translates into a longer half-life for the inhibitor.

WO 2006/045119 PCT/US2005/038282
9
[0033] Figure 15 provides a sample preparation procedure for a pharmacokinetic study. A
5 μl whole blood sample was drawn into a capillary at a specific time point, each sample was
extracted and anaylzed by LC/MS-MS.
[0034] Figure 16 shows the physical properties/parameters of compound 950.
[0035] Figure 17 shows graphs which illustrate the in vitro metabolism of 950 in (A)
human liver microsome (no NADPH), (B) S9 fractions, and (C) Liver microsomes both with
NADPH. Both rat and human microsomes were used for the 950 metabolism study. The
hydroxy metabolite was the major metabolite.
DETAILED DESCRIPTION OF THE INVENTION
Abbreviations and Definitions:
[0036] "cis-Epoxyeicosatrienoic acids" ("EETs") are biomediators synthesized by
cytochrome P450 epoxygenases.
[0037] "Epoxide hydrolases" ("EH;" EC 3.3.2.3) are enzymes in the alpha / beta hydrolase
fold family that add water to 3 membered cyclic ethers termed epoxides.
[0038] "Soluble epoxide hydrolase" ("sEH") is an enzyme which in endothelial, smooth
muscle and other cell types converts EETs to dihydroxy derivatives called
dihydroxyeicosatrienoic acids ("DHETs"). The cloning and sequence of the murine sEH is
set forth in Grant et al, J. Biol Chem. 268(23):17628-17633 (1993). The cloning, sequence,
and accession numbers of the human sEH sequence are set forth in Beetham et al., Arch.
Biochem. Biophys. 305(1): 197-201 (1993). The amino acid sequence of human sEH is also
set forth as SEQ ID NO.2 of U.S. Patent No. 5,445,956; the nucleic acid sequence encoding
the human sEH is set forth as nucleotides 42-1703 of SEQ ID NO:1 of that patent The
evolution and nomenclature of the gene is discussed in Beetham et al., DNA Cell Biol.
14(1):61-71 (1995). Soluble epoxide hydrolase represents a single highly conserved gene
product with over 90% homology between rodent and human (Arand et al., FEBS Lett.,
338:251-256 (1994)).
[0039] The terms "treat", "treating" and "treatment" refer to any method of alleviating or
abrogating a disease or its attendant symptoms.
[0040] The term "therapeutically effective amount" refers to that amount of the compound
being administered sufficient to prevent or decrease the development of one or more of the
symptoms of the disease, condition or disorder being treated.

WO 2006/045119 PCT/US2005/038282
10
[0041] The term "modulate" refers to the ability of a compound to increase or decrease the
function, or activity, of the associated activity (e.g., soluble epoxide hydrolase).
"Modulation", as used herein in its various forms, is meant to include antagonism and partial
antagonism of the activity associated with sEH. Inhibitors of sEH are compounds that, e.g.,
bind to, partially or totally block the enzyme's activity.
[0042] The term "compound" as used herein is intended to encompass not only the
specified molecular entity but also its pharmaceutically acceptable, pharmacologically active
derivatives, including, but not limited to, salts, prodrug conjugates such as esters and amides,
metabolites and the like.
[0043] The term "composition" as used herein is intended to encompass a product
comprising the specified ingredients in the specified amounts, as well as any product which
results, directly or indirectly, from combination of the specified ingredients in the specified
amounts. By "pharmaceutically acceptable" it is meant the carrier, diluent or excipient must
be compatible with the other ingredients of the formulation and not deleterious to the
recipient thereof.
[0044] The "subject" is defined herein to include animals such as mammals, including, but
not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats,
mice and the like. In some embodiments, the subject is a human.
[0045] As used herein, the term "sEH-mediated disease or condition" and the like refers to
a disease or condition characterized by less than or greater than normal, sEH activity. A
sEH-mediated disease or condition is one in which modulation of sEH results in some effect
on the underlying condition or disease (e.g., a sEH inhibitor or antagonist results in some
improvement in patient well-being in at least some patients).
[0046] "Parenchyma" refers to the tissue characteristic of an organ, as distinguished from
associated connective or supporting tissues.
[0047] "Chronic Obstructive Pulmonary Disease" or "COPD" is also sometimes known as
"chronic obstructive airway disease", "chronic obstructive lung disease", and "chronic
airways disease." COPD is generally defined as a disorder characterized by reduced maximal
expiratory flow and slow forced emptying of the lungs. COPD is considered to encompass
two related conditions, emphysema and chronic bronchitis. COPD can be diagnosed by the
general practitioner using art recognized techniques, such as the patient's forced vital capacity
("FVC"), the maximum volume of air that can be forceably expelled after a maximal

WO 2006/045119 PCT/US2005/038282
11
inhalation. In the offices of general practitioners, the FVC is typically approximated by a 6
second maximal exhalation through a spirometer. The definition, diagnosis and treatment of
COPD, emphysema, and chronic bronchitis are well known in the art and discussed in detail
by, for example, Honig and Ingram, in Harrison's Principles of Internal Medicine, (Fauci et
al., Eds.), 14th Ed., 1998, McGraw-Hill, New York, pp. 1451-1460 (hereafter, "Harrison's
Principles of Internal Medicine").
[0048] "Emphysema" is a disease of the lungs characterized by permanent destructive
enlargement of the airspaces distal to the terminal bronchioles without obvious fibrosis.
[0049] "Chronic bronchitis" is a disease of the lungs characterized by chronic bronchial
secretions which last for most days of a month, for three months a year, for two years.
[0050] As the names imply, "obstructive pulmonary disease" and "obstructive lung disease"
refer to obstructive diseases, as opposed to restrictive diseases. These diseases particularly
include COPD, bronchial asthma and small airway disease.
[0051] "Small airway disease." There is a distinct minority of patients whose airflow
obstruction is due, solely or predominantly to involvement of the small airways. These are
defined as airways less than 2 mm in diameter and correspond to small cartilaginous bronchi,
terminal bronchioles and respiratory bronchioles. Small airway disease (SAD) represents
luminal obstruction by inflammatory and fibrotic changes that increase airway resistance.
The obstruction may be transient or permanent.
[0052] The "interstitial lung diseases (ILDs)" are a group of conditions involving the
alveolar walls, perialveolar tissues, and contiguous supporting structures. As discussed on
the website of the American Lung Association, the tissue between the air sacs of the lung is
the interstitium, and this is the tissue affected by fibrosis in the disease. Persons with the
disease have difficulty breathing in because of the stiffness of the lung tissue but, in contrast
to persons with obstructive lung disease, have no difficulty breathing out. The definition,
diagnosis and treatment of interstitial lung diseases are well known in the art and discussed in
detail by, for example, Reynolds, H.Y., in Harrison's Principles of Internal Medicine, supra,
at pp. 1460-1466. Reynolds notes that, while ILDs have various initiating events, the
immunopathological responses of lung tissue are limited and the ILDs therefore have
common features.
[0053] "Idiopathic pulmonary fibrosis," or "IPF," is considered the prototype ILD.
Although it is idiopathic in that the cause is not known, Reynolds, supra, notes that the term
refers to a well defined clinical entity.

WO 2006/045119 PCT/US2005/038282
12
[0054] "Bronchoalveolar lavage," or "BAL," is a test which permits removal and
examination of cells from the lower respiratory tract and is used in humans as a diagnostic
procedure for pulmonary disorders such as IPF. In human patients, it is usually performed
during bronchoscopy.
[0055] As used herein, the term "alkyl" refers to a saturated hydrocarbon radical which may
be straight-chain or branched-chain (for example, ethyl, isopropyl, t-amyl, or 2,5-
dimethylhexyl). This definition applies both when the term is used alone and when it is used
as part of a compound term, such as "aralkyl," "alkylamino" and similar terms. In some
embodiments, alkyl groups are those containing 1 to 24 carbon atoms. All numerical ranges
in this specification and claims are intended to be inclusive of their upper and lower limits.
Lower alkyl refers to those alkyl groups having 1 to 4 carbon atoms. Additionally, the alkyl
and heteroalkyl groups may be attached to other moieties at any position on the alkyl or
heteroalkyl radical which would otherwise be occupied by a hydrogen atom (such as, for
example, 2-pentyl, 2-methylpent-l-yl and 2-propyloxy). Divalent alkyl groups may be
referred to as "alkylene", and divalent heteroalkyl groups may be referred to as
"heteroalkylene" such as those groups used as linkers in the present invention. The alkyl,
alkylene, and heteroalkyl moieties may also be optionally substituted with halogen atoms, or
other groups such as oxo, cyano, nitro, alkyl, alkylamino, carboxyl, hydroxyl, alkoxy,
aryloxy, and the like.
[0056] The terms "cycloalkyl" and "cycloalkenyl" refer to a saturated hydrocarbon ring and
includes bicyclic and polycyclic rings. Similarly, cycloalkyl and cycloalkenyl groups having
a heteroatom (e.g. N, O or S) in place of a carbon ring atom may be referred to as
"heterocycloalkyl" and heterocycloalkylene," respectively. Examples of cycloalkyl and
heteroaryl groups are, for example, cyclohexyl, norbornyl, adamantly, morpholinyl,
thiomorpholinyl, dioxothiomorphorinyl, and the like. The cycloalkyl and heterocycloalkyl
moieties may also be optionally substituted with halogen atoms, or other groups such as nitro,
alkyl, alkylamino, carboxyl, alkoxy, aryloxy and the like. In some embodiments, cycloalkyl
and cycloalkenyl moieties are those having 3 to 12 carbon atoms in the ring (e.g., cyclohexyl,
cyclooctyl, norbornyl, adamantyl, and the like). In some embodiments, heterocycloalkyl and
heterocycloalkylene moieties are those having 1 to 3 hetero atoms in the ring (e.g.,
morpholinyl, Momorpholinyl, dioxothiomorphonnyl, piperidinyl and the like). Additionally,
the term "(cycloalkyl)alkyl" refers to a group having a cycloalkyl moiety attached to an alkyl
moiety. Examples are cyclohexylmethyl, cyclohexylethyl and cyclopentylpropyl.

WO 2006/045119 PCT/US2005/038282
13
[0057] The term "alkenyl" as used herein refers to an alkyl group as described above which
contains one or more sites of unsaturation that is a double bond. Similarly, the term
"alkynyl" as used herein refers to an alkyl group as described above which contains one or
more sites of unsaturation that is a triple bond.
[0058] The term "alkoxy" refers to an alkyl radical as described above which also bears an
oxygen substituent which is capable of covalent attachment to another hydrocarbon radical
(such as, for example, methoxy, ethoxy, aryloxy and t-butoxy).
[0059] The term "aryl" refers to an aromatic carbocyclic substituent which may be a single
ring or multiple rings which are fused together, linked covalently or linked to a common
group such as an ethylene or methylene moiety. Similarly, aryl groups having a heteroatom
(e.g. N, O or S) in place of a carbon ring atom are referred to as "heteroaryl". Examples of
aryl and heteroaryl groups are, for example, phenyl, naphthyl, biphenyl, diphenylmethyl, 2,2-
diphenyl-1-ethyl, thienyl, pyridyl and quinoxalyl. The aryl and heteroaryl moieties may also
be optionally substituted with halogen atoms, or other groups such as nitro, alkyl, alkylamino,
carboxyl, alkoxy, phenoxy and the like. Additionally, the aryl and heteroaryl groups may be
attached to other moieties at any position on the aryl or heteroaryl radical which would
otherwise be occupied by a hydrogen atom (such as, for example, 2-pyridyl, 3-pyridyl and 4-
pyridyl). Divalent aryl groups are "arylene", and divalent heteroaryl groups are referred to as
"heteroarylene" such as those groups used as linkers in the present invention.
[0060] The terms "arylalkyl", "arylalkenyl" and "aryloxyalkyl" refer to an aryl radical
attached directly to an alkyl group, an alkenyl group, or an oxygen which is attached to an
alkyl group, respectively. For brevity, aryl as part of a combined term as above, is meant to
include heteroaryl as well.
[0061] The terms "halo" or "halogen," by themselves or as part of another substituent,
mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally,
terms such as "haloalkyl," are meant to include monohaloalkyl and polyhaloalkyl. For
example, the term "C1-C6 haloalkyl" is mean to include trifluoromethyl, 2,2,2-trifluoroethyl,
4-chlorobutyl, 3-bromopropyl, and the like.
[0062] The term "hetero" as used in a "heteroatom-containing alkyl group" (a "heteroalkyl"
group) or a "heteroatom-containing aryl group" (a "heteroaryl" group) refers to a molecule,
linkage or substituent in which one or more carbon atoms are replaced with an atom other
than carbon, e.g., nitrogen, oxygen, sulfur, phosphorus or silicon, typically nitrogen, oxygen

WO 2006/045119 PCT/US2005/038282
14
or sulfur or more thatnone non-carbon atom (e.g., sulfonamide). Similarly, the term
"heteroalkyl" refers to an alkyl substituent that is heteroatom-containing, the term
"heterocyclic" refers to a cyclic substituent that is heteroatom-containing, the terms
"heteroaryl" and heteroaromatic" respectively refer to "aryl" and "aromatic" substituents that
are heteroatom-containing, and the like. Examples of heteroalkyl groups include alkoxyaryl,
alkylsulfanyl-substituted alkyl, N-alkylated amino alkyl, and the like. Examples of heteroaryl
substituents include pyrrolyl, pyrrolidinyl, pyridinyl, quinolinyl, indolyl, pyrimidinyl,
imidazolyl, 1,2,4-triazolyl, tetrazolyl, etc., and examples of heteroatom-containing alicyclic
groups are pyrrolidino, morpholino, piperazino, piperidino, etc.
[0063] The term "hydrophobic radical" or "hydrophobic group" refers to a group which
lowers the water solubility of a molecule. In some embodiments, hydrophobic radicals are
groups containing at least 3 carbon atoms.
[0064] The term "carboxylic acid analog" refers to a variety of groups having an acidic
moiety that are capable of mimicking a carboxylic acid residue. Examples of such groups are
sulfonic acids, sulflnic acids, phosphoric acids, phosphonic acids, phosphinic acids,
sulfonamides, and heterocyclic moieties such as, for example, imidazoles, triazoles and
tetrazoles.
[0065] The term "substituted" refers to the replacement of an atom or a group of atoms of a
compound with another atom or group of atoms. For example, an atom or a group of atoms
may be substituted with one or more of the following substituents or groups: halo, cyano,
nitro, alkyl, alkylamino, hydroxyalkyl, haloalkyl, carboxyl, hydroxyl, alkoxy, alkoxyalkoxy,
haloalkoxy, thioalkyl, aryl, aryloxy, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl optionally
substituted with 1 or more, preferably 1 to 3, substituents selected from halo, halo alkyl and
alkyl, aralkyl, heteroaralkyl, alkenyl containing 1 to 2 double bonds, alkynyl containing 1 to 2
triple bonds, alk(en)(yn)yl groups, halo, cyano, hydroxy, haloalkyl and polyhaloalkyl,
preferably halo lower alkyl, especially trifiuoromethyl, formyl, alkylcarbonyl, arylcarbonyl
that is optionally substituted with 1 or more, preferably 1 to 3, substituents selected from
halo, halo alkyl and alkyl, heteroarylcarbonyl, carboxy, alkoxycarbonyl, aryloxycarbonyl,
aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, arylaminocarbonyl,
diarylaminocarbonyl, aralkylaminocarbonyl, alkoxy, aryloxy, perfluoroalkoxy, alkenyloxy,
alkynyloxy, arylalkoxy, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, arylaminoalkyl,
amino, alkylamino, dialkylamino, arylamino, alkylarylamino, alkylcarbonylamino,
arylcarbonylamino, azido, nitro, mercapto, alkylthio, arylthio, perfluoroalkylthio, thiocyano,

WO 2006/045119 PCT/US2005/038282
15
isothiocyano, alkylsulfinyl, alkylsulfonyl, arylsulfinyl, arylsulfonyl, aminosulfonyl,
alkylaminosulfonyl, dialkylaminosulfonyl and arylaminosulfonyl. When the term
"substituted" appears prior to a list of possible substituted groups, it is intended that the term
apply to every member of that group.
[0066] The term "unsubstituted" refers to a native compound that lacks replacement of an
atom or a group of atoms.
General:
[0067] The present invention derives from the discovery that 1,3-disubstituted ureas (or the
corresponding amides or carbamates, also referred to as the primary pharmacophore) can be
further functionalized to provide more potent sEH inhibitors with improved physical
properties. As described herein, the introduction of secondary and/or tertiary
pharmacophores can increase water solubility and oral availability of sEH inhibitors (see
Figure 2). The combination of the three pharmacophores (see the compounds of Table 18)
provides a variety of compounds of increased water solubility.
[0068] The discovery of the secondary and tertiary pharmacophores has also led to the
employment of combinatorial chemistry approaches for establishing a wide spectrum of
compounds having sEH inhibitory activity. The polar pharmacophores divide the molecule
into domains each of which can be easily manipulated by common chemical approaches in a
combinatorial manner, leading to the design and confirmation of novel orally available
therapeutic agents for the treatment of diseases such as hypertension and vascular
inflammation. The agents of the present invention treat such diseases while simultaneously
increasing sodium excretion, reducing vascular and renal inflammation, and reducing male
erectile dysfunction As shown below (see Example 51 and Figure 13), alterations in
solubility, bioavailability and pharmacological properties leads to compounds that can alter
the regulatory lipids of experimental animals increasing the relative amounts of epoxy
arachidonate derivatives when compared either to their diol products or to the
proinflammatory and hypertensive hydroxyeicosatetraenoic acids (HETEs). Since epoxy
arachidonates are anti-hypertensive and anti-inflammatory, altering the lipid ratios can lead to
reduced blood pressure and reduced vascular and renal inflammation. This approach has
been validated in a patient approaching end stage renal disease (ESRD) where even a brief
oral treatment with low doses compound 800 altered the serum profile of regulatory lipids in
a positive manner. This resulted in reduced systolic and diastolic blood pressure, a dramatic

WO 2006/045119 PCT/US2005/038282
16
reduction in blood urea nitrogen (an indicator of renal inflammation) and dramatically
reduced serum levels of C reactive protein (a common indicator of vascular inflammation).
[0069] Without intending to be bound by theory, and with reference to Figures 2, 3, 4 and
5, it is believed that the left side of the primary pharmacophore or R (in Figure 2) can be
varied to obtain optimal properties as can the primary pharmacophore, which contains groups
able to hydrogen bond to the catalytic aspartic acid on one side and the catalytic tyrosines on
the other (see Figure 3). The right side of the primary pharmacophore is effectively divided
into 4 segments: a spacer separating the primary and secondary pharmacophore (termed L1 in
the present invention), the secondary pharmacophore (termed P2 in the present invention)
and a tertiary pharmacophore (P3) flanked by a spacer (L2) and finally a terminating group Z
(collectively provided with the tertiary pharmacophore as P3). The spacer between the
primary and secondary pharmacophores, is optimally 3 atom units in length, while the
secondary pharmacophore can be, for example, a ketone, carbonate, amide, carbamate, urea,
ether/polyether, ester or other functionality able to form a hydrogen bond with the enzyme
approximately 7.5 angstroms from the carbonyl of the primary pharmacophore. The
identified tertiary pharmacophore consists of a polar group located approximately six to
eleven carbon units from the primary pharmacophore (see Figure 2). A conserved asparagine
residue (Asn471, see Figures 4 and 5) is thought to provide the site of interaction between the
protein and the polar functionality located at this tertiary site. While, in the rodent a
threonine (Thr468) is also in an appropriate position for hydrogen bonding, residue 468 is a
methionine in the human enzyme (Figure 5). As with the secondary pharmacophore, this
group improves water solubility of sEH inhibitors as well as the specificity for the sEH, and a
wide diversity of functionalities such as an ester, amide, carbamate, or similar functionalities
capable of donating or accepting a hydrogen bond similarly can contribute to this polar
group. For example, in pharmaceutical chemistry heterocyclic groups are commonly used to
mimic carbonyls as hydrogen bond donors and acceptors. Of course the primary, secondary
and tertiary pharmacophore groups can be combined in a single molecule with suitable
spacers to improve activity or present the inhibitor as a prodrug.
[0070] Figure 11 illustrates the binding interaction for structural evaluation of conserved
hydrogen bond donors in the sEH substrate binding pocket with linear distances to the
primary pharmacophore noted. The table below provides specific distances to residues
provided in Figures 4 and 5.

WO 2006/045119 PCT/US2005/038282
17
Table
[0071] Linear distances of hydrophylic residues to the carbonyl carbon of the bound urea
Residue Distance Conserved
from Urea Carbon
Asp333 4.7Å +
Tyr465 O 4.5Å +
Tyr381O 4.6Å +
Trp334NRing 7.1Å +
G1n382N 8.2Å +
Tyr465 NBack Bone 10.5Å +
Tyr4b8 14.9Å Met in Human
Asn471N 15.2Å +
Asn471 O 16.7Å +
*Note Figure 11 distances are measured linearly from the carbonyl oxygen to the alternate pharmacophores.
This Table measures 3 dimensional distances from carbonyl carbon of the primary phaxmacophore to amino
acids which could hydrogen bond with the inhibitor.
Methods of Inhibiting Soluble Epoxide Hvdrolases:
[0072] In view of the above, the present invention provides, in one aspect, a method for
inhibiting a soluble epoxide hydrolase, comprising contacting the soluble epoxide hydrolase
with an inhibiting amount of a compound having a formula selected from the group
consisting of:

and their pharmaceutically acceptable salts, wherein the symbol R1 is a member selected from
the group consisting of substituted or unsubstituted alkyl, substituted or unsubstituted
heteroalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted
cycloalkylheteroalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted
arylheteroalkyl, substituted or unsubstituted C5-C12 cycloalkyl, substituted or unsubstituted
aryl, substituted or unsubstituted heteroaryl and combinations thereof, wherein said
cycloalkyl portions are monocyclic or polycyclic; P is a primary pharmacophore selected
from the group consisting of -OC(O)O-, -OC(O)CH2-, CH2C(O)O-, -OC(O)-, -C(O)O-, -

WO 2006/045119 PCT/US2005/038282
18
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-NHC(O)NH-,
-OC(O)NH-, -NHC(O)O-,-NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, -
CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -NON-, -CH2C(O)NH-, -NHC(O)CH2-,
-C(O)NH-,

-NHC(O)-,
a secondary pharmacophore selected from the group consisting of -NH-, -OC(O)O-,-C(O)-,
-CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-,-NHC(NH)NH-, -NHC(NH)CH2-, -
CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-, -C(O)NH-, -NHC(O)-; -
NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, -

WO 2006/045119 PCT/US2005/038282
19
CH2C(NH)S-, -N=C=N-,

P3 is a tertiary pharmacophore selected from the group consisting of C2-C6 alkenyl, C2-C6
alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, -O(CH2CH2O)q-R2, - OR2,
-C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(O)OR2, -C(O)R2, -C(O)OR2
and carboxylic acid analogs, wherein R2 is a member selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl. In the above formula, the subscripts n and m
are each independently 0 or 1, and at least one of n or m is 1, and the subscript q is 0 to 6.
[0073] Turning next to the linking groups, the symbol L1 represents a first linker that is
selected from the group consisting of substituted or unsubstituted C1-C6 alkylene, substituted
or unsubstituted C3-C6 cycloalkylene, substituted or unsubstituted arylene and substituted or
unsubstituted heteroarylene; the symbol L2 represents a second linker selected from the group
consisting of substituted or unsubstituted C1-C12 alkylene, substituted or unsubstituted C3-C6
cycloalkylene, substituted or unsubstituted arylene, substituted or unsubstituted
heteroarylene; an ammo acid, a dipeptide and a dipeptide analog; and combinations thereof;
or is H when m is 0. Preferably, the compounds are other than 1 l-(3-cyclohexylureido)-

WO 2006/045119 PCT/US2005/038282
20
undecanoic acid, ll-(3-cyclohexylureido)-undecanoic acid methyl ester, 11-(3-
cyclohexylureido)-undecanoic acid amide, 12-(3-cyclohexylureido)-dodecanoic acid and 12-
(3-adamantan-l-yl-ureido)-dodecanoic acid.
[0074] In a first group of embodiments, R1 is selected from the group consisting of
substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or
unsubstituted cycloalkylalkyl, substituted or unsubstituted cycloalkylheteroalkyl, substituted
or unsubstituted arylalkyl and substituted or unsubstituted arylheteroalkyl. In another group
of embodiments, R1 is selected from C5-C12 cycloalkyl, phenyl and naphthyl. More
preferably, R1 is selected from C6-C10 cycloalkyl and phenyl. In some embodiments, are
those embodiments in which R1 is cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, adamantyl,
noradamantyl, and phenyl, wherein the phenyl group is either unsubstituted or substituted
with from one to three substituents selected from halogen, lower alkyl, lower halo alkyl,
lower alkoxy, C3-C5 cycloalkyl and cyano.
[0075] Returning to formula (I), P1 is preferably selected from -NHC(O)NH-,
-OC(O)NH- and -NHC(O)O-. Most preferably, P1 is -NHC(O)NH-. In other embodiments,
P1 is selected from the group consisting of -OC(O)O-, -OC(O)CH2-, CH2C(O)O-, -OC(O)-, -
C(O)O-, -NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-
NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, -
CH2C(NH)S-, -NON-, -NHC(O)CH2-,

WO 2006/045119 PCT/US2005/038282
21

[0076] Turning next to the first linking group, L1 is preferably selected from substituted or
unsubstituted C1-C6 alkylene, wherein the substituents are selected to impart desired
properties to the overall composition. For example, in some embodiments in which R1 is a
particularly hydrophobic residue, L1 may preferably have substituents that are hydrophilic to
offset to some degree the lack of aqueous solubility normally associated with very
hydrophobic compounds. As a result, in some embodiments, L1 will have one or two
hydroxy moieties as substituents, preferably only one hydroxy moiety substituents. In other
embodiments, L1 will be an alkylene, arylene or cycloalkylene linker having the length
indicated above, wherein one or more of the hydrogen atoms are replaced with fluorine atoms
to impart other attractive properties, such as facilitating the compound's use in stents so that it
is slowly released from the stent to then inhibit the soluble epoxide hydrolase. Other
examples of substituents, include but are not limited to, halo, cyano, nitro, alkyl, alkylamino,
carboxyl, hydroxyl, alkoxy, aryloxy, and the like. Further are embodiments in which L1 is
C2-C5 alkylene, more preferably C2-C4 alkylene, still more preferably C2-C3 alkylene, and
most preferably an ethylene linkage. Where L1 is C3-C6 cycloalkylene, it is more preferably
cyclohexyl that can be linked in a 1,3 or 1,4 manner. In certain embodiments, L1 is selected
to provide spacing between the first pharmacophore carbonyl moiety (in P1) and the second

WO 2006/045119 PCT/US2005/038282
22
pharmacophore carbonyl moiety (in P2) of about 7.5 ± 2 angstroms and more preferably,
about 7.5 ± 1 angstroms.
[0077] The secondary pharmacophore, P2, when present (n is 1) is selected from the group
consisting of-NH-, -OC(O)O-,-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(0)O, -OC(O)-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)-; -NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, -
CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -N=C=N-,

More preferably, P2 is selected from -C(O)-, -O(CH2CH2O)q-, -C(0)O, -OC(O)-, -OC(O)O-,
-OC(O)NH- and -C(O)NH-. Most preferably, P2 is selected from -C(O)-, -O(CH2CH2O)q-,
and -C(O)O-. In another embodiment, P2 is preferably selected from the group consisting of
-NH-, -OC(O)O-, -NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(S)NH-, -
NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -
N=C=N-,

WO 2006/045119 PCT/US2005/038282
23

[0078] The second linking group, L2 is selected from substituted or unsubstituted C1-C12
alkylene, substituted or unsubstituted arylene, and combinations thereof. For those
embodiments in which a secondary pharmacophore (P2) is not present, the Unking group L2
will be combined with L1 to provide spacing between the primary pharmacophore and the
tertiary pharmacophore preferably of about ≥2, and ≤12 carbon atoms. Accordingly, when
L1 is an alkylene or part of a cycloalkylene linkage of from 1 to 4 carbon atoms, and P2 is not
present, L2 will preferably be an alkylene linkage of from 1 to 8 carbon atoms, more
preferably, 4 to 8 carbon atoms, and most preferably 5, 6, 7 or 8 carbon atoms. For those
embodiments in which a tertiary pharmacophore (P3) is not present, the linking group L2 may
be H orwill terminate with hydrogen or a substituent selected as described for L1 above. In
such embodiments, the arylene group need not be divalent. In some embodiments, L2 will
comprise an arylene group, preferably a phenylene group that can be linked in a 1,2 or 1,3 or
1,4 manner, preferably in a 1,3 or 1,4 manner. As with L1, the alkylene portions of L2 can be
substituted or unsubstituted. The substituents are selected as described for L1 above.
[0079] The tertiary pharmacophore, P3, is a tertiary pharmacophore selected from the group
consisting of C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl,
-O(CH2CH2O)q-R2, - OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-

WO 2006/045119 PCT/US2005/038282
24
C(O)OR2, -C(O)R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is a member
selected from the group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl,
substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl;
substituted or unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl. In certain
embodiments, R2 is H, methyl, ethyl, propyl, allyl, 3-propynyl, butyl, 2-propyl, 1,1-
dimethylethyl, 2-butyl, 2-methyl-1 -propyl, adamantyl-methyl, benzyl, 2-chlorobenzyl and
naphthyknethyl. In one group of embodiments, P3 is -C(O)NHR2, -C(O)NHS(O)2R2,
-NHS(O)2R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is selected from hydrogen,
unsubstituted C1-C4 alkyl, and unsubstituted C3-C8 cycloalkyl. Still more preferably, R2 is H,
Me or Et. In some embodiments, P3 is -C(O)OR2 and carboxylic acid analogs, wherein R2 is
selected from hydrogen, Me or Et. In other embodiments, P3 is preferably selected from the
group consisting of is selected from the group consisting of C2-C6 alkenyl, heterocyclyl, OR2,
-OC2-C4alkyl-C(O)OR2 and -C(O)R2, wherein R2 is a member selected from the group
consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted
C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl
and substituted or unsubstituted aryl C1-C4 alkyl.
[0080] With the embodiments provided above, certain combinations of embodiments
represent particular embodiments. While all combinations of the groups represent additional
embodiments of the invention, particular embodiments include those wherein P1 is selected
from -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; P2 is selected from -C(O)O-, -OC(O)-,
-O(CH2CH2O)q-, -C(O)NH- and -NHC(O)-; m is 0 and L1 is selected from unsubstituted C1-
C6 alkylene. In another group of particular embodiments, P1 is selected from -NHC(O)NH-,
-OC(O)NH- and -NHC(O)O; P2 is selected from -C(O)O-, -OC(O)-, -O(CH2CH2O)q-,
-C(O)NH- and -NHC(O)-; n and m are each 1; L1 is selected from unsubstituted C1-C6
alkylene; L2 is selected from substituted or unsubstituted C1-C6 alkylene; and P3 is selected
from -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, and -C(O)0R2, wherein R2 is a member
selected from the group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl,
substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl;
substituted or unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl. Still other
particular embodiments are those in which the compound has formula (I), wherein P1 is
selected from -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; n is 0; m is 1; L1 is selected from
unsubstituted C1-C6 alkylene; L2 is selected from substituted or unsubstituted C1-C6 alkylene;
and P3 is selected from -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, and -C(O)OR2, wherein

WO 2006/045119 PCT/US2005/038282
25
R2 is a member selected from the group consisting of hydrogen, substituted or unsubstituted
C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted
heterocyclyl; substituted or unsubstituted aryl and substituted or unsubstituted aryl C1-C4
alkyl.
[0081] In one embodiment, the compound has the formula:

wherein R is a member selected from the group consisting of alkyl, aryl, alkylaryl,
cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl; and L2 is selected from the group consisting of phenylene
or methylenephenylene, heteroarylene, optionally substituted with from 1 to 2 substituents
each independently selected from the group consisting of halo and haloalkyl. Within this
embodiment, the compound has the formula:

[0082] In other embodiments, the compound has the formula:

wherein R2 is selected from the group consisting of substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl. Within this embodiment, the compound
preferably has the formula:

wherein R2 is substituted or unsubstituted aryl; and more preferably has the formula:

WO 2006/045119 PCT/US2005/038282
26

wherein R1 is a member selected from the group consisting of alkyl, aryl, alkylaryl,
cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl.
[0083] In one embodiment, compounds for use in this aspect of the invention are those
compounds provided in the Tables below, particularly Tables 5, 8-10 and 18.
[0084] In another group of embodiments the compounds of formula (I), as noted above,
contain an amino acid or dipeptide component which can be a dipeptide analog. The amino
acid residues, by themselves or as part of a dipeptide, are denoted by single-letter or three-
letter designations following conventional practices. The designations for gene-encoded
amino acids are as follows (amino acid, one letter symbol, three letter symbol): Alanine, A,
Ala; Arginine, R, Arg; Asparagine, N, Asn; Aspartic acid, D, Asp; Cysteine, C, Cys;
Glutamine, Q, G1n; Glutamic acid, E, Glu; Glycine, G, Gly; Histidine, H, His; Isoleucine, I,
lie; Leucine, L, Leu; Lysine, K, Lys; Methionine, M, Met; Phenylalanine, F, Phe; Proline, P,
Pro; Serme, S, Ser; Threonine, T, Thr; Tryptophan, W, Trp; Tyrosine, Y, Tyr; and Valine, V,
Val. Commonly encountered amino acids which are not gene-encoded may also be used in
the present invention. These amino acids and their abbreviations include ornithine (Orn); t-
butylglycine (t-BuG); phenylglycine (PhG); cyclohexylalanine (Cha); norleucine (Nle); 2-
naphthylalanine (2-Nal); 1-naphthylalanine (1-Nal); 2-thienylaniHne (2-Thi); N-
methylisoleucine (N-Melle), homoarginine (Har), Na-methylargmine (N-MeArg) and
sarcosine (Sar). All of the amino acids used in the present invention may be either the D- or
L-isomer.
[0085] In one embodiment, compounds of the invention are those in which L2 is selected
from the group consisting of substituted or unsubstituted C3-C6 cycloalkylene, substituted or
unsubstituted arylene, substituted or unsubstituted heteroarylene. In other embodiments, L2 is
preferably an amino acid or a dipeptide. Preferably, the dipeptide has a Tyr, His, Lys, Phe or
Trp residue directly attached to P2.

WO 2006/045119 PCT/US2005/038282
27
[0086] Other compounds for use in the present invention are those in which R1, P1 and L1
are selected from the groupings as described above for formula (I). Particular compounds of
formula (I) are those in which R1 is selected from C5-C12 cycloalkyl and phenyl. More
particularly, R1 is selected from C6-C10 cycloalkyl and phenyl. Other embodiments are those
embodiments in which R1 is cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, adamantly or
noradamantyl. P1 is preferably a urea (-NHC(O)NH-) or carbamate (-OC(O)NH-), more
preferably a urea. L1 is preferably a substituted or unsubstituted C2-C5 alkylene, more
preferably C2-C4 alkylene, still more preferably an ethylene or propylene linkage.
[0087] For those embodiments in which L2 is a single amino acid, L2 is preferably selected
from Ala, Arg, Asp, Cys, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Tip, Tyr and
Val. More preferably, L2 is selected from His, I1e, Lys, Phe, Trp and Tyr in which the amino
acid is linked to P2 in a manner to afford an amide linkage and terminal carboxylic acid
group. Of course, one of skill in the art will appreciate that these amino acids are meant to
refer to their corresponding methyl or ethyl esters, as well as their carboxamide derivatives
(e.g., terminal -C(O)NH2)- Most preferably, the compounds are those provided in Table 11.
[0088] For those embodiments in which L1 is a dipeptide, P2 is preferably attached to a Tyr,
His, Lys, Phe or Trp residue, with the remaining amino acid being selected from the gene-
encoded amino acids, their D-isomers or analogs thereof (e.g., hydroxy acids such as lactic
acid and the like). Still more prefereably, L2 is selected from TyrAla, TyrArg, TyrAsp,
TyrGly, Tyrlle, TyrLeu, TyrLys, TyrMet, TyrPhe, TyrPro, TyrSer, TyrThr, TyrTrp, TyrTyr
and TyrVal. More preferably, L2 is selected from TyrArg, TyrAsp, TyrMet, TyrPhe, TyrSer,
TyrTrp, TyrTyr and TyrVal. in which the Tyr amino acid is linked to P2 in a manner to afford
an amide linkage. As above, these dipeptides are also meant to refer to their corresponding
methyl or ethyl esters, as well as their carboxamide derivatives (e.g., terminal -C(O)NH2).
Most preferably, the compounds are those provided in Table 12.
Assays to Monitor Soluble Epoxide Hydrolase Activity:
[0089] Additionally, the present invention provides a variety of assays and associated
methods for monitoring soluble epoxide hydrolase activity, particularly the activity that has
been modulated by the administration of one or more of the compounds provided above.
[0090] In one group of embodiments, the invention provides methods for reducing the
formation of a biologically active diol produced by the action of a soluble epoxide hydrolase,

WO 2006/045119 PCT/US2005/038282
28
the method comprising contacting the soluble epoxide hydrolase with an amount of a
compound of formula (I) above, sufficient to inhibit the activity of the soluble epoxide
hydrolase and reduce the formation of the biologically active diol.
[0091] In another group of embodiments, the invention provides methods for stabilizing
biologically active epoxides in the presence of a soluble epoxide hydrolase, the method
comprising contacting the soluble epoxide hydrolase with an amount of a compound of
formula (I), sufficient to inhibit the activity of the soluble epoxide hydrolase and stabilize the
biologically active epoxide.
[0092] In each of these groups of embodiments, the methods can be carried out as part of
an in vitro assay or the methods can be carried out in vivo by monitoring blood liters of the
respective biologically active epoxide or diol.
[0093] Epoxides and diols of some fatty acids are biologically important chemical
mediators and are involved in several biological processes. The strongest biological data
support the action of oxylipins as chemical mediators between the vascular endothelium and
vascular smooth muscle. Accordingly, the epoxy lipids are anti-inflammatory and anti-
hypertensive. Additionally, the lipids are thought to be metabolized by beta-oxidation, as
well as by epoxide hydration. The soluble epoxide hydrolase is considered to be the major
enzyme involved in the hydrolytic metabolism of these oxylipins. The compounds of
formula (I) can inhibit the epoxide hydrolase and stabilize the epoxy lipids both in vitro and
in vivo. This activity results in a reduction of hypertension in four separate rodent models.
Moreover, the inhibitors show a reduction in renal inflammation associated with and
independent of the hypertensive models.
[0094] More particularly, the present invention provides methods for monitoring a variety
of lipids in both the arachidonate and linoleate cascade simultaneously in order to address the
biology of the system. A GLC-MS system or a LC-MS method can be used to monitor over
740 analytes in a highly quantitative fashion in a single injection. The analytes include the
regioisomers of the arachidonate epoxides (EETs), the diols (DHETs), as well as other P450
products including HETEs. Characteristic products of the cyclooxygenase, lipoxygenase, and
peroxidase pathways in both the arachidonate and linoleate series can also be monitored.
Such methods are particularly useful as being predictive of certain disease states. The
oxylipins can be monitored in mammals following the administration of inhibitors of epoxide

WO 2006/045119 PCT/US2005/038282
29
hydrolase. Generally, EH inhibitors increase epoxy lipid concentrations at the expense of
diol concentrations in body fluids and tissues.
[0095] Other compounds for use in this aspect of the invention are those inhibitors of
formula (I) in which the primary pharmacophore is separated from a tertiary pharmacophore
by a distance that approximates the distance between the terminal carboxylic acid and an
epoxide functional group in the natural substrate.
Methods of Treating Diseases Modulated by Soluble Epoxide Hvdrolases:
[0096] In another aspect, the present invention provides methods of treating diseases,
especially those modulated by soluble epoxide hydrolases (sEH). The methods generally
involve administering to a subject in need of such treatment an effective amount of a
compound having a formula (I) above. The dose, frequency and timing of such administering
will depend in large part on the selected therapeutic agent, the nature of the condition being
treated, the condition of the subject including age, weight and presence of other conditions or
disorders, the formulation being administered and the discretion of the attending physician.
Preferably, the compositions and compounds of the invention and the pharmaceutically
acceptable salts thereof are administered via oral, parenteral, subcutaneous, intramuscular,
intravenous or topical routes. Generally, the compounds are administered in dosages ranging
from about 2 mg up to about 2,000 mg per day, although variations will necessarily occur
depending, as noted above, on the disease target, the patient, and the route of administration.
Dosages are administered orally in the range of about 0.05 mg/kg to about 20 mg/kg, more
preferably in the range of about 0.05 mg/kg to about 2 mg/kg, most preferably in the range of
about 0.05 mg/kg to about 0.2 mg per kg of body weight per day. The dosage employed for
the topical administration will, of course, depend on the size of the area being treated.
[0097] It has previously been shown that inhibitors of soluble epoxide hydrolase ("sEH")
can reduce hypertension. See, e.g., U.S. Patent No. 6,351,506. Such inhibitors can be useful
in controlling the blood pressure of persons with undesirably high blood pressure, including
those who suffer from diabetes.
[0098] In some embodiments, compounds of formula (I) are administered to a subject in
need of treatment for hypertension, specifically renal, hepatic, or pulmonary hypertension;
inflammation, specifically renal inflammation, vascular inflammation, and lung

WO 2006/045119 PCT/US2005/038282
30
inflammation; adult respiratory distress syndrome; diabetic complications; end stage renal
disease; Raynaud syndrome and arthritis.
Methods for Inhibiting Progression of Kidney Deterioration (Nephropathv) and
Reducing Blood Pressnre:
[0099] In another aspect of the invention, the compounds of the invention can reduce
damage to the kidney, and especially damage to kidneys from diabetes, as measured by
albuminuria. The compounds of the invention can reduce kidney deterioration (nephropathy)
from diabetes even in individuals who do not have high blood pressure. The conditions of
therapeautic administration are as described above.
[0100] cis-Epoxyeicosantrienoic acids ("EETs") can be used in conjunction with the
compounds of the invention to further reduce kidney damage. EETs, which are epoxides of
arachidonic acid, are known to be effectors of blood pressure, regulators of inflammation,
and modulators of vascular permeability. Hydrolysis of the epoxides by sEH diminishes this
activity. Inhibition of sEH raises the level of EETs since the rate at which the EETs are
hydrolyzed into DHETs is reduced. Without wishing to be bound by theory, it is believed
that raising the level of EETs interferes with damage to kidney cells by the microvasculature
changes and other pathologic effects of diabetic hyperglycemia. Therefore, raising the EET
level in the kidney is believed to protect the kidney from progression from microalbummuria
to end stage renal disease.
[0101] EETs are well known in the art. EETs useful in the methods of the present
invention include 14,15-EET, 8,9-EET and 11,12-EET, and 5,6 EETs, in that order of
preference. Preferably, the EETs are administered as the methyl ester, which is more stable.
Persons of skill will recognize that the EETs are regioisomers, such as 8S,9R- and 14R,15S-
EET. 8,9-EET, 11,12-EET, and 14R,15S-EET, are commercially available from, for
example, Sigma-Aldrich (catalog nos. E5516, E5641, and E5766, respectively, Sigma-
Aldrich Corp., St. Louis, MO).
[0102] EETs produced by the endothelium have anti-hypertensive properties and the EETs
11,12-EET and 14,15-EET may be endothelium-derived hyperpolarizing factors (EDHFs).
Additionally, EETs such as 11,12-EET have profibrinolytic effects, anti-inflammatory actions
and inhibit smooth muscle cell proliferation and migration. In the context of the present

WO 2006/045119 PCT/US2005/038282
31
invention, these favorable properties are believed to protect the vasculature and organs during
renal and cardiovascular disease states.
[0103] It is now believed that sEH activity can be inhibited sufficiently to increase the
levels of EETs and thus augment the effects of administering sEH inhibitors by themselves.
This permits EETs to be used in conjunction with one or more sEH inhibitors to reduce
nephropathy in the methods of the invention. It further permits EETs to be used in
conjunction with one or more sEH inhibitors to reduce hypertension, or inflammation, or
both. Thus, medicaments of EETs can be made which can be administered in conjunction
with one or more sEH inhibitors, or a medicament containing one or more sEH inhibitors can
optionally contain one or more EETs.
[0104] The EETs can be administered concurrently with the sEH inhibitor, or following
administration of the sEH inhibitor. It is understood that, like all drugs, inhibitors have half
lives defined by the rate at which they are metabolized by or excreted from the body, and that
the inhibitor will have a period following administration during which it will be present in
amounts sufficient to be effective. If EETs are administered after the inhibitor is
administered, therefore, it is desirable that the EETs be administered during the period during
which the inhibitor will be present in amounts to be effective to delay hydrolysis of the EETs.
Typically, the EET or EETs will be administered within 48 hours of administering an sEH
inhibitor. Preferably, the EET or EETs are administered within 24 hours of the inhibitor, and
even more preferably within 12 hours. In increasing order of desirability, the EET or EETs
are administered within 10, 8, 6,4, 2, hours, 1 hour, or one half hour after administration of
the inhibitor. Most preferably, the EET or EETs are administered concurrently with the
inhibitor.
[0105] In some embodiments, the EETs, the compound of the invention, or both, are
provided in a material that permits them to be released over time to provide a longer duration
of action. Slow release coatings are well known in the pharmaceutical art; the choice of the
particular slow release coating is not critical to the practice of the present invention.
[0106] EETs are subject to degradation under acidic conditions. Thus, if the EETs are to be
administered orally, it is desirable that they are protected from degradation in the stomach.
Conveniently, EETs for oral administration may be coated to permit them to passage the
acidic environment of the stomach into the basic environment of the intestines. Such
coatings are well known in the art. For example, aspirin coated with so-called "enteric

WO 2006/045119 PCT/US2005/038282
32
coatings" is widely available commercially. Such enteric coatings may be used to protect
EETs during passage through the stomach. An exemplary coating is set forth in the
Examples.
[0107] While the anti-hypertensive effects of EETs have been recognized, EETs have not
been administered to treat hypertension because it was thought endogenous sEH would
hydro! yse the EETs too quickly for them to have any useful effect. Surprisingly, it was found
during the course of the studies underlying the present invention that exogenously
administered inhibitors of sEH succeeded in inhibiting sEH sufficiently that levels of EETs
could be further raised by the administration of exogenous EETs. These findings underlie the
co-administration of sEH inhibitors and of EETs described above with respect to inhibiting
the development and progression of nephropathy. This is an important improvement in
augmenting treatment. While levels of endogenous EETs are expected to rise with the
inhibition of sEH activity caused by the action of the sEH inhibitor, and therefore to result in
at least some improvement in symptoms or pathology, it may not be sufficient in all cases to
inhibit progression of kidney damage fully or to the extent intended. This is particularly true
where the diseases or other factors have reduced the endogenous concentrations of EETs
below those normally present in healthy individuals. Administration of exogenous EETs in
conjunction with a sEH inhibitor is therefore expected to be beneficial and to augment the
effects of the sEH inhibitor in reducing the progression of diabetic nephropathy.
[0108] The present invention can be used with regard to any and all forms of diabetes to the
extent that they are associated with progressive damage to the kidney or kidney function.
The chronic hyperglycemia of diabetes is associated with long-term damage, dysfunction,
and failure of various organs, especially the eyes, kidneys, nerves, heart, and blood vessels.
The long-term complications of diabetes include retinopathy with potential loss of vision;
nephropathy leading to renal failure; peripheral neuropathy with risk of foot ulcers,
amputation, and Charcot joints.
[0109] In addition, persons with metabolic syndrome are at high risk of progression to type
2 diabetes, and therefore at higher risk than average for diabetic nephropathy. It is therefore
desirable to monitor such individuals for microalbuminuria, and to administer a sEH inhibitor
and, optionally, one or more EETs, as an intervention to reduce the development of
nephropathy. The practitioner may wait until microalbuminuria is seen before beginning the
intervention. As noted above, a person can be diagnosed with metabolic syndrome without

WO 2006/045119 PCT/US2005/038282
33
having a blood pressure of 130/85 or higher. Both persons with blood pressure of 130/85 or
higher and persons with blood pressure below 130/85 can benefit from the administration of
sEH inhibitors and, optionally, of one or more EETs, to slow the progression of damage to
their kidneys. In some embodiments, the person has metabolic syndrome and blood pressure
below 130/85.
[0110] Dyslipidemia or disorders of lipid metabolism is another risk factor for heart
disease. Such disorders include an increased level of LDL cholesterol, a reduced level of
HDL cholesterol, and an increased level of triglycerides. An increased level of serum
cholesterol, and especially of LDL cholesterol, is associated with an increased risk of heart
disease. The kidneys are also damaged by such high levels. It is believed that high levels of
triglycerides are associated with kidney damage. In particular, levels of cholesterol over 200
mg/dL, and especially levels over 225 mg/dL, would suggest that sEH inhibitors and,
optionally, EETs, should be administered. Similarly, triglyceride levels of more than 215
mg/dL, and especially of 250 mg/dL or higher, would indicate that administration of sEH
inhibitors and, optionally, of EETs, would be desirable. The administration of compounds of
the present invention with or without the EETs, can reduce the need to administer statin drugs
(HMG-CoA reductase inhibitors) to the patients, or reduce the amount of the statins needed.
In some embodiments, candidates for the methods, uses and compositions of the invention
have triglyceride levels over 215 mg/dL and blood pressure below 130/85. In some
embodiments, the candidates have triglyceride levels over 250 mg/dL and blood pressure
below 130/85. In some embodiments, candidates for the methods, uses and compositions of
the invention have cholesterol levels over 200 mg/dL and blood pressure below 130/85. In
some embodiments, the candidates have cholesterol levels over 225 mg/dL and blood
pressure below 130/85.
Methods of Inhibiting the Proliferation of Vascular Smooth Muscle Cells:
[0111] In other embodiments, compounds of formula (I) inhibit proliferation of vascular
smooth muscle (VSM) cells without significant cell toxicity, (e.g. specific to VSM cells).
Because VSM cell proliferation is an integral process in the pathophysiology of
atherosclerosis, these compounds are suitable for slowing or inhibition atherosclerosis. These
compounds are useful to subjects at risk for atherosclerosis, such as individuals who have had

WO 2006/045119 PCT/US2005/038282
34
a heart attack or a test result showing decreased blood circulation to the heart. The conditions
of therapeautic administration are as described above.
[0112] The methods of the invention are particularly useful for patients who have had
percutaneous intervention, such as angioplasty to reopen a narrowed artery, to reduce or to
slow the narrowing of the reopened passage by restenosis. In some embodiments, the artery
is a coronary artery. The compounds of the invention can be placed on stents in polymeric
coatings to provide a controlled localized release to reduce restenosis. Polymer compositions
for implantable medical devices, such as stents, and methods for embedding agents in the
polymer for controlled release, are known in the art and taught, for example, in U.S. Patent
Nos. 6,335,029; 6,322,847; 6,299,604; 6,290,722; 6,287,285; and 5,637,113. In some
embodiments, the coating releases the inhibitor over a period of time, preferably over a
period of days, weeks, or months. The particular polymer or other coating chosen is not a
critical part of the present invention.
[0113] The methods of the invention are useful for slowing or inhibiting the stenosis or
restenosis of natural and synthetic vascular grafts. As noted above in connection with stents,
desirably, the synthetic vascular graft comprises a material which releases a compound of the
invention over time to slow or inhibit VSM proliferation and the consequent stenosis of the
graft. Hemodialysis grafts are a particular embodiment.
[0114] In addition to these uses, the methods of the invention can be used to slow or to"
inhibit stenosis or restenosis of blood vessels of persons who have had a heart attack, or
whose test results indicate that they are at risk of a heart attack.
[0115] In one group of embodiments, compounds of the invention are administered to
reduce proliferation of VSM cells in persons who do not have hypertension. In another group
of embodiments, compounds of the invention are used to reduce proliferation of VSM cells in
persons who are being treated for hypertension, but with an agent that is not an sEH inhibitor.
[0116] The compounds of the invention can be used to interfere with the proliferation of
cells which exhibit inappropriate cell cycle regulation, hi one important set of embodiments,
the cells are cells of a cancer. The proliferation of such cells can be slowed or inhibited by
contacting the cells with a compound of the invention. The determination of whether a
particular compound of the invention can slow or inhibit the proliferation of cells of any
particular type of cancer can be determined using assays routine in the art.

WO 2006/045119 PCT/US2005/038282
35
[0117] In addition to the use of the compounds of the invention, the levels of EETs can be
raised by adding EETs. VSM cells contacted with both an EET and a compound of the
invention exhibited slower proliferation than cells exposed to either the EET alone or to the a
compound of the invention alone. Accordingly, if desired, the slowing or inhibition of VSM
cells of a compound of the invention can be enhanced by adding an EET along with a
compound of the invention. In the case of stents or vascular grafts, for example, this can
conveniently be accomplished by embedding the EET in a coating along with a compound of
the invention so that both are released once the stent or graft is in position.
Methods of Inhibiting the Progression of Obstructive Pulmonary Disease, Interstitial
Lung Disease, or Asthma:
[0118] Chronic obstructive pulmonary disease, or COPD, encompasses two conditions,
emphysema and chronic bronchitis, which relate to damage caused to the lung by air
pollution, chronic exposure to chemicals, and tobacco smoke. Emphysema as a disease
relates to damage to the alveoli of the lung, which results in loss of the separation between
alveoli and a consequent reduction in the overall surface area available for gas exchange.
Chronic bronchitis relates to irritation of the bronchioles, resulting in excess production of
mucin, and the consequent blocking by mucin of the airways leading to the alveoli. While
persons with emphysema do not necessarily have chronic bronchitis or vice versa, it is
common for persons with one of the conditions to also have the other, as well as other lung
disorders.
[0119] Some of the damage to the lungs due to COPD, emphysema, chronic bronchitis, and
other obstructive lung disorders can be inhibited or reversed by administering inhibitors of
the enzyme known as soluble epoxide hydrolase, or "sEH". The effects of sEH inhibitors can
be increased by also administering EETs. The effect is at least additive over administering
the two agents separately, and may indeed be synergistic.
[0120] The studies reported herein show that EETs can be used in conjunction with sEH
inhibitors to reduce damage to the lungs by tobacco smoke or, by extension, by occupational
or environmental irritants. These findings indicate that the co-administration of sEH
inhibitors and of EETs can be used to inhibit or slow the development or progression of
COPD, emphysema, chronic bronchitis, or other chronic obstructive lung diseases which
cause irritation to the lungs.

WO 2006/045119 PCT/US2005/038282
36
[0121] Animal models of COPD and humans with COPD have elevated levels of
immunomodulatory lymphocytes and neutrophils. Neutrophils release agents that cause
tissue damage and, if not regulated, will over time have a destructive effect. Without wishing
to be bound by theory, it is believed that reducing levels of neutrophils reduces tissue damage
contributing to obstructive lung diseases such as COPD, emphysema, and chronic bronchitis.
Administration of sEH inhibitors to rats in an animal model of COPD resulted in a reduction
in the number of neutrophils found in the lungs. Administration of EETs in addition to the
sEH inhibitors also reduced neutrophil levels. The reduction in neutrophil levels in the
presence of sEH inhibitor and EETs was greater than in the presence of the sEH inhibitor
alone.
[0122] While levels of endogenous EETs are expected to rise with the inhibition of sEH
activity caused by the action of the sEH inhibitor, and therefore to result in at least some
improvement in symptoms or pathology, it may not be sufficient in all cases to inhibit
progression of COPD or other pulmonary diseases. This is particularly true where the
diseases or other factors have reduced the endogenous concentrations of EETs below those
normally present in healthy individuals. Administration of exogenous EETs in conjunction
with an sEH inhibitor is therefore expected to augment the effects of the sEH inhibitor in
inhibiting or reducing the progression of COPD or other pulmonary diseases.
[0123] In addition to inhibiting or reducing the progression of chronic obstructive airway
conditions, the invention also provides new ways of reducing the severity or progression of
chronic restrictive airway diseases. While obstructive airway diseases tend to result from the
destruction of the lung parenchyma, and especially of the alveoE, restrictive diseases tend to
arise from the deposition of excess collagen in the parenchyma. These restrictive diseases
are commonly referred to as "interstitial lung diseases", or "ILDs", and include conditions
such as idiopathic pulmonary fibrosis. The methods, compositions and uses of the invention
are useful for reducing the severity or progression of ILDs, such as idiopathic pulmonary
fibrosis. Macrophages play a significant role in stimulating interstitial cells, particularly
fibroblasts, to lay down collagen. Without wishing to be bound by theory, it is believed that
neutrophils are involved in activating macrophages, and that the reduction of neutrophil
levels found in the studies reported herein demonstrate that the methods and uses of the
invention will also be applicable to reducing the severity and progression of ELDs.

WO 2006/045119 PCT/US2005/038282
37
[0124] In some embodiments, the ILD is idiopathic pulmonary fibrosis. In other
embodiments, the ILD is one associated with an occupational or environmental exposure.
Exemplars of such ILDs, are asbestosis, silicosis, coal worker's pneumoconiosis, and
berylliosis. Further, occupational exposure to any of a number of inorganic dusts and organic
dusts is believed to be associated with mucus hypersecretion and respiratory disease,
including cement dust, coke oven emissions, mica, rock dusts, cotton dust, and grain dust (for
a more complete list of occupational dusts associated with these conditions, see Table 254-1
of Speizer, "Environmental Lung Diseases," Harrison's Principles of Internal Medicine, infra,
at pp. 1429-1436). In other embodiments, the ILD is sarcoidosis of the lungs. ILDs can also
result from radiation in medical treatment, particularly for breast cancer, and from connective
tissue or collagen diseases such as rheumatoid arthritis and systemic sclerosis. It is believed
that the methods, uses and compositions of the invention can be useful in each of these
interstitial lung diseases.
[0125] In another set of embodiments, the invention is used to reduce the severity or
progression of asthma. Asthma typically results in mucin hypersecretion, resulting in partial
airway obstruction. Additionally, irritation of the airway results in the release of mediators
which result in airway obstruction. While the lymphocytes and other immunomodulatory
cells recruited to the lungs in asthma may differ from those recruited as a result of COPD or
an ILD, it is expected that the invention will reduce the influx of immunomodulatory cells,
such as neutrophils and eosinophils, and ameliorate the extent of obstruction. Thus, it is
expected that the administration of sEH inhibitors, and the administration of sEH inhibitors in
combination with EETs, will be useful in reducing airway obstruction due to asthma.
[0126] In each of these diseases and conditions, it is believed that at least some of the
damage to the lungs is due to agents released by neutrophils which infiltrate into the lungs.
The presence of neutrophils in the airways is thus indicative of continuing damage from the
disease or condition, while a reduction in the number of neutrophils is indicative of reduced
damage or disease progression. Thus, a reduction in the number of neutrophils in the airways
in the presence of an agent is a marker that the agent is reducing damage due to the disease or
condition, and is slowing the further development of the disease or condition. The number of
neutrophils present in the lungs can be determined by, for example, bronchoalveolar lavage.
Prophvlatic and therapeutic methods to reduce stroke damage

WO 2006/045119 PCT/US2005/038282
38
[0127] Inhibitors of soluble epoxide hydrolase ("sEH") and EETs administered in
conjunction with inhibitors of sEH have been shown to reduce brain damage from strokes.
Based on these results, we expect that inhibitors of sEH taken prior to an ischernic stroke will
reduce the area of brain damage and will likely reduce the consequent degree of impairment.
The reduced area of damage should also be associated with a faster recovery from the effects
of the stroke.
[0128] While the pathophysiologies of different subtypes of stroke differ, they all cause
brain damage. Hemorrhagic stroke differs from ischemic stroke in that the damage is largely
due to compression of tissue as blood builds up in the confined space within the skull after a
blood vessel ruptures, whereas in ischemic stroke, the damage is largely due to loss of
oxygen supply to tissues downstream of the blockage of a blood vessel by a clot. Ischemic
strokes are divided into thrombotic strokes, in which a clot blocks a blood vessel in the brain,
and embolic strokes, in which a clot formed elsewhere in the body is carried through the
blood stream and blocks a vessel there. But, in both hemorrhagic stroke and ischemic stroke,
the damage is due to the death of brain cells. Based on the results observed in our studies,
however, we would expect at least some reduction in brain damage in all types of stroke and
in all subtypes.
[0129] A number of factors associated with an increased risk of stroke. Given the results of
the studies underlying the present invention, sEH inhibitors administered to persons with any
one or more of the following conditions or risk factors-.high blood pressure, tobacco use,
diabetes, carotid artery disease, peripheral artery disease, atrial fibrillation, transient ischemic
attacks (TIAs), blood disorders such as high red blood cell counts and sickle cell disease,
high blood cholesterol, obesity, alcohol use of more than one drink a day for women or two
drinks a day for men, use of cocaine, a family history of stroke, a previous stroke or heart
attack, or being elderly, will reduce the area of brain damaged of a stroke. With respect to
being elderly, the risk of stroke increases for every 10 years. Thus, as an individual reaches
60, 70, or 80, administration of sEH inhibitors has an increasingly larger potential benefit.
As noted in the next section, the administration of EETs in combination with one or more
sEH inhibitors can be beneficial in further reducing the brain damage.
[0130] In some uses and methods, the sEH inhibitors and, optionally, EETs, are
administered to persons who use tobacco, have carotid artery disease, have peripheral artery
disease, have atrial fibrillation, have had one or more transient ischemic attacks (TIAs), have
a blood disorder such as a high red blood cell count or sickle cell disease, have high blood
cholesterol, are obese, use alcohol in excess of one drink a day if a woman or two drinks a

WO 2006/045119 PCT/US2005/038282
39
day if a man, use cocaine, have a family history of stroke, have had a previous stroke or heart
attack and do not have high blood pressure or diabetes, or are 60, 70, or 80 years of age or
more and do not have hypertension or diabetes.
[0131] Clot dissolving agents, such as tissue plasminogen activator (tPA), have been shown
to reduce the extent of damage from ischemic strokes if administered in the hours shortly
after a stroke. tPA, for example, is approved by the FDA for use in the first three hours after
a stroke. Thus, at least some of the brain damage from a stoke is not instantaneous, but
occurs over a period of time or after a period of time has elapsed after the stroke. It is
therefore believed that administration of sEH inhibitors, optionally with EETs, can also
reduce brain damage if administered within 6 hours after a stroke has occurred, more
preferably within 5, 4, 3, or 2 hours after a stroke has occurred, with each successive shorter
interval being more preferable. Even more preferably, the inhibitor or inhibitors are
administered 2 hours or less or even 1 hour or less after the stroke, to maximize the reduction
in brain damage. Persons of skill are well aware of how to make a diagnosis of whether or
not a patient has had a stroke. Such determinations are typically made in hospital emergency
rooms, following standard differential diagnosis protocols and imaging procedures.
[0132] In some uses and methods, the sEH inhibitors and, optionally, EETs, are
administered to persons who have had a stroke within the last 6 hours who: use tobacco, have
carotid artery disease, have peripheral artery disease, have atrial fibrillation, have had one or
more transient ischemic attacks (TIAs), have a blood disorder such as a high red blood cell
count or sickle cell disease, have high blood cholesterol, are obese, use alcohol in excess of
one drink a day if a woman or two drinks a day if a man, use cocaine, have a family history
of stroke, have had a previous stroke or heart attack and do not have high blood pressure or
diabetes, or are 60, 70, or 80 years of age or more and do not have hypertension or diabetes.
[0133] The conditions of therapeautic administration for all of these indications are as
described above.
Combination Therapy
[0134] As noted above, the compounds of the present invention will, in some instances, be
used in combination with other therapeutic agents to bring about a desired effect. Selection
of additional agents will, in large part, depend on the desired target therapy (see, e.g., Turner,
N. etal. Prog. Drug Res. (1998) 51: 33-94; Haffner, S. Diabetes Care (1998) 21: 160-178;

WO 2006/045119 PCT/US2005/038282
40
and DeFronzo, R. et al. (ed's.), Diabetes Reviews (1997) Vol. 5 No. 4). A number of studies
have investigated the benefits of combination therapies with oral agents {see, e.g., Mahler, R.,
J. Clin. Endocrinol Metab. (1999) 84: 1165-71; United Kingdom Prospective Diabetes Study
Group: UKPDS 28, Diabetes Care (1998) 21: 87-92; Bardin, C. W.,(ed.), Current Therapy In
Endocrinology And Metabolism, 6th Edition (Mosby - Year Book, Inc., St. Louis, MO 1997);
Chiasson, J. et al., Ann. Intern. Med. (1994) 121: 928-935; Coniff, R. et al, Clin. Ther.
(1997) 19: 16-26; Coniff, R. et al., Am. J. Med. (1995) 98: 443-451; and Iwamoto, Y. et al.,
Diabet. Med. (1996) 13 365-370; Kwiterovich, P. Am. J. Cardiol (1998) 82(12A): 3U-17U).
Combination therapy includes administration of a single pharmaceutical dosage formulation
which contains a compound having the general structure of formula 1 and one or more
additional active agents, as well as administration of a compound of formula 1 and each
active agent in its own separate pharmaceutical dosage formulation. For example, a
compound of formula 1 and one or more angiotensin receptor blockers, angiotensin
converting enzyme inhibitors, calcium channel blockers, diuretics, alpha blockers, beta
blockers, centrally acting agents, vasopeptidase inhibitors, renin inhibitors, endothelin
receptor agonists, AGE crosslink breakers, sodium/potassium ATPase inhibitors, endothelin
receptor agonists, endothelin receptor antagonists, angiotensin vaccine, and the like; can be
administered to the human subject together in a single oral dosage composition, such as a
tablet or capsule, or each agent can be administered in separate oral dosage formulations.
Where separate dosage formulations are used, a compound of formula 1 and one or more
additional active agents can be administered at essentially the same time (i.e., concurrently),
or at separately staggered times (i.e., sequentially). Combination therapy is understood to
include all these regimens.
Compounds for Inhibiting Soluble Epoxide Hydrolases:
[0135] In addition to the methods provided above, the present invention provides in another
aspect, compounds that can inhibit the activity of soluble epoxide hydrolases. In particular,
the present invention provides compounds having a formula selected from formula (I) above.
Preferably, the compounds are other than 1 l-(3-cyclohexylureido)-undecanoic acid, 11-(3-
cyclohexylureido)-undecanoic acid methyl ester, 1 l-(3-cyclohexylureido)-undecanoic acid
amide, 12-(3-cyclohexylureido)-dodecanoic acid and 12-(3-adamantan-l-yl-ureido)-
dodecanoic acid.

WO 2006/045119 PCT/US2005/038282
41
[0136] In one embodiment, compounds are those compounds described above as for the
recited uses.
Methods of Preparation
[0137] The compounds of the present invention can be prepared by a variety of methods as
outlined generally in the schemes below.
Scheme 1 - Introduction of a secondary pharmacophore (ketone)
[0138] Scheme 1 illustrates general methods that can be used for preparation of compounds
of the invention having a secondary pharmacophore that is a ketone functional group. While
the scheme is provided for the synthesis of l-(3-chlorophenyl)-3-(4-oxodecyl)urea, one of
skill in the art will understand that a number of commercially available isocyanates could be
used in place of 3-chlorophenyl isocyanate, and that shorter or longer analogs of ethyl 4-
aminobutyric acid or hexylbromide could also be employed.
Scheme 1: Synthesis of l-(3-chlorophenyl)-3-(4-oxodecyl)urea (794).

Scheme 1: Synthesis of l-(3-chlorophenyI)-3-(4-oxodecyl)urea (794): (a) Benzophenone inline, CH2Cl2. rt; (b)
DIBAL, THF, -78°C; (c) Mg/I2, hexylbromide, THF, rt; (d) acetic anhydride, DMSO, rt; (e) 1N HCl/dioxane, rt;
(f) 3-chlorophenyl isocyanate, TEA, DMF, rt.
[0139] As shown in Scheme 1, ethyl 4-aminobutyrate hydrochloride (available from
Aldrich Chemical Co., Milwaukee, Wisconsin, USA) is combined with benzophenone irnine
at room temperature to provide intermediate (i). DIBAL reduction of the ester group
provides an unisolated aldehyde moiety that is then reacted with a suitable Grignard reagent
(prepared in situ) to provide intermediate alcohol (ii). Oxidation of the alcohol moiety to a
ketone provides (iii) which can then be deprotected to form the amino-ketone (iv). Reaction
of (iv) with a suitable isocyanate provides the target compound (794). Substitution of

WO 2006/045119 PCT/US2005/038282
42
3-chlorophenyl isocyanate with, for example, adamantyl isocyanate or cyclohexyl isocyanate
(also available from Aldrich Chemical Co.) provides other compounds of the invention.

Scheme 2: Syntheses of l-(aryl or alkyl)-3-(3-alkylated proply)ureas: (a) aryl or alkyl isocyanate, DMF, rt; (b)
bromopentane, K2CO3, Nal, acetonitrile, reflux; (c) di-t-butyl dicarbonate, dioxane, 50°C; (d) pentylamine,
isobutyl chloroformate, NMM, DMF, rt; (e) 4M hydrochloric acid, dioxane; (f) 3-chlorophenyi isocyanate,
TEA, DMF, rt.
[0140] As shown in Scheme 2, a variety of compounds having a secondary pharmacophore
that is either an ester or amide functional group can be prepared. Beginning with 4-
aminobutyric acid, treatment with a suitable cycloalkyl or aryl isocyanate provides the urea
intermediates shown as (v), wherein R is 3-chlorophenyl, cyclohexyl or 1-adamantyl. Of
course other suitable isocyanates can also be employed to provide desired urea intermediates.
Esterification via alkylation of the carboxylic acid present in (v) with, for example, pentyl
bromide provides the target compounds 767, 772 and 789. A variety of suitable alkyl halides
can be used to prepare other compounds of the invention. The second path illustrated in
Scheme 2 can be used to prepare compounds such as 768, as well as those compounds having
a primary pharmacophore that is a carbamate. Accordingly, treatment of 4-aminobutyric acid
with di-t-butyl dicarbonate provides the t-butyl carbamate acid (vi) that is converted to a
desired amide (vii) using pentylamine, for example, in a mild procedure employing isobutyl
chloroformate, and N-methyl morpholine (NMM). Removal of the carbamate protecting
group (as it is used in this instance) followed by formation of a urea with a suitable
isocyanate (shown here as 3-chlorophenyl isocyanate) provides the target compounds (e.g.,
768).

WO 2006/045119 PCT/US2005/038282
43

Scheme 3: Syntheses of l-(3-chlorophenyl)-3-(2-alkylated ethyl)ureas: (a) 3-chlorophenyl isocyanate, DMF, rt;
(b) heptanoic anhydride (761), chlorofonnic acid pentyl ester (760), or pentyl isocyanate (762), TEA, DMF, rt;
(c) di-t-butyl dicarbonate, dioxane, rt; (d) heptanoic anhydride (765), chloroformic acid pentyl ester (777), or
pentyl isocyanate (766), DMF, rt; (e) 4M HC1, dioxane; (f) 3-chlorophenyl isocyanate, TEA, DMF, rt.
[0141] Scheme 3 illustrates a variety of methods for introducing secondary
phannacophores that are esters, amide, ureas, carbonates and carbamates, from readily
accessible starting materials. In A, ethanolamine is treated with a suitable isocyanate to
introduce a primary pharmacophore that is a urea and form intermediate (viii). Treatment of
(viii) with an anhydride, a chloro formic acid ester or an isocyanate provides compounds such
as 761,760 and 762, respectively. Similar methodology in employed in B, with the addition
of protection/deprotection steps. Accordingly, ethylenediamine is monoprotected as a r-butyl
carbamate. The free amine is then converted to a secondary pharmacophore that is an amide,
carbamate or urea using reactants and conditions similar to those employed in "A" to provide
intermediates (x). Deprotection of (x) and reaction with a suitable isocyanate provides the
target compounds 765, 777 and 766. Again, use of isocyanates other than 3-chlorophenyl
isocyanate leads to other compounds of the invention, while substitution of certain reactants
used, for example, in the conversion of (ix) to (x) can provide still other compounds of the
invention.

WO 2006/045119 PCT/US2005/038282
44

Scheme 4: Syntheses of l-(l-adamantyl)-3-(ll-alkylated undecyl)ureas: (a) adamantyl isocyanate, chloroform,
reflux; (b) alkyl or aryl halide, K2CO3, Nal, acetonitrile, reflux; (c) alcohol or amine, isobutyl chloroformate,
TEA, DMF, rt; (d) t-butanol, EDCI, DMAP, metirylene chloride, rt.
[0142] Scheme 4 illustrates pathways for the introduction of a tertiary pharmacophore that
is an ester or an amide functional group. In each case, a carboxylic acid group is converted to
the desired ester or amide. As shown in Scheme 4,12-aminododecanoic acid (Aldrich
Chemical Co.) is converted to urea (687) upon treatment with adamantyl isocyanate. One of
skill in the art will appreciate that a variety of alkyl, aryl and cycloalkyl isocyanates can be
similarly employed to form other ureas as the primary pharmacophore. Similarly,
11-aminoundecanoic acid or another long chain amino fatty acid could be used in place of
12-aminododecanoic acid. The carboxylic acid moiety can then be esterified or converted to
an amide moiety following standard procedures to provide, for example, 780-785, 788 and
800-804 (as esters) and 786, 787, 792 and 793 (as esters and amides).
[0143] Scheme 5 illustrates pathways for the synthesis of cis- or trans-phenoxy or
benzyloxy-cyclohexyl compounds. In each case, trans-4-aminocyclohexanol hydrochloride is
converted to the desired isomer of the phenoxy or benzyloxy derivative. The alcohol moiety
can be alkylated with an appropriately substituted benzyl halide following standard
procedures to provide, the corresponding benzyl ether. Likewise, the alcohol moiety can be
alkylated with an appropriately substituted phenol in the presence of triphenylphosphine
following standard procedures to provide, the corresponding phenyl ether. As shown in
Scheme 5, each isomer can be converted to the corresponding urea upon treatment with an

WO 2006/045119 PCT/US2005/038282
45
appropriately substituted isocyanate, e.g. adamantyl isocyanate. One of skill in the art will
appreciate that a variety of alkyl, aryl and cycloalkyl isocyanates can be similarly employed
to form other ureas as the primary pharmacophore.

Scheme 5: Syntheses of cis and trans-phenoxy-and benzyloxy-cyclohexyl compounds.
Combinatorial library synthesis
[0144] A 192-member urea library has been constructed by a known two-step solid-phase
synthesis (Tetrahedron Letters 2003,44, 6099-6102), which employs a PS-Indole-CHO resin.
As shown in Scheme 6, four amines and 48 isocyanates have been used as building blocks to
find the optimal left side of urea for sEH inhibitors. Resin-bound secondary amines can be
obtained by a reductive amination with amines using sodium cyanoborohydride in the
presence of triethylorthoformate. The reaction with isocyanates gives the desired resin-bound
ureas, which can be cleaved from the resin by very mild acidic condition by using 1% TFA in
dichloromethane at room temperature. The 192 urea compounds were prepared in 20-50%
overall yields from the indole aldehyde resin. The identification and purity of the library
could be determined by LC-MS. Purities were above 80%. Compounds with purites below
this level were removed from the library.

WO 2006/045119 PCT/US2005/038282
46

[0145] As the polyether compounds of the invention increase the ease of formulation, oral
availability and serum half life of the compounds, another aspect of the present invention is to
provide a method of increasing ease of formulation, oral availability, or serum half-life of a
compound comprising covalently attaching a polyether substituent to a compound.
[0146] The following examples are provided to illustrate the invention and are not intended
to limit any aspect of the invention as set forth above or in the claims below.
EXAMPLES
[0147] All melting points were determined with a Thomas-Hoover apparatus (A.H. Thomas
Co.) and are uncorrected. Mass spectra were measured by LC-MS (Waters 2790). 'H-NMR
spectra were recorded on QE-300 spectrometer, using tetramethylsilane as an internal
standard. Signal multiplicities are represented as signlet (s), doublet (d), double doublet (dd),
triplet (t), quartet (q), quintet (quint), multiplet (m), broad (br) and broad singlet (brs).
Synthetic methods are described for representative compounds.
[0148] Lower case bolded Roman numerals in the examples below refer to the
corresponding intermediates in Schemes 1-4 above. Compounds numbers are also used as
provided in the Schemes as well as in the Tables below.

WO 2006/045119 PCT/US2005/038282
47
Example 1
Synthesis of 1-(3-chlorophenyl)-3-(4-oxodecyl)urea (794)
[0149] 1.00g (5.52 mmol) of benzophenone imine, 0.94 g (5.52 mmol) of ethyl 4-
aminobutyrate hydrochloride, and 20 mL of methylene chloride were stirred at room
temperature for 24 hr. The reaction mixture was filtered to remove NH4CI and evaporated to
dryness. The benzophenone Schiff base of ethyl 4-aminobutyrate (i) was extracted with ether
(20 mL), and the ether solution was washed with water (20 mL), dried over sodium sulfate
(Na2SO4), and concentrated. The residue was purified by column chromatography on silica
gel eluting with hexane and ethyl acetate (5:1) to give i (1.00 g, 61%) as an oil. To the
solution of the benzophenone Schiff base (i) in 20 mL of tetrahydrofuran (THF) was added
3.7 mL of 1M diisobutylaluminium hydride (DIBAL) solution in pentane (3.73 mmol) at
-78°C under nitrogen, and the reaction was stirred for 2 hr at the temperature. To 0.10 g of
magnesium turning (4.07 mmol) and I2 (catalytic amount) in THF (10 mL) was added 0.48
mL of hexylbromide (3.39 mmol) at room temperature under nitrogen. After stirring for 1 hr,
this reaction solution was added dropwise to the above reaction mixture at -78°C, and the
solution was allowed to warm to room temperature with stirring. After stirring for 5 hr at
room temperature, 10 mL of NaHCO3 aqueous solution was added to the reaction, then the
alkylated alcohol (ii) was extracted with ether (20 mL), and the ether solution was washed
with water (20 mL), dried over Na2SO4, and concentrated to give 0.26 g (60 %) of the alcohol
product (ii).
[0150] Acetic anhydride (2mL) was added to a solution of ii (0.77 mmol) in 5 mL of
dimethyl sulfoxide (DMSO). The mixture was allowed to stand at room temperature for 12
hr. and concentrated. The residue was extracted with ether (20 mL), and the ether was washed
with water (20 mL), dried over Na2SO4, and evaporated to provide 0.26 g (100 %) of the
ketone compound (iii). To a solution of iii in dioxane (5 mL) was added 1mL of 1N HC1 in
dioxane at room temperature. The reaction mixture was stirred for 2 hr and concentrated to
give keto amine hydrochloride (iv). Then iv was dissolved in 5 mL of dimethylformamide
(DMF) and treated with triethylamine (TEA, 0.27 mL, 1.95 mmol) and a solution of 3-
chlorophenyl isocyanate (0.10 mL, 0.78 mmol) in DMF (3 mL) at room temperature. After
stirring for 5 hr, the product was extracted with ether (30 mL), and the ether was washed with
water (30 mL), dried over Na2SO4, and evaporated to dryness. The residue was purified by

WO 2006/045119 PCT/US2005/038282
48
column chromatography on silica gel eluting hexane and ethyl acetate (3:1) to afford 75 mg
(30%) of 794. δ(CDC13): 0.88 (3H, t, /= 6.9 Hz), 1.21-1.29 (6H, m), 1.53-1.58 (2H, m), 1.81
(2H, quint, J = 6.9 Hz), 2.43 (2H, t, J=6.9 Hz), 2.49 (2H, t, J = 6.9 Hz), 3.23 (2H, t, /= 6.9
Hz), 5.10 (1H, s), 6.93 (1H, s), 6.98-7.02 (1H, m), 7.10-7.23 (2H, m), 7.49 (1H, s), [M + H]+
325.21
Example 2
Synthesis ofl-(3-chlorophenyl)-3-(3-pentoxycarbonylpropyl)urea (767)
[0151] To a suspension of 4-aminobutyric acid (1.41 g, 13.7 mol) in DMF (25 mL) was
added 3-chlorophenyl isocyanate (0.70 g, 4.56 mmol; cyclohexyl isocyanate for 772 and 1-
adamantyl isocyanate for 789) at room temperature. The reaction mixture was stirred for 24
hr. Then ethyl acetate (30 mL) and 1N HC1 aqueous solution (30 mL) were added into the
reaction, and the ethyl acetate layer dissolving the acid product was collected. The product
was extracted with ethyl acetate (20 mL) two more times from the aqueous layer. The
combined organic solution was dried over Na2SO4, and evaporated. The residue was purified
using column chromatography on silica gel eluting hexane and ethyl acetate (1:1) to give 0.88
g (75%) of urea acid (v). A mixture of v (0.50 g, 1.95 mmol), potassium carbonate (K2CO3,
0.54 g, 3.90 mmol), bromopentane (0.37 mL, 2.92 mmol), and sodium iodide (60 mg, 0.39
mmol) in DMF (20 mL) was stirred at room temperature for 20 hr. Then the product was
extracted with ether (20 mL), and the ether was washed with 1N NaOH aqueous solution (20
mL) and brine (20 mL), dried over Na2SO4, and evaporated to afford 0.59 g (92%) of 767.
δ(CDC13): 0.90 (3H, t, J= 6.9 Hz), 1.26-1.34 (4H, m), 1.62-1.65 (2H, m), 1.88 (2H, quint, J=
6.9 Hz), 2.41 (2H, t, J= 6.9 Hz), 3.30 (2H, t, J= 6.9 Hz), 4.08 (2H, t, J= 6.9 Hz), 4.96 (1H,
s), 6.62 (1H, s), 7.01-7.04 (1H, m), 7.18-7.22 (2H, m), 7.47 (1H, s), [M + H]+ 326.90
The following compounds were prepared in a similar manner:
l-Cyclohexyl-3-(3-pentoxycarbonylpropyl)urea (772)
[0152] δ(CDC13): 0.89 (3H, t, .7=6.9 Hz), 1.04-1.21 (2H, m), 1.29-1.43 (4H,m), 1.58-1.74
(6H, m), 1.82 (2H, quint, J= 6.9 Hz), 2.37 (2H, t, J= 6.9 Hz), 3.17-3.24 (2H, m), 3.46-3.48
(1H, m), 4.07 (2H, t, J= 6.9 Hz), 4.29 (1H, s), 4.47 (1H, s), [M + H]+ 299.24
1-(1 -Adamantyl)-3-(3-pentoxycarbonylpropyl)urea (789)

WO 2006/045119 PCT/US2005/038282
49
[0153] δ(CDC13): 0.92 (3H, t, j= 6.9 Hz),1.29-1.43 (4H, m), 1.64-1.69 (m, 10H), 1.83
(2H, quint, /= 6.9 Hz), 1.94-1.98 (6H, m), 2.06-2.09 (3H, m), 2.37 (2H, t, J= 6.9 Hz), 3.20
(2H, t, J= 6.9 Hz), 4.06-4.14 (3H, m), 4.30 (1H, s), [M + H]+ 251.26.
Example 3
Synthesis of l-(3-chlorophenyl)-3-(3-pentylaminocarbonylpropyl)urea (768)
[0154] To a suspension of 4-aminobutyric acid (2.84 g, 27.5 mmol) in DMF (30 mL) was
added TEA (3.86 mL, 27.5 mmol). To this mixture, di-t-butyl dicarbonate (2.00 g, 9.17
mmol) was added with stirring. The reaction mixture was heated to 50°C for 12 hr, and then
stirred with ice-cold dilute hydrochloric acid (15 mL) for 10 min. The t-butoxycarbonylated
amino acid (vi) was immediately extracted with ether (2 X 30 mL). The organic extract was
dried over Na2SO4 and evaporated to give 1.00 g (54%) of vi as an oil.
[0155] A solution of vi and 4-methyl morpholine (NMM, 0.54 mL, 4.92 mmol) in DMF
(10 mL) was treated at room temperature with isobutyl chloroformate (0.64 mL, 4.92 mmol).
After 30 min, pentylamine (0.57 mL, 4.92 mmol) was added. The reaction mixture was
stirred for 12 hr. The solvent was evaporated, and the residue was partitioned between ethyl
acetate (25 mL) and water (25 mL). The ethyl acetate layer was washed with 5% NaHCO3
(10 mL) and brine (20 mL) and dried over Na2SO4, and evaporated. The residue was
chromatographed on silica gel eluting hexane and ethyl acetate (2:1) to give 0.33 g (33%) of
t-butoxycarbonylated amino amide (vii). To a solution of vii in dioxane (10 mL) was treated
with 4M hydrochloric acid (2mL) in dioxane, and the mixture was stirred for lhr at room
temperature. Then the solvent was evaporated to dryness, and the residual solid was
dissolved in DMF (10 mL) and treated with TEA (0.51 mL, 3.63 mmol) and 3-chlorophenyl
isocyanate (0.15 mL, 1.21 mmol) at room temperature. After stirring for 5 hr, the product
was extracted with ether (30 mL), and the ether was washed with water (30 mL), dried over
Na2SO4, and evaporated to dryness. The residue was purified by column chromatography on
silica gel eluting hexane and ethyl acetate (3:1) to afford 0.39 g (100%) of 768. 5(CDC13):
0.89 (t, 3H, J= 6.9 Hz), 1.26-1.28 (4H, m), 1.46-1.50 (2H, m), 1.86 (2H, quint, J= 6.9 Hz),
2.30 (t, 2H, J= 6.9 Hz), 3.23 (t, 2H, J= 6.9 Hz), 3.30 (t, 2H, J= 6.9 Hz), 5.87 (1H, s), 6.06
(1H} s), 6.93-6.97 (1H, m), 7.12-7.23 (2H, m), 7.49 (1H, m),7.73 (1H, s), [M + H]+ 326.16.

WO 2006/045119 PCT/US2005/038282
50
Example 4
Synthesis of l-(3-chlorophenyl)-3-(2-hexylcarbonyloxyethyl)urea (761)
[0156] To a solution of 2-aminoethanol (2.98 g, 48.8 mmol) in DMF (30 mL) was added 3-
chlorophenol isocyanate (2.50 g, 16.3 mmol) at 0°C. The reaction mixture was stirred for 5
hr at room temperature. The solvent was evaporated, and the residue was partitioned between
ether (30 mL) and 1N hydrochloric acid (20 mL), and the ether layer was washed with brine,
dried over Na2SO4 and evaporated. The residue was purified by column chromatography on
silica gel eluting hexane and ethyl acetate (1:1) to provide 1.49 g (40%) of urea alcohol (viii)
as a white solid.
[0157] To a solution of viii (1.00g, 4.60 mmol) and TEA (0.97 mL, 6.90 mmol) in DMF
(15 mL) was added a solution of heptanoic anhydride (2.23 g, 9.20 mmol) in DMF (5 mL) at
room temperature. The reaction was stirred for 12 hr, and the solvent was evaporated. The
residue was partitioned between ether (30 mL) and cold 1N hydrochloric acid (20 mL). The
ether layer was washed with brine, dried over Na2SO4, and evaporated. The residual solid
was purified using silica gel column chromatography (hexane : ethyl acetate = 3:1) to afford
1.05 g (70%) of 761. δ(CDC13): 0.87 (t, 3H, J= 6.9 Hz), 1.20-1.29 (6H, m), 1.60-1.62 (2H,
m), 2.22-2.29 (2H, m), 3.50-3.55 (2H, m), 4.09-4.20 (2H, m), 5.32 (1H, s), 7.01-7.06 (2H,
m), 7.16-7.22 (2H, m), 7.40 (1H, s), [M + H]+ 327.15
[0158] Compounds 760 and 762 were prepared in the same manner as that used for
compound 761 from chloroformic acid pentyl ester and pentyl isocyanate in place of
heptanoic anhydride, respectively.
1 -(3-chlorophenyl)-3-(2-pentoxycarbonyloxyethyl)urea (760)
[0159] δ(CDC13): 0.91 (t, 3H, J= 6.9 Hz), 1.25-1.36 (4H, m), 1.63-1.67 (2H, m), 3.55-3.60
(2H, m), 4.14 (3H, t, J= 6.9 Hz), 4.25-4.28 (2H, m), 5.11 (1H, s), 6.50 (1H, s), 7.02-7.05
(1H, m), 7.19-7.23 (2H, m), 7.42 (1H, s), [M + H]+ 329.09
l-(3-chlorophenyl)-3-(2-pentylaminocarbonyloxyethyl)urea(762)
[0160] 1δ(CDC13): 0.87 (3H, t, J= 6.9 Hz), 1.30-1.33 (4H, m), 1.46-1.50 (2H, m), .3.12-
3.19 (2H, m), 3.50-3.52 (2H, m), 4.17-4.20 (2H, m), 4.83 (1H, s), 5.47 (1H, s), 6.96 (1H, s),
6.98-7.02 (lH,,m), 7.18-7.21 (2H, m), 7.44 (1H, s), [M + H]+ 328.20.

WO 2006/045119 PCT/US2005/038282
51
Example 5
Synthesis of l-(S-chlorophenyl)-3-(2-hexylcarbonylaminoeihyl)urea (765)
[0161] A solution of di-t-butyl dicarbonate (0.50 g, 2.29 mmol) in dioxane (20 mL) was
added over a period of 1 hr to a solution of 1,2-diaminoethane (1.10 g, 18.3 mmol) in dioxane
(20 mL). The mixture was allowed to stir for 22 hr and the solvent was evaporated to
dryness. Water (30 mL) was added to the residue and the insoluble bis-substituted product
was removed by filtration. The filtrate was extracted with methylene chloride (3 X 30 mL)
and the methylene chloride evaporated to yield ix as an oil (0.35 g, 95%).
[0162] A solution of heptanoic anhydride (0.91 g, 3.75 mmol; chloroformic acid pentyl
ester for 777 and pentyl isocyanate for 766) and ix (0.50 g, 3.13 mmol) in DMF (20 mL) was
stirred for 2 hr at room temperature. Then the solvent was evaporated. The residue was
partitioned between ether (30 mL) and water (30 mL). The ether layer was dried over
Na2SO4 and evaporated. The residue was purified by using column chromatography on silica
gel eluting hexane and ethyl acetate (1:1) to get 0.57 g (67%) of alkylated N-t-
butoxycarbonyl amine (x).
[0163] To a solution of x in dioxane (10 mL) was treated with 4M hydrochloric acid (2mL)
in dioxane, and the mixture was stirred for 1hr at room temperature. Then the solvent was
evaporated to dryness, and the residual solid was dissolved in DMF (10 mL) and treated with
TEA (0.58 mL, 4.19 mmol) and 3-chlorophenyl isocyanate (0.32 g, 2.10 mmol) at room
temperature. After stirring for 5 hr, the product was extracted with ether (30 mL), and the
ether was washed with water (30 mL), dried over Na2SO4, and evaporated to dryness. The
residue was purified by column chromatography on silica gel eluting hexane and ethyl acetate
(1:1) to afford 0.68 g (100%) of 765. δ(CDC13): 0.84 (t, 3H, J= 6.9 Hz), 1.16-1.25 (6H, m),
1.55-5.61 (2H, m), 2.21-2.24 (2H, m), 3.31-3.40 (4H, m), 6.27 (1H, s), 6.90-6.95 (2H, m),
7.18-7.20 (2H, m), 7.56 (1H, s), 8.07 (IB, s), [M + H]+ 326.25
The following compounds were prepared in a similar manner:
l-(3-chlorophenyl)-3-(2-pentoxycarbonylaminoethyl)urea (777)
[0164] δ(CDC13): 0.88 (3H, t, J= 6.9 Hz), 1.28-1.32 (4H, m), 1.44-1.49 (2H, m), 3.23-3.33
(4H, m), 3.95-3.97 (2H, m), 6.01 (1H, s), 6.34 (1H, s), 6.87-6.91 (1H, m), 7.18-7.26 (2H, m),
7.78 (1H, s), 8.21 (1H, s), [M + H]+ 328.22

WO 2006/045119 PCT/US2005/038282
52
l-(3-chlorophenyl)-3-(2-pentylaminocarbonylamiiioethyl)urea (766)
[0165] δ(Acetone): 0.87 (3H, t, J= 6.9 Hz), 1.27-1.30 (4H, m), 2.04-2.06 (2H, m), 3.02-
3.05 (2H, m), 3.20-3.22 (2H, m), 5.74 (2H, s), 6.22 (1H, s), 7.23-7.29 (2H, m), 7.82-7.87 (2H,
m), 8.67 (1H, s), [M + H]+ 327.10.
Example 6
Synthesis of l-(l-adamantyl)-3-(12-dodecanoic acid)urea (687)
[0166] A mixture of 1-adamantyl isocyanate (1.30 g, 7.34 mmol) and 12-aminododecanoic
acid (1.46 g, 6.77 mmol) in chloroform (30 mL) was refluxed for 10 hr. The solvent was
removed by evaporation, and the residue was washed with ethyl acetate (20 mL) to provide
2.66 g (100%) of urea acid product as a white solid. δ(CDC13): 1.20-1.36 (16H, m), 1.42-1.48
(2H, m), 1.57-1.65 (6H, m), 1.82-1.90 (6H, m), 1.94-1.98 (3H, m), 2.18 (2H, t, J= 6.9 Hz),
2.86-2.92 (2H, m), 3.45 (1H, bs), 5.43 (1H, s), 5.587 (1H, t, /= 5.4 Hz), [M + H]+ 393.28,
mp 140°C.
Example 7
Synthesis of' l-(l-adamantyl)-3-(ll-methoxycarbonylundecyl)urea (780)
[0167] To a mixture of compound 687 (0.15 g, 0.38 mmol), K2CO3 (64 mg, 0.46 mmol),
and iodomethane (54 mg, 0.38 mmol) in acetonitrile (20 mL) was refluxed for 10 hr. Then
the reaction mixture was filtered, and the filtrate was washed with brine (20 mL), dried over
Na2SO4, and evaporated. The residue was purified using column chromatography on silica
gel eluting hexane and ethyl acetate (3:1) to afford 0.14 g (92%) of 780 as a white solid.
δ(CDCb): 1.19-1.34 (12H, m), 1.41-1.48 (2H, m), 1.58-1.62 (4H, m), 1.63-1.75 (6H, m),
1.93-2.00 (6H, m), 2.04-2.07 (3H, m), 2.30 (2H, t, /= 6.9 Hz), 3.06-3.12 (2H, m), 3.67 (3H,
s), 4.00 (1H, s), 4.06 (1H, s), [M + H]+ 407.22, mp 75°C
[0168] Compounds 780,784, 783, 781, 788, 800, 785, 801, 802, 803, 804, and 782 were
prepared in the same manner using corresponding halides in a range of 30-95% yield.
l-(l-Adamantyl)-3-(ll-ethoxycarbonylundecyl)urea (784)
[0169] δ(CDC13): 1.21-1.38 (12H, m), 1.42-1.68 (15H, m), 1.96 (6H, bs), 2.06 (3H, m),
2.30 (2H, t, J= 6.9 Hz), 3.06-3.12 (2H, m), 3.97-4.01 (2H, bs), 4.12 (2H, q), [M + H]+
421.46, mp82°C

WO 2006/045119 PCT/US2005/038282
53
l-(l-Adamantyl)-3-(ll-propoxycarbonylundecyl)urea (783)
[0170] δ(CDC13): 0.94 (3H, t, J= 6.9 Hz ), 1.19-1.34 (12H, m), 1.41-1.48 (2H, m), 1.58-
1.62 (4H, m), 1.63-1.75 (8H, m), 1.93-2.00 (6H, m), 2.04-2.07 (3H, m), 2.30 (2H, t, /= 6.9
Hz), 3.06-3.12 (2H, m), 3.95-4.05 (4H, m), [M + H]+ 435.52, mp 86°C
1 -(] -Adamantyl)-3-(l 1 -allyloxycarbonylundecyl)urea (781)
[0171] δ(CDC13): 1.19-1.34 (12H, m), 1.41-1.48 (2H, m), 1.58-1.73 (13H, m), 1.93-2.00
(6H, m), 2.04-2.07 (3H, m), 2.33 (2H, t, J= 6.9 Hz), 3.06-3.12 (2H, m), 3.99 (1H, s), 4.04
(1H, s), 4.57-4.59 (2H, m), [M + H]+ 433.43, mp 81°C
l-(l-Adamantyl)-3-(l 1 -propagyloxycarbonylundecyl)urea (788)
[0172] δ(CDC13): 1.24-1.31 (12H, m), 1.44-1.46 (2H, m), 1.58-1.67 (11H, m), 1.94-1.98
(6H, m). 2.05-2.07 (3H, m), 2.35 (2H, t, J= 6.9 Hz), 3.05-3.12 (2H, m), 3.99 (1H, s), 4.04
(1H, s), 4.67 (2H, s), [M + H]+ 431.67, mp 79°C
l-(l-Adamantyl)-3-(ll-butoxycarbonylundecyl)urea (800)
[0173] δ(CDC13): 0.95 (3H, t, J= 6.9 Hz), 1.23-1.35 (12H, m), 1.44-1.52 (4H, m), 1.57-
1.61 (4H, m), 1.66-1.69 (6H, m), 1.96-2.00 (8H, m), 2.07-2.09 (3H, m), 2.30 (2H, t, /= 6.9
Hz), 3.09-3.13 (2H, m), 4.02-4.10 (4H, m), [M + H]+ 449.34
l-(l-Adamantyl)-3-(ll-iso-propoxycarbonylundecyl)urea (785)
[0174] δ(CDC13): 1.19-1.26 (18H, m), 1.41-1.48 (2H, m), 1.58-1.62 (4H, m), 1.63-1.75
(6H, m), 1.94-2.00 (6H, m), 2.03-2.07 (3H, m), 2.30 (2H, t, J= 6.9 Hz), 3.06-3.12 (2H, m),
3.67 (3H, s), 4.00 (1H, s), 4.06 (1H, s), 4.94-5.04 (1H, m), [M + H]+ 435.33, mp 90°C
1-(1 -Adamantyl)-3-(l 1 -sec-butoxycarbonylundecyl)urea (802)
[0175] δ(CDC13): 0.89 (3H, t, /= 6.9 Hz), 1.19 (3H, d, J= 6.9 Hz), 1.23-1.35 (12H, m),
1.44-1.50 (2H, m), 1.57-1.61 (4H, m), 1.66-1.72 (8H, m), 1.96-2.00 (6H, m), 2.07-2.09 (3H,
m), 2.27 (2H, t, /= 6.9 Hz), 3.09-3.13 (2H, m), 4.00 (1H, s), 4.05 (1H, s), 4.91-4.96 (1H, m);
and [M + H]+ 449.29, mp 65°C
l-(l-Adamantyl)-3-(ll-isobutoxycarbonylundecyl)wea (803)
[0176] δ(CDC13): 0.93 (6H, d, /= 6.9 Hz), 1.23-1.35 (12H, m), 1.45-1.47 (2H, m), 1.56-
1.58 (4H, m), 1.65-1.68 (6H, m), 1.94-1.97 (7H, m), 2.06-2.08 (3H, m), 2.31 (2H, t, /= 6.9
Hz), 3.07-3.11 (2H, m), 3.85 (2H, d, J= 6.9 Hz), 3.99 (1H, s), 4.03 (1H, s), [M + H]+449.32,
mp 91°C.

WO 2006/045119 PCT/US2005/038282
54
l-(l-Adamantyl)-3-(ll-benzyloxycarbonylundecyl)urea (804)
[0177] δ(CDC13): 1.24-1.28 (12H, m), 1.44-1.48 (2H, m), 1.63-1.68 (10H, m), 1.94-1.97
(6H, m), 2.05-2.07 (3H, m), 2.34 (2H, t, /= 6.9 Hz), 3.05-3.13 (2H, m), 4.04 (1H, s), 4.09
(1H, s), 5.12 (2H, s), 7.33-7.37 (5H, m), [M + H]+ 483.33, mp 49°C
l-(l-Adamantyl)-3-(ll-(2-chlorobenzyl)oxycarbonylundecyl)urea (782)
[0178] δ(CDC13): 1.24-1.28 (12H, m), 1.44-1.48 (2H, m), 1.63-1.68 (10H, m), 1.94-1.97
(6H, m), 2.05-2.07 (3H, m), 2.39 (2H, t, J= 6.9 Hz), 3.07-3.13 (2H, m), 4.00 (1H, s), 4.06
(1H, s), 5.23 (2H, s), 7.27-7.30 (3H, m), 7.39-7.42 (1H, m), [M + H]+ 517.05, mp 48°C.
Example 8
Synthesis of l-(l-adamantyl)-3-(ll-(l-adamantyl)methyloxycarbonylundecyl)urea (786)
[0179] . A solution of 687 (0.15, 0.38 mmol) and TEA (96 mg, 0.96 mmol) in DMF (10 mL)
was treated at room temperature with isobutyl chloroformate (52 mg, 0.38 mmol). After 30
min, a solution of adamantanemethanol (64 mg, 0.38 mmol) in DMF (2mL) was added. The
reaction mixture was stirred for 12 hr. The solvent was evaporated, and the residue was
partitioned between ethyl acetate (25 mL) and water (25 mL). The ethyl acetate layer was
washed with 5% NaHCO3 (10 mL) and brine (20 mL) and dried over Na2SO4, and
evaporated. The residue was chromatographed on silica gel eluting hexane and ethyl acetate
(5:1) to give 72 mg (35%) of 786 as a white solid. δ(CDC13): 1.23-1.33 (15H, m), 1.48-1.71
(21H, m), 1.90-1.96 (8H, m), 2.04-2.06 (3H, m), 2.31 (2H, t, J= 6.9 Hz), 3.05-3.12 (2H, m),
3.67 (2H, s), 4.00 (1H, s), 4.05 (1H, s), [M + H]+ 541.33, mp 68°C
[0180] Compound 792, 793 and 787 were prepared in this manner using ethylamine,
isopropylamine, and 1-naphthalenemethanol, respectively, instead of adamantanemethanol.
l-(l-Adamantyl)-3-(ll-ethylaminocarbonylundecyl)urea (792)
[0181] δ(CDC13): 1.14 (3H, t, /= 6.9 Hz), 1.24-1.31 (12H, m), 1.43-1.46 (2H, m), 1.58-
1.66 (10H, m), 1.94-1.98 (6H, m), 2.05-2.07 (3H, m), 2.15 (2H, t, /= 6.9 Hz), 3.06-3.12 (2H,
m), 3.25-3.13 (2H, m), 4.05 (1H, s), 4.12 (1H, s), 5.43 (1H, s), [M + H]+ 420.48, mp 119°C
l-(l-Adamantyl)-3-(ll-isopropylaminocarbonylundecyl)urea (793)
[0182] δ(CDC13): 1.14 (6H, d, J= 6.9 Hz), 1.24-1.31 (12H, m), 1.43-1.46 (2H, m), 1.61-
1.69 (10H, m), 1.94-1.98 (6H, m), 2.07-2.18 (5H, m), 3.07-3.13 (2H, m), 4.03-4.10 (2H, m),
4.14 (1H, s), 5.26 (1H, s), [M + H]+ 434.50, mp 115°C

WO 2006/045119 PCT/US2005/038282
55
1 -(1 -Adamantyl) -3-(l 1 -(1 -naphthyl)methoxycarbonylundecyl)urea (787)
[0183] δ(CDC13): 1.20-1.27 (12H, m), 1.43-1.46 (2H, m), 1.61-1.67 (10H, m), 1.96-2.06
(6H, m), 2.14-2.16 (2H, m), 2.35 (2H, t, J= 6.9 Hz), 3.06-3.10 (2H, m), 4.02(lH, s), 4.08
(1H, s), 5.57 (2H, s), 7.43-7.56 (4H, m), 7.84-7.87 (2H, m), 7.90 (8.02 (1H, m), [M + H]+
533.59.
Example 9
Synthesis of l-(l-Adamantyl)-3-(l l-t-butoxycarbonylundecyl)urea (801)
[0184] To a solution of compound 687 (0.10 g, 0.25 mmol), N,N-dmethylammopyridine
(DMAP, 10 mg, 0.13 mmol), and J-butanol (23 mg, 0.31 mmol) in methylene chloride (20
mL) was added l-(3-(dimemylammo)propyl)-3-ethylcarbodmnide hydrochloride (EDCI, 50
mg, 0.25 mmol) at room temperature. The mixture was stirred for 20 hr. The solvent was
evaporated, and the residue was partitioned between ether (30 mL) and water (30 mL). The
ether layer was dried over Na2SO4 and evaporated. Purification of the residue by silica gel
column chromatography eluting hexane and ethyl acetate (3:1) provided 21 mg (18 %) of t-
butyl ester as a white solid.
[0185] δ(CDC13): 1.23-1.35 (12H, m), 1.44-1.50 (2H, m), 1.57-1.61 (13H, m), 1.66-1.72
(6H, m), 1.96-2.00 (6H, m), 2.07-2.09 (3H, m), 2.27 (2H, t, J= 6.9 Hz), 3.09-3.13 (2H, m),
3.96 (1H, s), 4.01 (1H, s), [M + H]+ 449.36, mp 150°C.
Example 10
Synthesis of4-(3-Cyclohexyl-ureido)-butyric acid (632).
[0186] To a cold solution of 4-aminobutyric acid (2.16 g, 21 mmol) and catalytic amount of
DBU in 22 mL of 1.0 N NaOH, 2.5 g (20 mmol) of cyclohexyl isocyanate were added in one
time. The mixture was strongly mixed at room temperature overnight. The reaction was then
acidified with concentrated HC1. The formed white solid was collected by filtration. The
mixture was purified by chromatography on a silica column (8x3 cm). Elution with a
mixture 50:50:1 of hexane:ethyl acetate: acetic acid gave the pure targeted product. The
resulting white crystal (3.46 g; yield: 76%) had a mp of 153.0-154.0 °C. [M + H]+ 281.18.

WO 2006/045119 PCT/US2005/038282
56
Example 11
Synthesis of 2-[4-(3-Cyclohexyl-ureido)-butyrylammo]-3-(4-hydroxy-phenyl)-propionic acid
(632-Tyr).
[0187] To a solution of 632 (0.45 g, 2.0 mmol) and l-ethyl-3-(3-(dimethylamino)-propyI)
carbodiimide (0.5 g, 2.2 mmol) in 15 mL of DMF, 0.53 g (2.3 mmol) of tyrosine methyl ester
and 2.4 mmol of diisopropylethylamine were added. The mixture was heated at 60°C for 6h.
Then, 50 mL of 0.1 N NaOH were added and the mixture was left at room temperature
overnight. The reaction mixture was then acidified with concentrated HC1 and extracted
twice with a 2:1 mixture of chloroformrmethanol. The organic phases were pooled, dried and
evaporated. The residue was purified by chromatography on a silica column (5x4 cm).
Elution with a 75:25:1 mixture of ethyl acetate:methanol:acetic acid yielded 140 mg (yield:
18%) of the target product as a brown oily liquid. LC-MS-ES negative mode: 390.3 (100%,
[M-H]-), 290.9 (10%, (M-C6H10N]), 264.9 (5%, [M-C7H12NO]); positive mode: 392.5
(40%, [M+H]+), 264.95 (100%, [M-C7H10NO]+).
Example 12
Synthesis of 4-(3-Adamantan-l-yl-ureido) butyric Acid Methyl Ester (883).
[0188] The title compound was prepared by a procedure described in /. Med. Chem. 2004,
47,2110. To a suspension of 4-aminobutyric acid (2.79 g, 27.1 mmol) in DMF (40 mL) was
added 1-adamantyl isocyanate (1.20 g, 6.77 mmol) at room temperature. The reaction mixture
was stirred for 24 h. Then 1 N HC1 aqueous solution (40 mL) was added into the reaction,
and the mixture was stirred for 30 min. The solid crystalline product was filtered and washed
with water (20 mL) and ethyl acetate (20 mL). The resulting solid was dried in a vacuum
oven to give 1.90 g (100%) of 4-(3-adamantan-l-yl-ureido)butyric acid 822 as a white solid:
1H NMR (CD3OD): 1.66-1.75 (8H, m), 1.94-1.97 (6H, m), 2.05-2.07 (3H, m), 2.30 (2H, t, J
= 6.9 Hz), 3.08 (2H, q, J = 6.9 Hz), 3.32 (2H, s); LC-MS (ESI) m/z calcd for C15H24N2O3 [M
+ H]+ 281.18, found [M + H]+ 281.25; mp 165 C. Anal. (Cis^^Os) C, H, N.
[0189] A mixture of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (0.15 g, 0.54 mmol),
K2CO3 (0.09 g, 0.64 mmol), and iodomethane (0.04 mL, 0.59 mmol) in DMF (20 mL) was
stirred at room temperature for 20 h. Then the product was extracted with ether (20 mL), and
the ether was washed with 1 N NaOH aqueous solution (20 mL) and brine (20 mL), dried
over Na2SO4, and evaporated to afford 0.15 g (95%) of 883: lH NMR (CDCl3) 1.66-1.68

WO 2006/045119 PCT/US2005/038282
57
(6H, m), 1.81 (2H, quint, J = 6.9 Hz), 1.94-1.97 (6H, m), 2.05-2.07 (3H, m), 2.37 (2H, t, J =
6.9 Hz), 3.16 (2H, q, J = 6.9 Hz), 3.68 (3H, s), 4.09 (1H, s), 4.25 (1H, s); LC-MS (ESI) m/z
calcd for C16H26N2O3 [M + H]+ 295.19, found [M + H]+ 295.24; mp 114 C. Anal.
(C16H26N2O3)C,H,N.
[0190] Compounds 857, 876, 858, 877, and 878 were prepared in the same manner using
the corresponding ethyl bromoalkanoates instead of iodomethane to yield 30-95%.
Example 13
Synthesis of 4-(3-Adamantan-l-yl-ureido) butyric Acid 3,7-Dimethyl-oct-6-enyl Ester (798).
[0191] To a solution of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (0.10 g, 0.36 mmol),
4-(dimethylamino)pyridine (DMAP; 44 mg, 0.36 mmol), and 3,7-dimethyl-oct-6-en-l-ol (61
mg, 0.39 mmol) in methylene chloride (20 mL) was added l-[3-(dimethylamino)propyI]-3-
ethylcarbodiimide hydrochloride (EDCI; 75 mg, 0.39 mmol) at room temperature. After
stirring for 12 h, the reaction mixture was washed with 1 N NaOH aqueous solution (15 mL)
and water (30 mL), and the organic layer was dried over Na2SO4 and concentrated. The
residue was purified by column chromatography on silica gel eluting with hexane and ethyl
acetate (3:1) to give 798 (97 mg, 65%) as a solid: lH NMR (CDC13) 0.91 (3H, d, J = 6.9 Hz),
1.34-1.37 (2H, m), 1.56-1.60 (9H, m), 1.67-1.69 (8H, m), 1.81 (2H, quint, J = 6.9 Hz), 1.94-
1.97 (6H, m), 2.05-2.07 (3H, m), 2.35 (2H, t, J = 6.9 Hz), 3.16 (2H, q, J = 6.9 Hz), 4.05 (1H,
s), 4.11 (2H, t, J = 6.9 Hz), 4.21 (1H, s), 5.09 (1H, t, J = 6.9 Hz); LC-MS (ESI) m/z calcd for
C25H42N2O3 [M + H]+ 419.32, found [M + H]+ 419.22; mp 49 C. Anal. Calcd for
C25H42N2O3: C, 71.73; H, 10.11; N, 6.69. Found: C, 70.27; H, 9.83; N, 6.39.
Example 14
Synthesis of 8-[4-(3-Adamantan-l-yl-ureido)butyryloxy]octanoic Acid Ethyl Ester (879).
[0192] To a solution of 8-bromooctanoic acid (0.20 g, 0.89 mmol), DMAP (0.12 g, 0.99
mmol), and ethanol (0.05 g, 0.99 mmol) in methylene chloride (20 mL) was added EDCI
(0.19 g, 0.99 mmol) at room temperature. After stirring for 12 h, the reaction mixture was
washed with 1 N NaOH aqueous solution (15 mL) and water (30 mL), and the organic layer
was dried over Na2SO4 and evaporated to give 8-bromooctanoic acid ethyl ester (0.17 g,
75%). This bromide reacted with 4-(3-adamantan-l-yl-ureido)butyric acid 822 in the same
manner as that used for the preparation of 883 to provide 879 (0.19 g, 65%) as a solid: 1H

WO 2006/045119 PCT/US2005/038282
58
NMR (CDCl3) 1.26 (3H, t, J = 6.9 Hz), 1.32-1.35 (6H, m), 1.59-1.66 (10H, m), 1.82 (2H,
quint, J = 6.9 Hz), 1.94-1.97 (6H, m), 2.05-2.07 (3H, m), 2.28 (2H, t, J = 6.9 Hz), 2.36 (2H, t,
J = 6.9 Hz), 3.16 (2H, q, J = 6.9 Hz), 4.05-4.14 (5H, m), 4.31 (1H, s); LC-MS (ESI) m/z calcd
for C25H42N2O5 [M + H]+ 451.31, found [M + H]+ 451.20; mp 58-59 C. Anal. (C25H42N2O5)
C, H, N.
Example 15
Synthesis of 10-[4-(3-Adamantan-l-yl-ureido)butyryloxy]decanoic Acid Ethyl Ester (880).
[0193] A mixture of 10-hydroxydecanoic acid (0.25 g, 1.33 mmol; 11 -hydroxyundecanoic
acid for compound 881 and 12-hydroxydodecanoic acid for compound 882), ethyl bromide
(0.16 g, 1.46 mmol), and lithium carbonate (0.11 g, 1.46 mmol) in DMF (25 mL) was stirred
at 70 C for 6 h. Then the product was extracted with ether (30 mL), and the ether solution
was washed with 1 N NaOH aqueous solution (20 mL) and water (30 mL), dried over
Na2SO4, and concentrated. The residue was purified by column chromatography on silica gel
eluting with hexane and ethyl acetate (3:1) to give 10-hydroxydecanoic acid ethyl ester (80
mg, 28%). This alcohol was coupled with 4-(3-adamantan-l-yl-ureido)butyric acid 822 by
using EDCI/DMAP coupling reagent to give 880 (0.11 g, 60%) as a solid: 1H NMR (CDCl3)
1.24-1.32 (13H, m), 1.62-1.68 (10H, m), 1.80 (2H, quint, J = 6.9 Hz), 1.94-1.97 (6H, m),
2.05-2.07 (3H, m), 2.28 (2H, t, J = 6.9 Hz), 2.36 (2H, t, J = 6.9 Hz), 3.16 (2H, q, J = 6.9 Hz),
4.05-4.14 (5H, m), 4.25 (1H, s); LC-MS (ESI) m/z calcd for C27H46N2O5 [M + H]+ 479.34,
found [M + H]+ 479.29; mp 60-61 C. Anal. Calcd for C27H46N2O5: C, 67.75; H, 9.69; N, 5.85.
Found: C, 68.33; H, 9.92; N, 5.97.
[0194] Compound 4-(3-adamantan-1 -yl-ureido)butyric acid 822 was coupled with 11-
hydroxyundecanoic acid ethyl ester and 12-hydroxydodecanoic acid ethyl ester prepared from
corresponding acids to get compounds 881 and 882, respectively.
Example 16
Synthesis of 4-[4-(3-Adamantan-l-yl-ureido)butyryloxymethyl]benzoic Acid Ethyl Ester
(849).
[0195] A mixture of 4-formylbenzoic acid (1.00 g, 6.66 mmol), bromoethane (1.09 g, 9.99
mmol), and K2CO3 (1.10 g, 7.99 mmol) in acetonitrile (30 mL) was refluxed for 6 h. After
evaporation of the solvent, 4-formylbenzoic acid ethyl ester was extracted with ether (30

WO 2006/045119 PCT/US2005/038282
59
mL), and the organic solution was washed with 1 N NaOH aqueous solution (20 mL) and
water (30 mL), dried over Na2SO4, and concentrated to give the ethyl ester product (0.65 g,
55%). Without further purification, to a solution of the ester was added sodium borohydride
(NaBELt; 0.05 g, 3.65 mmol) in ethanol (20 mL) at 0 C. After stirring for 5 h at room
temperature, the product was extracted with ether (30 mL), and the ether solution was washed
with water (30 mL), dried over Na2SO4, and concentrated. The residue was purified by using
column chromatography on silica gel eluting with hexane and ethyl acetate (3:1) to give 4-
hydroxymethylbenzoic acid ethyl ester (0.30 g, 46%) as an oil.
[0196] To a solution of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (1.23 g, 0.83 mmol),
DMAP (0.05 g, 0.42 mmol), and the above alcohol (0.15 g, 0.83 mmol) in methylene chloride
(30 mL) was added EDCI (0.16 g, 0.83 mmol) at room temperature. After stirring for 12 h,
the reaction mixture was washed with 1 N NaOH aqueous solution (15 mL) and water (30
mL), and the organic layer was dried over Na2SO4 and concentrated. Then the residue was
purified by column chromatography on silica gel eluting hexane and ethyl acetate (5:1) to
provide 849 (0.28 g, 75%) as a white solid: 1H NMR (CDC13) 1.40 (3H, t, J = 6.9 Hz), 1.66-
1.68 (6H, m), 1.84 (2H, quint, J = 6.9 Hz), 1.94-1.96 (6H, m), 2.05-2.07 (3H, m), 2.44 (2H, t,
J = 6.9 Hz), 3.17 (2H, q, J = 6.9 Hz),.4.02 (1H, s), 4.17 (1H, s), 4.38 (2H, q, J = 6.9 Hz), 5.17
(2H, s), 7.40 (2H, d, J = 7.8 Hz), 8.00 (2H, d, J = 7.8 Hz); LC-MS (ESI) m/z calcd for
C25H34N2O5 [M + H]+ 443.25, found [M + H]+ 443.25; mp 96-99 C. Anal. (C25H34N2O5) C,
H,N.
Example 17
Synthesis of 4-(3-Adamantan-l-yl-ureido)butyric Acid 4-Ethoxycarbonylmethylphenyl Ester
(852).
[0197] To a solution of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (0.15 g, 0.54 mmol),
DMAP (0.07 g, 0.54 mmol), and 4-hydroxyphenylacetic acid (0.09 g, 0.59 mmol) in
methylene chloride (20 mL) was added EDCI (0.11 g, 0.59 mmol) at room temperature. After
stirring for 12 h, the reaction mixture was washed with water (20 mL), and the methylene
chloride solution dissolving the product was dried over Na2SO4 and concentrated to give
conjugated product. This crude mixture in DMF (30 mL) was treated with bromoethane (0.15
g, 1.34 mmol) and K2CO3 (0.18 g, 1.34 mmol) at room temperature and stirred for 12 h at
room temperature. The ethyl ester product was extracted with ether (30 mL), and the ether
solution was washed with 1 N NaOH aqueous solution (20 mL) and water (30 mL), dried

WO 2006/045119 PCT/US2005/038282
60
over Na2SO4, and concentrated. The residue was purified by column chromatography on
silica gel eluting hexane and ethyl acetate (5:1) to give 852 (47 ing, 20%) as a white solid: 1H
NMR (CDC13) 1.40 (3H, t, J = 6.9 Hz), 1.66-1.68 (6H, m), 1.89-1.95 (8H, m), 2.05-2.07 (3H,
m), 2.62 (2H, t, J = 6.9 Hz), 3.25 (2H, q, J = 6.9 Hz), 3.60 (2H, s), 4.07 (1H, s), 4.16 (2H, q, J
= 6.9 Hz), 4.29 (1H, s), 7.08-7.10 (2H, m), 7.28-7.30 (2H, m); LC-MS (ESI) m/z calcd for
C25H34N2O5 [M + H]+ 443.25, found [M + H]+ 443.25; mp 95-97 C. Anal. (C25H34N2O5) C,
H,N.
[0198] Compound 851 was prepared in the same manner by using 4-hydroxyphenylacrylic
acid instead of 4-hydroxyphenylacetic acid.
Example 18
Synthesis ofN-[12-(3-Adamantan-l-yl-ureido)dodecanoyl]methanesulfonamide (848)
[0199] To a solution of compound 687 (0.2 g, 0.51 mmol) and N-hydroxysuccinimide (60
mg, 0.56 mmol) in THF (10 mL) was added 1,3-dicyclohexylcarbodiimide (0.12 g, 0.56
mmol) at room temperature. The reaction mixture was stirred for 12 hrs and filtered. And
then, the filtrate was purified by column chromatography (hexane:ethyl acetate = 1:1) to give
2,5-dioxopyrrolidinyl ester (I) (0.18 g, 0.37 mmol) in 72% yield. To this intermediate (I)
dissolved in HMPA (10 mL) was added portionwise 4-dimethylarninopyridine (54 mg, 0.44
mmol; DMAP) and methanesulfonamide (0.35 g, 3.7 mmol). After stirring for 2hrs at 90°C,
the product was extracted with ether (30 mL) and washed with water (30 mL). The organic
solution was dried over magnesium sulfate and evaporated, and then the residue was purified
using column chromatography eluting with hexane and ethyl acetate (1:1) to afford
compound 848 (0.16 g, 0.34 mmol) in 92% yield. 1H NMR 5 (CDCl3) 1.23-1.35 (12H, m),
1.44-1.52 (4H, m), 1.57-1.61 (2H, m), 1.65-1.69 (6H, m), 1.92-1.98 (6H, m), 2.07-2.09 (3H,
m), 2.38 (2H, t, J = 6.9 Hz), 3.11 (2H, q, J = 6.9 Hz), 3.20 (3H, s), 4.40 (1H, s), 4.48 (1H, s),
10.52 (1H, s), LC-MS (ESI) m/z calcd for C24H43N3O4S [M + H]+ 469.30, found [M + H]+,
mp 103°C, Anal. (C24H43N3O4S) C, H, N.
[0200] Compound 914 was synthesized with the same method used for the preparation of
compound 687 using benzenesulfonamide instead of methanesulfonamide. 1H NMR 8
(CDCl3) 1.23-1.35 (12H, m), 1.44-1.52 (4H, m), 1.57-1.61 (2H, m), 1.65-1.69 (6H, m), 1.94-
1.98 (6H, m), 2.06-2.09 (3H, m), 2.28 (2H, t, J = 6.9 Hz), 3.10 (2H, q, J = 6.9 Hz), 4.39 (1H,
s), 4.93 (1H, s), 5.45 (1H, s), 7.50-7.55 (2H, m), 7.60-7.62 (1H, m), 7.80-7.83 (1H, m), 8.05-

WO 2006/045119 PCT/US2005/038282
61
8.08 (1H, m), LC-MS (ESI) m/z calcd for C29H45N3O4S [M + H]+ 532.31, found [M + H]+
532.34, mp 100°C, Anal. (C29H45N3O4S) C, H, N.
Example 19
Synthesis of2-[12-3-(Adamantan-l-yl-ureido)dodecanoylamino]decanoic acid (1001)
[0201] Sodium metal (3.9 g, 0.17 mol) was dissolved in ethanol (100 mL) under an inert
atmosphere in a round bottom flask fitted with a water condenser. Diethyl acetamido
malonate (30.4 g, 0.14 mol) was then added followed by 1-bromooctane (36.7 g, 0.19 mol).
The solution was refluxed overnight under an inert atmosphere. The reaction mixture was
poured onto crushed ice (600 mL) and stirred. The aminodiester product precipitated and was
collected by filtration. The crude product was then refluxed overnight in a solution HClrDMF
(9:1, 200 mL). The precipitated product was collected by filtration, washed with ice water,
and air dried in a vacuum desiccator to afford the a-amino acid hydrochloride in >90% crude
yield. The crude amino acid (3.0 g, 24.8 mmol) was then dissolved in methanol (100 mL) and
cooled to 0°C. Thionyl chloride (5.0 mL, 25.8 mmol) was added dropwise, and the reaction
was stirred at 0°C for 10 minutes and then refluxed overnight. The reaction was cooled to
room temperature and the volatiles removed under reduced pressure, and the crude product
was triturated in methanol to afford racemic methyl 2-amino decanoate, 4.44g, 89% yield.
[0202] Compound 687 (1.04 g, 2.65 mmol) and HBTU (1.0 g, 2.64 mmol) were dissolved
in THF (60 mL). DIEA (0.5 mL, 2.87 mmol), DMF (~ 2 mL), and methyl 2-amino decanoate
(1.26 g, 5.30 mmol) were added, and the solution was stirred under N2 at room temperature
overnight. The yellow oil produced was diluted with 5 % citric acid (100 mL) and extracted
with ethyl acetate (3 x 50 mL). Organic layers were combined and washed with 5 % citric
acid (2 x 50 mL), saturated sodium bicarbonate (NaHCO3) (2 x 50 mL), and brine (1 x 50
mL). The organic layer was dried over magnesium sulfate and evaporated to yield an oil. The
crude product was purified by column chromatography with 1-2 % MeOH/DCM to yield a
methyl ester as a yellow oil(0.77 g, 66 % yield). The methyl ester(0.77 g, 1.34 mmol) was
dissolved in DME (25 mL) and water (10 mL). Solid LiOH●H2O (0.33 g, 7.86 mmol) was
added, and the solution was stirred at room temperature overnight. Reaction mixture was
acidified (pH = 4) with 5 % citric acid (~ 20 mL), and the product was extracted with ethyl
acetate (3 x 30 mL). Combined organic layers were washed with brine (30 mL), dried over
MgSCU, filtered, and evaporated to yield compound 1001 as a yellow oil (0.34 g, 45 % yield).

WO 2006/045119 PCTYUS2005/038282
62
1H NMR 8 (CDC13) 0.82 (3H, t, J = 7 Hz), 1.16-1.23 (28H, bm), 1.41 (2H, m), 1.57 (2H, bm),
1.60 (6H, bs), 1.89 (6H, bs), 2.01 (3H, bs), 2.18 (2H, t, J = 7Hz), 2.98 (2H, m), 4.52 (1H, dd,
J = 7.3,13.3 Hz), 6.46 (2H, d, J = 7.6 Hz) 8.16 (1H, bs). 13C NMR 5 (CDCI3) 14.0, 22.5,
25.0, 25.6, 26.7, 28.85,28.00,29.06, 29.10, 29.13, 29.24, 29.31,29.44, 29.56, 31.73, 32.22,
36.27, 36.41,40.68, 42.29, 50.96, 52.31,158.70, 173.65,175.39. LC-MS (ESI) m/z calcd for
C33H59N3O4 [M + H]+ 562.45, found [M + H]+ 562.51.
Example 20
Synthesis of 12-(3-Adamantan-l-yl-ureido)dodecanoic acid [l-(2,3,4-trihydroxy-5-
hydroxymethyl-cyclohexylcarbamoyl)nonyl]amide (1002)
[0203] 1,2,3,4,6-Penta-O-acetyl-a-D-glucopyranose (5.00 g, 12.8 mmol) was dissolved in
10-15 mL dry CH2CI2 under an inert atmosphere. Trimethylsilyl azide (4.24 mL, 32.1 mmol)
and tin (IV) chloride (0.75 mL, 6.41 mmol) were added, and the reaction was stirred for 18
hours at room temperature. The reaction mixture was diluted with CH2CI2 (30 mL) and
washed twice with saturated NaHCC>3 (20 mL) and with brine (20 mL). The organic phase
was dried, filtered and concentrated to leave 2,3,4,6-tetra-O-acetyl-a-D-glucopyranosyl azide
as a white solid (3.92 g, 82%). 2,3,4,6-tetra-O-acetyl-a-D-glucopyranosyl azide (0.30 g, 80
mmol) was then dissolved in dry THF (15 mL), and Pd/C was added (~ 5 %) to this solution.
The mixture was stirred under H2 overnight. TLC revealed the reduction was completed
(Rfazide = 0.80, Rfamine = 0.10). In a separate flask, compound 23 (0.17 g, 0.30 mmol) and
0.5 M HBTU in DMF (1.20 mL, 0.60 mmol) were combined, followed by the addition of
DMF (~ 2 mL) and DIEA (104 ΜL, 0.60 mmol). This solution was then added to the reaction
flask, and the reaction mixture was stirred under N2 overnight. The resulting suspension was
filtered through celite which was washed well with ethyl acetate (~ 100 mL), and the filtrate
was washed with 5 M HC1 (50 mL), saturated NaHCO3 (2 x 50 mL), and brine (1 x 50 mL).
The organic solution was dried over magnesium sulfate, filtered, and concentrated. The
residue was purified using a silica gel column chromatography with 10 % MeOH/DCM to
yield V (0.16 g, 59.33). 1H NMR δ (CDCl3) 0.78 (3H, t, J = 7 Hz), 1.16 - 1.23 (32H, brm),
1.35 (2H, m) 1.52 (2H, m)1.57 (6H, brs), 1.86 (6H, brs,), 1.91-2.00 (15H, m), 2.14 (2H, m),
2.99 (2H, t, J = 7Hz), 3.75 (2H, m), 3.95-4.07 (3H, m), 4.15-4.29 (4H, m), 4.43-4.38 (1H, m)
4.80 (1H, dd, J = 5,11 Hz), 4.84-4.89 (2H, m), 4.95-5.02 (3H, m), 5.12-5.24 (3H, m), 5.35
(1H, d, J = 3 Hz), 5.47 (1H, t, J = 9.8 Hz), 6.28 (1H, dd, J = 7.6,19 Hz), 6.60 (1H, d, J = 9.3

WO 2006/045119 PCT/US2005/038282
63
Hz), 7.18 (1H, d, J = 9.1 Hz), 7.34 (1H, d, J = 9.1 Hz). LC-MS (ESI) m/z calcd for
C47H78N4O12 [M + H]+ 891.56, found [M + H]+ 891.69.
[0204] To a solution of V (0.16 g, 0.18 mmol) in MeOH (15 mL) was added sodium metal
(0.05 g), and the mixture was stirred at room temperature overnight. The reaction mixture
was filtered through amberlite and washed with MeOH (~ 150 mL). The solvent was
evaporated under vacuum to produce compound 1002 as a fine white powder (0.12 g, 95 %).
LC-MS (ESI) m/z calcd for C39H70N4O8 [M + H]+ 724.50, found [M + H]+ 724.6.
[0205] Compound 798 was prepared with the same method as that used for the preparation
of compound 1002 from compound 687 instead of compound 1001.
Example 21
Synthesis of5-(3-Chloro-phenylcarbamoyl)pentanoic Acid Pentyl Ester (987).
[0206] To a solution of adipic acid (0.5 g, 3.42 mmol) and DMAP (0.42 g, 3.42 mmol) in
dichloromethane (30 mL) and DMF (3 mL) was added 3-chloroaniline (0.44 g, 3.42 mmol) at
room temperature. After stirring 10 min, l-[3-(dimemylammo)propyl]-3-emylcarbodiimide
hydrochloride (0.65 g, 3.42 mmol; EDCI) was added portionwise to the mixture at room
temperature. The reaction was stirred for 12 h. A 1 N aqueous HC1 solution (20 mL) was
poured into the reaction mixture, and 5-(3-chlorophenylcarbomoyl)pentanoic acid was
extracted with dichloromethane (30 mL). The organic solution was washed with water (50
mL), dried over Na2SO4, and concentrated. This residue was used for the next reaction
without further purification.
[0207] To the solution of the above carbamoylpentanoic acid (0.72 g, 2.80 mmol) in DMF
(15 mL) was added K2CO3 (0.58 g, 4.21 mmol) and 1-bromopentane (0.64 g, 4.21 mmol) at
room temperature. After stirring for 12 h, the product was extracted with ether (30 mL), and
the ether solution was washed with an aqueous solution of 1 N NaOH (15 mL) and water (40
mL), dried over Na2SO4, and concentrated. The residue was purified using silica gel column
chromatography (hexane:ethyl acetate = 5:1) to afford 987 (0.59 g, 65%): 1H NMR (CDCl3)
0.91 (3H, t, J = 6.9 Hz), 1.29-1.37 (4H, m), 1.60-1.66 (2H, m), 1.70-1.78 (4H, m), 2.35-2.43
(4H, m), 4.08 (H, t, J = 6.9 Hz), 7.05-7.09 (1H, m), 7.21-7.23 (1H, m), 7.37-7.40 (1H, m),
7.52-7.55 (1H, m), 7.68 (1H, s); LC-MS (ESI) m/z calcd for C17H24CINO3 [M + H]+ 326.14,
found [M + H]+ 326.16, mp 82 C. Anal. (C17H24CINO3) C, H, N.
[0208] Compound 13 was prepared with the same method used for the preparation of
compound 987 using adamantylamine instead of 3-chloroaniline: 1H NMR (CDCl3) 0.91

WO 2006/045119 PCT/US2005/038282
64
(3H, t, J = 6.9 Hz), 1.29-1.43 (4H, m), 1.64-1.69 (12H, m), 1.94-1.98 (6H, m), 2.06-2.13 (5H,
m), 2.32 (2H, t, J = 6.9 Hz), 4.06 (H, t, J = 6.9 Hz), 5.16 (1H, s); LC-MS (ESI) m/z calcd for
C21H35NO3 [M + H]+ 350.26, found [M + H]+ 350.30. Anal. (C21H35NO3) C, H, N.
Example 22
Synthesis of 4-(2-Chloro-pheny)acethylaminolbutyric Acid Peniyl Ester (988).
[0209] To a solution of 3-chlorophenylacetic acid (0.5 g, 2.93 mmol) and DMAP (0.36 g,
2.93 mmol) in dichloromethane (30 mL) was added ethyl 4-aminobutyrate hydrochloride
(0.49 g, 2.93 mmol) at room temperature. After stirring for 10 min, EDCI (0.56 g, 2.93
mmol) was added portionwise to the mixture at room temperature. The reaction was stirred
for 12 h. A 1 N aqueous HC1 solution (20 mL) was poured into the reaction mixture, and 4-
[2-(3-chlorophenyl)acetylamino]butyric acid ethyl ester was extracted with ether (30 mL).
The ether solution was washed with water (50 mL), dried over Na2SO4, and concentrated. To
the residue dissolved in ethanol (10 mL) was added 1 N aqueous NaOH solution (6 mL), and
after 12 h of stirring at room temperature, the product was extracted with dichloromethane
(30 mL). The organic solution was washed with water (30 mL), dried over Na2SO4, and
concentrated to give 4-[2-(3-chlorophenyl)acetylamino]butyric acid (0.6 g, 80%). A mixture
of this acid (0.6 g, 2.35 mmol), K2CO3 (0.49 g, 3.52 mmol), and 1-bromopentane (0.53 g,
3.52 mmol) in DMF (20 mL) was stirred overnight at room temperature. The product was
extracted with ether (40 mL), and the ether solution was washed with water (50 mL), dried
over Na2SO4, and concentrated. The residue was purified using silica gel column
chromatography (hexane:ethyl acetate = 3:1) to afford 988 as an oil (0.74 g, 97%): 1H NMR
(CDCl3) 0.91 (3H, t, J = 6.9 Hz), 1.26-1.33 (4H, m), 1.59-1.63 (2H, m), 1.80 (2H, quint, J =
6.9 Hz), 2.31 (2H, t, J = 6.9 Hz), 3.27 (2H, q, J = 6.9 Hz), 3.52 (2H, s), 4.04 (2H, t, J = 6.9
Hz), 5.72 (1H, s), 7.13-7.17 (2H, m), 7.27-7.30 (2H, m); LC-MS (ESI) m/z calcd for
C7H24CINO3 [M + H]+ 326.14, found [M + H]+ 326.15. Anal. (C17H24ClNO3) C, H, N.
[0210] Compounds 837 and 1068 were prepared with the same method used for the
preparation of compound 988 using 1-adamantylacetic acid or adamantane-1-carboxylic acid
instead of 3-chlorophenylacetic acid. Compound 837: 1H NMR (CDC13) 0.91 (3H, t, J = 6.9
Hz), 1.29-1.35 (4H, m), 1.35-1.72 (14H, m), 1.84 (2H, quint, J = 6.9 Hz), 1.91 (2H, s), 1.97
(3H, m), 2.37 (2H, t, J = 6.9 Hz), 3.29 (2H, q, J = 6.9 Hz), 4.07 (2H, t, J = 6.9 Hz), 5.66 (1H,
s); LC-MS (ESI) m/z calcd for C21H35NO3 [M + H]+ 350.26, found [M + H]+ 350.29. Anal.
(C2,H35NO3)C,H,N.

WO 2006/045119 PCT/US2005/038282
65
[0211] Compound 1068: 1H NMR (CDC13) 0.91 (3H, t, J = 6.9 Hz), 1.29-1.35 (4H, m),
1.60-1.86 (16H, m), 2.03 (3H, m), 2.35 (2H, t, J = 6.9 Hz), 3.29 (2H, q, J = 6.9 Hz), 4.07 (2H,
t, J = 6.9 Hz), 5.85 (1H, s); LC-MS (ESI) m/z calcd for C20H33NO3 [M + H]+ 336.25, found
[M + H]+ 336.34. Anal. (C20H33NO3) C, H, N.
Example 23
Synthesis of 4-(3-Chloro-phenylcarbamoyloxy)butyric Acid Pentyl Ester (825).
[0212] To a solution of succinic anhydride (3.58 g, 35.7 mmol) and DMAP (4.16 g, 34.0
mmol) in DMF (40 mL) was added pentanol (3.0 g, 34.0 mmol) at room temperature under
nitrogen. After stirring for 12 h, succinic acid pentyl ester was extracted with ether (40 mL),
and the ether solution was washed with 1 N aqueous HC1 solution (20 mL) and water (40
mL), dried over sodium sulfate (Na2SO4), and concentrated. The residue was purified by
column chromatography on silica gel eluting with hexane and ethyl acetate (3:1) to give the
succinic acid pentyl ester (6.07 g, 95%) as an oil. To the solution of this acid in
tetrahydrofuran (THF, 60 mL) was added 1 M BH3-THF complex (64.53 mL, 64.5 mmol) at -
10C under nitrogen, and the reaction mixture was allowed to warm to room temperature with
stirring. After stirring for 12 h at room temperature, 5% NaHCC"3 aqueous solution (50 mL)
was added to the reaction and then the reduced alcohol (T) was extracted with ethyl acetate
(50 mL). The ethyl acetate solution was dried over Na2SO4 and concentrated to give 4-
Hydroxybutyric acid pentyl ester (5.06 g, 90%).
[0213] 4-Hydroxybutyric acid pentyl ester (100 mg, 0.57 mmol) was added to a solution of
3-chlorophenyl isocyanate (88 mg, 0.57 mmol) and triethylamine (0.12 mL, 0.86 mmol;
TEA) in DMF (15 mL) at room temperature. The mixture was allowed to stand at room
temperature for 12 h, the product was extracted with ether (20 mL), and the ether solution
was washed with 1 N aqueous HC1 solution (20 mL) and water (30 mL), dried over Na2SO4,
and concentrated. The residue was purified by column chromatography on silica gel eluting
with hexane and ethyl acetate (5:1) to afford 9 (94 mg, 50%) as a solid: 1H NMR (CDCl3)
0.90 (3H, t, J = 6.9 Hz), 1.26-1.34 (4H, m), 1.62-1.65 (2H, m), 1.71 (2H, quint, J = 6.9 Hz),
2.66 (2H, t, J = 6.9 Hz), 2.74 (2H, q, J = 6.9 Hz), 4.10 (2H, t, J = 6.9 Hz), 7.05-7.08 (1H, m),
7.18-7.22 (3H, m), 7.35 (1H, s); LC-MS (ESI) m/z calcd for C16H22ClNO4 [M + H]+ 328.12,
found [M + Hf 328.13; mp 82 C. Anal. (C16H22CINO4) C, H, N.

WO 2006/045119 PCTYUS2005/038282
66
Example 24
Synthesis of l-(3-Chloro-phenyl)-3-(l-hydroxymethyl-pentyl)-urea (978).
[0214] 2-aminohexanol hydrochloride (211 mg, 1.37 mmol), m - chlorophenyl isocyanate
(211 mg, 1.37 mmol) were combined in THF (10 mL) with triethylamine (228 uL) and stirred
over night. The solvent was removed and the residue chromatographed on silica gel to give
343 mg of the target product (93 %). 1H NMR (300 MHz, CDCl3/MeOH d4 1:1) 8 = 7.50 -
7.47 (m, 1H), 7.20 - 7.10 (m, 2H), 6.93 (dt, J = 6.6,1.6 Hz, 1H), 5.76 (d, J = 8.2 Hz, 1H),
3.75 (br, 1H), 3.65 - 3.45 (m, 2H), 1.60-1.20 (m, 6H), 1.87 (m, 3H).
[0215] Compound 977 were prepared in the same manner using aminoalcohol and
cyclohexylisocyanate.
Synthesis ofl-(3-Chloro-phenyl)-3-(l-hydroxymethyl-butyl)-urea (977)
[0216] !H NMR (300 MHz, CDC13) 8 = 5.41 (m, 2H), 3.8-3.2 (m, 4H), 2.00 - 0.90 (m, 20
H)
Example 25
Synthesis of(4-Butyl-4,5-dihydro-oxazol-2-yl)-(3-chloro-phenyl)-amine(980).
[0217] Compound 978 (50 mg) was treated with POC13 (1.0 mL). This was stirred
overnight. The solvent was removed via reduced pressure distillation. The residue was washd
with aqueous sodium bicarbonate and then dissolved in EtOAc. The solvent was dried,
filtered and evaporated. The residue was dissolved in acetone (1 mL) and refluxed with water
(2 mL) for 1.5 hrs. The solution was extracted with EtOAc, the organic layer evaporated and
the residue chromatographed on silica to give the target compound. 1H NMR (300 MHz,
CDCI3) 6= 7.38 (s, 1H), 7.15 (d, J=8.0 Hz, 1H), 7.05 (br, 1H), 7.00 (d, 7.5 Hz, 1H), 4.47 (m,
1H), 3.95 (m, 2H), 1.8-1.2 (m, 6H), 0.95 (m, 3H). ESMS = 224.9 m/z (M+H)
Example 26
Synthesis of (994).
[0218] Methylaminopentanoate hydrochloride (720 mg, 4.3 mmol) and
benzophenoneimine (722 mg, 4.3 mmol) were stirred together in dichloromethane for 18 hrs.
At this point, the reaction was washed with aqueous sodium bicarbonate and the organic layer

WO 2006/045119 PCT/US2005/038282
67
dried with MgS04 and evaporated to leave 1.3 g of a thick oil. The product (530 mg, 1.8
mmol) was then dissolved in THF (15 mL) and cooled (under N2) to -78°C. DBAL ( 2.0 mL,
1 M solution) was added dropwise and the reaction stirred for lhr. When TLC indicated the
absence of the ester starting material and the presence of a aldehyde (via DNP stain), 1.3 eq
of butyl magnesium bromide was added to the reaction. This was allowed to warm to room
temperature over 2.5 hrs. The reaction was quenched with bicarbonate solution and the
organic layer was dried and evaporated to give the imino alcohol (350 mg) as an oil. The
imino alcohol (150 mg) was stirred with aqueous HC1 (1M, 1mL) and diethyl ether (5 mL)
overnight. The aqueous layer was evaporated and 1 equivalent of 1-adamantylisocyanate was
added as a solution in dichloromethane (5 ml) and triethylamine (0.5 mL). This was stirred
overnight. The crude reaction was chromatographed on silica gel (1:1 EtOAc rhexanes) to
give the product as an oil (35 mg). 1H NMR (300 MHz, CDC13) δ= 4.22 (br, 1H), 4.08 (br,
1H), 3.91 (br, 1H), 3.10 (m, 1H), 2.30 (br, 2H), 2.2 -1.0 (br m, 32 H)
Example 27
Synthesis of (996).
[0219] Dodecylamine (500 mg, 2.7 mmol) was added to a biphasic system of
dichloromethane (20 mL) and aqueous bicarbonate (20 mL, satd). The mixture was stirred
while triphosgene (264 mg, 0.9 mmol) was added. The reaction was stirred for 2 hrs. The
organic layer was removed and filtered through a plug of silica gel using EtOAc as the eluent.
Evaporation of the solvent lead to 564 mg of the corresponding isocyanate. The isocyanate
(leq) was combined with the amine (15 mg, 0.048 mmol) in dichloromethane. This was
stirred overnight. The reaction was loaded onto silica gel and chromatographed using EtOAc
to yield the product (22 mg, 89%) as an orange solid. 1H NMR (300 MHz, CDC13) δ= 7.69
(d, J=8.3 Hz, 1H), 5.81 (d, J = 8.3 Hz, 1H), 5.15 (d, J=6.72 Hz, 1H), 4.70 (m, 1H), 4.60 (m,
1H), 4.00 - 3.5 (br, 2H), 3.30 (t, J = 6.7 Hz, 2H), 3.17 (q, J=6.6 Hz, 2H), 2.4-2.0 (m, 2H), 1.9
- 0.9 (br m, 32H). ESMS = 523 m/z (M+H)
Example 28
Synthesis of (997).
[0220] 12-aminododecanaol (50 mg) was stirred in dichloromethane with 1-
adamantylisocyanate (44mg) overnight. The reaction was evaporated and

WO 2006/045119 PCT/US2005/038282
68
carbonyldiimidazole was added in 2 mL acetonitrile. This was refluxed 5 hrs. The solvent
was removed in vacuo and the solid was partitioned between dichloromethane and water. The
organic layer was washed repeatedly with water to yield the target compound (22mg). 1H
NMR (300 MHz, CDC13) δ = 8.17 (s, IE), 7.45 (s, 1H), 7.05 (s, 1H), 4.41 (t, J = 7.5 Hz, 2H),
4.32 (br, 1H), 4.21 (br, 1H), 3.08 (q, J = 6.7 Hz, 2 H), 2.0 - 1.0 (m, 35H).
Example 29
General procedure for the synthesis of trans-benzyloxy isomers: Synthesis of trans-1-(4-
Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7] decan-1-yl-urea 1032.
Synthesis oftrans-l-(4-Hydroxy-cyclohexyl)-3-tricyclo[3.3.L13,7]decan-1-yl-urea 1039
[0221] To a solution of 1-adamantyl isocyanate (10 g, 56.4 mmol) and trans-4-
aminocyclohexanol hydrochloride (10.3 g, 67.7 mmol) in DMF (300 mL) was added
triethylamine (6.9 g, 67.7 mmol) at 0 oC. The reaction mixture was stirred overnight. The
reaction mixture was poured into water, and the resulting precipitates were collected and
washed with water. The crude product was recrystallized from methanol/water. Yield: 15.5
g (94% of theory). M.P.: 254 °C. 1H NMR (300 MHz, DMSO-d6): δ 5.48 (d. J = 9 Hz, 1H),
5.38 (s, 1H), 4.48 (d, J = 4.7 Hz, 1H), 3.42-3.28 (m, 1H), 3.28-3.13 (m, 1H), 2.02-1.93 (m,
3H), 1.87-1.68 (m, 9H), 1.63-1.54 (m, 7H), 1.24-0.93 (4H).
Synthesis of trans-l-(4-Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-1-yl-urea 1032
[0222] To a solution of compound 1 in DMF (10 mL) was added 60% sodium hydride in
oil (60 mg, 1.5 mmol) at 0 °C. After 10 min, benzyl bromide (0.20 g, 1.2 mmol) was
introduced and then warmed up to room temperature and stirred overnight. The reaction was
quenched by adding water and the resulting white precipitates were collected and washed
with water. The solids were recrystallized from DCM/hexanes. Yield: 0.35 g (92% of
theory). M.P.:244°C. 1HNMR(300MHz,CDCl3): δ 7.40-7.23 (m, 5H), 4.52 (s, 2H),
4.10-3.92 (m, 2H), 3.58-3.41 (m, 1H), 3.37-3.24 (m, 1H), 2.11-1.81 (m, 13H), 1.50-1.33 (m,
6H), 1.50-1.33 (m, 2H), 1.17-0.99 (m, 2H).

WO 2006/045119 PCT/US2005/038282
69
Example 30
General procedure for the synthesis of cis-benzyloxy isomers: Synthesis ofcis-l-(4-
Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-l-yl-urea 1078
Synthesis of trans-4-Nitro-benzoic acid 4-(l,3-dioxo-l,3-dihydro-isoindol-2-yl)-cyclohexyl
ester
[0223] To a solution of trans-2-(4-Hydroxy-cyclohexyl)-isoindole-l,3-dione (38 g, 154.9
mmol), triphenylphosphine(65 g, 248 mmol), and 4-nitrobenzoic acid (41.4 g, 248 mmol) in
1500 mL of THF was added dropwise diisopropyl azodicarboxylate (50.1 g, 248 mmol) at
room temperature. The reaction mixture was stirred overnight. The solvent was evaporated,
and the resulting solid was recrystallized from methanol. Yield: 53 g (86.7% of theory). 1H
NMR (300 MHz, CDC13): δ 8.40-8.36 (m, 4H), 7.79 (ddd, J = 0.12, 0.02, and 0.02 Hz, 4H),
5.39 (s, 1H), 4.37-4.22 (m, 1H), 2.82-2.65 (m, 2H), 2.27-2.16 (m, 2H), 1.84-1.65 (m, 4H).
Synthesis of trans-4-Nitro-benzoic acid 4-amino-cyclohexyl ester
[0224] 35 wt% Hydrazine hydrate (0.93 g, 10.1 mmol) was added to a solution of the above
compound (2.0 g, 5.1 mmol) in DCM (50 mL) followed by MeOH (50 mL) at room
temperature. The reaction mixture was allowed to stir overnight. The resulting white
precipitates were filtered off and the solvent was removed in vacuo. The resulting white
solids were dissolved in aqueous 1N HCI solution and washed with DCM. Aqueous layer
was basified with excess IN NaOH solution and then extracted with DCM. After drying with
MgSO4, the solvent was evaporated affording crude trans-4-Nitro-benzoic acid 4-amino-
cyclohexyl ester as a solid, which was used in the next step without further purification.
Yield: 1.1 g (89% of theory). 1H NMR (DMSO-d6) δ 8.26 (dd, J = 43.5 and 8.8 Hz, 4H),
6.72 (d, J = 7.3 Hz, 2H), 5.08 (s, 1H), 2.00-1.36 (m, 9H).

WO 2006/045119 PCT/US2005/038282
70
Synthesis of trans-4-Nitro-benzoic acid 4-(3-tricyclo[3.3.L13,7]decan-l-yl-ureido)-
cyclohexyl ester 1076
[0225] To a solution the above compound (1.33 g, 5.1 mmol) in DMF was added 1-
adamantyl isocyanate (0.82 g, 4.6 mmol) followed by triethylamine (0.47 g, 4.6 mmol) at 0
°C. The reaction mixture was stirred overnight. The reaction mixture was poured into water,
and the resulting precipitates were collected and washed with water. The crude product was
recrystallized from DCM/hexanes. Yield: 1.83 g (90% of theory). M.P.:124°C. 1H NMR
(300 MHz, CDC13): δ 8.24 (dd, J = 28.7 and 9.1 Hz, 4H), 5.23 (s, 1H), 4.13 (d, J = 7.2 Hz,
1H), 4.05 (s, 1H), 3.75-3.61 (m, 1H), 2.17-1.41 (m, 23H).
Synthesis of cis-l-(4-Hydroxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-l-yl-urea 1077
[0226] To a solution of ester 1076 (1 g, 2.3 mmol) in THF (100 mL) was added IN NaOH
solution (4.6 mL, 4.6 mmol) at room temperature. The reaction mixture was stirred
overnight, at which time the reaction was quenched by addition of IN HC1 solution (5.5 mL).
The resulting white precipitate was collected by filtration and recrystallized from
methanol/water. Yield: 0.63 g (95% of theory). M.P.: 205 °C. 1H NMR (300 MHz, DMSO-
d6): δ 5.67 (d, J = 8.2 Hz, 1H), 5.45 (s, 1H), 4.41 (s, 1H), 3.63-3.51 (m, 1H), 3.46-3.36 (m,
1H), 2.00-1.92 (m, 3H), 1.87-1.72 (m, 6H), 1.66-1.28 (m, 14H).
Synthesis ofcis-l-(4-Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-1-yl-wea 1078
[0227] Compound 1078 (2.22 g, 60%) was synthesized from compound 1077 (0.29 g, 1
mmol) by the same method) as that described for compound 1032 with benzyl bromide (0.20
g, 1.2 mmol) and 60% sodium hydride (0.06 g, 1.5 mmol). Yield: 0.35 g (92% of theory).
M.P.: 181 °C. 1H NMR (300 MHz, CDC13): δ 7.43-7.24 (m, 5H), 4.49 (s, 2H), 4.11 (d, J =
8.3 Hz, 1H), 4.02 (s, 1H), 3.66-3.51 (m, 2H), 2.23-1.07 (m, 23H).
Example 31

WO 2006/045119 PCT/US2005/038282
71
General procedure for the synthesis of cis-phenoxy isomers: Synthesis ofcis-l-[4-(4-
Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7] decan-1-yl-urea 1135
Synthesis of cis-2-[4-(4-Fluoro-phenoxy)-cyclohexyl]-isoindole-l,3-dione
[0228] To a solution of trans-2-(4-Hydroxy-cyclohexyl)-isoindole-l,3-dione 3 (1.0 g, 4.1
mmol), triphenylphosphine (1.3 g, 4.9 mmol), and 4-fluorophenol (0.55g, 4.9 mmol) in 40
mL of THF was added dropwise at room temperature diisopropyl azodicarboxylate (0.99 g,
4.9 mmol). The reaction mixture was stirred overnight. The solvent was evaporated after 12
h, and the resulting solid was purified by recrystallization from methanol. Yield: 1.1 g (80%
of theory). 1H NMR (300 MHz, CDC13): δ 7.84-7.80 (m, 2H), 7.71-7.67 (m, 2H), 6.98-6.94
(m, 4H), 4.51 (s, 1H), 4.26-4.12 (m, 1H), 2.76 - 2.60 (m, 2H), 2.18 (d, J = 13 Hz, 2H), 1.79-
1.49 (m,4H).
Synthesis ofcis-l-[4-(4-Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7]decan-1-yl-urea
1135
[0229] 35 wt% Hydrazine hydrate (0.27 g, 2.9 mmol) was added to a solution of the above
compound (0.5 g, 1.5 mmol) in DCM (15 mL) followed by MeOH (15 mL) at room
temperature. The reaction mixture was allowed to stir overnight The resulting white
precipitates were filtered off and the solvent was removed in vacuo. The resulting white
solids were dissolved in aqueous 1N HC1 solution and washed with DCM. Aqueous layer
was basified with excess 1N NaOH solution and then extracted with DCM. After drying with
MgSO4, the solvent was evaporated affording crude cis-4-(4-Fluoro-phenoxy)-
cyclohexylamine 10 as a solid, which was used in the next step without further purification.
[0230] To a solution compound 10 in DMF was added 1-adamantyl isocyanate (0.16 g,
0.91 mmol) followed by triethylamine (0.10 g, 1.0 mmol) at 0 °C. The reaction mixture was
stirred overnight. The reaction mixture was poured into water, and the resulting precipitates
were collected and washed with water. The crude product was recrystallized from
DCM/hexanes. Yield: 0.31 g (88% of theory). M.P.: 207 °C. 1H NMR (300 MHz, CDCl3):
δ 6.98-6.91 (m, 2H), 6.84-6.78 (m, 2H), 4.34 (s, 1H), 4.30 (d, J = 9.8 Hz, 1H), 4.20 (s, 1H),
3.71-3.56 (m, 1H), 2.13 -1.44 (m, 23H).

WO 2006/045119 PCT/US2005/038282
72
Example 32
General procedure for the synthesis of trans-phenoxy isomers: Synthesis of trans-1-[4-(4-
Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7]decan-l-yl-urealVi6.
Synthesis of cis-2-(4-Hydroxy-cyclohexyl)-isoindole-l,3-dione
[0231] IN NaOH solution (19 mL, 19 mmol) was added at room temperature to a solution
of ester 4 (5 g, 12.7 mmol) in THF (100 mL). The mixture was stirred overnight at room
temperature, at which time the reaction was quenched by addition of IN HC1 solution (40
mL). The solvent was removed under reduced pressure, and the resulting white precipitate
formed was collected by filtration and dissolved in DMF. After adding triethylamine (6.5 g,
64 mmol) at room temperature, the reaction mixture heated at 150 °C for 30 min in the
microwave. After cooling to r.t., the reaction mixture was poured into water and then
extracted with ether. The organic layer was washed with water thoroughly. After drying
with MgSO4, the solvent was removed in vacuo. The resulting white solids were
recrystallized from DCM/hexanes. Yield: 1.9 g (60% of theory). 1H NMR (300 MHz,
CDC13): δ 7.76 (ddd, J = 37.6, 5.4 and 3.1 Hz, 4H), 4.21-4.07 (m, 2H), 2.72-2.55 (m, 2H),
1.96 (d, J = 13.9 Hz, 2H), 1.73-1.50 (m, 4H).
Synthesis of trans-2-[4-(4-Fluoro-phenoxy)-cyclohexyl]-isoindole-1,3-dione
[0232] Synthesis of trans-2-[4-(4-Fluoro-phenoxy)-cyclohexyl]-isoindole-l,3-dione was
synthesized from the above compound (1 g, 4.1 mmol) by the same method as that described
with DIAD (0.99g, 4.9 mmol), PPh3 (1.3 g, 4.9 mmol), and 4-fluorophenol (0.55g, 4.9
mmol). Yield: 0.56 g (40% of theory). 1H NMR (300 MHz, CDCl3): δ 7.77 (ddd, J = 37.6,
5.4 and 3.1 Hz, 4H), 7.00-6.84 (m, 4H), 4.30-4.15 (m, 2H), 2.48-2.31 (m, 2H), 2.26 (d, J =
11.4 Hz, 2H), 1.89-1.77 (m, 2H), 1.65-1.49 (m, 4H).
Synthesis oftrans-l-[4-(4-Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7]decan-l-yl-
urea 1136

WO 2006/045119 PCT/US2005/038282
73
[0233] Compound 1136was synthesized (0.5 g, 1.5 mmol) by the same method as that
described above with 35 wt% hydrazine (0.27g, 2.95 mmol) in 15 mL of DCM and 15 mL of
MeOH followed by the reaction with 1-adamantyl isocyanate (0.12g, 0.67 mmol) and
triethylamine (0.07g, 0.74 mmol) in DMF (5 mL). Yield: 0.24 g (93% of theory). M.P.:
243 °C. 1HNMR (300 MHz, CDC13): δ 6.98-6.91 (m, 2H), 6.85-6.79 (m, 2H), 4.12-3.94 (m,
3H), 3.66-3.51 (m, 1H), 2.17-1.88 (m, 12H), 1.73-1.45 (m, 9H), 1.28-1.11 (m, 2H).
Example 33
Combinatorial synthesis of urea inhibitors
[0234] A mixture of PS-Indole-CHO resin (0.5 g, 0.46 mmol), THF (3 mL), TEOF (3 mL)
and the primary amine (1.0 mmol) was agitated at ambient temperature for 4 h. Then, a
solution of NaBH3CN (1.0 mL, 1 M) in THF and acetic acid (O.lmL) were added. The
resulting mixture was stirred for 2h. The supernatant liquid was drained off and the resin
washed with THF (8 mL*2), MeOH (8 mL*3) and DCM (8 mLx2).
[0235] To a suspension of the resin-bound secondary amine (0.1 g) in DCM (2 mL) was
added an isocyanate (0.5 mmol). The resulting mixture was agitated overnight at ambient
temperature. The supernatant liquid is drained off and the resin washed with DMF, MeOH
and DCM.
[0236] The resin-bound product was suspended hi 1 % (v/v) TFA (3 mL, ca. 4 mol equiv.)
and the mixture agitated at ambient temperature for 4 h. The color of the resin became deep
purple. The supernatant liquid was collected and the resin was washed with DCM (2x2 mL).
The combined solution was concentrated to afford pure products in excellent yields.
Example 34
[0237] This example provides assays and illustrates the inhibition of mouse and human
soluble epoxide hydrolases by compounds of the invention having a secondary
pharmacophore that is a carboxylic acid or carboxylic methyl ester functional group.
Enzyme preparation
[0238] Recombinant mouse sEH and human sEH were produced in a baculovirus
expression system and purified by affinity chromatography. The preparations were at least
97% pure as judged by SDS-PAGE and scanning densitometry. No detectable esterase or
glutathione transferase activity, which can interfere with this sEH assay, was observed.

WO 2006/045119 PCT/US2005/038282
74
Protein concentration was quantified by using the Pierce BCA assay using Fraction V bovine
serum albumin as the calibrating standard.
IC50 Assay conditions
[0239] IC50 values were determined in one of two method. One method uses racemic 4-
mtophenyl-trans-2,3-epoxy-3-phenylpropyl carbonate as substrate. Enzymes (0.12 fiM
mouse sEH or 0.24 μM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer, 0.1 M pH 7.4, at 30°C before substrate introduction([S] = 40 μM). Activity
was assessed by measuring the appearance of the 4-nitrophenolate anion at 405 nm at 30°C
during 1 min (Spectramax 200; Molecular Devices). Assays were performed in triplicate.
IC50 is a concentration of inhibitor, which reduces enzyme activity by 50%, and was
determined by regression of at least five datum points with a minimum of two points in the
linear region of the curve on either side of the IC50. The curve was generated from at least
three separate runs, each in triplicate, to obtain the standard deviation (SD) given in Table 1
thru Table 4.
[0240] Other IC50 values were determined using the procedure described in Analytical
Biochemistry 343 66-75 (2005) using cyano(6-methoxy-naphthalen-2-yl)methyl trans-[(3-
phenyloxiran-2-yl)methyl] carbonate as a substrate (see Table 18b). Enzymes (0.88 nM for
murine and 0.96 nM for human sEH) were incubated with inhibitors ([T] = 0.5-10,000 nM)
for 5 min in BisTris-HCl buffer (25 mM, pH 7.0, containing 0.1 mg/ml of BSA) at 30 °C
prior to substrate introduction ([S] = 51M). Enzyme activity was measured by monitoring the
appearance of 6-methoxy-2-naphthaldehyde. Assays were performed in triplicate. By
definition, IC50 values are concentrations of inhibitor that reduce enzyme activity by 50%.
IC50 values were determined by regression of at least five datum points, with a minimum of
two datum points in the linear region of the curve on either side of the IC50 values. The curve
was generated from at least three separate runs, each in triplicate.
[0241] Assays were conducted with the compounds indicated in Table 1, as described
above.

WO 2006/045119 PCTYUS2005/038282
75
Table 1: Inhibition of mouse and human sBH by l-cyclohexyl-3-n-(substituted)alkylureas

a Enzymes (0.12 uM mouse sEH and 0.24 uM human sEH) were incubated with
inhibitors for 5 min in sodium phosphate buffer (pH 7.4) at 30°C before substrate
introduction ([S] = 40μM). Results are means ± SD of three separate experiments.
[0242] As can be seen from the above table, the conversion of a carboxylic acid function to
its methyl ester (549, 635, and 774) increased inhibition potency for both mouse and human
i
sEHs. Moreover, the methyl ester of butanoic acid (774) showed 8-100 fold higher activity
than the esters of acetic and propanoic acids (549 and 635) for both enzymes, indicating that
a polar functional group located three carbon units (carbonyl on the fourth carbon, about 7.5
angstroms from the urea carbonyl) from the carbonyl of the primary urea pharmacophore can
be effective for making potent sEH inhibitors of improved water solubility, hi addition, the

WO 2006/045119 PCT/US2005/038282
76
distance from the carbonyl of the primary urea pharmacophore to the secondary ester
pharmacophore in compound 854 is about 8.9 A showing that the secondary pharmacophore
may be located about 7 A to about 9 A from the carbonyl of the primary urea pharmacophore
group.
Example 35
[0243] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention having a secondary pharmacophore, with
comparison to compounds having only a primary pharmacophore. As can be seen from the
results in Table 2, the activity is relatively consistent.
[0244] Assays were conducted with the compounds indicated in Table 2, according to
established protocols (see, above).
Table 2: Inhibition of mouse and human sEH by l-cycloalkyl-3-alkylureasa


WO 2006/045119 PCT/US2005/038282
77

Enzymes (0.12 μM mouse sbn and 0.24 uM human sEH) were incubated with inhibitors for 5 mm in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40μM). Results are means ± SD of three
separate experiments.
[0245] As shown in the above table, the substitution at R with a cyclohexyl (772) or
adamantyl (789) increased inhibitor potency 10-fold over the 3-chlorophenyl analog (767, see
Table 3 below). Furthermore, these compounds functionalized with a polar group were as
active and potent as non-functionalized lipophilic inhibitors (for example, 791, 790,297, and
686) for both murine and human enzymes. Adding polar groups to compounds generally
increases their water solubility, and this was the case when one compares compounds 772 or
789 to 791 and 790. In addition, stripping water of hydration out of the enzyme catalytic site
requires about the same amount of energy that is gained by forming a new hydrogen bond
between the inhibitor and the enzyme. Thus addition of polar groups which hydrogen bond
to a target enzyme does not dramatically increase potency if the inhibitor is already potent.
However, the presence of an additional polar group can be expected to dramatically increase
specificity by decreasing hydrophobic binding to biological molecules other than the primary
target (sEH). In this way combining several active pharmacophores into a single molecule
often has a massive increase in specificity and biological activity in complex biological
systems.
Example 36
[0246] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention having a secondary pharmacophore that is a
ketone, amide, alcohol, carbonate, carbamate, urea, carboxylate ester functional group.
[0247] Based on the initial activity shown in Table 1, urea compounds were prepared
having a polar carbonyl group located approximately 7.5 angstroms from the carbonyl of the
primary urea pharmacophore to improve water solubility of lipophilic sEH inhibitors (192

WO 2006/045119 PCT/US2005/038282
78
and 686). The table below shows various functionalities such as ketone, ester, amide,
carbonate, carbamate, and urea which contribute a carbonyl group, and are termed as the
secondary pharmacophores. To determine the effect for each of the secondary
pharmacophores, a 3-chlorophenyl group was held constant as one of substituents of the urea
pharmacophore. The 3-chlorophenyl group is also particularly useful for monitoring
chemical reactions quickly via chromatography. After optimizing the secondary
pharmacophore, the aryl substituent can be replaced by a cyclohexyl, adamantyl or other
group leading to more potent inhibitors.
[0248] Assays were conducted with the compounds indicated in Table 3, according to
established protocols (see, above).
Table 3: Inhibition of mouse and human sEH by l-(3-chlorophenyl)-3-(2-alkylated
ethyl)ureasa


WO 2006/045119 PCT/US2005/038282
79

a Enzymes (0.12 μM mouse sEH and 0.24 uM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40μM). Results are means ± SD of three
separate experiments.
[0249] When the left of the carbonyl (X) is a methylene carbon, the best inhibition was
obtained if a methylene carbon (ketone, 794) or oxygen (ester, 767) is present in the right
position (Y). The ester bond can be stabilized by stearic hindrance of the alcohol or acid
moiety or both (805). The presence of nitrogen (amide, 768) reduced the activity. In
compounds with an oxygen in the left of the carbonyl group, a > 10-fold drop in activity was
observed and there was not any change in tide activity even if the right position, Y, was
modified with a methylene carbon (ester, 761), oxygen (carbonate, 760), or nitrogen
(carbamate, 762), respectively. All compounds (765,777, and 766) with nitrogen in the left
position had lower activities than 794 or 767. Comparing compounds 767 and 761, the
presence of a methylene carbon around the carbonyl showed a very different effect on the
inhibition activity. The compound with a methylene carbon in the left of the carbonyl (767)
showed a 20-fold better inhibition than that in the right (761). While the rank-order potency
of this inhibitor series was equivalent with mouse and human sEH, a 3-5-fold higher
inhibition potency was observed for the murine enzyme.
Example 37
[0250] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention having no secondary pharmacophore, but having a
tertiary pharmacophore that is an amide or a carboxylate ester functional group (with alkyl,
alkenyl, alkynyl, cycloalkyl and arylalkyl ester groups).
[0251] Compound 687, having a carboxylic acid group at the end of twelve carbon chain,
was found to be an excellent inhibitor of both the mouse and human enzymes. Additionally,
an ester found to be a suitable secondary pharmacophore. As a result, a variety of ester
derivatives having a carbonyl group located eleven carbon units from the urea
pharmacophore were synthesized and evaluated to examine contributions of a tertiary
pharmacophore.

WO 2006/045119 PCT/US2005/038282
80
[0252] Assays were conducted with the compounds indicated in Table 4, according to
established protocols (see, above).
Table 4: Inhibition of mouse and human sEH by l-(l-adamantyl)-3-(l 1-alkylated undecyl)-
ureasa


WO 2006/045119 PCT/US2005/038282
81
a Enzymes (0.12 μM mouse sEH and 0.24 μM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40μM). Results are means ± SD of three
separate experiments.
[0253] While the presence of a polar group at the end of a shorter chain reduced inhibition
potency for both enzymes (see Table 1), when the carboxylic acid was modified to esters with
various aliphatic groups (780,784,783,781,788,800,785,801,802, and 803) inhibition
potencies were as high as that of the acid (687) for both enzymes. Ethyl (792) and isopropyl
(793) amide derivatives were also potent inhibitors. Compounds with methyl-branched
aliphatic chains were also potent (785, 801,802, 803, and 793). Still further, larger bulky
group such as 1-adamantylmethyl (786), benzyl (804), 2-chlorobenzyl (782) or 2-
naphthylmethyl (787) provided good levels of activity, although slightly reduced (1.5-3-fold)
for both enzymes. These results identified an additional site within the sEH inhibitor
structure which allows the inclusion of a third polar function, i.e. a tertiary pharmacophore.
Example 38
[0254] In order to further explore the effects of functional groups which improve water
solubility on the bioavailability and potency of the inhibitors, seven amide derivatives of
compound 687 with various functionalities were synthesized. As shown in Table 5, alkyl,
sulfonyl, lipoamino acid, and glucosylamide derivatives were prepared, and their inhibition
potency on sEHs, melting point, and solubility in water and oil were examined. For mouse
sEH, there was no change in the inhibition activity when the acid function of compound 687
was substituted by an alkyl (792 and 793), sulfonyl (848 and 914), lipoamino acid (1001), or
glucosyl (1002 and 798) groups. Interestingly, for human sEH, significant differences in
inhibition potencies, which were measured by the spectrometric and fluorescent based assays,
were observed when replacing the acid of compound 687 by the substituents. While
introduction of the ethyl amide group (792) resulted in a 1.5-fold decrease in potency, the
isopropyl analogue (793) exhibited very similar potency to ADUA (687). The corresponding
ester derivatives 784 and 785 in Table 5 exhibited a 2-3-fold improvement in inhibition of the
human enzyme. Two sulfonylamides with a methyl (848) or phenyl (914) group improved the
inhibition potency of compound 4 about 1.5-fold. On the other hand, a 25-fold and 3-fold
decrease in potency was observed when the sulfonylamide was replaced by a lipoamino acid
(1001) or glucosyl groups (1002 and 798), respectively. This suggests that such bulky, and in
the case of glucose, highly polar functional groups are not effective in retaining the inhibition

WO 2006/045119 PCT/US2005/038282
82
potency on human sEH. Comparing compounds 1002 and 798, similar inhibition on the
human enzyme was shown in these two compounds although an octyl group is present
between the ADUA and sugar moieties of compound 1002, suggesting that relatively
lipophilic alkyl groups located around the amide function do not alter the binding activity of
inhibitors to the enzyme. Melting points of most of the amides in Table 5 were measured in a
range of 100-140°C, which was similar to 114°C of the acid compound 687, while the ester
derivates of compound 687 in Table 5 showed 23-66°C lower melting points than that of
compound 687. An exception was compound 1001 for which we were unable to obtain a
crystalline solid at room temperature. These results imply that the amide function is not as
useful in reducing the melting point of AUDA derives as the esters. As with the esters
prepared for this study, the amides failed to result in a large increase in potency on the target
enzyme. Thus any advantage of these amides would result from improvements in ease of
formulation, oral availability and pharmacokinetics. When water solubility of three
compounds (848,1001, and 1002) was compared to that of compound 4, a 3-fold lower or
similar solubility was shown in compounds with a rnethanesulfonyl (848) or glucosyl (1002)
group, respectively. Compound 1001, the lipoamino acid conjugate, surprisingly had a 2.5-
fold better water solubility than compound 687. The 25-fold decrease in the inhibitory
potency of compound 1001 compared to 687 indicates that with compound 1001 the
biological activity will be attributed largely to the production of AUDA rather than being due
to both AUDA and its ester or amide. In addition, no valuable improvement in oil solubility
was obtained in the amides. Although significant improvements in the properties of the
compounds were not observed in the seven amide derivatives, their relative stability and
inhibition potency is sufficient to encourage the further exploration of other amide
compounds to develop bioavailable inhibitors with improved physical properties. We have
reported that modification of the urea pharmacophore of potent sEH inhibitors to an amide
functionality does not dramatically alter the inhibition potency and that at least a 10-fold
improvement hi water solubility and a decrease in the melting points of these amide inhibitors
is observed.( Kim, et al. J. Med. Chem. 2005, 48, 3621-3629). This suggests that the
inhibition potency and physical properties of the amide derivatives in Table 5 might be
improved with the modification of the urea function to the corresponding amide
pharmacophore.

WO 2006/045119 PCT/US2005/038282
83
Table 5. Inhibition of mouse and human sEH by 12-(3-adamantan-l-yl-ureido)dodecanoic
acid alkyl or sulfonylamide derivatives

a Spectrometric-based assay: enzymes (0.12 μM mouse sEH or 0.24 uM human sEH) were
incubated with inhibitors for 5 min in sodium phosphate buffer (200 uL; pH 7.4) at 30°C
before substrate introduction ([S] = 40 uM), results are means ± SD of three separate
experiments.
b Fluorescent-based assay: enzymes (0.88 nM mouse sEH or 0.96 nM human sEH) were
incubated with inhibitors for 5 min in Bis-Tris/HCl buffer (25mM; pH 7.0) at 30°C before
substrate introduction ([S] = 5 uM), results are means ± SD of three separate experiments
c Melting point

WO 2006/045119 PCT/US2005/038282
84
Example 39
[0255] This example provides assays and illustrates the inhibition of mouse and human
soluble epoxide hydrolases by compounds of the invention having a both a secondary and
tertiary pharmacophore that is a carboxylic ester functional group.
[0256] Assays were conducted with the compounds indicated in Table 6, according to
established protocols (see, above).
Table 6. Inhibition of mouse and human sEH by 4-(3-adamantan-l-yl-ureido)butyryloxy
comoounds

No. n TAa Mouse sEHb Human sEHb MP(°C) cLogP0
lC50 (μM) IC90 (μM) IC50 (μM) IC90 (μM)
857 1 8 0.05±0.01 0.11±0.01 0.39±0.01 9±2 123 0.98±0.47
876 2 9 0.05±0.01 0.63±0.02 0.54±0.05 9±2 95-97 1.27±0.47
858 3 10 0.05±0.01 0.16±0.01 0.12±0.01 5.0±0.1 89-91 1.55±0.47
877 4 11 0.05±0.01 0.10±0.01 0.13±0.01 1.5±0.1 84-86 1.97±0.47
878 6 13 0.05±0.01 0.13±0.01 0.12+0.01 0.81±0.01 65-67 2.81±0.47
879 7 14 0.05±0.01 0.16±0.02 0.11±0.01 0.72±0.01 58-59 3.22± .47
880 9 16 0.05±0.01 0.26±0.03 0.10±0.01 0.68±0.01 60-61 4.06±0.47
881 10 17 0.05±0.01 0.35±0.05 0.10±0.01 1.2±0.1 54-55 4.48±0.47
882 11 18 0.05±0.01 0.63+0.04 0.10±0.01 1.8±0.2 64-65 4.89±0.47
a The total number of atoms extending from the carbonyl group of the primary urea pharmacophore, TA = n + 7
b Enzymes (0.12 μM mouse sEH and 0.24 uM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40 μM). Results are means ± SD of three
separate experiments.
c cLog P: calculated log P by Crippen's method by using CS ChemDraw 6.0 version

WO 2006/045119 PCT/US2005/038282
85
[0257] As can be seen from the above table, in increasing the distance between the
secondary ester phannacphore and the tertiary ester pharmacaphore (549, 635, and 774)
increased inhibition potency for human sEHs but mouse EH activity remained relatively
consistent.
Example 40
[0258] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having a secondary ether
pharmacophore.
[0259] Adamantyl-urea compounds were prepared having a polar ether group located
various distances from the carbonyl of the primary urea pharmacophore. These compounds
were prepared to improve water solubility of lipophilic sEH inhibitors (192 and 686). As can
be seen from the results in Table 7, the activity is relatively consistent.
[0260] Assays were conducted with the compounds indicated in Table 7, according to
established protocols (see, above).
Table 7. Inhibition of mouse and human sEH by alkyl ether derivatives


WO 2006/045119 PCT/US2005/038282
86

[0261] As shown in the above table, these compounds fiinctionalized with a single ether
group could be as active and potent as non-functionalized lipophilic inhibitors (790, see Table
2 above) for both murine and human enzymes. Adding a polar ether group to these
compounds increased their water solubility (compare compound 866-870 with 790). The
distance from the carbonyl of the primary urea pharmacophore to the secondary ether
. pharmacophore in compound 869 is about 8.9 A showing that the secondary pharmacophore
may be located about 7 A to about 9 A from the carbonyl of the primary urea pharmacophore
group.
Example 41
[0262] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having a secondary ether or
polyether pharmacophore, with comparison to compounds further including a tertiary
pharmacophore.
[0263] Because compounds having a ether secondary pharmacophore were found to be
suitable inhibitors of both the mouse and human enzymes, a variety of polyether derivatives
were synthesized and evaluated along with contributions of a tertiary pharmacophore. As can
be seen from the results in Table 8, the activity is relatively consistent.
[0264] Assays were conducted with the compounds indicated in Table 8, according to
established protocols (see, above).

WO 2006/045119 PCT/US2005/038282
87
Table 8. Inhibition of mouse and human sEH by substituted ether derivatives


WO 2006/045119 PCT7US2005/038282
88

[0265] Compounds with from two to four ether groups (908, 950, and 952) had inhibition
potencies that were as high as non-functionalized lipophilic inhibitors (790, see Table 2
above) for both murine and human enzymes, as well as increased water solubility and
improved pharmacokinetics (See Figures 14-24). Including a tertiary pharmacophore were
also potent inhibitors but did not further increase their activity (compare compounds 913 and
940 with 908 and compound 951 with 950).
Example 42
[0266] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having a primary amide
pharmacophore.
[0267] Adamantyl-amide compounds were prepared having a polar secondary
pharmacophore group located various distances from the carbonyl of the primary amide
pharmacophore.
[0268] Assays were conducted with the compounds indicated in Table 9, according to
established protocols (see, above).

WO 2006/045119 PCT7US2005/038282
89
Table 9. Inhibition of mouse and human sEH by adamantyl-amide derivatives

[0269] As shown in the above table, these compounds functionalized with a amide group
could be as active and potent as urea inhibitors for both murine and human enzymes. The
nitrogen to the right of the amide carbonyl group is important for activity.

WO 2006/045119 PCTYUS2005/038282
90
Example 43
[0270] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having an arylene or cycloalkylene
linker.
[0271] Because compounds having an alkylene linker between the primary and secondary
pharmacophore were found to be excellent inhibitors of both the mouse and human enzymes,
a variety of admantyl-urea derivatives having a phenyl or cyclohexyl spacer between a
primary urea and secondary pharmacophore were synthesized and evaluated to examine the
contributions of the linker.
[0272] Assays were conducted with the compounds indicated in Table 10, according to
established protocols (see, above).
Table 10. Inhibition of mouse and human sEH by substituted phenyl and cyclohexyl
derivatives


WO 2006/045119 PCT7US2005/038282
91


WO 2006/045119 PCT/US2005/038282
92

[0273] Compounds with alkylene and arylene linker groups (859 and 861) had inhibition
potencies that were higher than compounds with alkylene linkers (789, see Table 2 above,
and 868, see Table 7 above) for both murine and human enzymes, independent of the
topography {compare compound 859 with 860 and compound 861 with 863) or type of the
secondary phannacophore {compare compounds 860 and 863 with 909).
Example 44
[0274] This example illustrates the inhibition of mouse soluble epoxide hydrolases by
compounds of the invention (formula (T)) having a secondary phannacophore, and further
including a mono amino acid moiety. This example further illustrates the use of a
combinatorial approach toward compound preparation and evaluation.
[0275] The utility of a combinatorial approach is illustrated by using the butanoic acid
derivatives from Table 11 and Table 12 to form amide bonds with one or more natural or
synthetic amino acids. This approach rapidly leads to a large number of compounds that are
highly active and can be recognized by the intestinal peptide uptake system. As shown
above, polar groups could be incorporated into one of the alkyl groups of the dialkyl-urea
sEH inhibitors without loss of activity, when placed at an appropriate distance from the urea
function. These modifications give the new inhibitors better solubility and availability. To
expand this assessment of inhibitor structure refinement a semi-combinatorial approach was
used with amino acids. Because amino acids are simple bifunctional synthons with a wide
variety of side chains, mono and di-peptidic derivatives of 4-(3-cyclohexyl-ureido)-butyric
acid 625 were synthesized. This parent compound (acid 625) was selected due to its low
inhibition of sEH. Furthermore, to make the peptidic bond, reactants were used, such as 1-
ethyl-3-(3-(dimethylamino)-propyl) carbodiimide, that themselves or their reaction product,
such as l-efhyl-3-(3-dimethylamino)-propyl urea, are not inhibitors of sEH. Therefore, any
inhibition observed was derived from the targeted peptidic derivatives. This approach allows
the preparation of compounds on an analytical scale (10 μmol) without purification of the

WO 2006/045119 PCT/US2005/038282
93
products. The presence of the desired products was confirmed by LC-MS and the ratio of the
LC-MS peak of the desire compounds with the starting material was used to estimate the
reaction yield. Because each inhibitor presents a single carboxyl group for negative mode
ionization, the estimation of yield is reasonably quantitative.
[0276] Syntheses of amino acid derivatives of 4-(3-cyclohexyl-ureido)-butyric acid (632)
were performed at analytical scale. Reactions were performed in 2mL glass vials for each
amino acid. To 100 μL of a solution of 632 in DMF at 100 mM (10 μmol), 200 μL of a
solution of l-ethyl-3-(3-(dimethylamino)-propyl) carbodiimide in DMF at 100 mM (20
μmol) was added. After 15 minutes reaction at room temperature, 400 μL of amino acid
methyl ester solution at 100 mM (40 umol) in 90:10 DMF:1 N NaOH was added. The
reaction was strongly mixed at 40°C overnight. Three hundred microliters of 1 N NaOH was
then added and allowed to react overnight at 40°C. Product formation was confirmed for
each amino acid using electrospray-ionization mass spectrometry (ESI-MS). Reaction
solutions were used directly for inhibitor potency measurement with a theoretical
concentration of 10 mM.
[0277] Assays were conducted with the compounds indicated in Table 11, according to
established protocols (see, above).
Table 11: Inhibition of mouse sEH by mono-amino acid derivatives of 4-(3-cyclohexyl-
ureido)- butyric acid (632).


WO 2006/045119 PCT/US2005/038282
94
Glycine 285.2 286.6 >50
Histidine 365.2 366.6 1.9 ±0.2
Isoleucine 341.2 342.7 18±3
Leucine 341.2 342.7 >50
Lysine 356.3 357.7 2.2 ±0.5
Methionine 359.2 360.7 >50
Phenylalanine 375.2 376.7 5.6 ± 0.4
Proline 325.2 326.7 >50
Serine 315.2 316.7 >50
Threonine 329.2 330.7 >50
Tryptophane 414.2 415.8 1.6 ±0.2
Tyrosine 391.2 392.8 0.59 ± 0.03
Valine 327.2 328.7 >50
Results are means ± SD of three separate experiments.
[0278] Significant improvement of the inhibition potency was observed for the aromatic
derivatives (phenylalanine, tryptophane and tyrosine), histidine and lysine. Again, without
intending to be bound by theory, it is believed that the specificity of the interaction of the
enzyme with the five peptidic inhibitors listed results from specific pi-pi stacking between
tryptophane 334 (Trp334) located in close proximity to the secondary pharmacophore, and the
aromatic moieties with four of the five amino acids above. This interaction should alter the
fluorescence spectrum of the enzyme. For the lysine derivative, because reaction can occur
with the side chain amino group, the resulting product could resemble the alkyl derivatives
synthesized above with the acid function playing the role of the third pharmacophore.
Example 45
[0279] This example illustrates the inhibition of mouse soluble epoxide hydrolases by
compounds of the invention (formula (I)) having a secondary pharmacophore, and further
including a dipeptide moiety.

WO 2006/045119 PCT/US2005/038282
95
[0280] Compounds in the amino acid derivative series, 625-Tyr, showed an inhibition
potency in the hundreds of nanomolar range, prompting the evaluation of the effect of adding
a second amino acid.
[0281] In a manner similar to that described above, syntheses of amino acid derivatives of
2-[4-(3-Cyclohexyl-ureido)-butyrylamino}-3- (4-hydroxy-phenyl)-propionic acid (632-Tyr)
that are examples of dipetide derivatives of 632 were done on an analytical scale. Synthesis
was performed as described above for the derivatives of 632, simply substituting this
compound by 632-Tyr. Product formation was confirmed by ESI-MS.
[0282] Assays were conducted with the compounds indicated in Table 12, according to
established protocols (see, above).
Table 12: Inhibition of mouse sEH by mono-amino acid derivatives of 4-(3-cyclohexyl-
ureido)- butyryl-tyrosine.


WO 2006/045119 PCT/US2005/038282
96
Methionine 522.8 521.2 2 0.05 2.0
Phenylalanine 538.7 537.5 1 0.05 1.6
Proline 488.6 487.4 1 0.06 6.3
Serine 478.6 477.3 1 0.07 3.3
Threonine 492.6 491.3 4 0.12 12.5
Tryptophane 577.7 576.4 1 0.05 1.0
Tyrosine 554.7 553.4 5 0.05 2.5
Valine 490.6 489.4 2 0.05 3.1
Results are means ± SD of three separate experiments.
[0283] Significant improvement of inhibition potency was observed for almost all the
derivatives tested except for alanine, isoleucine, leucine and threonine. These results indicate
that the enzyme has a narrower specificity close to the catalytic center than toward the end of
the active site tunnel. The inhibition potency found for the best dipeptidic derivatives are
similar to those found for the corresponding alkyl inhibitors (see, C. Morisseau, et al.,
Biochem. Pharm. 63:1599-1608 (2002)), indicating that such peptide-mimics are excellent
inhibitors of sEH. Because of the presence of the amino acid derivatives in their structure,
these compounds have excellent water solubility. Furthermore, because of the presence of
active small peptide transport system in the gut, the dipeptidic urea derivatives will be
absorbed in the gut by such systems as observed for several peptide derivative drugs (see, E.
Walter, et al., Pharm. Res. 12: 360-365 (1995) and K. Watanabe, et al., Biol. Pharm. Bull 25:
1345-1350 (2002)), giving these compounds excellent bioavailability.
Example 46
[0284] This example provides studies directed to the metabolic stability of certain
inhibitors of sEH.
[0285] To evaluate the metabolic stability of these inhibitors, the microsomal and NADPH
dependent metabolism of a number of potent sEH inhibitors was evaluated. The rates of
metabolism among the compounds varied dramatically, however the appearance of an
omega-terminal acid was observed for all inhibitors containing n-alkane substitutions. When

WO 2006/045119 PCT/US2005/038282
97
tested, the potent alkyl derivatives (e.g. 686) are rapidly metabolized in microsomal
preparations by P450 dependents processes (see Figure 6), while the omega acid analogs (e.g.
687) were stable (see Figure 7). The first step in the metabolic transformation of the n-alkyl
to n-alkanoic acid derivatives is an NAPDH dependent process carried out by cytochrome
P450 dependent omega hydroxylation in rodent and human hepatic tissue preparations (see
Figure 8). The metabolites identified along this metabolic route are provided in Table 13.
When in vivo metabolism was evaluated, evidence for the beta-oxidation of the alkanoic acid
derivatives was also found (see Figure 9). Together, these data indicate that P450 omega
hydroxylation can result in the rapid in vivo metabolic inactivation and excretion of these
inhibitors.
Table 13: Structure of metabolites formed from compound 686.

Example 47
[0286] This example provides the structures of compounds of the invention designed to
slow esterase dependent inactivation, block beta-oxidation, block cytochrome P450
dependent omega hydroxylation, or inhibit cytochrome P450 omega hydrolase.
[0287] Beta-oxidation can be blocked in a variety of ways, for example with an alpha
halogen or alpha branched alkyl group (806), cyclopropane (807) or aromatic groups (808),
or by replacing the acid or ester functional groups with alternate functionalities, such as
sulfonamides (809 and 810), which mimic ester and acid functional groups yet provide
metabolic stability in vivo. Similarly in pharmacology heterocyclic groups are used for
hydrogen bond donors and acceptors to mimic carboxylic acids and esters (811). In addition,
P450 omega hydroxylation can be blocked by including acetylene (812), trifluoromethyl
(813), or aryl (814) groups at the terminus of the alkyl chain. This series of inhibitors also

WO 2006/045119 PCT/US2005/038282
98
illustrates that with both the secondary and tertiary phannacophore, replacement can be made
for the carbonyl with other functionalities as hydrogen bond donors and acceptors.
Table 14: Structures of sEH inhibitors designed to prevent beta-oxidation and P450 omega
5 hydroxylation.

Example 48
[0288] This example illustrates a comparison of cyclohexyl and adamantyl groups in
stability and solubility.
[0289] Another consistent observation during the metabolism studies was that the
adamantyl substituent (both 192 and 686 substituted) provided compounds having improved
stability (see Figure 6). Surprisingly the adamantyl compounds were approximately 2x more
soluble than the corresponding cyclohexyl derivatives (772 vs. 789, 791 vs. 790, and 297 vs.
686 see Table 2 for structures). Surprisingly, the LC-MS/MS analyses producing collision
induced dissociation of compounds containing the adamantyl substituent provided extremely

WO 2006/045119 PCT/US2005/038282
99
high abundance ions, which dramatically enhanced the analytical sensitivity for these
inhibitors (see Table 15 below). This enhanced sensitivity is a distinct advantage for drug
metabolism studies using either in vivo or in vitro systems. Moreover, adamantane represents
the smallest diamond nucleus and the adamantyl substituents not only yield compounds of
improved metabolic stability and pharmacokinetic parameters, but also compounds that are
very easy to detect.
Table 15: Calibration curves and detections limit (DL) of inhibitors analyzed by HPLC-
MS/MS.

Example 49
[0290] This example provides the pharmacokinetic studies carried out using compounds of
the present invention.
[0291] The pharmacokinetic properties of some of the most potent sEH inhibitors was
evaluated following oral gavage in mice. As noted above, the use of 1-adamantyl urea
inhibitors afforded exquisite sensitivity, allowing the determination of the determined
pharmacokinetic parameters from serial blood samples collected from individual mice (see
Table 17).

WO 2006/045119 PCT/US2005/038282
100
[0292] Animals. Male Swiss Webster mice, 6 weeks-old, were obtained from Charles
River (CA, USA). After 1-2 week acclimation period, healthy animals were assigned to
study groups based on body-weight stratified randomization procedure. The body weight of
animals used in all the experiments ranged from 28 g to 38 g. Mice were maintained on a 12
h light /12 h dark cycle under controlled temperature and humidity conditions, and food and
water available ad libid ran.
[0293] Administration and measurement. Pharmacokinetic studies in mice used a 5
mg/kg dose of sEH inhibitors dissolved in corn oil and 4% DMSO administered orally.
Serial tail bled blood samples (5-10 aL) were collected in heparinized 1.5 mL tubes at
various time points (0.5,1, 2, 3, 4, 5, 6, and 24 hr) after the administration for measuring
parent compounds and their metabolites by using LC-MS/MS: a Waters 2790 liquid
chromatograph equipped with a 30 X 2.1 mm 3 um C18 Xterra™ column (Waters) and a
Micromass Quattro Ultima triple quadrupole tandem mass spectrometer (Micromass,
Manchester, UK). To the collected samples were added l00μL of distilled water, 25 uL of
internal standard (500 ng/mL; l-cyclohexyl-3-tetradecylurea, CTU), and 500uL of ethyl
acetate. Then the samples were centrifuged at 6000 rpm for 5 min, and the ethyl acetate layer
was dried under nitrogen. The residue was reconstituted in 25 μL of methanol, and aliquots
(5μL) were injected onto the LC-MS/MS system.
[0294] Pharmacokinetic studies using a human subject employed doses of 0.1-1.0 mg/kg of
sEH inhibitors (800) or a 0.3 mg/kg dose of 687 dissolved in olive oil administered orally.
Serial bled blood samples (3-50 uL) were collected from finger tips into 50 uL heparinized
capillary tube at various time points (0.5,1, 2,4,6,12 and 24 hr) after administration. These
samples were used to measure parent compounds and their metabolites using LC-MS/MS as
described above for experiments with mice. Blood samples were added 400 μL of distilled
water and 25 μL of internal standard (500 ng/mL CTU), and vortexed. The blood samples
were then extracted with 500 μL of ethyl acetate twice and the ethyl acetate layer was dried
under nitrogen. The residue was reconstituted in 25 μL of methanol, and aliquots (10 μL)
were injected onto the LC-MS/MS system as described above. Biological end points came
from clinical chemistry samples run at The University of California Davis Clinical
Laboratory and a series of 6 inflammatory markers including C reactive protein were run
blind at the University of California Davis Department of Nephrology.

WO 2006/045119 PCT/US2005/038282
101
[0295] Analysis. Pharmacokinetics analysis was performed using SigmaPlot software
system (SPSS science, Chicago, IL). A one-compartment model was used for blood
concentration-time profiles for the oral gavage dosing and fits to the following equation (see,
Gibson, G.G. and Skett, P.: INTRODUCTION TO DRUG METABOLISM, SECOND ED.,
Chapman and Hall, New York 1994,199-210):
C = ae-bt
The half-life (t1/2) for the elimination phase was calculated by the following equation:
t1/2 = 0.693/b
The area under the concentration (AUC) was calculated by the following equation:
AUC = alb
Where:
- C = the total blood concentration at time t
- a = the extrapolated zero intercept
- b = the apparent first-order elimination rate constant
Table 17: Pharmacokinetic parameters of l-(l-adamantyl)-3-(l 1 -alkylated undecyl)ureasa


WO 2006/045119 PCT/US2005/038282
102

5 mg/kg dosing of compounds were administered orally to male Swill Webster mice, maximum
concentration,c time of maximum concentration,d area under concentration,c half-life.
[0296] The ester compounds were generally hydrolyzed to the acid compound (687) when
administered orally. An example of the time course of free acid appearance is shown in
Figure 10. When compound 687 was administered orally, it reached the maximum
concentration (2-fold higher than 686) in 30 min, while compound 686 reached its maximum
concentration in 2 h. Furthermore, the area under the curve (AUC) for 687 was 2-fold higher,
indicating an improvement in oral bioavailability. The maximum concentrations of primary
esters (780, 784, 783,781, 788, 800, 803 and 804) esters were 1.5-5-fold higher than 687, and
the AUC increased 1.2-2.3-fold for the ester compounds indicating higher bioavailabilities.
On the other hand, secondary esters (785 and 802) showed similar maximum concentrations
and bioavailabilities to those of 687 in mice, while the tertiary ester (801) displayed a 4-8-
fold decrease in maximum concentration and bioavailability. Accordingly, the alkylation of a
potent acid inhibitor (687) to form primary esters improves the oral availability of these
inhibitors.
Example 50
[0297] This example provides a table of structures for compounds of the invention having
all three pharmacophores present.

WO 2006/045119 PCT/US2005/038282
103
Tables 18a and b: Structures and inhibition of mouse and human sEH by other sEH
inhibitors containing the primary, secondary, and tertiary pharmacophores.
Table 18a


WO 2006/045119 PCT7US2005/038282
104


WO 2006/045119 PCTVUS2005/038282
105


WO 2006/045119 PCTYUS2005/038282
106


WO 2006/045119 PCT/US2005/038282
107


WO 2006/045119 PCT/US2005/038282
108


WO 2006/045119 PCT/US2005/038282
109


WO 2006/045119 PCT/US2005/038282
110


WO 2006/045119 PCT/US2005/038282
111


WO 2006/045119 PCT/US2005/038282
112
Table 18b._____________________________________________________________


WO 2006/045119 PCT/US2005/038282
113


WO 2006/045119 PCT7US2005/038282
114


WO 2006/045119 PCT/US2005/038282
115


WO 2006/045119 PCT/US2005/038282
116


WO 2006/045119 PCT/US2005/038282
117


WO 2006/045119 PCT/US2005/038282
118


WO 2006/045119 PCT/US2005/038282
119


WO 2006/045119 PCT/US2005/038282
120


WO 2006/045119 PCT/US2005/038282
121


WO 2006/045119 PCT/US2005/038282
122


WO 2006/045119 PCT/US2005/038282
123


WO 2006/045119 PCT/US2005/038282
124


WO 2006/045119 PCT/US2005/038282
125


WO 2006/045119 PCT/US2005/038282
126


WO 2006/045119 PCTYUS2005/038282
127


WO 2006/045119 PCT/US2005/038282
128


WO 2006/045119 PCT/US2005/038282
129


WO 2006/045119 PCT/US2005/038282
130


WO 2006/045119 PCT7US2005/038282
131


WO 2006/045119 PCT/US2005/038282
132


WO 2006/045119 PCT/US2005/038282
133


WO 2006/045119 PCT/US2005/038282
134


WO 2006/045119 PCT/US2005/038282
135


WO 2006/045119 PCT/US2005/038282
136


WO 2006/045119 PCT/US2005/038282
137


WO 2006/045119 PCT/US2005/038282
138


WO 2006/045119 PCT/US2005/038282
139


WO 2006/045119 PCT/US2005/038282
140


WO 2006/045119 PCT/US2005/038282
141


WO 2006/045119 PCT/US2005/038282
142


WO 2006/045119 PCT/US2005/038282
143


WO 2006/045119 PCT/US2005/038282
144


WO 2006/045119 PCT/US2005/038282
145


WO 2006/045119 PCT/US2005/038282
146


WO 2006/045119 PCT/US2005/038282
147


WO 2006/045119 PCT/US2005/038282
148


WO 2006/045119 PCT/US2005/038282
149


WO 2006/045119 PCT/US2005/038282
150

* Inhibition potencies were determined using a fluorescent based high-throughput assay. Inhibitors in solution at
10 mM in DMSO were serially diluted by 10-fold increment in Bis/Tris HC1 buffer (25 mM PH 7.0) containing
0.1 mg/mL of BSA (Buffer A). In black 96-well plates, 20pL of the inhibitor dilution or buffer were delivered in
every well, and then 130μL of Human sEH at ~0.4 μg/mL in solution in Buffer A were added to each well The
plate was then mixed and incubated at room temperature for 5 minutes. Fifty microliters of substrate ((3-Phenyl-
oxiranyl)-acetic acid cyano-(6-methoxy-naphthalen-2-yl)-methyl ester; PHOME) at 200μM in solution in 96:4
Buffer A:DMSO was then added to each well to give [S]final = 50μM and [E]final= ~4nM. The plate was then

WO 2006/045119 PCT/US2005/038282
151
mixed and incubated in the dark at room temperature (~25°C) for 90 min. Activity was measured by determining
the relative quantity of 6-methoxy-2-naphthaldehyde formed with an excitation wavelength of 316 nm and an
emission wavelength of 460 mn measured with a SpectraMax M-2 fluorometer (molecular Devices, Sunnyvale
CA). Results are not reported.
[0298] The primary urea pharmacophore can be varied (compound #) with amide or
carbamate functionality to improve physical properties of sEH inhibitors as well. The
carbonyls can be replaced by heterocyclic or acyclic hydrogen bond acceptors and donators
as shown in Table 14.
Example 51
[0299] This example shows the effect of sEH inhibitors on serum and urinary oxylipin
profiles in rodents.
[0300] The described soluble epoxide inhibitors have been shown to modulate the relative
abundance and amounts of epoxy and dihydroxy fatty acids formed in treated animals. One
such example of this alteration is provided in Figure 13. In this example, hypertension was
induced in one group of Sprague-Dawley rats by the infusion of angiotensin II (ANGII). A
second group of rats received both ANGII and a subcutaneous injection of the model sEH
inhibitor l-adamantyl-3-(dodecanoic acid) urea (i.e. compound 687). Urine samples were
collected for 24hr post exposure to compound 687 and analyzed for linoleate (Panel A) and
arachidonate (Panel B) derived epoxides and diols using LC/MS/MS. As shown in Figure 13,
ANGII exposure decreased the concentration of both linoleate (EpOMEs) and arachidonate
(EETs) derived epoxides and increased arachidonate derived diols (DHETs) but not linoleate
derived diols (DHOMEs). hi the case of both lipid classes, treating animals with compound
687 resulted in an increase in urinary epoxides, as well as a decrease in diol concentrations.
Example 52
[0301] This example illustrates the effect of certain compounds of the invention on
members of the arachidonic acid cascade.
[0302] For epoxy fatty acid hydrolysis, the soluble epoxide hydrolase prefers substrates
with epoxide moieties that are more distant from the carboxyl terminal. Specifically the
substrate preference decreases in the order of 14,15-EET > 11,12-EET > 8,9-EET >>> 5,6-
EET for the epoxides of arachidonic acid. Independently, the relative substrate turnover of

WO 2006/045119 PCT7US2005/038282
152
the epoxy arachidonates were calculated at 0.1:8.1:14.3 when a 1:1:2 mixture of 8,9-, 11,12-,
and 14,15-EET fatty acid was hydrolyzed to 30% by rat renal cortex cytosol. By considering
the primary pharmacophore of the urea to be a transition-state analog of epoxide hydrolysis,
inhibitors have now been developed which incorporate long aliphatic acids. These
compounds are better substrate and transition state mimics than those incorporating shorter
aliphatic acids. Accordingly, optimal soluble epoxide hydrolase inhibitors can be obtained by
producing compounds with aliphatic acid substituents (i.e. a tertiary pharmacophore) which
are separated from the primary pharmacophore by an equivalent distance as the terminal acid
is separated from the epoxide in optimal substrates. Within the enzyme active site, epoxy
fatty acids have been predicted to exist in an extended or pseudo-linear confirmation.
Therefore, both the epoxy fatty acids and the aliphatic acid containing urea structures were
approximated as two dimensional linear representations and measurements were made on
each species. The critical measurements taken were distances (in angstroms) from the
carboxylate hydroxyl to the urea carbonyl and the urea nitrogens.
[0303] The distance of the carboxylate to the urea function of l-cyclohexyl-3-octanoic acid
is similar to the distance of the epoxide to the carboxylate in 8,9-EET. Therefore, the
calculated inhibitor potencies were normalized to this compound, resulting in a ranked
inhibitor potency. We then correlated epoxide to carbonyl distance with respect to relative
substrate turnover rate to establish a correlative regression. By plotting the relative inhibitor
potency on this graph we find that the distances of the carboxyl to the N' -nitrogen correlate
best with the carboxyl to epoxide oxygen distance. These data further highlight the similarity
between inhibitor and substrate interaction with the soluble epoxide hydrolase.
Programs:
[0304] All structures were drawn and exported as MDL MOL files using
ACD/ChemSketch v 4.55 (5/06/2000) Advanced Chemistry Development Inc., Toronto,
Ontario, Canada). Distance measurements were made on the corresponding MOL file image
using ACD/3D v 4.52 (4/10/2000). Structural optimizations were not used.
Table 19 provides results for this analysis (see also, Figure 12).
Table 19: Linear distances between the primary and secondary pharmacophores of a series of sEH
inhibitors and their rank order potencies with the mouse (MsEH) and human sEHs (HsEH) are
shown in comparison with the epoxide to free acid distances and relative turnover rate of the four
arachidonic acid epoxides with the rat sEH.

WO 2006/045119 PCT/US2005/038282
153


WO 2006/045119 PCT/US2005/038282
154
WHAT IS CLAIMED IS:
1. A compound having a formula:

and their pharmaceutically acceptable salts, wherein
R1 is a member selected from the group consisting of substituted or unsubstituted
alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted
cycloalkylalkyl, substituted or unsubstituted cycloalkylheteroalkyl, substituted
or unsubstituted arylalkyl, substituted or unsubstituted arylheteroalkyl,
substituted or unsubstituted C5-C12 cycloalkyl, substituted or unsubstituted
aryl, substituted or unsubstituted heteroaryl and combinations thereof, wherein
said cycloalkyl portions are monocyclic or polycyclic;
P1 is a primary pharmacophore selected from the group consisting of -C(O)O-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-
,-NHC(O)NH-,--OC(O)NH-, -NHC(O)O-, -NHC(S)NH-, -CH2C(O)NH-,
-NHC(O)CH2-, -C(O)NH-, -NHC(O)-, and

P2 is a secondary pharmacophore selected from the group consisting of -NH-, -
-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -NHC(NH)NH-, -
NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)- and -NHC(S)NH-;
P3 is a tertiary pharmacophore selected from the group consisting of C2-C6 alkenyl,
C2-C6 alkynyl,C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl,
-O(CH2CH2O)q-R2, OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-
C4alkyl-C(O)OR2, -C(O)R2, -C(0)OR2 and carboxylic acid analogs, wherein
R2 is a member selected from the group consisting of hydrogen, substituted or
unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl,
substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl;

WO 2006/045119 PCT/US2005/038282
155
the subscripts n and m are each independently 0 or 1, and at least one of n or m is 1,
and the subscript q is 0 to 6;
L1 is a first linker selected from the group consisting of substituted or unsubstituted
C1-C6 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, substituted
or unsubstituted arylene and substituted or unsubstituted heteroarylene;
L2 is a second linker selected from the group consisting of substituted or unsubstituted
C1-C12 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, substituted
or unsubstituted arylene, substituted or unsubstituted heteroarylene; an amino
acid, a dipeptide and a dipeptide analog; and combinations thereof; or is H
when m is 0.
2. The compound in accordance with claim 1, wherein wherein R1 is
selected from the group consisting of substituted or unsubstituted alkyl, substituted or
unsubstituted heteroalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or
unsubstituted cycloalkylheteroalkyl, substituted or unsubstituted arylalkyl and substituted or
unsubstituted arylheteroalkyl.
3. The compound in accordance with claim 2, wherein R1 is a member
selected from the group consisting of alkyl, heteroalkyl, cycloalkylalkyl, arylalkyl and
arylheteroalkyl, each of which is optionally substituted with from 1 to 2 substitutuents
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, niiro,
haloalkoxy, thioalkyl, phenyl..
4. The compound in accordance with claim 3, wherein R1 is a member
selected from the group consisting of cycloalkyl and aryl, each of which is optionally
substituted with from 1 to 2 substituents each independently selected from the group
consisting of alkyl, halo, haloalkyl, alkoxy, nitro, haloalkoxy, thioalkyl, phenyl.
5. The compound in accordance with claim 4, wherein R1 is selected from
the group consisting of C5-C12 cycloalkyl, phenyl and naphthyl.
6. The compound in accordance with any of the preceeding claims,
wherein P1 is selected from the group consisting of -NHC(O)NH-NHC(NH)NH-, -
NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-NHC(S)NH-,-NHC(O)CH2-,
CH2C(O)NH-, NHC(O)CH2, NHC(O)- and -C(O)NH-.

WO 2006/045119 PCT/US2005/038282
156
7. The compound in accordance with claim 6, wherein P1 is selected from
the group consisting of-NHC(O)NH-, -C(O)NH- and -NHC(O) -.
8. The compound in accordance any one of the proceeding claims,
wherein P2 is selected from the group consisting of -NB-, -OC(O)O-, -O(CH2CH2O)q-, -
NHC(NH)NH-5 -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(S)NH-, -NHC(S)CH2-,
CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -NON-,

9. The compound in accordance with claim 8, wherein P2 is selected from
the group consisting of -NH-, -C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)-; and -NHC(S)NH~.
10. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of-NHC(O)NH-, -C(O)NH- and -NHC(O) -; P2 is selected from the
group consisting of-NH-,-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)-, and -NHC(S)NH-; m is 0, n is 1 and L1 is selected from the group
consisting of unsubstituted C1-C6 alkylene, substituted or unsubstituted C3-C6 cycloalkylene,

WO 2006/045119 PCT/US2005/038282
157
and substituted or unsubstituted arylene; and L2 is selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C12 alkylene, substituted or unsubstituted C3-C12
cycloalkylene, substituted or unsubstituted arylene and substituted or unsubstituted
heteroarylene.
11. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -C(O)NH- and -NHC(O) -; P2 is selected from the
group consisting of-CH(OH)-, -C(O)O-5 -O(CH2CH2O)q-! -OC(O)-, -C(O)NH- and
-NHC(O)-; n and m are each 1; L1 is selected from the group consisting of unsubstituted C1-
C6 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, and substituted or
unsubstituted arylene; L2 is selected from the group consisting of substituted or unsubstituted
C1-C12 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, and substituted or
unsubstituted arylene; and P3 is selected from the group consisting of C2-C6 alkenyl, C2-C6
alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, OR2, -C(O)NHR2, -C(O)NHS(O)2R2,
-NHS(O)2R2, -OC2-C4alkyl-C(O)OR2, -C(O)R2, -C(O)OR2 and carboxylic acid analogs,
wherein R2 is a member selected from the group consisting of hydrogen, substituted or
unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or
unsubstituted heterocyclyl; substituted or unsubstituted aryl and substituted or unsubstituted
aryl C1-C4 alkyl.
12. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; n is 0; m is 1; L1 is
selected from the group consisting of unsubstituted C1-C6 alkylene, substituted or
unsubstituted C3-C6 cycloalkylene, and substituted or unsubstituted arylene; L2 is selected
from the group consisting of substituted or unsubstituted C1-C6 alkylene, substituted or
unsubstituted C3-C12 cycloalkylene, and substituted or unsubstituted arylene and substituted
or unsubstituted heteroarylene; and P3 is selected from the group consisting of C2-C6 alkenyl,
C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, OR2, -C(O)NHR2,
-C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(O)OR2, -C(O)R2, -C(O)OR2 and
carboxylic acid analogs, wherein R2 is a member selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl.

WO 2006/045119 PCT/US2005/038282
158
13. The compound in accordance with claim 1, wherein P3 is selected from
the group consisting of C2-C6 alkenyl, heterocyclyl, OR2, -OC2-C4alkyl-C(O)OR2 and
-C(O)R2, wherein R2 is a member selected from the group consisting of hydrogen, substituted
or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or
unsubstituted heterocyclyl; substituted or unsubstituted aryl and substituted or unsubstituted
aryl C1-C4 alkyl.
14. The compound in accordance with claim 1, wherein P3 is selected from
the group consisting of haloalkoxy, morpholino, dioxothiomorpholino,.
15. The compound in accordance with claim 1, wherein L2 is selected from
the group consisting of substituted or unsubstituted C3-C6 cycloalkylene, substituted or
unsubstituted arylene and substituted or unsubstituted heteroarylene or is H when m is 0.
16. The compound in accordance with claim 15, wherein L2 is selected
from the group consisting of substituted or unsubstituted C3-C6 cycloalkylene and substituted
or unsubstituted heteroarylene or is H when m is 0.
17. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; P2 is selected from the
group consisting of -C(O)O-, -CH(OH)-, -O(CH2CH2O)q-, -OC(O)-, -C(O)NH- and
-NHC(O)-; n and m are each 1; L1 is selected from the group consisting of unsubstituted C1-
C6 alkylene, substituted or unsubstituted C3-C6cycloalkylene, and substituted or
unsubstituted arylene; L2 is selected from the group consisting of substituted or unsubstituted
C1-C6 alkylene; and P3 is selected from the group consisting of -C(O)NHR2,
-C(O)NHS(O)2R2, -NHS(O)2R2, and -C(O)OR2, wherein R2 is a member selected from the
group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl.
18. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; n is 0; m is 1; L1 is
selected from the group consisting of unsubstituted C1-C6 alkylene, substituted or
unsubstituted C3-C6cycloalkylene, and substituted or unsubstituted arylene; L2 is selected
from the group consisting of substituted or unsubstituted C1-C6 alkylene; and P3 is selected

WO 2006/045119 PCT/US2005/038282
159
from the group consisting of C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl,
heterocyclyl, OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(0)OR2,
-C(O)R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is a member selected from the
group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl.
19. The compound in accordance with claim 1, wherein R1 is a member
selected from the group consisting of C5-C12 cycloalkyl, wherein said cycloalkyl portions are
monocyclic or polycyclic; P1 is selected from the group consisting of -NHC(O)NH-; P2 is
selected from the group consisting of -O(CH2CH2O)q- and -C(O)O-; P3 is selected from the
group consisting of C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl,
heterocyclyl, OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(O)OR2,
-C(O)R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is a member selected from the
group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl; m is 1 and q is 0 to 6; L1
is selected from the group consisting of substituted or unsubstituted C1-C6 alkylene,
substituted or unsubstituted C3-C6 cycloalkylene, and substituted or unsubstituted arylene;
and L2 is selected from the group consisting of substituted or unsubstituted C1-C12 alkylene.
20. The compound in accordance with claim 1, wherein L2 is a dipeptide or
dipeptide analog.
21. The compound in accordance with claim 20, wherein L2 is a dipeptide
having an N-terminal residue selected from the group consisting of Tyr, His, Lys, Phe and
Trp, and a C-terminal residue selected from the group consisting of Ala, Arg, Asp, Gly, He,
Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr and Val.
22. The compound in accordance with claim 1, having the formula:

wherein R1 is a member selected from the group consisting of alkyl, aryl,
ilkylaryl, cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each

WO 2006/045119 PCT/US2005/038282
160
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl;
L2 is selected from the group consisting of phenylene or methylenephenylene,
heteroarylene, optionally substituted with from 1 to 2 substituents each independently
selected from the group consisting of halo and haloalkyl.
23. The compound in accordance with claim 22, having the formula:

24. The compound in accordance with claim 22, having the formula:

25. The compound in accordance with claim 1, having the formula:

wherein R2 is selected from the group consisting of substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl.
26. The compound in accordance with claim 25, having the formula:

wherein R2 is substituted or unsubstituted aryl.
27. The compound in accordance with claim 26, having the formula:


WO 2006/045119 PCT/US2005/038282
161
wherein R1 is a member selected from the group consisting of alkyl, aryl,
alkylaryl, cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl.
28. The compound having the formula 972, 973, 975,1003,1004,1005,
1006,1011 described in Table 8, the formula 960, 961,981,982,983,984,985,1009,1014
described in Table 10, and the compounds in Tables 9 and 18 and their pharmaceutically
acceptable salts.
29. A method for inhibiting a soluble epoxide hydrolase, comprising
contacting said soluble epoxide hydrolase with an inhibiting amount of a compound of any of
claims 1 to 28.
30. A method of treating diseases modulated by soluble epoxide
hydrolases, said method comprising administering to a subject in need of such treatment an
effective amount of a compound of any of claims 1 to 28.
31. The method in accordance with claim 30, wherein said disease is
selected from the group consisting of hypertension, inflammation, adult respiratory distress
syndrome; diabetic complications; end stage renal disease; Raynaud syndrome and arthritis.
32. The method in accordance with claim 31, wherein said treatment
increases sodium excretion, reduces vascular and renal inflammation, and reduces male
erectile dysfunction.
- 33. The method in accordance with claim 31, wherein said hypertension is
selected from the group consisting of renal hypertension, pulmonary hypertension and hepatic
hypertension.
34. The method in accordance with claim 31, wherein said inflammation is
selected from the group consisting of renal inflammation, vascular inflammation, and lung
inflammation.
35. A method for reducing renal deterioration in a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28.

WO 2006/045119 PCT/US2005/038282
162
36. The method in accordance with claim 35, wherein said renal
deterioration is present in said subject afflicted with diabetes, hypertension or an
inflammatory disorder.
37. A method for inhibiting progression of nephropathy in a subject, said
method comprising administering to said subject an effective amount of a compound of any
of claims 1 to 28.
38. The method in accordance with claim 37 wherein the subject is (a) a
person with diabetes mellitus whose blood pressure is 130/85 or less, (b) a person with
metabolic syndrome whose blood pressure is 130/85 or less, (c) a person with a triglyceride
level over 215 mg/dL, or (d) a person with a cholesterol level over 200 mg/dL.
39. A method for reducing blood pressure in a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28. .
40. The method in accordance with claim 39, said method further
comprising administering to said subject an effective amount of a cis-epoxyeicosantrienoic
acid.
41. The method in accordance with claim 40, wherein said cis-
epoxyeicosantrienoic acid is administered with said compound having formula (I).
42. A method of increasing vasodilation hi a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28.
43. A method of inhibiting the progression of obstructive pulmonary
disease, an interstitial lung disease, or asthma in a subject, said method comprising
administering to said subject an effective amount of a compound of any of claims 1 to 28.
44. The method in accordance with claim 43, wherein said obstructive
pulmonary disease is selected from the group consisting of chronic obstructive pulmonary
disease, emphysema, and chronic bronchitis.

WO 2006/045119 PCT/US2005/038282
163
45. The method in accordance with claim 43, wherein said interstitial lung
disease is idiopathic pulmonary fibrosis or is one associated with exposure to dust.
46. The method in accordance with claim 43, said method further
comprising administering to said subject an effective amount of a cis-epoxyeicosantrienoic
acid.
47. The method in accordance with claim 46, wherein said cis-
epoxyeicosantrienoic acid is administered with said compound having formula (I).
48. A method of reducing vascular inflammation in a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28.
49. A method of reducing renal inflammation in a subject, said method
comprising administering to a subject an effective amount of a compound of any of claims 1
to 28.
50. A method of regulating endothelial cell function in a subject, said
method comprising administering to said subject an effective amount of a compound of any
of claims 1 to 28.
51. A method of decreasing endothelial cell inflammation in a subject, said
method comprising administering to said subject an effective amount of a compound of any
of claims 1 to 28.
52. A pharmaceutical composition comprising a pharmaceutically
acceptable excipient and a compound of any of claims 1 to 28.
53. A method for stabilizing biologically active epoxides in the presence of
a soluble epoxide hydrolase, said method comprising contacting said soluble epoxide
hydrolase with an amount of a compound of any of claims 1 to 28, sufficient to inhibit the
activity of said soluble epoxide hydrolase and stabilize said biologically active epoxide.
54. The method in accordance with claim 53, wherein said contacting is
conducted in an in vitro assay.

WO 2006/045119 PCT/US2005/038282
164
55. The method in accordance with claim 53, wherein said contacting is
conducted in vivo.
56. The method for reducing the formation of a biologically active diol
produced by the action of a soluble epoxide hydrolase, said method comprising contacting
said soluble epoxide hydrolase with an amount of a compound of any of claims 1 to 28,
sufficient to inhibit the activity of said soluble epoxide hydrolase and reduce the formation of
said biologically active diol.
57. The method in accordance with claim 56, wherein said contacting is
conducted in an in vitro assay.
58. The method in accordance with claim 56, wherein said contacting is
conducted in vivo.
59. A method for monitoring the activity of a soluble epoxide hydrolase,
said method comprising contacting said soluble epoxide hydrolase with an amount of a
compound of any of claims 1 to 28 sufficient to produce a detectable change in fluorescence
of said soluble epoxide hydrolase by interacting with one or more tryptophan residues present
in the catalytic site of said sEH.
60. The method in accordance with claim 59, wherein said compound has
an aryl group present in one or more components selected from the group consisting of R1,
L1,P3 and L2.
61. A method of increasing ease of formulation, oral availability, or serum
half-life of a compound comprising covalently attaching a polyether substituent to said
compound.
WO 2006/045119 PCT/US2005/038282
IMPROVED INHIBITORS FOR THE SOLUBLE EPOXIDE
HYDROLASE
CROSS-REFERENCE TO RELATED APPLICATION
[0001] This application claims the benefit of U.S. Patent Application No. 60/651,487, filed
October 20, 2004, the content of which is incorporated herein by reference.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER
FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] The U.S. Government has certain rights to the invention pursuant to contract
ES02710 awarded by the National Institutes of Health.
REFERENCE TO A "SEQUENCE LISTING," A TABLE, OR A COMPUTER
PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK.
[0003] NOT APPLICABLE
BACKGROUND OF THE INVENTION
[0004] Epoxide hydrolases (EHs, EC 3.3.2.3) catalyze the hydrolysis of epoxides or arene
oxides to their corresponding diols by the addition of water (see, Oesch, F., et al.,
Xenobiotica 1973, 3, 305-340). Some EHs play an important role in the metabolism of a
variety of compounds including hormones, chemotherapeutic drugs, carcinogens,
environmental pollutants, mycotoxins, and other harmful foreign compounds.
[0005] There are two well-studied EHs, microsomal epoxide hydrolase (mEH) and soluble
epoxide hydrolase (sEH). These en2ymes are very distantly related, have different
subcellular localization, and have different but partially overlapping substrate selectivities.
The soluble and microsomal EH forms are known to complement each other in degrading
some plant natural products (see, Hammock, B.D., et al., COMPREHENSIVE
TOXICOLOGY. Oxford: Pergamon Press 1977,283-305 and Fretland, A.J., et al., Chem.
Biol. Intereract 2000,129, 41-59).
[0006] The major role of the sEH is in the metabolism of lipid epoxides including the
metabolism of arachidonic acid (see, Zeldin, D.C., et al., J. Biol. Chem. 1993, 268, 6402-

WO 2006/045119 PCT/US2005/038282
2
6407), linoleic (see, Moghaddam, M.F., et al, Nat. Med. 1991,3, 562-567) acid, some of
which are endogenous chemical mediators (see, Carroll, M.A., et al., Thorax 2000,55, S13-
16). Epoxides of arachidonic acid (epoxyeicosatrienoic acids or EETs) and other lipid
epoxides and diols are known effectors of blood pressure (see, Capdevila, J.H., et al., J. Lipid.
Res. 2000, 41,163-181), and modulators of vascular permeability (see, Oltman, C.L., et al.,
Circ Res. 1998, 83, 932-939). The vasodilatory properties of EETs are associated with an
increased open-state probability of calcium-activated potassium channels leading to
hyperpolarization of the vascular smooth muscle (see Fisslthaler, B., et al., Nature 1999,401,
493-497). Hydrolysis of the arachidonate epoxides by sEH diminishes this activity (see,
Capdevila, J.H., et al., J. Lipid. Res. 2000, 41,163-181). sEH hydrolysis of EETs also
regulates their incorporation into coronary endothelial phospholipids, suggesting a regulation
of endothelial function by sEH (see, Weintraub, N.L., et al., Am. J. Physiol. 1992, 277,
H2098-2108), It has recently been shown that treatment of spontaneous hypertensive rats
(SHRs) with selective sEH inhibitors significantly reduces their blood pressure (see, Yu, Z.,
et al., Circ. Res. 2000, 87, 992-998). In addition, male knockout sEH mice have significantly
lower blood pressure than wild-type mice (see Sinal, C.J., et al., J. Biol. Chem. 2000,275,
40504-405010), further supporting the role of sEH in blood pressure regulation.
[0007] The EETs have also demonstrated anti-inflammatory properties in endothelial cells
(see, Node, K., et al., Science 1999, 285,1276-1279 and Campbell, W.B. Trends Pharmacol.
Sci. 2000, 21,125-127). In contrast, diols derived from epoxy-linoleate (leukotoxin) perturb
membrane permeability and calcium homeostasis (see, Moghaddam, M.F., et al., Nat. Med.
1997, 3, 562-567), which results in inflammation that is modulated by nitric oxide synthase
and endothelin-1 (see, Ishizaki, T.? et al., Am. J. Physiol. 1995,269, L65-70 and Ishizaki, T.,
et al., J. Appl. Physiol. 1995, 79, 1106-1611). Micromolar concentrations of leukotoxin
reported in association with inflammation and hypoxia (see, Dudda, A., et al., Chem. Phys.
Lipids 1996, 82, 39-51), depress mitochondrial respiration in vitro (see, Sakai, T., et al., Am.
J. Physiol. 1995, 269, L3 26-3 31), and cause mammalian cardiopulmonary toxicity in vivo
(see, Ishizaki, T., et al., Am. J. Physiol. 1995, 269, L65-70; Fukushima, A., et al., Cardiovasc.
Res. 1988, 22, 213-218; and Ishizaki, T., et al., Am. J. Physiol. 1995, 268, L123-128).
Leukotoxin toxicity presents symptoms suggestive of multiple organ failure and acute
respiratory distress syndrome (ARDS) (see, Ozawa, T. et al., Am. Rev. Respir. Dis. 1988,
137, 535-540). In both cellular and organismal models, leukotoxin-mediated toxicity is
dependent upon epoxide hydrolysis (see, Moghaddam, M.F., et al., Nat. Med. 1997, 3, 562-

WO 2006/045119 PCT/US2005/038282
3
567; Morisseau, C., et al., Proc. Natl. Acad. Sci USA 1999, 96, 8849-8854; and Zheng, J., et
al., Am. J. Respir. CellMol. Biol. 2001, 25,434-438), suggesting a role for sEH in the
regulation of inflammation and vascular permeability. The bioactivity of these epoxy-fatty
acids suggests that inhibition of vicnal-dihydroxy-lipid biosynthesis may have therapeutic
value, making sEH a promising pharmacological target.
[0008] Recently, 1,3-disubstituted ureas, carbamates, and amides have been reported as
new potent and stable inhibitors of sEH (Figure 1). See, U.S. Patent No. 6,150,415.
Compounds 192 and 686 are representative structures for this type of inhibitors (Figure 1).
These compounds are competitive tight-binding inhibitors with nanomolar KJ values that
interact stoichiometrically with purified recombinant sEH (see, Morisseau, C, et al., Proc.
Natl. Acad. Sci. USA 1999, 96, 8849-8854). Based on the X-ray crystal structure, the urea
inhibitors were shown to establish hydrogen bonds and to form salt bridges between the urea
function of the inhibitor and residues of the sEH active site, mimicking features encountered
in the reaction coordinate of epoxide ring opening by this enzyme (see, Argiriadi, M.A., et
al., Proc. Natl. Acad. Sci. USA 1999, 96,10637-10642 and Argiriadi, M.A., et al., J. Biol.
Chem. 2000, 275,15265-15270). These inhibitors efficiently reduced epoxide hydrolysis in
several in vitro and in vivo models (see, Yu, Z., et al., Circ. Res. 2000, 87, 992-998;
Morisseau, C, et al., Proc. Natl. Acad. Sci. USA 1999, 96, 8849-8854; and Newman, J.W., et
al., Environ. Health Perspect. 2001, 109, 61-66). Despite the high activity associated with
these inhibitors, there exists a need for compounds possessing similar or increased activities,
with improved solubility and pharmacokinetic properties to facilitate formulation and
delivery.
[0009] Surprisingly, the present invention provides such compounds along with methods
for their use and compositions that contain them.
BRIEF SUMMARY OF THE INVENTION
[0010] In one aspect, the present invention provides a method for inhibiting a soluble
epoxide hydrolase, comprising contacting the soluble epoxide hydrolase with an inhibiting
amount of a compound having a formula selected from the group consisting of:


WO 2006/045119 PCT/US2005/038282
4
and their pharmaceutically acceptable salts, wherein the symbol; R1 is a member selected
from the group consisting of substituted or unsubstituted alkyi, substituted or unsubstituted
heteroalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted
cycloalkylheteroalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted
arylheteroalkyl, substituted or unsubstituted C5-C12 cycloalkyl, substituted or unsubstituted
aryl, substituted or unsubstituted heteroaryl and combinations thereof, wherein said
cycloalkyl portions are monocyclic or polycyclic; P1 is a primary pharmacophore selected
from the group consisting of -OC(O)O-, -OC(O)CH2-, CH2C(O)O-, -OqO)-, -C(O)O-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-NHC(O)NH-,
-OC(O)NH-, -NHC(O)O-,-NHC(S)NH-, -NHC(S)CH2, CH2C(S)NH-, -SC(O)CH2-, -
CH2C(O)S-, -SC(NH)CH2-; - CH2C(NH)S-, -NON-, -CH2C(O)NH-, -NHC(O)CH2-;
-C(O)NH-, -NHC(O)-,

P2 is a secondary pharmacophore selected from the group consisting of -NH-, -OC(O)O-
,-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -NHC(NH)NH-, -NHC(NH)CH2-,
-CH2C(NH)NH-, -NHC(O)KB-, -OC(O)NH-, -NHC(O)O-, -C(O)NH-, -NHC(O)-; -
NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, -

WO 2006/045119 PCT/US2005/038282
5
CH2C(NH)S-, -N=C=N-,

P3 is a tertiary pharmacophore selected from the group consisting of C2-C6 alkenyl, C2-C6
alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, -O(CH2CH2O)q-R2, - OR2,
-C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4a1kyl-C(O)OR2, -C(O)R2, -C(O)OR2
and carboxylic acid analogs, wherein R2 is a member selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl; L1 is a first linker selected from the group
consisting of substituted or unsubstituted C1-C6 alkylene, substituted or unsubstituted C3-C6
cycloalkylene, substituted or unsubstituted arylene and substituted or unsubstituted
heteroarylene; L2 is a second linker selected from the group consisting of substituted or
unsubstituted C1-C12 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, substituted
or unsubstituted arylene, substituted or unsubstituted heteroarylene; an amino acid, a
dipeptide and a dipeptide analog; and combinations thereof; or is selected from the group
consisting of H and CH3 when m is 0. In the above formulae, the subscripts n and m are each
independently 0 or 1, and at least one of n or m is 1, and the subscript q is 0 to 6. When n is
0, then L1 and L2 are combined. When m is 0, then L2 can also be H.

WO 2006/045119 PCT/US2005/038282
6
[0011] Turning next to the linking groups, the symbol L1 represents a first linker that is a
substituted or unsubstituted C1-C6 alkylene, C3-C6-cycloalkylene, or an arylene or
heteroarylene group; the symbol L2 represents a second linker selected from substituted or
unsubstituted C1-C12 alkylene, substituted or unsubstituted arylene, an amino acid, a
dipeptide, a dipeptide analog, and combinations thereof; or is H when m is 0.
[0012] In a related aspect, the present invention provides methods of treating diseases
modulated by soluble epoxide hydrolases, the method comprising administering to a subject
in need of such treatment an effective amount of a compound having a formula selected from
formula (I), above.
[0013] In other aspects, the present invention provides methods of reducing renal
deterioration in a subject, the method comprising administering to the subject an effective
amount of a compound of formula (I), above.
[0014] In a related aspect, the present invention provides methods method for inhibiting
progression of nephropathy in a subject, the method comprising administering to the subject
an effective amount of a compound of formula (I), above.
[0015] In another aspect, the present invention provides for reducing blood pressure in a
subject, the method comprising administering to the subject an effective amount of a
compound of formula (I), above.
[0016] In a related aspect, the present invention provides methods of inhibiting the
proliferation of vascular smooth muscle cells in a subject, the method comprising
administering to the subject an effective amount of a compound of formula (I), above.
[0017] In another aspect, the present invention provides methods of inhibiting the
progression of an obstructive pulmonary disease, an interstitial lung disease, or asthma in a
subject, the method comprising administering to the subject an effective amount of a
compound of formula (T), above. The obstructive pulmonary disease can be, for example,
chronic obstructive pulmonary disease ("COPD"), emphysema, or chronic bronchitis. The
interstitial lung disease can be, for example, idiopathic pulmonary fibrosis, or one associated
with occupational exposure to a dust
[0018] In yet another aspect, the present invention provides compounds having a formula
(I) above, as well as pharmaceutical compositions containing one or more of the subject
compounds.

WO 2006/045119 PCT/US2005/038282
7
BRIEF DESCRIPTION OF THE DRAWINGS
[0019] Figure 1 provides structures of known sEH inhibitors having only a primary
pharmacophore: l-adamantyl-3-cyclohexylurea (192), l-adamantyl-3-dodecylurea (686).
[0020] Figure 2 provides a structural diagram defining the sEH inhibitors primary,
secondary, and tertiary pharmacophores. The nomenclature used refers to the three
pharmacophores and two substituents (R and R' groups). The secondary and tertiary
pharmacophores located in the R' area are illustrated linearly from the primary
pharmacophore. The secondary pharmacophore generally consists of a polar carbonyl group
or a polar ether group. When the secondary pharmacophore is a carbonyl group, it is located
about 7.5 ± 1 A from the carbonyl of the primary pharmacophore, with either side of the
carbonyl (X and Y) being a CH2, O or NH. When the secondary pharmacophore is a ether
group it is preferably located about 1 carbon unit further from the carbonyl of the primary
pharmacophore. The tertiary pharmacophore is also a polar group located approximately 11
carbon units (17 ± 1 Å) from the carbonyl of the primary pharmacophore with the Z group as
an OH, or a substituted amine or alcohol or a heterocyclic or acyclic structure mimicing the
terminal ester or acid.
[0021] Figure 3 provides a hydrophobicity map of the mouse sEH substrate binding pocket
co-crystalyzed with the inhibitor l-cyclohexyl-3-dodecyl urea. A shading gradient indicates
degrees of hydrophobicity. A series of hydrophilic residues were observed on the "top" side
of the channel, while the "bottom" of the channel was very hydrophobic, with the exception
of the catalytic aspartate (Asp333). This structural analysis indicated that a number of
potential hydrogen bonding sites are observed in the substrate binding pocket of the soluble
epoxide hydrolase, primarily located on the surface opposite Asp333 (the catalytic nucleophile
which reacts with the substrate or binds to the primary pharmacophores).
[0022] Figure 4 provides mammalian soluble epoxide hydrolase protein sequence
alignments (residue 1-340).
[0023] Figure 5 provides mammalian soluble epoxide hydrolase protein sequence
alignments (residue 341-554).
[0024] Figure 6 is a graph illustrating the metabolic stabilities of l-adamantyl-3-dodecyl
urea (686) and 1-cyclohexyl- 3-dodecyl urea (297) in rat hepatic microsomes. Microsomes

WO 2006/045119 PCT/US2005/038282
8
were incubated with 1 μM 686 or 297 in the presence of an NADPH generating system. Data
are expressed as mean ± SD of triplicate experiments.
[0025] Figure 7 is a graph illustrating.the metabolic stabilities of 686 and 687 in rat hepatic
microsomes as described above.
[0026] Figure 8 is a series of graphs illustrating the metabolic conversion of 1 -adamantyl-
3-dodecyl urea (686) in microsomal preparations from rat, mouse, and human hepatic tissues.
The metabolites identified are the omega hydroxyl (686-M1), the omega aldehyde (686-M2),
the omega acid (687), and a mixture of monohydroxy adamantyl omega hydroxylated
compounds (686-M3). These structures are shown in Table 13.
[0027] Figure 9 provides a mass spectrum showing collision induced dissociation of a
dominant urinary metabolite of l-adamantyl-3-dodecyl urea (686) and the 3-dodecanoic acid
analog (687) suggesting that these compounds can ultimately enter beta-oxidation to produce
chain shortened inhibitors.
[0028] Figure 10 is a graph illustrating the blood concentration vs. time profiles of 687
after oral administration of 5 mg/kg of either 687 or 800 to mice. The ester compound delays
the time to achieve the maximum circulating dose, and increases the maximum circulating
concentration of 687 observed. This translates into a longer half-life for the inhibitor.
[0029] Figure 11 provides a structural evaluation of conserved hydrogen bond donors in
the sEH substrate binding pocket with linear distances to the primary pharmacophore noted
and further illustrating the effect of functional group distances on interactions with the
mammalian soluble epoxide hydrolases.
[0030] Figure 12 is a graph illustrating the relative substrate turnover/relative inhibitor
potency as a function of terminal carboxyl distance to either substrate epoxide of inhibitor 3-
position nitrogen.
[0031] Figure 13 is a bar graph showing the levels of urinary octadecanoids (A) and
urinary eicosanoids (B) in rats treated with angiotensin II in the presence of absence of 687.
[0032] Figure 14 is a graph showing blood concentration vs. time profiles of 950 after
single oral administration of 0.1 to 1.0 mg/kg of 950 to 70 kg rats. The presence of the
polyether secondary pharmacophore increases the maximum circulating concentration of 950
observed. This translates into a longer half-life for the inhibitor.

WO 2006/045119 PCT/US2005/038282
9
[0033] Figure 15 provides a sample preparation procedure for a pharmacokinetic study. A
5 μl whole blood sample was drawn into a capillary at a specific time point, each sample was
extracted and anaylzed by LC/MS-MS.
[0034] Figure 16 shows the physical properties/parameters of compound 950.
[0035] Figure 17 shows graphs which illustrate the in vitro metabolism of 950 in (A)
human liver microsome (no NADPH), (B) S9 fractions, and (C) Liver microsomes both with
NADPH. Both rat and human microsomes were used for the 950 metabolism study. The
hydroxy metabolite was the major metabolite.
DETAILED DESCRIPTION OF THE INVENTION
Abbreviations and Definitions:
[0036] "cis-Epoxyeicosatrienoic acids" ("EETs") are biomediators synthesized by
cytochrome P450 epoxygenases.
[0037] "Epoxide hydrolases" ("EH;" EC 3.3.2.3) are enzymes in the alpha / beta hydrolase
fold family that add water to 3 membered cyclic ethers termed epoxides.
[0038] "Soluble epoxide hydrolase" ("sEH") is an enzyme which in endothelial, smooth
muscle and other cell types converts EETs to dihydroxy derivatives called
dihydroxyeicosatrienoic acids ("DHETs"). The cloning and sequence of the murine sEH is
set forth in Grant et al, J. Biol Chem. 268(23):17628-17633 (1993). The cloning, sequence,
and accession numbers of the human sEH sequence are set forth in Beetham et al., Arch.
Biochem. Biophys. 305(1): 197-201 (1993). The amino acid sequence of human sEH is also
set forth as SEQ ID NO.2 of U.S. Patent No. 5,445,956; the nucleic acid sequence encoding
the human sEH is set forth as nucleotides 42-1703 of SEQ ID NO:1 of that patent The
evolution and nomenclature of the gene is discussed in Beetham et al., DNA Cell Biol.
14(1):61-71 (1995). Soluble epoxide hydrolase represents a single highly conserved gene
product with over 90% homology between rodent and human (Arand et al., FEBS Lett.,
338:251-256 (1994)).
[0039] The terms "treat", "treating" and "treatment" refer to any method of alleviating or
abrogating a disease or its attendant symptoms.
[0040] The term "therapeutically effective amount" refers to that amount of the compound
being administered sufficient to prevent or decrease the development of one or more of the
symptoms of the disease, condition or disorder being treated.

WO 2006/045119 PCT/US2005/038282
10
[0041] The term "modulate" refers to the ability of a compound to increase or decrease the
function, or activity, of the associated activity (e.g., soluble epoxide hydrolase).
"Modulation", as used herein in its various forms, is meant to include antagonism and partial
antagonism of the activity associated with sEH. Inhibitors of sEH are compounds that, e.g.,
bind to, partially or totally block the enzyme's activity.
[0042] The term "compound" as used herein is intended to encompass not only the
specified molecular entity but also its pharmaceutically acceptable, pharmacologically active
derivatives, including, but not limited to, salts, prodrug conjugates such as esters and amides,
metabolites and the like.
[0043] The term "composition" as used herein is intended to encompass a product
comprising the specified ingredients in the specified amounts, as well as any product which
results, directly or indirectly, from combination of the specified ingredients in the specified
amounts. By "pharmaceutically acceptable" it is meant the carrier, diluent or excipient must
be compatible with the other ingredients of the formulation and not deleterious to the
recipient thereof.
[0044] The "subject" is defined herein to include animals such as mammals, including, but
not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats,
mice and the like. In some embodiments, the subject is a human.
[0045] As used herein, the term "sEH-mediated disease or condition" and the like refers to
a disease or condition characterized by less than or greater than normal, sEH activity. A
sEH-mediated disease or condition is one in which modulation of sEH results in some effect
on the underlying condition or disease (e.g., a sEH inhibitor or antagonist results in some
improvement in patient well-being in at least some patients).
[0046] "Parenchyma" refers to the tissue characteristic of an organ, as distinguished from
associated connective or supporting tissues.
[0047] "Chronic Obstructive Pulmonary Disease" or "COPD" is also sometimes known as
"chronic obstructive airway disease", "chronic obstructive lung disease", and "chronic
airways disease." COPD is generally defined as a disorder characterized by reduced maximal
expiratory flow and slow forced emptying of the lungs. COPD is considered to encompass
two related conditions, emphysema and chronic bronchitis. COPD can be diagnosed by the
general practitioner using art recognized techniques, such as the patient's forced vital capacity
("FVC"), the maximum volume of air that can be forceably expelled after a maximal

WO 2006/045119 PCT/US2005/038282
11
inhalation. In the offices of general practitioners, the FVC is typically approximated by a 6
second maximal exhalation through a spirometer. The definition, diagnosis and treatment of
COPD, emphysema, and chronic bronchitis are well known in the art and discussed in detail
by, for example, Honig and Ingram, in Harrison's Principles of Internal Medicine, (Fauci et
al., Eds.), 14th Ed., 1998, McGraw-Hill, New York, pp. 1451-1460 (hereafter, "Harrison's
Principles of Internal Medicine").
[0048] "Emphysema" is a disease of the lungs characterized by permanent destructive
enlargement of the airspaces distal to the terminal bronchioles without obvious fibrosis.
[0049] "Chronic bronchitis" is a disease of the lungs characterized by chronic bronchial
secretions which last for most days of a month, for three months a year, for two years.
[0050] As the names imply, "obstructive pulmonary disease" and "obstructive lung disease"
refer to obstructive diseases, as opposed to restrictive diseases. These diseases particularly
include COPD, bronchial asthma and small airway disease.
[0051] "Small airway disease." There is a distinct minority of patients whose airflow
obstruction is due, solely or predominantly to involvement of the small airways. These are
defined as airways less than 2 mm in diameter and correspond to small cartilaginous bronchi,
terminal bronchioles and respiratory bronchioles. Small airway disease (SAD) represents
luminal obstruction by inflammatory and fibrotic changes that increase airway resistance.
The obstruction may be transient or permanent.
[0052] The "interstitial lung diseases (ILDs)" are a group of conditions involving the
alveolar walls, perialveolar tissues, and contiguous supporting structures. As discussed on
the website of the American Lung Association, the tissue between the air sacs of the lung is
the interstitium, and this is the tissue affected by fibrosis in the disease. Persons with the
disease have difficulty breathing in because of the stiffness of the lung tissue but, in contrast
to persons with obstructive lung disease, have no difficulty breathing out. The definition,
diagnosis and treatment of interstitial lung diseases are well known in the art and discussed in
detail by, for example, Reynolds, H.Y., in Harrison's Principles of Internal Medicine, supra,
at pp. 1460-1466. Reynolds notes that, while ILDs have various initiating events, the
immunopathological responses of lung tissue are limited and the ILDs therefore have
common features.
[0053] "Idiopathic pulmonary fibrosis," or "IPF," is considered the prototype ILD.
Although it is idiopathic in that the cause is not known, Reynolds, supra, notes that the term
refers to a well defined clinical entity.

WO 2006/045119 PCT/US2005/038282
12
[0054] "Bronchoalveolar lavage," or "BAL," is a test which permits removal and
examination of cells from the lower respiratory tract and is used in humans as a diagnostic
procedure for pulmonary disorders such as IPF. In human patients, it is usually performed
during bronchoscopy.
[0055] As used herein, the term "alkyl" refers to a saturated hydrocarbon radical which may
be straight-chain or branched-chain (for example, ethyl, isopropyl, t-amyl, or 2,5-
dimethylhexyl). This definition applies both when the term is used alone and when it is used
as part of a compound term, such as "aralkyl," "alkylamino" and similar terms. In some
embodiments, alkyl groups are those containing 1 to 24 carbon atoms. All numerical ranges
in this specification and claims are intended to be inclusive of their upper and lower limits.
Lower alkyl refers to those alkyl groups having 1 to 4 carbon atoms. Additionally, the alkyl
and heteroalkyl groups may be attached to other moieties at any position on the alkyl or
heteroalkyl radical which would otherwise be occupied by a hydrogen atom (such as, for
example, 2-pentyl, 2-methylpent-l-yl and 2-propyloxy). Divalent alkyl groups may be
referred to as "alkylene", and divalent heteroalkyl groups may be referred to as
"heteroalkylene" such as those groups used as linkers in the present invention. The alkyl,
alkylene, and heteroalkyl moieties may also be optionally substituted with halogen atoms, or
other groups such as oxo, cyano, nitro, alkyl, alkylamino, carboxyl, hydroxyl, alkoxy,
aryloxy, and the like.
[0056] The terms "cycloalkyl" and "cycloalkenyl" refer to a saturated hydrocarbon ring and
includes bicyclic and polycyclic rings. Similarly, cycloalkyl and cycloalkenyl groups having
a heteroatom (e.g. N, O or S) in place of a carbon ring atom may be referred to as
"heterocycloalkyl" and heterocycloalkylene," respectively. Examples of cycloalkyl and
heteroaryl groups are, for example, cyclohexyl, norbornyl, adamantly, morpholinyl,
thiomorpholinyl, dioxothiomorphorinyl, and the like. The cycloalkyl and heterocycloalkyl
moieties may also be optionally substituted with halogen atoms, or other groups such as nitro,
alkyl, alkylamino, carboxyl, alkoxy, aryloxy and the like. In some embodiments, cycloalkyl
and cycloalkenyl moieties are those having 3 to 12 carbon atoms in the ring (e.g., cyclohexyl,
cyclooctyl, norbornyl, adamantyl, and the like). In some embodiments, heterocycloalkyl and
heterocycloalkylene moieties are those having 1 to 3 hetero atoms in the ring (e.g.,
morpholinyl, Momorpholinyl, dioxothiomorphonnyl, piperidinyl and the like). Additionally,
the term "(cycloalkyl)alkyl" refers to a group having a cycloalkyl moiety attached to an alkyl
moiety. Examples are cyclohexylmethyl, cyclohexylethyl and cyclopentylpropyl.

WO 2006/045119 PCT/US2005/038282
13
[0057] The term "alkenyl" as used herein refers to an alkyl group as described above which
contains one or more sites of unsaturation that is a double bond. Similarly, the term
"alkynyl" as used herein refers to an alkyl group as described above which contains one or
more sites of unsaturation that is a triple bond.
[0058] The term "alkoxy" refers to an alkyl radical as described above which also bears an
oxygen substituent which is capable of covalent attachment to another hydrocarbon radical
(such as, for example, methoxy, ethoxy, aryloxy and t-butoxy).
[0059] The term "aryl" refers to an aromatic carbocyclic substituent which may be a single
ring or multiple rings which are fused together, linked covalently or linked to a common
group such as an ethylene or methylene moiety. Similarly, aryl groups having a heteroatom
(e.g. N, O or S) in place of a carbon ring atom are referred to as "heteroaryl". Examples of
aryl and heteroaryl groups are, for example, phenyl, naphthyl, biphenyl, diphenylmethyl, 2,2-
diphenyl-1-ethyl, thienyl, pyridyl and quinoxalyl. The aryl and heteroaryl moieties may also
be optionally substituted with halogen atoms, or other groups such as nitro, alkyl, alkylamino,
carboxyl, alkoxy, phenoxy and the like. Additionally, the aryl and heteroaryl groups may be
attached to other moieties at any position on the aryl or heteroaryl radical which would
otherwise be occupied by a hydrogen atom (such as, for example, 2-pyridyl, 3-pyridyl and 4-
pyridyl). Divalent aryl groups are "arylene", and divalent heteroaryl groups are referred to as
"heteroarylene" such as those groups used as linkers in the present invention.
[0060] The terms "arylalkyl", "arylalkenyl" and "aryloxyalkyl" refer to an aryl radical
attached directly to an alkyl group, an alkenyl group, or an oxygen which is attached to an
alkyl group, respectively. For brevity, aryl as part of a combined term as above, is meant to
include heteroaryl as well.
[0061] The terms "halo" or "halogen," by themselves or as part of another substituent,
mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally,
terms such as "haloalkyl," are meant to include monohaloalkyl and polyhaloalkyl. For
example, the term "C1-C6 haloalkyl" is mean to include trifluoromethyl, 2,2,2-trifluoroethyl,
4-chlorobutyl, 3-bromopropyl, and the like.
[0062] The term "hetero" as used in a "heteroatom-containing alkyl group" (a "heteroalkyl"
group) or a "heteroatom-containing aryl group" (a "heteroaryl" group) refers to a molecule,
linkage or substituent in which one or more carbon atoms are replaced with an atom other
than carbon, e.g., nitrogen, oxygen, sulfur, phosphorus or silicon, typically nitrogen, oxygen

WO 2006/045119 PCT/US2005/038282
14
or sulfur or more thatnone non-carbon atom (e.g., sulfonamide). Similarly, the term
"heteroalkyl" refers to an alkyl substituent that is heteroatom-containing, the term
"heterocyclic" refers to a cyclic substituent that is heteroatom-containing, the terms
"heteroaryl" and heteroaromatic" respectively refer to "aryl" and "aromatic" substituents that
are heteroatom-containing, and the like. Examples of heteroalkyl groups include alkoxyaryl,
alkylsulfanyl-substituted alkyl, N-alkylated amino alkyl, and the like. Examples of heteroaryl
substituents include pyrrolyl, pyrrolidinyl, pyridinyl, quinolinyl, indolyl, pyrimidinyl,
imidazolyl, 1,2,4-triazolyl, tetrazolyl, etc., and examples of heteroatom-containing alicyclic
groups are pyrrolidino, morpholino, piperazino, piperidino, etc.
[0063] The term "hydrophobic radical" or "hydrophobic group" refers to a group which
lowers the water solubility of a molecule. In some embodiments, hydrophobic radicals are
groups containing at least 3 carbon atoms.
[0064] The term "carboxylic acid analog" refers to a variety of groups having an acidic
moiety that are capable of mimicking a carboxylic acid residue. Examples of such groups are
sulfonic acids, sulflnic acids, phosphoric acids, phosphonic acids, phosphinic acids,
sulfonamides, and heterocyclic moieties such as, for example, imidazoles, triazoles and
tetrazoles.
[0065] The term "substituted" refers to the replacement of an atom or a group of atoms of a
compound with another atom or group of atoms. For example, an atom or a group of atoms
may be substituted with one or more of the following substituents or groups: halo, cyano,
nitro, alkyl, alkylamino, hydroxyalkyl, haloalkyl, carboxyl, hydroxyl, alkoxy, alkoxyalkoxy,
haloalkoxy, thioalkyl, aryl, aryloxy, cycloalkyl, cycloalkylalkyl, aryl, heteroaryl optionally
substituted with 1 or more, preferably 1 to 3, substituents selected from halo, halo alkyl and
alkyl, aralkyl, heteroaralkyl, alkenyl containing 1 to 2 double bonds, alkynyl containing 1 to 2
triple bonds, alk(en)(yn)yl groups, halo, cyano, hydroxy, haloalkyl and polyhaloalkyl,
preferably halo lower alkyl, especially trifiuoromethyl, formyl, alkylcarbonyl, arylcarbonyl
that is optionally substituted with 1 or more, preferably 1 to 3, substituents selected from
halo, halo alkyl and alkyl, heteroarylcarbonyl, carboxy, alkoxycarbonyl, aryloxycarbonyl,
aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, arylaminocarbonyl,
diarylaminocarbonyl, aralkylaminocarbonyl, alkoxy, aryloxy, perfluoroalkoxy, alkenyloxy,
alkynyloxy, arylalkoxy, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, arylaminoalkyl,
amino, alkylamino, dialkylamino, arylamino, alkylarylamino, alkylcarbonylamino,
arylcarbonylamino, azido, nitro, mercapto, alkylthio, arylthio, perfluoroalkylthio, thiocyano,

WO 2006/045119 PCT/US2005/038282
15
isothiocyano, alkylsulfinyl, alkylsulfonyl, arylsulfinyl, arylsulfonyl, aminosulfonyl,
alkylaminosulfonyl, dialkylaminosulfonyl and arylaminosulfonyl. When the term
"substituted" appears prior to a list of possible substituted groups, it is intended that the term
apply to every member of that group.
[0066] The term "unsubstituted" refers to a native compound that lacks replacement of an
atom or a group of atoms.
General:
[0067] The present invention derives from the discovery that 1,3-disubstituted ureas (or the
corresponding amides or carbamates, also referred to as the primary pharmacophore) can be
further functionalized to provide more potent sEH inhibitors with improved physical
properties. As described herein, the introduction of secondary and/or tertiary
pharmacophores can increase water solubility and oral availability of sEH inhibitors (see
Figure 2). The combination of the three pharmacophores (see the compounds of Table 18)
provides a variety of compounds of increased water solubility.
[0068] The discovery of the secondary and tertiary pharmacophores has also led to the
employment of combinatorial chemistry approaches for establishing a wide spectrum of
compounds having sEH inhibitory activity. The polar pharmacophores divide the molecule
into domains each of which can be easily manipulated by common chemical approaches in a
combinatorial manner, leading to the design and confirmation of novel orally available
therapeutic agents for the treatment of diseases such as hypertension and vascular
inflammation. The agents of the present invention treat such diseases while simultaneously
increasing sodium excretion, reducing vascular and renal inflammation, and reducing male
erectile dysfunction As shown below (see Example 51 and Figure 13), alterations in
solubility, bioavailability and pharmacological properties leads to compounds that can alter
the regulatory lipids of experimental animals increasing the relative amounts of epoxy
arachidonate derivatives when compared either to their diol products or to the
proinflammatory and hypertensive hydroxyeicosatetraenoic acids (HETEs). Since epoxy
arachidonates are anti-hypertensive and anti-inflammatory, altering the lipid ratios can lead to
reduced blood pressure and reduced vascular and renal inflammation. This approach has
been validated in a patient approaching end stage renal disease (ESRD) where even a brief
oral treatment with low doses compound 800 altered the serum profile of regulatory lipids in
a positive manner. This resulted in reduced systolic and diastolic blood pressure, a dramatic

WO 2006/045119 PCT/US2005/038282
16
reduction in blood urea nitrogen (an indicator of renal inflammation) and dramatically
reduced serum levels of C reactive protein (a common indicator of vascular inflammation).
[0069] Without intending to be bound by theory, and with reference to Figures 2, 3, 4 and
5, it is believed that the left side of the primary pharmacophore or R (in Figure 2) can be
varied to obtain optimal properties as can the primary pharmacophore, which contains groups
able to hydrogen bond to the catalytic aspartic acid on one side and the catalytic tyrosines on
the other (see Figure 3). The right side of the primary pharmacophore is effectively divided
into 4 segments: a spacer separating the primary and secondary pharmacophore (termed L1 in
the present invention), the secondary pharmacophore (termed P2 in the present invention)
and a tertiary pharmacophore (P3) flanked by a spacer (L2) and finally a terminating group Z
(collectively provided with the tertiary pharmacophore as P3). The spacer between the
primary and secondary pharmacophores, is optimally 3 atom units in length, while the
secondary pharmacophore can be, for example, a ketone, carbonate, amide, carbamate, urea,
ether/polyether, ester or other functionality able to form a hydrogen bond with the enzyme
approximately 7.5 angstroms from the carbonyl of the primary pharmacophore. The
identified tertiary pharmacophore consists of a polar group located approximately six to
eleven carbon units from the primary pharmacophore (see Figure 2). A conserved asparagine
residue (Asn471, see Figures 4 and 5) is thought to provide the site of interaction between the
protein and the polar functionality located at this tertiary site. While, in the rodent a
threonine (Thr468) is also in an appropriate position for hydrogen bonding, residue 468 is a
methionine in the human enzyme (Figure 5). As with the secondary pharmacophore, this
group improves water solubility of sEH inhibitors as well as the specificity for the sEH, and a
wide diversity of functionalities such as an ester, amide, carbamate, or similar functionalities
capable of donating or accepting a hydrogen bond similarly can contribute to this polar
group. For example, in pharmaceutical chemistry heterocyclic groups are commonly used to
mimic carbonyls as hydrogen bond donors and acceptors. Of course the primary, secondary
and tertiary pharmacophore groups can be combined in a single molecule with suitable
spacers to improve activity or present the inhibitor as a prodrug.
[0070] Figure 11 illustrates the binding interaction for structural evaluation of conserved
hydrogen bond donors in the sEH substrate binding pocket with linear distances to the
primary pharmacophore noted. The table below provides specific distances to residues
provided in Figures 4 and 5.

WO 2006/045119 PCT/US2005/038282
17
Table
[0071] Linear distances of hydrophylic residues to the carbonyl carbon of the bound urea
Residue Distance Conserved
from Urea Carbon
Asp333 4.7Å +
Tyr465 O 4.5Å +
Tyr381O 4.6Å +
Trp334NRing 7.1Å +
G1n382N 8.2Å +
Tyr465 NBack Bone 10.5Å +
Tyr4b8 14.9Å Met in Human
Asn471N 15.2Å +
Asn471 O 16.7Å +
*Note Figure 11 distances are measured linearly from the carbonyl oxygen to the alternate pharmacophores.
This Table measures 3 dimensional distances from carbonyl carbon of the primary phaxmacophore to amino
acids which could hydrogen bond with the inhibitor.
Methods of Inhibiting Soluble Epoxide Hvdrolases:
[0072] In view of the above, the present invention provides, in one aspect, a method for
inhibiting a soluble epoxide hydrolase, comprising contacting the soluble epoxide hydrolase
with an inhibiting amount of a compound having a formula selected from the group
consisting of:

and their pharmaceutically acceptable salts, wherein the symbol R1 is a member selected from
the group consisting of substituted or unsubstituted alkyl, substituted or unsubstituted
heteroalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted
cycloalkylheteroalkyl, substituted or unsubstituted arylalkyl, substituted or unsubstituted
arylheteroalkyl, substituted or unsubstituted C5-C12 cycloalkyl, substituted or unsubstituted
aryl, substituted or unsubstituted heteroaryl and combinations thereof, wherein said
cycloalkyl portions are monocyclic or polycyclic; P is a primary pharmacophore selected
from the group consisting of -OC(O)O-, -OC(O)CH2-, CH2C(O)O-, -OC(O)-, -C(O)O-, -

WO 2006/045119 PCT/US2005/038282
18
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-NHC(O)NH-,
-OC(O)NH-, -NHC(O)O-,-NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, -
CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -NON-, -CH2C(O)NH-, -NHC(O)CH2-,
-C(O)NH-,

-NHC(O)-,
a secondary pharmacophore selected from the group consisting of -NH-, -OC(O)O-,-C(O)-,
-CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-,-NHC(NH)NH-, -NHC(NH)CH2-, -
CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-, -C(O)NH-, -NHC(O)-; -
NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, -

WO 2006/045119 PCT/US2005/038282
19
CH2C(NH)S-, -N=C=N-,

P3 is a tertiary pharmacophore selected from the group consisting of C2-C6 alkenyl, C2-C6
alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, -O(CH2CH2O)q-R2, - OR2,
-C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(O)OR2, -C(O)R2, -C(O)OR2
and carboxylic acid analogs, wherein R2 is a member selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl. In the above formula, the subscripts n and m
are each independently 0 or 1, and at least one of n or m is 1, and the subscript q is 0 to 6.
[0073] Turning next to the linking groups, the symbol L1 represents a first linker that is
selected from the group consisting of substituted or unsubstituted C1-C6 alkylene, substituted
or unsubstituted C3-C6 cycloalkylene, substituted or unsubstituted arylene and substituted or
unsubstituted heteroarylene; the symbol L2 represents a second linker selected from the group
consisting of substituted or unsubstituted C1-C12 alkylene, substituted or unsubstituted C3-C6
cycloalkylene, substituted or unsubstituted arylene, substituted or unsubstituted
heteroarylene; an ammo acid, a dipeptide and a dipeptide analog; and combinations thereof;
or is H when m is 0. Preferably, the compounds are other than 1 l-(3-cyclohexylureido)-

WO 2006/045119 PCT/US2005/038282
20
undecanoic acid, ll-(3-cyclohexylureido)-undecanoic acid methyl ester, 11-(3-
cyclohexylureido)-undecanoic acid amide, 12-(3-cyclohexylureido)-dodecanoic acid and 12-
(3-adamantan-l-yl-ureido)-dodecanoic acid.
[0074] In a first group of embodiments, R1 is selected from the group consisting of
substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or
unsubstituted cycloalkylalkyl, substituted or unsubstituted cycloalkylheteroalkyl, substituted
or unsubstituted arylalkyl and substituted or unsubstituted arylheteroalkyl. In another group
of embodiments, R1 is selected from C5-C12 cycloalkyl, phenyl and naphthyl. More
preferably, R1 is selected from C6-C10 cycloalkyl and phenyl. In some embodiments, are
those embodiments in which R1 is cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, adamantyl,
noradamantyl, and phenyl, wherein the phenyl group is either unsubstituted or substituted
with from one to three substituents selected from halogen, lower alkyl, lower halo alkyl,
lower alkoxy, C3-C5 cycloalkyl and cyano.
[0075] Returning to formula (I), P1 is preferably selected from -NHC(O)NH-,
-OC(O)NH- and -NHC(O)O-. Most preferably, P1 is -NHC(O)NH-. In other embodiments,
P1 is selected from the group consisting of -OC(O)O-, -OC(O)CH2-, CH2C(O)O-, -OC(O)-, -
C(O)O-, -NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-
NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, -
CH2C(NH)S-, -NON-, -NHC(O)CH2-,

WO 2006/045119 PCT/US2005/038282
21

[0076] Turning next to the first linking group, L1 is preferably selected from substituted or
unsubstituted C1-C6 alkylene, wherein the substituents are selected to impart desired
properties to the overall composition. For example, in some embodiments in which R1 is a
particularly hydrophobic residue, L1 may preferably have substituents that are hydrophilic to
offset to some degree the lack of aqueous solubility normally associated with very
hydrophobic compounds. As a result, in some embodiments, L1 will have one or two
hydroxy moieties as substituents, preferably only one hydroxy moiety substituents. In other
embodiments, L1 will be an alkylene, arylene or cycloalkylene linker having the length
indicated above, wherein one or more of the hydrogen atoms are replaced with fluorine atoms
to impart other attractive properties, such as facilitating the compound's use in stents so that it
is slowly released from the stent to then inhibit the soluble epoxide hydrolase. Other
examples of substituents, include but are not limited to, halo, cyano, nitro, alkyl, alkylamino,
carboxyl, hydroxyl, alkoxy, aryloxy, and the like. Further are embodiments in which L1 is
C2-C5 alkylene, more preferably C2-C4 alkylene, still more preferably C2-C3 alkylene, and
most preferably an ethylene linkage. Where L1 is C3-C6 cycloalkylene, it is more preferably
cyclohexyl that can be linked in a 1,3 or 1,4 manner. In certain embodiments, L1 is selected
to provide spacing between the first pharmacophore carbonyl moiety (in P1) and the second

WO 2006/045119 PCT/US2005/038282
22
pharmacophore carbonyl moiety (in P2) of about 7.5 ± 2 angstroms and more preferably,
about 7.5 ± 1 angstroms.
[0077] The secondary pharmacophore, P2, when present (n is 1) is selected from the group
consisting of-NH-, -OC(O)O-,-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(0)O, -OC(O)-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)-; -NHC(S)NH-, -NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, -
CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -N=C=N-,

More preferably, P2 is selected from -C(O)-, -O(CH2CH2O)q-, -C(0)O, -OC(O)-, -OC(O)O-,
-OC(O)NH- and -C(O)NH-. Most preferably, P2 is selected from -C(O)-, -O(CH2CH2O)q-,
and -C(O)O-. In another embodiment, P2 is preferably selected from the group consisting of
-NH-, -OC(O)O-, -NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(S)NH-, -
NHC(S)CH2-, CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -
N=C=N-,

WO 2006/045119 PCT/US2005/038282
23

[0078] The second linking group, L2 is selected from substituted or unsubstituted C1-C12
alkylene, substituted or unsubstituted arylene, and combinations thereof. For those
embodiments in which a secondary pharmacophore (P2) is not present, the Unking group L2
will be combined with L1 to provide spacing between the primary pharmacophore and the
tertiary pharmacophore preferably of about ≥2, and ≤12 carbon atoms. Accordingly, when
L1 is an alkylene or part of a cycloalkylene linkage of from 1 to 4 carbon atoms, and P2 is not
present, L2 will preferably be an alkylene linkage of from 1 to 8 carbon atoms, more
preferably, 4 to 8 carbon atoms, and most preferably 5, 6, 7 or 8 carbon atoms. For those
embodiments in which a tertiary pharmacophore (P3) is not present, the linking group L2 may
be H orwill terminate with hydrogen or a substituent selected as described for L1 above. In
such embodiments, the arylene group need not be divalent. In some embodiments, L2 will
comprise an arylene group, preferably a phenylene group that can be linked in a 1,2 or 1,3 or
1,4 manner, preferably in a 1,3 or 1,4 manner. As with L1, the alkylene portions of L2 can be
substituted or unsubstituted. The substituents are selected as described for L1 above.
[0079] The tertiary pharmacophore, P3, is a tertiary pharmacophore selected from the group
consisting of C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl,
-O(CH2CH2O)q-R2, - OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-

WO 2006/045119 PCT/US2005/038282
24
C(O)OR2, -C(O)R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is a member
selected from the group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl,
substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl;
substituted or unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl. In certain
embodiments, R2 is H, methyl, ethyl, propyl, allyl, 3-propynyl, butyl, 2-propyl, 1,1-
dimethylethyl, 2-butyl, 2-methyl-1 -propyl, adamantyl-methyl, benzyl, 2-chlorobenzyl and
naphthyknethyl. In one group of embodiments, P3 is -C(O)NHR2, -C(O)NHS(O)2R2,
-NHS(O)2R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is selected from hydrogen,
unsubstituted C1-C4 alkyl, and unsubstituted C3-C8 cycloalkyl. Still more preferably, R2 is H,
Me or Et. In some embodiments, P3 is -C(O)OR2 and carboxylic acid analogs, wherein R2 is
selected from hydrogen, Me or Et. In other embodiments, P3 is preferably selected from the
group consisting of is selected from the group consisting of C2-C6 alkenyl, heterocyclyl, OR2,
-OC2-C4alkyl-C(O)OR2 and -C(O)R2, wherein R2 is a member selected from the group
consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted
C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl
and substituted or unsubstituted aryl C1-C4 alkyl.
[0080] With the embodiments provided above, certain combinations of embodiments
represent particular embodiments. While all combinations of the groups represent additional
embodiments of the invention, particular embodiments include those wherein P1 is selected
from -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; P2 is selected from -C(O)O-, -OC(O)-,
-O(CH2CH2O)q-, -C(O)NH- and -NHC(O)-; m is 0 and L1 is selected from unsubstituted C1-
C6 alkylene. In another group of particular embodiments, P1 is selected from -NHC(O)NH-,
-OC(O)NH- and -NHC(O)O; P2 is selected from -C(O)O-, -OC(O)-, -O(CH2CH2O)q-,
-C(O)NH- and -NHC(O)-; n and m are each 1; L1 is selected from unsubstituted C1-C6
alkylene; L2 is selected from substituted or unsubstituted C1-C6 alkylene; and P3 is selected
from -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, and -C(O)0R2, wherein R2 is a member
selected from the group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl,
substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl;
substituted or unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl. Still other
particular embodiments are those in which the compound has formula (I), wherein P1 is
selected from -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; n is 0; m is 1; L1 is selected from
unsubstituted C1-C6 alkylene; L2 is selected from substituted or unsubstituted C1-C6 alkylene;
and P3 is selected from -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, and -C(O)OR2, wherein

WO 2006/045119 PCT/US2005/038282
25
R2 is a member selected from the group consisting of hydrogen, substituted or unsubstituted
C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted
heterocyclyl; substituted or unsubstituted aryl and substituted or unsubstituted aryl C1-C4
alkyl.
[0081] In one embodiment, the compound has the formula:

wherein R is a member selected from the group consisting of alkyl, aryl, alkylaryl,
cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl; and L2 is selected from the group consisting of phenylene
or methylenephenylene, heteroarylene, optionally substituted with from 1 to 2 substituents
each independently selected from the group consisting of halo and haloalkyl. Within this
embodiment, the compound has the formula:

[0082] In other embodiments, the compound has the formula:

wherein R2 is selected from the group consisting of substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl. Within this embodiment, the compound
preferably has the formula:

wherein R2 is substituted or unsubstituted aryl; and more preferably has the formula:

WO 2006/045119 PCT/US2005/038282
26

wherein R1 is a member selected from the group consisting of alkyl, aryl, alkylaryl,
cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl.
[0083] In one embodiment, compounds for use in this aspect of the invention are those
compounds provided in the Tables below, particularly Tables 5, 8-10 and 18.
[0084] In another group of embodiments the compounds of formula (I), as noted above,
contain an amino acid or dipeptide component which can be a dipeptide analog. The amino
acid residues, by themselves or as part of a dipeptide, are denoted by single-letter or three-
letter designations following conventional practices. The designations for gene-encoded
amino acids are as follows (amino acid, one letter symbol, three letter symbol): Alanine, A,
Ala; Arginine, R, Arg; Asparagine, N, Asn; Aspartic acid, D, Asp; Cysteine, C, Cys;
Glutamine, Q, G1n; Glutamic acid, E, Glu; Glycine, G, Gly; Histidine, H, His; Isoleucine, I,
lie; Leucine, L, Leu; Lysine, K, Lys; Methionine, M, Met; Phenylalanine, F, Phe; Proline, P,
Pro; Serme, S, Ser; Threonine, T, Thr; Tryptophan, W, Trp; Tyrosine, Y, Tyr; and Valine, V,
Val. Commonly encountered amino acids which are not gene-encoded may also be used in
the present invention. These amino acids and their abbreviations include ornithine (Orn); t-
butylglycine (t-BuG); phenylglycine (PhG); cyclohexylalanine (Cha); norleucine (Nle); 2-
naphthylalanine (2-Nal); 1-naphthylalanine (1-Nal); 2-thienylaniHne (2-Thi); N-
methylisoleucine (N-Melle), homoarginine (Har), Na-methylargmine (N-MeArg) and
sarcosine (Sar). All of the amino acids used in the present invention may be either the D- or
L-isomer.
[0085] In one embodiment, compounds of the invention are those in which L2 is selected
from the group consisting of substituted or unsubstituted C3-C6 cycloalkylene, substituted or
unsubstituted arylene, substituted or unsubstituted heteroarylene. In other embodiments, L2 is
preferably an amino acid or a dipeptide. Preferably, the dipeptide has a Tyr, His, Lys, Phe or
Trp residue directly attached to P2.

WO 2006/045119 PCT/US2005/038282
27
[0086] Other compounds for use in the present invention are those in which R1, P1 and L1
are selected from the groupings as described above for formula (I). Particular compounds of
formula (I) are those in which R1 is selected from C5-C12 cycloalkyl and phenyl. More
particularly, R1 is selected from C6-C10 cycloalkyl and phenyl. Other embodiments are those
embodiments in which R1 is cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, adamantly or
noradamantyl. P1 is preferably a urea (-NHC(O)NH-) or carbamate (-OC(O)NH-), more
preferably a urea. L1 is preferably a substituted or unsubstituted C2-C5 alkylene, more
preferably C2-C4 alkylene, still more preferably an ethylene or propylene linkage.
[0087] For those embodiments in which L2 is a single amino acid, L2 is preferably selected
from Ala, Arg, Asp, Cys, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Tip, Tyr and
Val. More preferably, L2 is selected from His, I1e, Lys, Phe, Trp and Tyr in which the amino
acid is linked to P2 in a manner to afford an amide linkage and terminal carboxylic acid
group. Of course, one of skill in the art will appreciate that these amino acids are meant to
refer to their corresponding methyl or ethyl esters, as well as their carboxamide derivatives
(e.g., terminal -C(O)NH2)- Most preferably, the compounds are those provided in Table 11.
[0088] For those embodiments in which L1 is a dipeptide, P2 is preferably attached to a Tyr,
His, Lys, Phe or Trp residue, with the remaining amino acid being selected from the gene-
encoded amino acids, their D-isomers or analogs thereof (e.g., hydroxy acids such as lactic
acid and the like). Still more prefereably, L2 is selected from TyrAla, TyrArg, TyrAsp,
TyrGly, Tyrlle, TyrLeu, TyrLys, TyrMet, TyrPhe, TyrPro, TyrSer, TyrThr, TyrTrp, TyrTyr
and TyrVal. More preferably, L2 is selected from TyrArg, TyrAsp, TyrMet, TyrPhe, TyrSer,
TyrTrp, TyrTyr and TyrVal. in which the Tyr amino acid is linked to P2 in a manner to afford
an amide linkage. As above, these dipeptides are also meant to refer to their corresponding
methyl or ethyl esters, as well as their carboxamide derivatives (e.g., terminal -C(O)NH2).
Most preferably, the compounds are those provided in Table 12.
Assays to Monitor Soluble Epoxide Hydrolase Activity:
[0089] Additionally, the present invention provides a variety of assays and associated
methods for monitoring soluble epoxide hydrolase activity, particularly the activity that has
been modulated by the administration of one or more of the compounds provided above.
[0090] In one group of embodiments, the invention provides methods for reducing the
formation of a biologically active diol produced by the action of a soluble epoxide hydrolase,

WO 2006/045119 PCT/US2005/038282
28
the method comprising contacting the soluble epoxide hydrolase with an amount of a
compound of formula (I) above, sufficient to inhibit the activity of the soluble epoxide
hydrolase and reduce the formation of the biologically active diol.
[0091] In another group of embodiments, the invention provides methods for stabilizing
biologically active epoxides in the presence of a soluble epoxide hydrolase, the method
comprising contacting the soluble epoxide hydrolase with an amount of a compound of
formula (I), sufficient to inhibit the activity of the soluble epoxide hydrolase and stabilize the
biologically active epoxide.
[0092] In each of these groups of embodiments, the methods can be carried out as part of
an in vitro assay or the methods can be carried out in vivo by monitoring blood liters of the
respective biologically active epoxide or diol.
[0093] Epoxides and diols of some fatty acids are biologically important chemical
mediators and are involved in several biological processes. The strongest biological data
support the action of oxylipins as chemical mediators between the vascular endothelium and
vascular smooth muscle. Accordingly, the epoxy lipids are anti-inflammatory and anti-
hypertensive. Additionally, the lipids are thought to be metabolized by beta-oxidation, as
well as by epoxide hydration. The soluble epoxide hydrolase is considered to be the major
enzyme involved in the hydrolytic metabolism of these oxylipins. The compounds of
formula (I) can inhibit the epoxide hydrolase and stabilize the epoxy lipids both in vitro and
in vivo. This activity results in a reduction of hypertension in four separate rodent models.
Moreover, the inhibitors show a reduction in renal inflammation associated with and
independent of the hypertensive models.
[0094] More particularly, the present invention provides methods for monitoring a variety
of lipids in both the arachidonate and linoleate cascade simultaneously in order to address the
biology of the system. A GLC-MS system or a LC-MS method can be used to monitor over
740 analytes in a highly quantitative fashion in a single injection. The analytes include the
regioisomers of the arachidonate epoxides (EETs), the diols (DHETs), as well as other P450
products including HETEs. Characteristic products of the cyclooxygenase, lipoxygenase, and
peroxidase pathways in both the arachidonate and linoleate series can also be monitored.
Such methods are particularly useful as being predictive of certain disease states. The
oxylipins can be monitored in mammals following the administration of inhibitors of epoxide

WO 2006/045119 PCT/US2005/038282
29
hydrolase. Generally, EH inhibitors increase epoxy lipid concentrations at the expense of
diol concentrations in body fluids and tissues.
[0095] Other compounds for use in this aspect of the invention are those inhibitors of
formula (I) in which the primary pharmacophore is separated from a tertiary pharmacophore
by a distance that approximates the distance between the terminal carboxylic acid and an
epoxide functional group in the natural substrate.
Methods of Treating Diseases Modulated by Soluble Epoxide Hvdrolases:
[0096] In another aspect, the present invention provides methods of treating diseases,
especially those modulated by soluble epoxide hydrolases (sEH). The methods generally
involve administering to a subject in need of such treatment an effective amount of a
compound having a formula (I) above. The dose, frequency and timing of such administering
will depend in large part on the selected therapeutic agent, the nature of the condition being
treated, the condition of the subject including age, weight and presence of other conditions or
disorders, the formulation being administered and the discretion of the attending physician.
Preferably, the compositions and compounds of the invention and the pharmaceutically
acceptable salts thereof are administered via oral, parenteral, subcutaneous, intramuscular,
intravenous or topical routes. Generally, the compounds are administered in dosages ranging
from about 2 mg up to about 2,000 mg per day, although variations will necessarily occur
depending, as noted above, on the disease target, the patient, and the route of administration.
Dosages are administered orally in the range of about 0.05 mg/kg to about 20 mg/kg, more
preferably in the range of about 0.05 mg/kg to about 2 mg/kg, most preferably in the range of
about 0.05 mg/kg to about 0.2 mg per kg of body weight per day. The dosage employed for
the topical administration will, of course, depend on the size of the area being treated.
[0097] It has previously been shown that inhibitors of soluble epoxide hydrolase ("sEH")
can reduce hypertension. See, e.g., U.S. Patent No. 6,351,506. Such inhibitors can be useful
in controlling the blood pressure of persons with undesirably high blood pressure, including
those who suffer from diabetes.
[0098] In some embodiments, compounds of formula (I) are administered to a subject in
need of treatment for hypertension, specifically renal, hepatic, or pulmonary hypertension;
inflammation, specifically renal inflammation, vascular inflammation, and lung

WO 2006/045119 PCT/US2005/038282
30
inflammation; adult respiratory distress syndrome; diabetic complications; end stage renal
disease; Raynaud syndrome and arthritis.
Methods for Inhibiting Progression of Kidney Deterioration (Nephropathv) and
Reducing Blood Pressnre:
[0099] In another aspect of the invention, the compounds of the invention can reduce
damage to the kidney, and especially damage to kidneys from diabetes, as measured by
albuminuria. The compounds of the invention can reduce kidney deterioration (nephropathy)
from diabetes even in individuals who do not have high blood pressure. The conditions of
therapeautic administration are as described above.
[0100] cis-Epoxyeicosantrienoic acids ("EETs") can be used in conjunction with the
compounds of the invention to further reduce kidney damage. EETs, which are epoxides of
arachidonic acid, are known to be effectors of blood pressure, regulators of inflammation,
and modulators of vascular permeability. Hydrolysis of the epoxides by sEH diminishes this
activity. Inhibition of sEH raises the level of EETs since the rate at which the EETs are
hydrolyzed into DHETs is reduced. Without wishing to be bound by theory, it is believed
that raising the level of EETs interferes with damage to kidney cells by the microvasculature
changes and other pathologic effects of diabetic hyperglycemia. Therefore, raising the EET
level in the kidney is believed to protect the kidney from progression from microalbummuria
to end stage renal disease.
[0101] EETs are well known in the art. EETs useful in the methods of the present
invention include 14,15-EET, 8,9-EET and 11,12-EET, and 5,6 EETs, in that order of
preference. Preferably, the EETs are administered as the methyl ester, which is more stable.
Persons of skill will recognize that the EETs are regioisomers, such as 8S,9R- and 14R,15S-
EET. 8,9-EET, 11,12-EET, and 14R,15S-EET, are commercially available from, for
example, Sigma-Aldrich (catalog nos. E5516, E5641, and E5766, respectively, Sigma-
Aldrich Corp., St. Louis, MO).
[0102] EETs produced by the endothelium have anti-hypertensive properties and the EETs
11,12-EET and 14,15-EET may be endothelium-derived hyperpolarizing factors (EDHFs).
Additionally, EETs such as 11,12-EET have profibrinolytic effects, anti-inflammatory actions
and inhibit smooth muscle cell proliferation and migration. In the context of the present

WO 2006/045119 PCT/US2005/038282
31
invention, these favorable properties are believed to protect the vasculature and organs during
renal and cardiovascular disease states.
[0103] It is now believed that sEH activity can be inhibited sufficiently to increase the
levels of EETs and thus augment the effects of administering sEH inhibitors by themselves.
This permits EETs to be used in conjunction with one or more sEH inhibitors to reduce
nephropathy in the methods of the invention. It further permits EETs to be used in
conjunction with one or more sEH inhibitors to reduce hypertension, or inflammation, or
both. Thus, medicaments of EETs can be made which can be administered in conjunction
with one or more sEH inhibitors, or a medicament containing one or more sEH inhibitors can
optionally contain one or more EETs.
[0104] The EETs can be administered concurrently with the sEH inhibitor, or following
administration of the sEH inhibitor. It is understood that, like all drugs, inhibitors have half
lives defined by the rate at which they are metabolized by or excreted from the body, and that
the inhibitor will have a period following administration during which it will be present in
amounts sufficient to be effective. If EETs are administered after the inhibitor is
administered, therefore, it is desirable that the EETs be administered during the period during
which the inhibitor will be present in amounts to be effective to delay hydrolysis of the EETs.
Typically, the EET or EETs will be administered within 48 hours of administering an sEH
inhibitor. Preferably, the EET or EETs are administered within 24 hours of the inhibitor, and
even more preferably within 12 hours. In increasing order of desirability, the EET or EETs
are administered within 10, 8, 6,4, 2, hours, 1 hour, or one half hour after administration of
the inhibitor. Most preferably, the EET or EETs are administered concurrently with the
inhibitor.
[0105] In some embodiments, the EETs, the compound of the invention, or both, are
provided in a material that permits them to be released over time to provide a longer duration
of action. Slow release coatings are well known in the pharmaceutical art; the choice of the
particular slow release coating is not critical to the practice of the present invention.
[0106] EETs are subject to degradation under acidic conditions. Thus, if the EETs are to be
administered orally, it is desirable that they are protected from degradation in the stomach.
Conveniently, EETs for oral administration may be coated to permit them to passage the
acidic environment of the stomach into the basic environment of the intestines. Such
coatings are well known in the art. For example, aspirin coated with so-called "enteric

WO 2006/045119 PCT/US2005/038282
32
coatings" is widely available commercially. Such enteric coatings may be used to protect
EETs during passage through the stomach. An exemplary coating is set forth in the
Examples.
[0107] While the anti-hypertensive effects of EETs have been recognized, EETs have not
been administered to treat hypertension because it was thought endogenous sEH would
hydro! yse the EETs too quickly for them to have any useful effect. Surprisingly, it was found
during the course of the studies underlying the present invention that exogenously
administered inhibitors of sEH succeeded in inhibiting sEH sufficiently that levels of EETs
could be further raised by the administration of exogenous EETs. These findings underlie the
co-administration of sEH inhibitors and of EETs described above with respect to inhibiting
the development and progression of nephropathy. This is an important improvement in
augmenting treatment. While levels of endogenous EETs are expected to rise with the
inhibition of sEH activity caused by the action of the sEH inhibitor, and therefore to result in
at least some improvement in symptoms or pathology, it may not be sufficient in all cases to
inhibit progression of kidney damage fully or to the extent intended. This is particularly true
where the diseases or other factors have reduced the endogenous concentrations of EETs
below those normally present in healthy individuals. Administration of exogenous EETs in
conjunction with a sEH inhibitor is therefore expected to be beneficial and to augment the
effects of the sEH inhibitor in reducing the progression of diabetic nephropathy.
[0108] The present invention can be used with regard to any and all forms of diabetes to the
extent that they are associated with progressive damage to the kidney or kidney function.
The chronic hyperglycemia of diabetes is associated with long-term damage, dysfunction,
and failure of various organs, especially the eyes, kidneys, nerves, heart, and blood vessels.
The long-term complications of diabetes include retinopathy with potential loss of vision;
nephropathy leading to renal failure; peripheral neuropathy with risk of foot ulcers,
amputation, and Charcot joints.
[0109] In addition, persons with metabolic syndrome are at high risk of progression to type
2 diabetes, and therefore at higher risk than average for diabetic nephropathy. It is therefore
desirable to monitor such individuals for microalbuminuria, and to administer a sEH inhibitor
and, optionally, one or more EETs, as an intervention to reduce the development of
nephropathy. The practitioner may wait until microalbuminuria is seen before beginning the
intervention. As noted above, a person can be diagnosed with metabolic syndrome without

WO 2006/045119 PCT/US2005/038282
33
having a blood pressure of 130/85 or higher. Both persons with blood pressure of 130/85 or
higher and persons with blood pressure below 130/85 can benefit from the administration of
sEH inhibitors and, optionally, of one or more EETs, to slow the progression of damage to
their kidneys. In some embodiments, the person has metabolic syndrome and blood pressure
below 130/85.
[0110] Dyslipidemia or disorders of lipid metabolism is another risk factor for heart
disease. Such disorders include an increased level of LDL cholesterol, a reduced level of
HDL cholesterol, and an increased level of triglycerides. An increased level of serum
cholesterol, and especially of LDL cholesterol, is associated with an increased risk of heart
disease. The kidneys are also damaged by such high levels. It is believed that high levels of
triglycerides are associated with kidney damage. In particular, levels of cholesterol over 200
mg/dL, and especially levels over 225 mg/dL, would suggest that sEH inhibitors and,
optionally, EETs, should be administered. Similarly, triglyceride levels of more than 215
mg/dL, and especially of 250 mg/dL or higher, would indicate that administration of sEH
inhibitors and, optionally, of EETs, would be desirable. The administration of compounds of
the present invention with or without the EETs, can reduce the need to administer statin drugs
(HMG-CoA reductase inhibitors) to the patients, or reduce the amount of the statins needed.
In some embodiments, candidates for the methods, uses and compositions of the invention
have triglyceride levels over 215 mg/dL and blood pressure below 130/85. In some
embodiments, the candidates have triglyceride levels over 250 mg/dL and blood pressure
below 130/85. In some embodiments, candidates for the methods, uses and compositions of
the invention have cholesterol levels over 200 mg/dL and blood pressure below 130/85. In
some embodiments, the candidates have cholesterol levels over 225 mg/dL and blood
pressure below 130/85.
Methods of Inhibiting the Proliferation of Vascular Smooth Muscle Cells:
[0111] In other embodiments, compounds of formula (I) inhibit proliferation of vascular
smooth muscle (VSM) cells without significant cell toxicity, (e.g. specific to VSM cells).
Because VSM cell proliferation is an integral process in the pathophysiology of
atherosclerosis, these compounds are suitable for slowing or inhibition atherosclerosis. These
compounds are useful to subjects at risk for atherosclerosis, such as individuals who have had

WO 2006/045119 PCT/US2005/038282
34
a heart attack or a test result showing decreased blood circulation to the heart. The conditions
of therapeautic administration are as described above.
[0112] The methods of the invention are particularly useful for patients who have had
percutaneous intervention, such as angioplasty to reopen a narrowed artery, to reduce or to
slow the narrowing of the reopened passage by restenosis. In some embodiments, the artery
is a coronary artery. The compounds of the invention can be placed on stents in polymeric
coatings to provide a controlled localized release to reduce restenosis. Polymer compositions
for implantable medical devices, such as stents, and methods for embedding agents in the
polymer for controlled release, are known in the art and taught, for example, in U.S. Patent
Nos. 6,335,029; 6,322,847; 6,299,604; 6,290,722; 6,287,285; and 5,637,113. In some
embodiments, the coating releases the inhibitor over a period of time, preferably over a
period of days, weeks, or months. The particular polymer or other coating chosen is not a
critical part of the present invention.
[0113] The methods of the invention are useful for slowing or inhibiting the stenosis or
restenosis of natural and synthetic vascular grafts. As noted above in connection with stents,
desirably, the synthetic vascular graft comprises a material which releases a compound of the
invention over time to slow or inhibit VSM proliferation and the consequent stenosis of the
graft. Hemodialysis grafts are a particular embodiment.
[0114] In addition to these uses, the methods of the invention can be used to slow or to"
inhibit stenosis or restenosis of blood vessels of persons who have had a heart attack, or
whose test results indicate that they are at risk of a heart attack.
[0115] In one group of embodiments, compounds of the invention are administered to
reduce proliferation of VSM cells in persons who do not have hypertension. In another group
of embodiments, compounds of the invention are used to reduce proliferation of VSM cells in
persons who are being treated for hypertension, but with an agent that is not an sEH inhibitor.
[0116] The compounds of the invention can be used to interfere with the proliferation of
cells which exhibit inappropriate cell cycle regulation, hi one important set of embodiments,
the cells are cells of a cancer. The proliferation of such cells can be slowed or inhibited by
contacting the cells with a compound of the invention. The determination of whether a
particular compound of the invention can slow or inhibit the proliferation of cells of any
particular type of cancer can be determined using assays routine in the art.

WO 2006/045119 PCT/US2005/038282
35
[0117] In addition to the use of the compounds of the invention, the levels of EETs can be
raised by adding EETs. VSM cells contacted with both an EET and a compound of the
invention exhibited slower proliferation than cells exposed to either the EET alone or to the a
compound of the invention alone. Accordingly, if desired, the slowing or inhibition of VSM
cells of a compound of the invention can be enhanced by adding an EET along with a
compound of the invention. In the case of stents or vascular grafts, for example, this can
conveniently be accomplished by embedding the EET in a coating along with a compound of
the invention so that both are released once the stent or graft is in position.
Methods of Inhibiting the Progression of Obstructive Pulmonary Disease, Interstitial
Lung Disease, or Asthma:
[0118] Chronic obstructive pulmonary disease, or COPD, encompasses two conditions,
emphysema and chronic bronchitis, which relate to damage caused to the lung by air
pollution, chronic exposure to chemicals, and tobacco smoke. Emphysema as a disease
relates to damage to the alveoli of the lung, which results in loss of the separation between
alveoli and a consequent reduction in the overall surface area available for gas exchange.
Chronic bronchitis relates to irritation of the bronchioles, resulting in excess production of
mucin, and the consequent blocking by mucin of the airways leading to the alveoli. While
persons with emphysema do not necessarily have chronic bronchitis or vice versa, it is
common for persons with one of the conditions to also have the other, as well as other lung
disorders.
[0119] Some of the damage to the lungs due to COPD, emphysema, chronic bronchitis, and
other obstructive lung disorders can be inhibited or reversed by administering inhibitors of
the enzyme known as soluble epoxide hydrolase, or "sEH". The effects of sEH inhibitors can
be increased by also administering EETs. The effect is at least additive over administering
the two agents separately, and may indeed be synergistic.
[0120] The studies reported herein show that EETs can be used in conjunction with sEH
inhibitors to reduce damage to the lungs by tobacco smoke or, by extension, by occupational
or environmental irritants. These findings indicate that the co-administration of sEH
inhibitors and of EETs can be used to inhibit or slow the development or progression of
COPD, emphysema, chronic bronchitis, or other chronic obstructive lung diseases which
cause irritation to the lungs.

WO 2006/045119 PCT/US2005/038282
36
[0121] Animal models of COPD and humans with COPD have elevated levels of
immunomodulatory lymphocytes and neutrophils. Neutrophils release agents that cause
tissue damage and, if not regulated, will over time have a destructive effect. Without wishing
to be bound by theory, it is believed that reducing levels of neutrophils reduces tissue damage
contributing to obstructive lung diseases such as COPD, emphysema, and chronic bronchitis.
Administration of sEH inhibitors to rats in an animal model of COPD resulted in a reduction
in the number of neutrophils found in the lungs. Administration of EETs in addition to the
sEH inhibitors also reduced neutrophil levels. The reduction in neutrophil levels in the
presence of sEH inhibitor and EETs was greater than in the presence of the sEH inhibitor
alone.
[0122] While levels of endogenous EETs are expected to rise with the inhibition of sEH
activity caused by the action of the sEH inhibitor, and therefore to result in at least some
improvement in symptoms or pathology, it may not be sufficient in all cases to inhibit
progression of COPD or other pulmonary diseases. This is particularly true where the
diseases or other factors have reduced the endogenous concentrations of EETs below those
normally present in healthy individuals. Administration of exogenous EETs in conjunction
with an sEH inhibitor is therefore expected to augment the effects of the sEH inhibitor in
inhibiting or reducing the progression of COPD or other pulmonary diseases.
[0123] In addition to inhibiting or reducing the progression of chronic obstructive airway
conditions, the invention also provides new ways of reducing the severity or progression of
chronic restrictive airway diseases. While obstructive airway diseases tend to result from the
destruction of the lung parenchyma, and especially of the alveoE, restrictive diseases tend to
arise from the deposition of excess collagen in the parenchyma. These restrictive diseases
are commonly referred to as "interstitial lung diseases", or "ILDs", and include conditions
such as idiopathic pulmonary fibrosis. The methods, compositions and uses of the invention
are useful for reducing the severity or progression of ILDs, such as idiopathic pulmonary
fibrosis. Macrophages play a significant role in stimulating interstitial cells, particularly
fibroblasts, to lay down collagen. Without wishing to be bound by theory, it is believed that
neutrophils are involved in activating macrophages, and that the reduction of neutrophil
levels found in the studies reported herein demonstrate that the methods and uses of the
invention will also be applicable to reducing the severity and progression of ELDs.

WO 2006/045119 PCT/US2005/038282
37
[0124] In some embodiments, the ILD is idiopathic pulmonary fibrosis. In other
embodiments, the ILD is one associated with an occupational or environmental exposure.
Exemplars of such ILDs, are asbestosis, silicosis, coal worker's pneumoconiosis, and
berylliosis. Further, occupational exposure to any of a number of inorganic dusts and organic
dusts is believed to be associated with mucus hypersecretion and respiratory disease,
including cement dust, coke oven emissions, mica, rock dusts, cotton dust, and grain dust (for
a more complete list of occupational dusts associated with these conditions, see Table 254-1
of Speizer, "Environmental Lung Diseases," Harrison's Principles of Internal Medicine, infra,
at pp. 1429-1436). In other embodiments, the ILD is sarcoidosis of the lungs. ILDs can also
result from radiation in medical treatment, particularly for breast cancer, and from connective
tissue or collagen diseases such as rheumatoid arthritis and systemic sclerosis. It is believed
that the methods, uses and compositions of the invention can be useful in each of these
interstitial lung diseases.
[0125] In another set of embodiments, the invention is used to reduce the severity or
progression of asthma. Asthma typically results in mucin hypersecretion, resulting in partial
airway obstruction. Additionally, irritation of the airway results in the release of mediators
which result in airway obstruction. While the lymphocytes and other immunomodulatory
cells recruited to the lungs in asthma may differ from those recruited as a result of COPD or
an ILD, it is expected that the invention will reduce the influx of immunomodulatory cells,
such as neutrophils and eosinophils, and ameliorate the extent of obstruction. Thus, it is
expected that the administration of sEH inhibitors, and the administration of sEH inhibitors in
combination with EETs, will be useful in reducing airway obstruction due to asthma.
[0126] In each of these diseases and conditions, it is believed that at least some of the
damage to the lungs is due to agents released by neutrophils which infiltrate into the lungs.
The presence of neutrophils in the airways is thus indicative of continuing damage from the
disease or condition, while a reduction in the number of neutrophils is indicative of reduced
damage or disease progression. Thus, a reduction in the number of neutrophils in the airways
in the presence of an agent is a marker that the agent is reducing damage due to the disease or
condition, and is slowing the further development of the disease or condition. The number of
neutrophils present in the lungs can be determined by, for example, bronchoalveolar lavage.
Prophvlatic and therapeutic methods to reduce stroke damage

WO 2006/045119 PCT/US2005/038282
38
[0127] Inhibitors of soluble epoxide hydrolase ("sEH") and EETs administered in
conjunction with inhibitors of sEH have been shown to reduce brain damage from strokes.
Based on these results, we expect that inhibitors of sEH taken prior to an ischernic stroke will
reduce the area of brain damage and will likely reduce the consequent degree of impairment.
The reduced area of damage should also be associated with a faster recovery from the effects
of the stroke.
[0128] While the pathophysiologies of different subtypes of stroke differ, they all cause
brain damage. Hemorrhagic stroke differs from ischemic stroke in that the damage is largely
due to compression of tissue as blood builds up in the confined space within the skull after a
blood vessel ruptures, whereas in ischemic stroke, the damage is largely due to loss of
oxygen supply to tissues downstream of the blockage of a blood vessel by a clot. Ischemic
strokes are divided into thrombotic strokes, in which a clot blocks a blood vessel in the brain,
and embolic strokes, in which a clot formed elsewhere in the body is carried through the
blood stream and blocks a vessel there. But, in both hemorrhagic stroke and ischemic stroke,
the damage is due to the death of brain cells. Based on the results observed in our studies,
however, we would expect at least some reduction in brain damage in all types of stroke and
in all subtypes.
[0129] A number of factors associated with an increased risk of stroke. Given the results of
the studies underlying the present invention, sEH inhibitors administered to persons with any
one or more of the following conditions or risk factors-.high blood pressure, tobacco use,
diabetes, carotid artery disease, peripheral artery disease, atrial fibrillation, transient ischemic
attacks (TIAs), blood disorders such as high red blood cell counts and sickle cell disease,
high blood cholesterol, obesity, alcohol use of more than one drink a day for women or two
drinks a day for men, use of cocaine, a family history of stroke, a previous stroke or heart
attack, or being elderly, will reduce the area of brain damaged of a stroke. With respect to
being elderly, the risk of stroke increases for every 10 years. Thus, as an individual reaches
60, 70, or 80, administration of sEH inhibitors has an increasingly larger potential benefit.
As noted in the next section, the administration of EETs in combination with one or more
sEH inhibitors can be beneficial in further reducing the brain damage.
[0130] In some uses and methods, the sEH inhibitors and, optionally, EETs, are
administered to persons who use tobacco, have carotid artery disease, have peripheral artery
disease, have atrial fibrillation, have had one or more transient ischemic attacks (TIAs), have
a blood disorder such as a high red blood cell count or sickle cell disease, have high blood
cholesterol, are obese, use alcohol in excess of one drink a day if a woman or two drinks a

WO 2006/045119 PCT/US2005/038282
39
day if a man, use cocaine, have a family history of stroke, have had a previous stroke or heart
attack and do not have high blood pressure or diabetes, or are 60, 70, or 80 years of age or
more and do not have hypertension or diabetes.
[0131] Clot dissolving agents, such as tissue plasminogen activator (tPA), have been shown
to reduce the extent of damage from ischemic strokes if administered in the hours shortly
after a stroke. tPA, for example, is approved by the FDA for use in the first three hours after
a stroke. Thus, at least some of the brain damage from a stoke is not instantaneous, but
occurs over a period of time or after a period of time has elapsed after the stroke. It is
therefore believed that administration of sEH inhibitors, optionally with EETs, can also
reduce brain damage if administered within 6 hours after a stroke has occurred, more
preferably within 5, 4, 3, or 2 hours after a stroke has occurred, with each successive shorter
interval being more preferable. Even more preferably, the inhibitor or inhibitors are
administered 2 hours or less or even 1 hour or less after the stroke, to maximize the reduction
in brain damage. Persons of skill are well aware of how to make a diagnosis of whether or
not a patient has had a stroke. Such determinations are typically made in hospital emergency
rooms, following standard differential diagnosis protocols and imaging procedures.
[0132] In some uses and methods, the sEH inhibitors and, optionally, EETs, are
administered to persons who have had a stroke within the last 6 hours who: use tobacco, have
carotid artery disease, have peripheral artery disease, have atrial fibrillation, have had one or
more transient ischemic attacks (TIAs), have a blood disorder such as a high red blood cell
count or sickle cell disease, have high blood cholesterol, are obese, use alcohol in excess of
one drink a day if a woman or two drinks a day if a man, use cocaine, have a family history
of stroke, have had a previous stroke or heart attack and do not have high blood pressure or
diabetes, or are 60, 70, or 80 years of age or more and do not have hypertension or diabetes.
[0133] The conditions of therapeautic administration for all of these indications are as
described above.
Combination Therapy
[0134] As noted above, the compounds of the present invention will, in some instances, be
used in combination with other therapeutic agents to bring about a desired effect. Selection
of additional agents will, in large part, depend on the desired target therapy (see, e.g., Turner,
N. etal. Prog. Drug Res. (1998) 51: 33-94; Haffner, S. Diabetes Care (1998) 21: 160-178;

WO 2006/045119 PCT/US2005/038282
40
and DeFronzo, R. et al. (ed's.), Diabetes Reviews (1997) Vol. 5 No. 4). A number of studies
have investigated the benefits of combination therapies with oral agents {see, e.g., Mahler, R.,
J. Clin. Endocrinol Metab. (1999) 84: 1165-71; United Kingdom Prospective Diabetes Study
Group: UKPDS 28, Diabetes Care (1998) 21: 87-92; Bardin, C. W.,(ed.), Current Therapy In
Endocrinology And Metabolism, 6th Edition (Mosby - Year Book, Inc., St. Louis, MO 1997);
Chiasson, J. et al., Ann. Intern. Med. (1994) 121: 928-935; Coniff, R. et al, Clin. Ther.
(1997) 19: 16-26; Coniff, R. et al., Am. J. Med. (1995) 98: 443-451; and Iwamoto, Y. et al.,
Diabet. Med. (1996) 13 365-370; Kwiterovich, P. Am. J. Cardiol (1998) 82(12A): 3U-17U).
Combination therapy includes administration of a single pharmaceutical dosage formulation
which contains a compound having the general structure of formula 1 and one or more
additional active agents, as well as administration of a compound of formula 1 and each
active agent in its own separate pharmaceutical dosage formulation. For example, a
compound of formula 1 and one or more angiotensin receptor blockers, angiotensin
converting enzyme inhibitors, calcium channel blockers, diuretics, alpha blockers, beta
blockers, centrally acting agents, vasopeptidase inhibitors, renin inhibitors, endothelin
receptor agonists, AGE crosslink breakers, sodium/potassium ATPase inhibitors, endothelin
receptor agonists, endothelin receptor antagonists, angiotensin vaccine, and the like; can be
administered to the human subject together in a single oral dosage composition, such as a
tablet or capsule, or each agent can be administered in separate oral dosage formulations.
Where separate dosage formulations are used, a compound of formula 1 and one or more
additional active agents can be administered at essentially the same time (i.e., concurrently),
or at separately staggered times (i.e., sequentially). Combination therapy is understood to
include all these regimens.
Compounds for Inhibiting Soluble Epoxide Hydrolases:
[0135] In addition to the methods provided above, the present invention provides in another
aspect, compounds that can inhibit the activity of soluble epoxide hydrolases. In particular,
the present invention provides compounds having a formula selected from formula (I) above.
Preferably, the compounds are other than 1 l-(3-cyclohexylureido)-undecanoic acid, 11-(3-
cyclohexylureido)-undecanoic acid methyl ester, 1 l-(3-cyclohexylureido)-undecanoic acid
amide, 12-(3-cyclohexylureido)-dodecanoic acid and 12-(3-adamantan-l-yl-ureido)-
dodecanoic acid.

WO 2006/045119 PCT/US2005/038282
41
[0136] In one embodiment, compounds are those compounds described above as for the
recited uses.
Methods of Preparation
[0137] The compounds of the present invention can be prepared by a variety of methods as
outlined generally in the schemes below.
Scheme 1 - Introduction of a secondary pharmacophore (ketone)
[0138] Scheme 1 illustrates general methods that can be used for preparation of compounds
of the invention having a secondary pharmacophore that is a ketone functional group. While
the scheme is provided for the synthesis of l-(3-chlorophenyl)-3-(4-oxodecyl)urea, one of
skill in the art will understand that a number of commercially available isocyanates could be
used in place of 3-chlorophenyl isocyanate, and that shorter or longer analogs of ethyl 4-
aminobutyric acid or hexylbromide could also be employed.
Scheme 1: Synthesis of l-(3-chlorophenyl)-3-(4-oxodecyl)urea (794).

Scheme 1: Synthesis of l-(3-chlorophenyI)-3-(4-oxodecyl)urea (794): (a) Benzophenone inline, CH2Cl2. rt; (b)
DIBAL, THF, -78°C; (c) Mg/I2, hexylbromide, THF, rt; (d) acetic anhydride, DMSO, rt; (e) 1N HCl/dioxane, rt;
(f) 3-chlorophenyl isocyanate, TEA, DMF, rt.
[0139] As shown in Scheme 1, ethyl 4-aminobutyrate hydrochloride (available from
Aldrich Chemical Co., Milwaukee, Wisconsin, USA) is combined with benzophenone irnine
at room temperature to provide intermediate (i). DIBAL reduction of the ester group
provides an unisolated aldehyde moiety that is then reacted with a suitable Grignard reagent
(prepared in situ) to provide intermediate alcohol (ii). Oxidation of the alcohol moiety to a
ketone provides (iii) which can then be deprotected to form the amino-ketone (iv). Reaction
of (iv) with a suitable isocyanate provides the target compound (794). Substitution of

WO 2006/045119 PCT/US2005/038282
42
3-chlorophenyl isocyanate with, for example, adamantyl isocyanate or cyclohexyl isocyanate
(also available from Aldrich Chemical Co.) provides other compounds of the invention.

Scheme 2: Syntheses of l-(aryl or alkyl)-3-(3-alkylated proply)ureas: (a) aryl or alkyl isocyanate, DMF, rt; (b)
bromopentane, K2CO3, Nal, acetonitrile, reflux; (c) di-t-butyl dicarbonate, dioxane, 50°C; (d) pentylamine,
isobutyl chloroformate, NMM, DMF, rt; (e) 4M hydrochloric acid, dioxane; (f) 3-chlorophenyi isocyanate,
TEA, DMF, rt.
[0140] As shown in Scheme 2, a variety of compounds having a secondary pharmacophore
that is either an ester or amide functional group can be prepared. Beginning with 4-
aminobutyric acid, treatment with a suitable cycloalkyl or aryl isocyanate provides the urea
intermediates shown as (v), wherein R is 3-chlorophenyl, cyclohexyl or 1-adamantyl. Of
course other suitable isocyanates can also be employed to provide desired urea intermediates.
Esterification via alkylation of the carboxylic acid present in (v) with, for example, pentyl
bromide provides the target compounds 767, 772 and 789. A variety of suitable alkyl halides
can be used to prepare other compounds of the invention. The second path illustrated in
Scheme 2 can be used to prepare compounds such as 768, as well as those compounds having
a primary pharmacophore that is a carbamate. Accordingly, treatment of 4-aminobutyric acid
with di-t-butyl dicarbonate provides the t-butyl carbamate acid (vi) that is converted to a
desired amide (vii) using pentylamine, for example, in a mild procedure employing isobutyl
chloroformate, and N-methyl morpholine (NMM). Removal of the carbamate protecting
group (as it is used in this instance) followed by formation of a urea with a suitable
isocyanate (shown here as 3-chlorophenyl isocyanate) provides the target compounds (e.g.,
768).

WO 2006/045119 PCT/US2005/038282
43

Scheme 3: Syntheses of l-(3-chlorophenyl)-3-(2-alkylated ethyl)ureas: (a) 3-chlorophenyl isocyanate, DMF, rt;
(b) heptanoic anhydride (761), chlorofonnic acid pentyl ester (760), or pentyl isocyanate (762), TEA, DMF, rt;
(c) di-t-butyl dicarbonate, dioxane, rt; (d) heptanoic anhydride (765), chloroformic acid pentyl ester (777), or
pentyl isocyanate (766), DMF, rt; (e) 4M HC1, dioxane; (f) 3-chlorophenyl isocyanate, TEA, DMF, rt.
[0141] Scheme 3 illustrates a variety of methods for introducing secondary
phannacophores that are esters, amide, ureas, carbonates and carbamates, from readily
accessible starting materials. In A, ethanolamine is treated with a suitable isocyanate to
introduce a primary pharmacophore that is a urea and form intermediate (viii). Treatment of
(viii) with an anhydride, a chloro formic acid ester or an isocyanate provides compounds such
as 761,760 and 762, respectively. Similar methodology in employed in B, with the addition
of protection/deprotection steps. Accordingly, ethylenediamine is monoprotected as a r-butyl
carbamate. The free amine is then converted to a secondary pharmacophore that is an amide,
carbamate or urea using reactants and conditions similar to those employed in "A" to provide
intermediates (x). Deprotection of (x) and reaction with a suitable isocyanate provides the
target compounds 765, 777 and 766. Again, use of isocyanates other than 3-chlorophenyl
isocyanate leads to other compounds of the invention, while substitution of certain reactants
used, for example, in the conversion of (ix) to (x) can provide still other compounds of the
invention.

WO 2006/045119 PCT/US2005/038282
44

Scheme 4: Syntheses of l-(l-adamantyl)-3-(ll-alkylated undecyl)ureas: (a) adamantyl isocyanate, chloroform,
reflux; (b) alkyl or aryl halide, K2CO3, Nal, acetonitrile, reflux; (c) alcohol or amine, isobutyl chloroformate,
TEA, DMF, rt; (d) t-butanol, EDCI, DMAP, metirylene chloride, rt.
[0142] Scheme 4 illustrates pathways for the introduction of a tertiary pharmacophore that
is an ester or an amide functional group. In each case, a carboxylic acid group is converted to
the desired ester or amide. As shown in Scheme 4,12-aminododecanoic acid (Aldrich
Chemical Co.) is converted to urea (687) upon treatment with adamantyl isocyanate. One of
skill in the art will appreciate that a variety of alkyl, aryl and cycloalkyl isocyanates can be
similarly employed to form other ureas as the primary pharmacophore. Similarly,
11-aminoundecanoic acid or another long chain amino fatty acid could be used in place of
12-aminododecanoic acid. The carboxylic acid moiety can then be esterified or converted to
an amide moiety following standard procedures to provide, for example, 780-785, 788 and
800-804 (as esters) and 786, 787, 792 and 793 (as esters and amides).
[0143] Scheme 5 illustrates pathways for the synthesis of cis- or trans-phenoxy or
benzyloxy-cyclohexyl compounds. In each case, trans-4-aminocyclohexanol hydrochloride is
converted to the desired isomer of the phenoxy or benzyloxy derivative. The alcohol moiety
can be alkylated with an appropriately substituted benzyl halide following standard
procedures to provide, the corresponding benzyl ether. Likewise, the alcohol moiety can be
alkylated with an appropriately substituted phenol in the presence of triphenylphosphine
following standard procedures to provide, the corresponding phenyl ether. As shown in
Scheme 5, each isomer can be converted to the corresponding urea upon treatment with an

WO 2006/045119 PCT/US2005/038282
45
appropriately substituted isocyanate, e.g. adamantyl isocyanate. One of skill in the art will
appreciate that a variety of alkyl, aryl and cycloalkyl isocyanates can be similarly employed
to form other ureas as the primary pharmacophore.

Scheme 5: Syntheses of cis and trans-phenoxy-and benzyloxy-cyclohexyl compounds.
Combinatorial library synthesis
[0144] A 192-member urea library has been constructed by a known two-step solid-phase
synthesis (Tetrahedron Letters 2003,44, 6099-6102), which employs a PS-Indole-CHO resin.
As shown in Scheme 6, four amines and 48 isocyanates have been used as building blocks to
find the optimal left side of urea for sEH inhibitors. Resin-bound secondary amines can be
obtained by a reductive amination with amines using sodium cyanoborohydride in the
presence of triethylorthoformate. The reaction with isocyanates gives the desired resin-bound
ureas, which can be cleaved from the resin by very mild acidic condition by using 1% TFA in
dichloromethane at room temperature. The 192 urea compounds were prepared in 20-50%
overall yields from the indole aldehyde resin. The identification and purity of the library
could be determined by LC-MS. Purities were above 80%. Compounds with purites below
this level were removed from the library.

WO 2006/045119 PCT/US2005/038282
46

[0145] As the polyether compounds of the invention increase the ease of formulation, oral
availability and serum half life of the compounds, another aspect of the present invention is to
provide a method of increasing ease of formulation, oral availability, or serum half-life of a
compound comprising covalently attaching a polyether substituent to a compound.
[0146] The following examples are provided to illustrate the invention and are not intended
to limit any aspect of the invention as set forth above or in the claims below.
EXAMPLES
[0147] All melting points were determined with a Thomas-Hoover apparatus (A.H. Thomas
Co.) and are uncorrected. Mass spectra were measured by LC-MS (Waters 2790). 'H-NMR
spectra were recorded on QE-300 spectrometer, using tetramethylsilane as an internal
standard. Signal multiplicities are represented as signlet (s), doublet (d), double doublet (dd),
triplet (t), quartet (q), quintet (quint), multiplet (m), broad (br) and broad singlet (brs).
Synthetic methods are described for representative compounds.
[0148] Lower case bolded Roman numerals in the examples below refer to the
corresponding intermediates in Schemes 1-4 above. Compounds numbers are also used as
provided in the Schemes as well as in the Tables below.

WO 2006/045119 PCT/US2005/038282
47
Example 1
Synthesis of 1-(3-chlorophenyl)-3-(4-oxodecyl)urea (794)
[0149] 1.00g (5.52 mmol) of benzophenone imine, 0.94 g (5.52 mmol) of ethyl 4-
aminobutyrate hydrochloride, and 20 mL of methylene chloride were stirred at room
temperature for 24 hr. The reaction mixture was filtered to remove NH4CI and evaporated to
dryness. The benzophenone Schiff base of ethyl 4-aminobutyrate (i) was extracted with ether
(20 mL), and the ether solution was washed with water (20 mL), dried over sodium sulfate
(Na2SO4), and concentrated. The residue was purified by column chromatography on silica
gel eluting with hexane and ethyl acetate (5:1) to give i (1.00 g, 61%) as an oil. To the
solution of the benzophenone Schiff base (i) in 20 mL of tetrahydrofuran (THF) was added
3.7 mL of 1M diisobutylaluminium hydride (DIBAL) solution in pentane (3.73 mmol) at
-78°C under nitrogen, and the reaction was stirred for 2 hr at the temperature. To 0.10 g of
magnesium turning (4.07 mmol) and I2 (catalytic amount) in THF (10 mL) was added 0.48
mL of hexylbromide (3.39 mmol) at room temperature under nitrogen. After stirring for 1 hr,
this reaction solution was added dropwise to the above reaction mixture at -78°C, and the
solution was allowed to warm to room temperature with stirring. After stirring for 5 hr at
room temperature, 10 mL of NaHCO3 aqueous solution was added to the reaction, then the
alkylated alcohol (ii) was extracted with ether (20 mL), and the ether solution was washed
with water (20 mL), dried over Na2SO4, and concentrated to give 0.26 g (60 %) of the alcohol
product (ii).
[0150] Acetic anhydride (2mL) was added to a solution of ii (0.77 mmol) in 5 mL of
dimethyl sulfoxide (DMSO). The mixture was allowed to stand at room temperature for 12
hr. and concentrated. The residue was extracted with ether (20 mL), and the ether was washed
with water (20 mL), dried over Na2SO4, and evaporated to provide 0.26 g (100 %) of the
ketone compound (iii). To a solution of iii in dioxane (5 mL) was added 1mL of 1N HC1 in
dioxane at room temperature. The reaction mixture was stirred for 2 hr and concentrated to
give keto amine hydrochloride (iv). Then iv was dissolved in 5 mL of dimethylformamide
(DMF) and treated with triethylamine (TEA, 0.27 mL, 1.95 mmol) and a solution of 3-
chlorophenyl isocyanate (0.10 mL, 0.78 mmol) in DMF (3 mL) at room temperature. After
stirring for 5 hr, the product was extracted with ether (30 mL), and the ether was washed with
water (30 mL), dried over Na2SO4, and evaporated to dryness. The residue was purified by

WO 2006/045119 PCT/US2005/038282
48
column chromatography on silica gel eluting hexane and ethyl acetate (3:1) to afford 75 mg
(30%) of 794. δ(CDC13): 0.88 (3H, t, /= 6.9 Hz), 1.21-1.29 (6H, m), 1.53-1.58 (2H, m), 1.81
(2H, quint, J = 6.9 Hz), 2.43 (2H, t, J=6.9 Hz), 2.49 (2H, t, J = 6.9 Hz), 3.23 (2H, t, /= 6.9
Hz), 5.10 (1H, s), 6.93 (1H, s), 6.98-7.02 (1H, m), 7.10-7.23 (2H, m), 7.49 (1H, s), [M + H]+
325.21
Example 2
Synthesis ofl-(3-chlorophenyl)-3-(3-pentoxycarbonylpropyl)urea (767)
[0151] To a suspension of 4-aminobutyric acid (1.41 g, 13.7 mol) in DMF (25 mL) was
added 3-chlorophenyl isocyanate (0.70 g, 4.56 mmol; cyclohexyl isocyanate for 772 and 1-
adamantyl isocyanate for 789) at room temperature. The reaction mixture was stirred for 24
hr. Then ethyl acetate (30 mL) and 1N HC1 aqueous solution (30 mL) were added into the
reaction, and the ethyl acetate layer dissolving the acid product was collected. The product
was extracted with ethyl acetate (20 mL) two more times from the aqueous layer. The
combined organic solution was dried over Na2SO4, and evaporated. The residue was purified
using column chromatography on silica gel eluting hexane and ethyl acetate (1:1) to give 0.88
g (75%) of urea acid (v). A mixture of v (0.50 g, 1.95 mmol), potassium carbonate (K2CO3,
0.54 g, 3.90 mmol), bromopentane (0.37 mL, 2.92 mmol), and sodium iodide (60 mg, 0.39
mmol) in DMF (20 mL) was stirred at room temperature for 20 hr. Then the product was
extracted with ether (20 mL), and the ether was washed with 1N NaOH aqueous solution (20
mL) and brine (20 mL), dried over Na2SO4, and evaporated to afford 0.59 g (92%) of 767.
δ(CDC13): 0.90 (3H, t, J= 6.9 Hz), 1.26-1.34 (4H, m), 1.62-1.65 (2H, m), 1.88 (2H, quint, J=
6.9 Hz), 2.41 (2H, t, J= 6.9 Hz), 3.30 (2H, t, J= 6.9 Hz), 4.08 (2H, t, J= 6.9 Hz), 4.96 (1H,
s), 6.62 (1H, s), 7.01-7.04 (1H, m), 7.18-7.22 (2H, m), 7.47 (1H, s), [M + H]+ 326.90
The following compounds were prepared in a similar manner:
l-Cyclohexyl-3-(3-pentoxycarbonylpropyl)urea (772)
[0152] δ(CDC13): 0.89 (3H, t, .7=6.9 Hz), 1.04-1.21 (2H, m), 1.29-1.43 (4H,m), 1.58-1.74
(6H, m), 1.82 (2H, quint, J= 6.9 Hz), 2.37 (2H, t, J= 6.9 Hz), 3.17-3.24 (2H, m), 3.46-3.48
(1H, m), 4.07 (2H, t, J= 6.9 Hz), 4.29 (1H, s), 4.47 (1H, s), [M + H]+ 299.24
1-(1 -Adamantyl)-3-(3-pentoxycarbonylpropyl)urea (789)

WO 2006/045119 PCT/US2005/038282
49
[0153] δ(CDC13): 0.92 (3H, t, j= 6.9 Hz),1.29-1.43 (4H, m), 1.64-1.69 (m, 10H), 1.83
(2H, quint, /= 6.9 Hz), 1.94-1.98 (6H, m), 2.06-2.09 (3H, m), 2.37 (2H, t, J= 6.9 Hz), 3.20
(2H, t, J= 6.9 Hz), 4.06-4.14 (3H, m), 4.30 (1H, s), [M + H]+ 251.26.
Example 3
Synthesis of l-(3-chlorophenyl)-3-(3-pentylaminocarbonylpropyl)urea (768)
[0154] To a suspension of 4-aminobutyric acid (2.84 g, 27.5 mmol) in DMF (30 mL) was
added TEA (3.86 mL, 27.5 mmol). To this mixture, di-t-butyl dicarbonate (2.00 g, 9.17
mmol) was added with stirring. The reaction mixture was heated to 50°C for 12 hr, and then
stirred with ice-cold dilute hydrochloric acid (15 mL) for 10 min. The t-butoxycarbonylated
amino acid (vi) was immediately extracted with ether (2 X 30 mL). The organic extract was
dried over Na2SO4 and evaporated to give 1.00 g (54%) of vi as an oil.
[0155] A solution of vi and 4-methyl morpholine (NMM, 0.54 mL, 4.92 mmol) in DMF
(10 mL) was treated at room temperature with isobutyl chloroformate (0.64 mL, 4.92 mmol).
After 30 min, pentylamine (0.57 mL, 4.92 mmol) was added. The reaction mixture was
stirred for 12 hr. The solvent was evaporated, and the residue was partitioned between ethyl
acetate (25 mL) and water (25 mL). The ethyl acetate layer was washed with 5% NaHCO3
(10 mL) and brine (20 mL) and dried over Na2SO4, and evaporated. The residue was
chromatographed on silica gel eluting hexane and ethyl acetate (2:1) to give 0.33 g (33%) of
t-butoxycarbonylated amino amide (vii). To a solution of vii in dioxane (10 mL) was treated
with 4M hydrochloric acid (2mL) in dioxane, and the mixture was stirred for lhr at room
temperature. Then the solvent was evaporated to dryness, and the residual solid was
dissolved in DMF (10 mL) and treated with TEA (0.51 mL, 3.63 mmol) and 3-chlorophenyl
isocyanate (0.15 mL, 1.21 mmol) at room temperature. After stirring for 5 hr, the product
was extracted with ether (30 mL), and the ether was washed with water (30 mL), dried over
Na2SO4, and evaporated to dryness. The residue was purified by column chromatography on
silica gel eluting hexane and ethyl acetate (3:1) to afford 0.39 g (100%) of 768. 5(CDC13):
0.89 (t, 3H, J= 6.9 Hz), 1.26-1.28 (4H, m), 1.46-1.50 (2H, m), 1.86 (2H, quint, J= 6.9 Hz),
2.30 (t, 2H, J= 6.9 Hz), 3.23 (t, 2H, J= 6.9 Hz), 3.30 (t, 2H, J= 6.9 Hz), 5.87 (1H, s), 6.06
(1H} s), 6.93-6.97 (1H, m), 7.12-7.23 (2H, m), 7.49 (1H, m),7.73 (1H, s), [M + H]+ 326.16.

WO 2006/045119 PCT/US2005/038282
50
Example 4
Synthesis of l-(3-chlorophenyl)-3-(2-hexylcarbonyloxyethyl)urea (761)
[0156] To a solution of 2-aminoethanol (2.98 g, 48.8 mmol) in DMF (30 mL) was added 3-
chlorophenol isocyanate (2.50 g, 16.3 mmol) at 0°C. The reaction mixture was stirred for 5
hr at room temperature. The solvent was evaporated, and the residue was partitioned between
ether (30 mL) and 1N hydrochloric acid (20 mL), and the ether layer was washed with brine,
dried over Na2SO4 and evaporated. The residue was purified by column chromatography on
silica gel eluting hexane and ethyl acetate (1:1) to provide 1.49 g (40%) of urea alcohol (viii)
as a white solid.
[0157] To a solution of viii (1.00g, 4.60 mmol) and TEA (0.97 mL, 6.90 mmol) in DMF
(15 mL) was added a solution of heptanoic anhydride (2.23 g, 9.20 mmol) in DMF (5 mL) at
room temperature. The reaction was stirred for 12 hr, and the solvent was evaporated. The
residue was partitioned between ether (30 mL) and cold 1N hydrochloric acid (20 mL). The
ether layer was washed with brine, dried over Na2SO4, and evaporated. The residual solid
was purified using silica gel column chromatography (hexane : ethyl acetate = 3:1) to afford
1.05 g (70%) of 761. δ(CDC13): 0.87 (t, 3H, J= 6.9 Hz), 1.20-1.29 (6H, m), 1.60-1.62 (2H,
m), 2.22-2.29 (2H, m), 3.50-3.55 (2H, m), 4.09-4.20 (2H, m), 5.32 (1H, s), 7.01-7.06 (2H,
m), 7.16-7.22 (2H, m), 7.40 (1H, s), [M + H]+ 327.15
[0158] Compounds 760 and 762 were prepared in the same manner as that used for
compound 761 from chloroformic acid pentyl ester and pentyl isocyanate in place of
heptanoic anhydride, respectively.
1 -(3-chlorophenyl)-3-(2-pentoxycarbonyloxyethyl)urea (760)
[0159] δ(CDC13): 0.91 (t, 3H, J= 6.9 Hz), 1.25-1.36 (4H, m), 1.63-1.67 (2H, m), 3.55-3.60
(2H, m), 4.14 (3H, t, J= 6.9 Hz), 4.25-4.28 (2H, m), 5.11 (1H, s), 6.50 (1H, s), 7.02-7.05
(1H, m), 7.19-7.23 (2H, m), 7.42 (1H, s), [M + H]+ 329.09
l-(3-chlorophenyl)-3-(2-pentylaminocarbonyloxyethyl)urea(762)
[0160] 1δ(CDC13): 0.87 (3H, t, J= 6.9 Hz), 1.30-1.33 (4H, m), 1.46-1.50 (2H, m), .3.12-
3.19 (2H, m), 3.50-3.52 (2H, m), 4.17-4.20 (2H, m), 4.83 (1H, s), 5.47 (1H, s), 6.96 (1H, s),
6.98-7.02 (lH,,m), 7.18-7.21 (2H, m), 7.44 (1H, s), [M + H]+ 328.20.

WO 2006/045119 PCT/US2005/038282
51
Example 5
Synthesis of l-(S-chlorophenyl)-3-(2-hexylcarbonylaminoeihyl)urea (765)
[0161] A solution of di-t-butyl dicarbonate (0.50 g, 2.29 mmol) in dioxane (20 mL) was
added over a period of 1 hr to a solution of 1,2-diaminoethane (1.10 g, 18.3 mmol) in dioxane
(20 mL). The mixture was allowed to stir for 22 hr and the solvent was evaporated to
dryness. Water (30 mL) was added to the residue and the insoluble bis-substituted product
was removed by filtration. The filtrate was extracted with methylene chloride (3 X 30 mL)
and the methylene chloride evaporated to yield ix as an oil (0.35 g, 95%).
[0162] A solution of heptanoic anhydride (0.91 g, 3.75 mmol; chloroformic acid pentyl
ester for 777 and pentyl isocyanate for 766) and ix (0.50 g, 3.13 mmol) in DMF (20 mL) was
stirred for 2 hr at room temperature. Then the solvent was evaporated. The residue was
partitioned between ether (30 mL) and water (30 mL). The ether layer was dried over
Na2SO4 and evaporated. The residue was purified by using column chromatography on silica
gel eluting hexane and ethyl acetate (1:1) to get 0.57 g (67%) of alkylated N-t-
butoxycarbonyl amine (x).
[0163] To a solution of x in dioxane (10 mL) was treated with 4M hydrochloric acid (2mL)
in dioxane, and the mixture was stirred for 1hr at room temperature. Then the solvent was
evaporated to dryness, and the residual solid was dissolved in DMF (10 mL) and treated with
TEA (0.58 mL, 4.19 mmol) and 3-chlorophenyl isocyanate (0.32 g, 2.10 mmol) at room
temperature. After stirring for 5 hr, the product was extracted with ether (30 mL), and the
ether was washed with water (30 mL), dried over Na2SO4, and evaporated to dryness. The
residue was purified by column chromatography on silica gel eluting hexane and ethyl acetate
(1:1) to afford 0.68 g (100%) of 765. δ(CDC13): 0.84 (t, 3H, J= 6.9 Hz), 1.16-1.25 (6H, m),
1.55-5.61 (2H, m), 2.21-2.24 (2H, m), 3.31-3.40 (4H, m), 6.27 (1H, s), 6.90-6.95 (2H, m),
7.18-7.20 (2H, m), 7.56 (1H, s), 8.07 (IB, s), [M + H]+ 326.25
The following compounds were prepared in a similar manner:
l-(3-chlorophenyl)-3-(2-pentoxycarbonylaminoethyl)urea (777)
[0164] δ(CDC13): 0.88 (3H, t, J= 6.9 Hz), 1.28-1.32 (4H, m), 1.44-1.49 (2H, m), 3.23-3.33
(4H, m), 3.95-3.97 (2H, m), 6.01 (1H, s), 6.34 (1H, s), 6.87-6.91 (1H, m), 7.18-7.26 (2H, m),
7.78 (1H, s), 8.21 (1H, s), [M + H]+ 328.22

WO 2006/045119 PCT/US2005/038282
52
l-(3-chlorophenyl)-3-(2-pentylaminocarbonylamiiioethyl)urea (766)
[0165] δ(Acetone): 0.87 (3H, t, J= 6.9 Hz), 1.27-1.30 (4H, m), 2.04-2.06 (2H, m), 3.02-
3.05 (2H, m), 3.20-3.22 (2H, m), 5.74 (2H, s), 6.22 (1H, s), 7.23-7.29 (2H, m), 7.82-7.87 (2H,
m), 8.67 (1H, s), [M + H]+ 327.10.
Example 6
Synthesis of l-(l-adamantyl)-3-(12-dodecanoic acid)urea (687)
[0166] A mixture of 1-adamantyl isocyanate (1.30 g, 7.34 mmol) and 12-aminododecanoic
acid (1.46 g, 6.77 mmol) in chloroform (30 mL) was refluxed for 10 hr. The solvent was
removed by evaporation, and the residue was washed with ethyl acetate (20 mL) to provide
2.66 g (100%) of urea acid product as a white solid. δ(CDC13): 1.20-1.36 (16H, m), 1.42-1.48
(2H, m), 1.57-1.65 (6H, m), 1.82-1.90 (6H, m), 1.94-1.98 (3H, m), 2.18 (2H, t, J= 6.9 Hz),
2.86-2.92 (2H, m), 3.45 (1H, bs), 5.43 (1H, s), 5.587 (1H, t, /= 5.4 Hz), [M + H]+ 393.28,
mp 140°C.
Example 7
Synthesis of' l-(l-adamantyl)-3-(ll-methoxycarbonylundecyl)urea (780)
[0167] To a mixture of compound 687 (0.15 g, 0.38 mmol), K2CO3 (64 mg, 0.46 mmol),
and iodomethane (54 mg, 0.38 mmol) in acetonitrile (20 mL) was refluxed for 10 hr. Then
the reaction mixture was filtered, and the filtrate was washed with brine (20 mL), dried over
Na2SO4, and evaporated. The residue was purified using column chromatography on silica
gel eluting hexane and ethyl acetate (3:1) to afford 0.14 g (92%) of 780 as a white solid.
δ(CDCb): 1.19-1.34 (12H, m), 1.41-1.48 (2H, m), 1.58-1.62 (4H, m), 1.63-1.75 (6H, m),
1.93-2.00 (6H, m), 2.04-2.07 (3H, m), 2.30 (2H, t, /= 6.9 Hz), 3.06-3.12 (2H, m), 3.67 (3H,
s), 4.00 (1H, s), 4.06 (1H, s), [M + H]+ 407.22, mp 75°C
[0168] Compounds 780,784, 783, 781, 788, 800, 785, 801, 802, 803, 804, and 782 were
prepared in the same manner using corresponding halides in a range of 30-95% yield.
l-(l-Adamantyl)-3-(ll-ethoxycarbonylundecyl)urea (784)
[0169] δ(CDC13): 1.21-1.38 (12H, m), 1.42-1.68 (15H, m), 1.96 (6H, bs), 2.06 (3H, m),
2.30 (2H, t, J= 6.9 Hz), 3.06-3.12 (2H, m), 3.97-4.01 (2H, bs), 4.12 (2H, q), [M + H]+
421.46, mp82°C

WO 2006/045119 PCT/US2005/038282
53
l-(l-Adamantyl)-3-(ll-propoxycarbonylundecyl)urea (783)
[0170] δ(CDC13): 0.94 (3H, t, J= 6.9 Hz ), 1.19-1.34 (12H, m), 1.41-1.48 (2H, m), 1.58-
1.62 (4H, m), 1.63-1.75 (8H, m), 1.93-2.00 (6H, m), 2.04-2.07 (3H, m), 2.30 (2H, t, /= 6.9
Hz), 3.06-3.12 (2H, m), 3.95-4.05 (4H, m), [M + H]+ 435.52, mp 86°C
1 -(] -Adamantyl)-3-(l 1 -allyloxycarbonylundecyl)urea (781)
[0171] δ(CDC13): 1.19-1.34 (12H, m), 1.41-1.48 (2H, m), 1.58-1.73 (13H, m), 1.93-2.00
(6H, m), 2.04-2.07 (3H, m), 2.33 (2H, t, J= 6.9 Hz), 3.06-3.12 (2H, m), 3.99 (1H, s), 4.04
(1H, s), 4.57-4.59 (2H, m), [M + H]+ 433.43, mp 81°C
l-(l-Adamantyl)-3-(l 1 -propagyloxycarbonylundecyl)urea (788)
[0172] δ(CDC13): 1.24-1.31 (12H, m), 1.44-1.46 (2H, m), 1.58-1.67 (11H, m), 1.94-1.98
(6H, m). 2.05-2.07 (3H, m), 2.35 (2H, t, J= 6.9 Hz), 3.05-3.12 (2H, m), 3.99 (1H, s), 4.04
(1H, s), 4.67 (2H, s), [M + H]+ 431.67, mp 79°C
l-(l-Adamantyl)-3-(ll-butoxycarbonylundecyl)urea (800)
[0173] δ(CDC13): 0.95 (3H, t, J= 6.9 Hz), 1.23-1.35 (12H, m), 1.44-1.52 (4H, m), 1.57-
1.61 (4H, m), 1.66-1.69 (6H, m), 1.96-2.00 (8H, m), 2.07-2.09 (3H, m), 2.30 (2H, t, /= 6.9
Hz), 3.09-3.13 (2H, m), 4.02-4.10 (4H, m), [M + H]+ 449.34
l-(l-Adamantyl)-3-(ll-iso-propoxycarbonylundecyl)urea (785)
[0174] δ(CDC13): 1.19-1.26 (18H, m), 1.41-1.48 (2H, m), 1.58-1.62 (4H, m), 1.63-1.75
(6H, m), 1.94-2.00 (6H, m), 2.03-2.07 (3H, m), 2.30 (2H, t, J= 6.9 Hz), 3.06-3.12 (2H, m),
3.67 (3H, s), 4.00 (1H, s), 4.06 (1H, s), 4.94-5.04 (1H, m), [M + H]+ 435.33, mp 90°C
1-(1 -Adamantyl)-3-(l 1 -sec-butoxycarbonylundecyl)urea (802)
[0175] δ(CDC13): 0.89 (3H, t, /= 6.9 Hz), 1.19 (3H, d, J= 6.9 Hz), 1.23-1.35 (12H, m),
1.44-1.50 (2H, m), 1.57-1.61 (4H, m), 1.66-1.72 (8H, m), 1.96-2.00 (6H, m), 2.07-2.09 (3H,
m), 2.27 (2H, t, /= 6.9 Hz), 3.09-3.13 (2H, m), 4.00 (1H, s), 4.05 (1H, s), 4.91-4.96 (1H, m);
and [M + H]+ 449.29, mp 65°C
l-(l-Adamantyl)-3-(ll-isobutoxycarbonylundecyl)wea (803)
[0176] δ(CDC13): 0.93 (6H, d, /= 6.9 Hz), 1.23-1.35 (12H, m), 1.45-1.47 (2H, m), 1.56-
1.58 (4H, m), 1.65-1.68 (6H, m), 1.94-1.97 (7H, m), 2.06-2.08 (3H, m), 2.31 (2H, t, /= 6.9
Hz), 3.07-3.11 (2H, m), 3.85 (2H, d, J= 6.9 Hz), 3.99 (1H, s), 4.03 (1H, s), [M + H]+449.32,
mp 91°C.

WO 2006/045119 PCT/US2005/038282
54
l-(l-Adamantyl)-3-(ll-benzyloxycarbonylundecyl)urea (804)
[0177] δ(CDC13): 1.24-1.28 (12H, m), 1.44-1.48 (2H, m), 1.63-1.68 (10H, m), 1.94-1.97
(6H, m), 2.05-2.07 (3H, m), 2.34 (2H, t, /= 6.9 Hz), 3.05-3.13 (2H, m), 4.04 (1H, s), 4.09
(1H, s), 5.12 (2H, s), 7.33-7.37 (5H, m), [M + H]+ 483.33, mp 49°C
l-(l-Adamantyl)-3-(ll-(2-chlorobenzyl)oxycarbonylundecyl)urea (782)
[0178] δ(CDC13): 1.24-1.28 (12H, m), 1.44-1.48 (2H, m), 1.63-1.68 (10H, m), 1.94-1.97
(6H, m), 2.05-2.07 (3H, m), 2.39 (2H, t, J= 6.9 Hz), 3.07-3.13 (2H, m), 4.00 (1H, s), 4.06
(1H, s), 5.23 (2H, s), 7.27-7.30 (3H, m), 7.39-7.42 (1H, m), [M + H]+ 517.05, mp 48°C.
Example 8
Synthesis of l-(l-adamantyl)-3-(ll-(l-adamantyl)methyloxycarbonylundecyl)urea (786)
[0179] . A solution of 687 (0.15, 0.38 mmol) and TEA (96 mg, 0.96 mmol) in DMF (10 mL)
was treated at room temperature with isobutyl chloroformate (52 mg, 0.38 mmol). After 30
min, a solution of adamantanemethanol (64 mg, 0.38 mmol) in DMF (2mL) was added. The
reaction mixture was stirred for 12 hr. The solvent was evaporated, and the residue was
partitioned between ethyl acetate (25 mL) and water (25 mL). The ethyl acetate layer was
washed with 5% NaHCO3 (10 mL) and brine (20 mL) and dried over Na2SO4, and
evaporated. The residue was chromatographed on silica gel eluting hexane and ethyl acetate
(5:1) to give 72 mg (35%) of 786 as a white solid. δ(CDC13): 1.23-1.33 (15H, m), 1.48-1.71
(21H, m), 1.90-1.96 (8H, m), 2.04-2.06 (3H, m), 2.31 (2H, t, J= 6.9 Hz), 3.05-3.12 (2H, m),
3.67 (2H, s), 4.00 (1H, s), 4.05 (1H, s), [M + H]+ 541.33, mp 68°C
[0180] Compound 792, 793 and 787 were prepared in this manner using ethylamine,
isopropylamine, and 1-naphthalenemethanol, respectively, instead of adamantanemethanol.
l-(l-Adamantyl)-3-(ll-ethylaminocarbonylundecyl)urea (792)
[0181] δ(CDC13): 1.14 (3H, t, /= 6.9 Hz), 1.24-1.31 (12H, m), 1.43-1.46 (2H, m), 1.58-
1.66 (10H, m), 1.94-1.98 (6H, m), 2.05-2.07 (3H, m), 2.15 (2H, t, /= 6.9 Hz), 3.06-3.12 (2H,
m), 3.25-3.13 (2H, m), 4.05 (1H, s), 4.12 (1H, s), 5.43 (1H, s), [M + H]+ 420.48, mp 119°C
l-(l-Adamantyl)-3-(ll-isopropylaminocarbonylundecyl)urea (793)
[0182] δ(CDC13): 1.14 (6H, d, J= 6.9 Hz), 1.24-1.31 (12H, m), 1.43-1.46 (2H, m), 1.61-
1.69 (10H, m), 1.94-1.98 (6H, m), 2.07-2.18 (5H, m), 3.07-3.13 (2H, m), 4.03-4.10 (2H, m),
4.14 (1H, s), 5.26 (1H, s), [M + H]+ 434.50, mp 115°C

WO 2006/045119 PCT/US2005/038282
55
1 -(1 -Adamantyl) -3-(l 1 -(1 -naphthyl)methoxycarbonylundecyl)urea (787)
[0183] δ(CDC13): 1.20-1.27 (12H, m), 1.43-1.46 (2H, m), 1.61-1.67 (10H, m), 1.96-2.06
(6H, m), 2.14-2.16 (2H, m), 2.35 (2H, t, J= 6.9 Hz), 3.06-3.10 (2H, m), 4.02(lH, s), 4.08
(1H, s), 5.57 (2H, s), 7.43-7.56 (4H, m), 7.84-7.87 (2H, m), 7.90 (8.02 (1H, m), [M + H]+
533.59.
Example 9
Synthesis of l-(l-Adamantyl)-3-(l l-t-butoxycarbonylundecyl)urea (801)
[0184] To a solution of compound 687 (0.10 g, 0.25 mmol), N,N-dmethylammopyridine
(DMAP, 10 mg, 0.13 mmol), and J-butanol (23 mg, 0.31 mmol) in methylene chloride (20
mL) was added l-(3-(dimemylammo)propyl)-3-ethylcarbodmnide hydrochloride (EDCI, 50
mg, 0.25 mmol) at room temperature. The mixture was stirred for 20 hr. The solvent was
evaporated, and the residue was partitioned between ether (30 mL) and water (30 mL). The
ether layer was dried over Na2SO4 and evaporated. Purification of the residue by silica gel
column chromatography eluting hexane and ethyl acetate (3:1) provided 21 mg (18 %) of t-
butyl ester as a white solid.
[0185] δ(CDC13): 1.23-1.35 (12H, m), 1.44-1.50 (2H, m), 1.57-1.61 (13H, m), 1.66-1.72
(6H, m), 1.96-2.00 (6H, m), 2.07-2.09 (3H, m), 2.27 (2H, t, J= 6.9 Hz), 3.09-3.13 (2H, m),
3.96 (1H, s), 4.01 (1H, s), [M + H]+ 449.36, mp 150°C.
Example 10
Synthesis of4-(3-Cyclohexyl-ureido)-butyric acid (632).
[0186] To a cold solution of 4-aminobutyric acid (2.16 g, 21 mmol) and catalytic amount of
DBU in 22 mL of 1.0 N NaOH, 2.5 g (20 mmol) of cyclohexyl isocyanate were added in one
time. The mixture was strongly mixed at room temperature overnight. The reaction was then
acidified with concentrated HC1. The formed white solid was collected by filtration. The
mixture was purified by chromatography on a silica column (8x3 cm). Elution with a
mixture 50:50:1 of hexane:ethyl acetate: acetic acid gave the pure targeted product. The
resulting white crystal (3.46 g; yield: 76%) had a mp of 153.0-154.0 °C. [M + H]+ 281.18.

WO 2006/045119 PCT/US2005/038282
56
Example 11
Synthesis of 2-[4-(3-Cyclohexyl-ureido)-butyrylammo]-3-(4-hydroxy-phenyl)-propionic acid
(632-Tyr).
[0187] To a solution of 632 (0.45 g, 2.0 mmol) and l-ethyl-3-(3-(dimethylamino)-propyI)
carbodiimide (0.5 g, 2.2 mmol) in 15 mL of DMF, 0.53 g (2.3 mmol) of tyrosine methyl ester
and 2.4 mmol of diisopropylethylamine were added. The mixture was heated at 60°C for 6h.
Then, 50 mL of 0.1 N NaOH were added and the mixture was left at room temperature
overnight. The reaction mixture was then acidified with concentrated HC1 and extracted
twice with a 2:1 mixture of chloroformrmethanol. The organic phases were pooled, dried and
evaporated. The residue was purified by chromatography on a silica column (5x4 cm).
Elution with a 75:25:1 mixture of ethyl acetate:methanol:acetic acid yielded 140 mg (yield:
18%) of the target product as a brown oily liquid. LC-MS-ES negative mode: 390.3 (100%,
[M-H]-), 290.9 (10%, (M-C6H10N]), 264.9 (5%, [M-C7H12NO]); positive mode: 392.5
(40%, [M+H]+), 264.95 (100%, [M-C7H10NO]+).
Example 12
Synthesis of 4-(3-Adamantan-l-yl-ureido) butyric Acid Methyl Ester (883).
[0188] The title compound was prepared by a procedure described in /. Med. Chem. 2004,
47,2110. To a suspension of 4-aminobutyric acid (2.79 g, 27.1 mmol) in DMF (40 mL) was
added 1-adamantyl isocyanate (1.20 g, 6.77 mmol) at room temperature. The reaction mixture
was stirred for 24 h. Then 1 N HC1 aqueous solution (40 mL) was added into the reaction,
and the mixture was stirred for 30 min. The solid crystalline product was filtered and washed
with water (20 mL) and ethyl acetate (20 mL). The resulting solid was dried in a vacuum
oven to give 1.90 g (100%) of 4-(3-adamantan-l-yl-ureido)butyric acid 822 as a white solid:
1H NMR (CD3OD): 1.66-1.75 (8H, m), 1.94-1.97 (6H, m), 2.05-2.07 (3H, m), 2.30 (2H, t, J
= 6.9 Hz), 3.08 (2H, q, J = 6.9 Hz), 3.32 (2H, s); LC-MS (ESI) m/z calcd for C15H24N2O3 [M
+ H]+ 281.18, found [M + H]+ 281.25; mp 165 C. Anal. (Cis^^Os) C, H, N.
[0189] A mixture of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (0.15 g, 0.54 mmol),
K2CO3 (0.09 g, 0.64 mmol), and iodomethane (0.04 mL, 0.59 mmol) in DMF (20 mL) was
stirred at room temperature for 20 h. Then the product was extracted with ether (20 mL), and
the ether was washed with 1 N NaOH aqueous solution (20 mL) and brine (20 mL), dried
over Na2SO4, and evaporated to afford 0.15 g (95%) of 883: lH NMR (CDCl3) 1.66-1.68

WO 2006/045119 PCT/US2005/038282
57
(6H, m), 1.81 (2H, quint, J = 6.9 Hz), 1.94-1.97 (6H, m), 2.05-2.07 (3H, m), 2.37 (2H, t, J =
6.9 Hz), 3.16 (2H, q, J = 6.9 Hz), 3.68 (3H, s), 4.09 (1H, s), 4.25 (1H, s); LC-MS (ESI) m/z
calcd for C16H26N2O3 [M + H]+ 295.19, found [M + H]+ 295.24; mp 114 C. Anal.
(C16H26N2O3)C,H,N.
[0190] Compounds 857, 876, 858, 877, and 878 were prepared in the same manner using
the corresponding ethyl bromoalkanoates instead of iodomethane to yield 30-95%.
Example 13
Synthesis of 4-(3-Adamantan-l-yl-ureido) butyric Acid 3,7-Dimethyl-oct-6-enyl Ester (798).
[0191] To a solution of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (0.10 g, 0.36 mmol),
4-(dimethylamino)pyridine (DMAP; 44 mg, 0.36 mmol), and 3,7-dimethyl-oct-6-en-l-ol (61
mg, 0.39 mmol) in methylene chloride (20 mL) was added l-[3-(dimethylamino)propyI]-3-
ethylcarbodiimide hydrochloride (EDCI; 75 mg, 0.39 mmol) at room temperature. After
stirring for 12 h, the reaction mixture was washed with 1 N NaOH aqueous solution (15 mL)
and water (30 mL), and the organic layer was dried over Na2SO4 and concentrated. The
residue was purified by column chromatography on silica gel eluting with hexane and ethyl
acetate (3:1) to give 798 (97 mg, 65%) as a solid: lH NMR (CDC13) 0.91 (3H, d, J = 6.9 Hz),
1.34-1.37 (2H, m), 1.56-1.60 (9H, m), 1.67-1.69 (8H, m), 1.81 (2H, quint, J = 6.9 Hz), 1.94-
1.97 (6H, m), 2.05-2.07 (3H, m), 2.35 (2H, t, J = 6.9 Hz), 3.16 (2H, q, J = 6.9 Hz), 4.05 (1H,
s), 4.11 (2H, t, J = 6.9 Hz), 4.21 (1H, s), 5.09 (1H, t, J = 6.9 Hz); LC-MS (ESI) m/z calcd for
C25H42N2O3 [M + H]+ 419.32, found [M + H]+ 419.22; mp 49 C. Anal. Calcd for
C25H42N2O3: C, 71.73; H, 10.11; N, 6.69. Found: C, 70.27; H, 9.83; N, 6.39.
Example 14
Synthesis of 8-[4-(3-Adamantan-l-yl-ureido)butyryloxy]octanoic Acid Ethyl Ester (879).
[0192] To a solution of 8-bromooctanoic acid (0.20 g, 0.89 mmol), DMAP (0.12 g, 0.99
mmol), and ethanol (0.05 g, 0.99 mmol) in methylene chloride (20 mL) was added EDCI
(0.19 g, 0.99 mmol) at room temperature. After stirring for 12 h, the reaction mixture was
washed with 1 N NaOH aqueous solution (15 mL) and water (30 mL), and the organic layer
was dried over Na2SO4 and evaporated to give 8-bromooctanoic acid ethyl ester (0.17 g,
75%). This bromide reacted with 4-(3-adamantan-l-yl-ureido)butyric acid 822 in the same
manner as that used for the preparation of 883 to provide 879 (0.19 g, 65%) as a solid: 1H

WO 2006/045119 PCT/US2005/038282
58
NMR (CDCl3) 1.26 (3H, t, J = 6.9 Hz), 1.32-1.35 (6H, m), 1.59-1.66 (10H, m), 1.82 (2H,
quint, J = 6.9 Hz), 1.94-1.97 (6H, m), 2.05-2.07 (3H, m), 2.28 (2H, t, J = 6.9 Hz), 2.36 (2H, t,
J = 6.9 Hz), 3.16 (2H, q, J = 6.9 Hz), 4.05-4.14 (5H, m), 4.31 (1H, s); LC-MS (ESI) m/z calcd
for C25H42N2O5 [M + H]+ 451.31, found [M + H]+ 451.20; mp 58-59 C. Anal. (C25H42N2O5)
C, H, N.
Example 15
Synthesis of 10-[4-(3-Adamantan-l-yl-ureido)butyryloxy]decanoic Acid Ethyl Ester (880).
[0193] A mixture of 10-hydroxydecanoic acid (0.25 g, 1.33 mmol; 11 -hydroxyundecanoic
acid for compound 881 and 12-hydroxydodecanoic acid for compound 882), ethyl bromide
(0.16 g, 1.46 mmol), and lithium carbonate (0.11 g, 1.46 mmol) in DMF (25 mL) was stirred
at 70 C for 6 h. Then the product was extracted with ether (30 mL), and the ether solution
was washed with 1 N NaOH aqueous solution (20 mL) and water (30 mL), dried over
Na2SO4, and concentrated. The residue was purified by column chromatography on silica gel
eluting with hexane and ethyl acetate (3:1) to give 10-hydroxydecanoic acid ethyl ester (80
mg, 28%). This alcohol was coupled with 4-(3-adamantan-l-yl-ureido)butyric acid 822 by
using EDCI/DMAP coupling reagent to give 880 (0.11 g, 60%) as a solid: 1H NMR (CDCl3)
1.24-1.32 (13H, m), 1.62-1.68 (10H, m), 1.80 (2H, quint, J = 6.9 Hz), 1.94-1.97 (6H, m),
2.05-2.07 (3H, m), 2.28 (2H, t, J = 6.9 Hz), 2.36 (2H, t, J = 6.9 Hz), 3.16 (2H, q, J = 6.9 Hz),
4.05-4.14 (5H, m), 4.25 (1H, s); LC-MS (ESI) m/z calcd for C27H46N2O5 [M + H]+ 479.34,
found [M + H]+ 479.29; mp 60-61 C. Anal. Calcd for C27H46N2O5: C, 67.75; H, 9.69; N, 5.85.
Found: C, 68.33; H, 9.92; N, 5.97.
[0194] Compound 4-(3-adamantan-1 -yl-ureido)butyric acid 822 was coupled with 11-
hydroxyundecanoic acid ethyl ester and 12-hydroxydodecanoic acid ethyl ester prepared from
corresponding acids to get compounds 881 and 882, respectively.
Example 16
Synthesis of 4-[4-(3-Adamantan-l-yl-ureido)butyryloxymethyl]benzoic Acid Ethyl Ester
(849).
[0195] A mixture of 4-formylbenzoic acid (1.00 g, 6.66 mmol), bromoethane (1.09 g, 9.99
mmol), and K2CO3 (1.10 g, 7.99 mmol) in acetonitrile (30 mL) was refluxed for 6 h. After
evaporation of the solvent, 4-formylbenzoic acid ethyl ester was extracted with ether (30

WO 2006/045119 PCT/US2005/038282
59
mL), and the organic solution was washed with 1 N NaOH aqueous solution (20 mL) and
water (30 mL), dried over Na2SO4, and concentrated to give the ethyl ester product (0.65 g,
55%). Without further purification, to a solution of the ester was added sodium borohydride
(NaBELt; 0.05 g, 3.65 mmol) in ethanol (20 mL) at 0 C. After stirring for 5 h at room
temperature, the product was extracted with ether (30 mL), and the ether solution was washed
with water (30 mL), dried over Na2SO4, and concentrated. The residue was purified by using
column chromatography on silica gel eluting with hexane and ethyl acetate (3:1) to give 4-
hydroxymethylbenzoic acid ethyl ester (0.30 g, 46%) as an oil.
[0196] To a solution of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (1.23 g, 0.83 mmol),
DMAP (0.05 g, 0.42 mmol), and the above alcohol (0.15 g, 0.83 mmol) in methylene chloride
(30 mL) was added EDCI (0.16 g, 0.83 mmol) at room temperature. After stirring for 12 h,
the reaction mixture was washed with 1 N NaOH aqueous solution (15 mL) and water (30
mL), and the organic layer was dried over Na2SO4 and concentrated. Then the residue was
purified by column chromatography on silica gel eluting hexane and ethyl acetate (5:1) to
provide 849 (0.28 g, 75%) as a white solid: 1H NMR (CDC13) 1.40 (3H, t, J = 6.9 Hz), 1.66-
1.68 (6H, m), 1.84 (2H, quint, J = 6.9 Hz), 1.94-1.96 (6H, m), 2.05-2.07 (3H, m), 2.44 (2H, t,
J = 6.9 Hz), 3.17 (2H, q, J = 6.9 Hz),.4.02 (1H, s), 4.17 (1H, s), 4.38 (2H, q, J = 6.9 Hz), 5.17
(2H, s), 7.40 (2H, d, J = 7.8 Hz), 8.00 (2H, d, J = 7.8 Hz); LC-MS (ESI) m/z calcd for
C25H34N2O5 [M + H]+ 443.25, found [M + H]+ 443.25; mp 96-99 C. Anal. (C25H34N2O5) C,
H,N.
Example 17
Synthesis of 4-(3-Adamantan-l-yl-ureido)butyric Acid 4-Ethoxycarbonylmethylphenyl Ester
(852).
[0197] To a solution of 4-(3-adamantan-l-yl-ureido)butyric acid 822 (0.15 g, 0.54 mmol),
DMAP (0.07 g, 0.54 mmol), and 4-hydroxyphenylacetic acid (0.09 g, 0.59 mmol) in
methylene chloride (20 mL) was added EDCI (0.11 g, 0.59 mmol) at room temperature. After
stirring for 12 h, the reaction mixture was washed with water (20 mL), and the methylene
chloride solution dissolving the product was dried over Na2SO4 and concentrated to give
conjugated product. This crude mixture in DMF (30 mL) was treated with bromoethane (0.15
g, 1.34 mmol) and K2CO3 (0.18 g, 1.34 mmol) at room temperature and stirred for 12 h at
room temperature. The ethyl ester product was extracted with ether (30 mL), and the ether
solution was washed with 1 N NaOH aqueous solution (20 mL) and water (30 mL), dried

WO 2006/045119 PCT/US2005/038282
60
over Na2SO4, and concentrated. The residue was purified by column chromatography on
silica gel eluting hexane and ethyl acetate (5:1) to give 852 (47 ing, 20%) as a white solid: 1H
NMR (CDC13) 1.40 (3H, t, J = 6.9 Hz), 1.66-1.68 (6H, m), 1.89-1.95 (8H, m), 2.05-2.07 (3H,
m), 2.62 (2H, t, J = 6.9 Hz), 3.25 (2H, q, J = 6.9 Hz), 3.60 (2H, s), 4.07 (1H, s), 4.16 (2H, q, J
= 6.9 Hz), 4.29 (1H, s), 7.08-7.10 (2H, m), 7.28-7.30 (2H, m); LC-MS (ESI) m/z calcd for
C25H34N2O5 [M + H]+ 443.25, found [M + H]+ 443.25; mp 95-97 C. Anal. (C25H34N2O5) C,
H,N.
[0198] Compound 851 was prepared in the same manner by using 4-hydroxyphenylacrylic
acid instead of 4-hydroxyphenylacetic acid.
Example 18
Synthesis ofN-[12-(3-Adamantan-l-yl-ureido)dodecanoyl]methanesulfonamide (848)
[0199] To a solution of compound 687 (0.2 g, 0.51 mmol) and N-hydroxysuccinimide (60
mg, 0.56 mmol) in THF (10 mL) was added 1,3-dicyclohexylcarbodiimide (0.12 g, 0.56
mmol) at room temperature. The reaction mixture was stirred for 12 hrs and filtered. And
then, the filtrate was purified by column chromatography (hexane:ethyl acetate = 1:1) to give
2,5-dioxopyrrolidinyl ester (I) (0.18 g, 0.37 mmol) in 72% yield. To this intermediate (I)
dissolved in HMPA (10 mL) was added portionwise 4-dimethylarninopyridine (54 mg, 0.44
mmol; DMAP) and methanesulfonamide (0.35 g, 3.7 mmol). After stirring for 2hrs at 90°C,
the product was extracted with ether (30 mL) and washed with water (30 mL). The organic
solution was dried over magnesium sulfate and evaporated, and then the residue was purified
using column chromatography eluting with hexane and ethyl acetate (1:1) to afford
compound 848 (0.16 g, 0.34 mmol) in 92% yield. 1H NMR 5 (CDCl3) 1.23-1.35 (12H, m),
1.44-1.52 (4H, m), 1.57-1.61 (2H, m), 1.65-1.69 (6H, m), 1.92-1.98 (6H, m), 2.07-2.09 (3H,
m), 2.38 (2H, t, J = 6.9 Hz), 3.11 (2H, q, J = 6.9 Hz), 3.20 (3H, s), 4.40 (1H, s), 4.48 (1H, s),
10.52 (1H, s), LC-MS (ESI) m/z calcd for C24H43N3O4S [M + H]+ 469.30, found [M + H]+,
mp 103°C, Anal. (C24H43N3O4S) C, H, N.
[0200] Compound 914 was synthesized with the same method used for the preparation of
compound 687 using benzenesulfonamide instead of methanesulfonamide. 1H NMR 8
(CDCl3) 1.23-1.35 (12H, m), 1.44-1.52 (4H, m), 1.57-1.61 (2H, m), 1.65-1.69 (6H, m), 1.94-
1.98 (6H, m), 2.06-2.09 (3H, m), 2.28 (2H, t, J = 6.9 Hz), 3.10 (2H, q, J = 6.9 Hz), 4.39 (1H,
s), 4.93 (1H, s), 5.45 (1H, s), 7.50-7.55 (2H, m), 7.60-7.62 (1H, m), 7.80-7.83 (1H, m), 8.05-

WO 2006/045119 PCT/US2005/038282
61
8.08 (1H, m), LC-MS (ESI) m/z calcd for C29H45N3O4S [M + H]+ 532.31, found [M + H]+
532.34, mp 100°C, Anal. (C29H45N3O4S) C, H, N.
Example 19
Synthesis of2-[12-3-(Adamantan-l-yl-ureido)dodecanoylamino]decanoic acid (1001)
[0201] Sodium metal (3.9 g, 0.17 mol) was dissolved in ethanol (100 mL) under an inert
atmosphere in a round bottom flask fitted with a water condenser. Diethyl acetamido
malonate (30.4 g, 0.14 mol) was then added followed by 1-bromooctane (36.7 g, 0.19 mol).
The solution was refluxed overnight under an inert atmosphere. The reaction mixture was
poured onto crushed ice (600 mL) and stirred. The aminodiester product precipitated and was
collected by filtration. The crude product was then refluxed overnight in a solution HClrDMF
(9:1, 200 mL). The precipitated product was collected by filtration, washed with ice water,
and air dried in a vacuum desiccator to afford the a-amino acid hydrochloride in >90% crude
yield. The crude amino acid (3.0 g, 24.8 mmol) was then dissolved in methanol (100 mL) and
cooled to 0°C. Thionyl chloride (5.0 mL, 25.8 mmol) was added dropwise, and the reaction
was stirred at 0°C for 10 minutes and then refluxed overnight. The reaction was cooled to
room temperature and the volatiles removed under reduced pressure, and the crude product
was triturated in methanol to afford racemic methyl 2-amino decanoate, 4.44g, 89% yield.
[0202] Compound 687 (1.04 g, 2.65 mmol) and HBTU (1.0 g, 2.64 mmol) were dissolved
in THF (60 mL). DIEA (0.5 mL, 2.87 mmol), DMF (~ 2 mL), and methyl 2-amino decanoate
(1.26 g, 5.30 mmol) were added, and the solution was stirred under N2 at room temperature
overnight. The yellow oil produced was diluted with 5 % citric acid (100 mL) and extracted
with ethyl acetate (3 x 50 mL). Organic layers were combined and washed with 5 % citric
acid (2 x 50 mL), saturated sodium bicarbonate (NaHCO3) (2 x 50 mL), and brine (1 x 50
mL). The organic layer was dried over magnesium sulfate and evaporated to yield an oil. The
crude product was purified by column chromatography with 1-2 % MeOH/DCM to yield a
methyl ester as a yellow oil(0.77 g, 66 % yield). The methyl ester(0.77 g, 1.34 mmol) was
dissolved in DME (25 mL) and water (10 mL). Solid LiOH●H2O (0.33 g, 7.86 mmol) was
added, and the solution was stirred at room temperature overnight. Reaction mixture was
acidified (pH = 4) with 5 % citric acid (~ 20 mL), and the product was extracted with ethyl
acetate (3 x 30 mL). Combined organic layers were washed with brine (30 mL), dried over
MgSCU, filtered, and evaporated to yield compound 1001 as a yellow oil (0.34 g, 45 % yield).

WO 2006/045119 PCTYUS2005/038282
62
1H NMR 8 (CDC13) 0.82 (3H, t, J = 7 Hz), 1.16-1.23 (28H, bm), 1.41 (2H, m), 1.57 (2H, bm),
1.60 (6H, bs), 1.89 (6H, bs), 2.01 (3H, bs), 2.18 (2H, t, J = 7Hz), 2.98 (2H, m), 4.52 (1H, dd,
J = 7.3,13.3 Hz), 6.46 (2H, d, J = 7.6 Hz) 8.16 (1H, bs). 13C NMR 5 (CDCI3) 14.0, 22.5,
25.0, 25.6, 26.7, 28.85,28.00,29.06, 29.10, 29.13, 29.24, 29.31,29.44, 29.56, 31.73, 32.22,
36.27, 36.41,40.68, 42.29, 50.96, 52.31,158.70, 173.65,175.39. LC-MS (ESI) m/z calcd for
C33H59N3O4 [M + H]+ 562.45, found [M + H]+ 562.51.
Example 20
Synthesis of 12-(3-Adamantan-l-yl-ureido)dodecanoic acid [l-(2,3,4-trihydroxy-5-
hydroxymethyl-cyclohexylcarbamoyl)nonyl]amide (1002)
[0203] 1,2,3,4,6-Penta-O-acetyl-a-D-glucopyranose (5.00 g, 12.8 mmol) was dissolved in
10-15 mL dry CH2CI2 under an inert atmosphere. Trimethylsilyl azide (4.24 mL, 32.1 mmol)
and tin (IV) chloride (0.75 mL, 6.41 mmol) were added, and the reaction was stirred for 18
hours at room temperature. The reaction mixture was diluted with CH2CI2 (30 mL) and
washed twice with saturated NaHCC>3 (20 mL) and with brine (20 mL). The organic phase
was dried, filtered and concentrated to leave 2,3,4,6-tetra-O-acetyl-a-D-glucopyranosyl azide
as a white solid (3.92 g, 82%). 2,3,4,6-tetra-O-acetyl-a-D-glucopyranosyl azide (0.30 g, 80
mmol) was then dissolved in dry THF (15 mL), and Pd/C was added (~ 5 %) to this solution.
The mixture was stirred under H2 overnight. TLC revealed the reduction was completed
(Rfazide = 0.80, Rfamine = 0.10). In a separate flask, compound 23 (0.17 g, 0.30 mmol) and
0.5 M HBTU in DMF (1.20 mL, 0.60 mmol) were combined, followed by the addition of
DMF (~ 2 mL) and DIEA (104 ΜL, 0.60 mmol). This solution was then added to the reaction
flask, and the reaction mixture was stirred under N2 overnight. The resulting suspension was
filtered through celite which was washed well with ethyl acetate (~ 100 mL), and the filtrate
was washed with 5 M HC1 (50 mL), saturated NaHCO3 (2 x 50 mL), and brine (1 x 50 mL).
The organic solution was dried over magnesium sulfate, filtered, and concentrated. The
residue was purified using a silica gel column chromatography with 10 % MeOH/DCM to
yield V (0.16 g, 59.33). 1H NMR δ (CDCl3) 0.78 (3H, t, J = 7 Hz), 1.16 - 1.23 (32H, brm),
1.35 (2H, m) 1.52 (2H, m)1.57 (6H, brs), 1.86 (6H, brs,), 1.91-2.00 (15H, m), 2.14 (2H, m),
2.99 (2H, t, J = 7Hz), 3.75 (2H, m), 3.95-4.07 (3H, m), 4.15-4.29 (4H, m), 4.43-4.38 (1H, m)
4.80 (1H, dd, J = 5,11 Hz), 4.84-4.89 (2H, m), 4.95-5.02 (3H, m), 5.12-5.24 (3H, m), 5.35
(1H, d, J = 3 Hz), 5.47 (1H, t, J = 9.8 Hz), 6.28 (1H, dd, J = 7.6,19 Hz), 6.60 (1H, d, J = 9.3

WO 2006/045119 PCT/US2005/038282
63
Hz), 7.18 (1H, d, J = 9.1 Hz), 7.34 (1H, d, J = 9.1 Hz). LC-MS (ESI) m/z calcd for
C47H78N4O12 [M + H]+ 891.56, found [M + H]+ 891.69.
[0204] To a solution of V (0.16 g, 0.18 mmol) in MeOH (15 mL) was added sodium metal
(0.05 g), and the mixture was stirred at room temperature overnight. The reaction mixture
was filtered through amberlite and washed with MeOH (~ 150 mL). The solvent was
evaporated under vacuum to produce compound 1002 as a fine white powder (0.12 g, 95 %).
LC-MS (ESI) m/z calcd for C39H70N4O8 [M + H]+ 724.50, found [M + H]+ 724.6.
[0205] Compound 798 was prepared with the same method as that used for the preparation
of compound 1002 from compound 687 instead of compound 1001.
Example 21
Synthesis of5-(3-Chloro-phenylcarbamoyl)pentanoic Acid Pentyl Ester (987).
[0206] To a solution of adipic acid (0.5 g, 3.42 mmol) and DMAP (0.42 g, 3.42 mmol) in
dichloromethane (30 mL) and DMF (3 mL) was added 3-chloroaniline (0.44 g, 3.42 mmol) at
room temperature. After stirring 10 min, l-[3-(dimemylammo)propyl]-3-emylcarbodiimide
hydrochloride (0.65 g, 3.42 mmol; EDCI) was added portionwise to the mixture at room
temperature. The reaction was stirred for 12 h. A 1 N aqueous HC1 solution (20 mL) was
poured into the reaction mixture, and 5-(3-chlorophenylcarbomoyl)pentanoic acid was
extracted with dichloromethane (30 mL). The organic solution was washed with water (50
mL), dried over Na2SO4, and concentrated. This residue was used for the next reaction
without further purification.
[0207] To the solution of the above carbamoylpentanoic acid (0.72 g, 2.80 mmol) in DMF
(15 mL) was added K2CO3 (0.58 g, 4.21 mmol) and 1-bromopentane (0.64 g, 4.21 mmol) at
room temperature. After stirring for 12 h, the product was extracted with ether (30 mL), and
the ether solution was washed with an aqueous solution of 1 N NaOH (15 mL) and water (40
mL), dried over Na2SO4, and concentrated. The residue was purified using silica gel column
chromatography (hexane:ethyl acetate = 5:1) to afford 987 (0.59 g, 65%): 1H NMR (CDCl3)
0.91 (3H, t, J = 6.9 Hz), 1.29-1.37 (4H, m), 1.60-1.66 (2H, m), 1.70-1.78 (4H, m), 2.35-2.43
(4H, m), 4.08 (H, t, J = 6.9 Hz), 7.05-7.09 (1H, m), 7.21-7.23 (1H, m), 7.37-7.40 (1H, m),
7.52-7.55 (1H, m), 7.68 (1H, s); LC-MS (ESI) m/z calcd for C17H24CINO3 [M + H]+ 326.14,
found [M + H]+ 326.16, mp 82 C. Anal. (C17H24CINO3) C, H, N.
[0208] Compound 13 was prepared with the same method used for the preparation of
compound 987 using adamantylamine instead of 3-chloroaniline: 1H NMR (CDCl3) 0.91

WO 2006/045119 PCT/US2005/038282
64
(3H, t, J = 6.9 Hz), 1.29-1.43 (4H, m), 1.64-1.69 (12H, m), 1.94-1.98 (6H, m), 2.06-2.13 (5H,
m), 2.32 (2H, t, J = 6.9 Hz), 4.06 (H, t, J = 6.9 Hz), 5.16 (1H, s); LC-MS (ESI) m/z calcd for
C21H35NO3 [M + H]+ 350.26, found [M + H]+ 350.30. Anal. (C21H35NO3) C, H, N.
Example 22
Synthesis of 4-(2-Chloro-pheny)acethylaminolbutyric Acid Peniyl Ester (988).
[0209] To a solution of 3-chlorophenylacetic acid (0.5 g, 2.93 mmol) and DMAP (0.36 g,
2.93 mmol) in dichloromethane (30 mL) was added ethyl 4-aminobutyrate hydrochloride
(0.49 g, 2.93 mmol) at room temperature. After stirring for 10 min, EDCI (0.56 g, 2.93
mmol) was added portionwise to the mixture at room temperature. The reaction was stirred
for 12 h. A 1 N aqueous HC1 solution (20 mL) was poured into the reaction mixture, and 4-
[2-(3-chlorophenyl)acetylamino]butyric acid ethyl ester was extracted with ether (30 mL).
The ether solution was washed with water (50 mL), dried over Na2SO4, and concentrated. To
the residue dissolved in ethanol (10 mL) was added 1 N aqueous NaOH solution (6 mL), and
after 12 h of stirring at room temperature, the product was extracted with dichloromethane
(30 mL). The organic solution was washed with water (30 mL), dried over Na2SO4, and
concentrated to give 4-[2-(3-chlorophenyl)acetylamino]butyric acid (0.6 g, 80%). A mixture
of this acid (0.6 g, 2.35 mmol), K2CO3 (0.49 g, 3.52 mmol), and 1-bromopentane (0.53 g,
3.52 mmol) in DMF (20 mL) was stirred overnight at room temperature. The product was
extracted with ether (40 mL), and the ether solution was washed with water (50 mL), dried
over Na2SO4, and concentrated. The residue was purified using silica gel column
chromatography (hexane:ethyl acetate = 3:1) to afford 988 as an oil (0.74 g, 97%): 1H NMR
(CDCl3) 0.91 (3H, t, J = 6.9 Hz), 1.26-1.33 (4H, m), 1.59-1.63 (2H, m), 1.80 (2H, quint, J =
6.9 Hz), 2.31 (2H, t, J = 6.9 Hz), 3.27 (2H, q, J = 6.9 Hz), 3.52 (2H, s), 4.04 (2H, t, J = 6.9
Hz), 5.72 (1H, s), 7.13-7.17 (2H, m), 7.27-7.30 (2H, m); LC-MS (ESI) m/z calcd for
C7H24CINO3 [M + H]+ 326.14, found [M + H]+ 326.15. Anal. (C17H24ClNO3) C, H, N.
[0210] Compounds 837 and 1068 were prepared with the same method used for the
preparation of compound 988 using 1-adamantylacetic acid or adamantane-1-carboxylic acid
instead of 3-chlorophenylacetic acid. Compound 837: 1H NMR (CDC13) 0.91 (3H, t, J = 6.9
Hz), 1.29-1.35 (4H, m), 1.35-1.72 (14H, m), 1.84 (2H, quint, J = 6.9 Hz), 1.91 (2H, s), 1.97
(3H, m), 2.37 (2H, t, J = 6.9 Hz), 3.29 (2H, q, J = 6.9 Hz), 4.07 (2H, t, J = 6.9 Hz), 5.66 (1H,
s); LC-MS (ESI) m/z calcd for C21H35NO3 [M + H]+ 350.26, found [M + H]+ 350.29. Anal.
(C2,H35NO3)C,H,N.

WO 2006/045119 PCT/US2005/038282
65
[0211] Compound 1068: 1H NMR (CDC13) 0.91 (3H, t, J = 6.9 Hz), 1.29-1.35 (4H, m),
1.60-1.86 (16H, m), 2.03 (3H, m), 2.35 (2H, t, J = 6.9 Hz), 3.29 (2H, q, J = 6.9 Hz), 4.07 (2H,
t, J = 6.9 Hz), 5.85 (1H, s); LC-MS (ESI) m/z calcd for C20H33NO3 [M + H]+ 336.25, found
[M + H]+ 336.34. Anal. (C20H33NO3) C, H, N.
Example 23
Synthesis of 4-(3-Chloro-phenylcarbamoyloxy)butyric Acid Pentyl Ester (825).
[0212] To a solution of succinic anhydride (3.58 g, 35.7 mmol) and DMAP (4.16 g, 34.0
mmol) in DMF (40 mL) was added pentanol (3.0 g, 34.0 mmol) at room temperature under
nitrogen. After stirring for 12 h, succinic acid pentyl ester was extracted with ether (40 mL),
and the ether solution was washed with 1 N aqueous HC1 solution (20 mL) and water (40
mL), dried over sodium sulfate (Na2SO4), and concentrated. The residue was purified by
column chromatography on silica gel eluting with hexane and ethyl acetate (3:1) to give the
succinic acid pentyl ester (6.07 g, 95%) as an oil. To the solution of this acid in
tetrahydrofuran (THF, 60 mL) was added 1 M BH3-THF complex (64.53 mL, 64.5 mmol) at -
10C under nitrogen, and the reaction mixture was allowed to warm to room temperature with
stirring. After stirring for 12 h at room temperature, 5% NaHCC"3 aqueous solution (50 mL)
was added to the reaction and then the reduced alcohol (T) was extracted with ethyl acetate
(50 mL). The ethyl acetate solution was dried over Na2SO4 and concentrated to give 4-
Hydroxybutyric acid pentyl ester (5.06 g, 90%).
[0213] 4-Hydroxybutyric acid pentyl ester (100 mg, 0.57 mmol) was added to a solution of
3-chlorophenyl isocyanate (88 mg, 0.57 mmol) and triethylamine (0.12 mL, 0.86 mmol;
TEA) in DMF (15 mL) at room temperature. The mixture was allowed to stand at room
temperature for 12 h, the product was extracted with ether (20 mL), and the ether solution
was washed with 1 N aqueous HC1 solution (20 mL) and water (30 mL), dried over Na2SO4,
and concentrated. The residue was purified by column chromatography on silica gel eluting
with hexane and ethyl acetate (5:1) to afford 9 (94 mg, 50%) as a solid: 1H NMR (CDCl3)
0.90 (3H, t, J = 6.9 Hz), 1.26-1.34 (4H, m), 1.62-1.65 (2H, m), 1.71 (2H, quint, J = 6.9 Hz),
2.66 (2H, t, J = 6.9 Hz), 2.74 (2H, q, J = 6.9 Hz), 4.10 (2H, t, J = 6.9 Hz), 7.05-7.08 (1H, m),
7.18-7.22 (3H, m), 7.35 (1H, s); LC-MS (ESI) m/z calcd for C16H22ClNO4 [M + H]+ 328.12,
found [M + Hf 328.13; mp 82 C. Anal. (C16H22CINO4) C, H, N.

WO 2006/045119 PCTYUS2005/038282
66
Example 24
Synthesis of l-(3-Chloro-phenyl)-3-(l-hydroxymethyl-pentyl)-urea (978).
[0214] 2-aminohexanol hydrochloride (211 mg, 1.37 mmol), m - chlorophenyl isocyanate
(211 mg, 1.37 mmol) were combined in THF (10 mL) with triethylamine (228 uL) and stirred
over night. The solvent was removed and the residue chromatographed on silica gel to give
343 mg of the target product (93 %). 1H NMR (300 MHz, CDCl3/MeOH d4 1:1) 8 = 7.50 -
7.47 (m, 1H), 7.20 - 7.10 (m, 2H), 6.93 (dt, J = 6.6,1.6 Hz, 1H), 5.76 (d, J = 8.2 Hz, 1H),
3.75 (br, 1H), 3.65 - 3.45 (m, 2H), 1.60-1.20 (m, 6H), 1.87 (m, 3H).
[0215] Compound 977 were prepared in the same manner using aminoalcohol and
cyclohexylisocyanate.
Synthesis ofl-(3-Chloro-phenyl)-3-(l-hydroxymethyl-butyl)-urea (977)
[0216] !H NMR (300 MHz, CDC13) 8 = 5.41 (m, 2H), 3.8-3.2 (m, 4H), 2.00 - 0.90 (m, 20
H)
Example 25
Synthesis of(4-Butyl-4,5-dihydro-oxazol-2-yl)-(3-chloro-phenyl)-amine(980).
[0217] Compound 978 (50 mg) was treated with POC13 (1.0 mL). This was stirred
overnight. The solvent was removed via reduced pressure distillation. The residue was washd
with aqueous sodium bicarbonate and then dissolved in EtOAc. The solvent was dried,
filtered and evaporated. The residue was dissolved in acetone (1 mL) and refluxed with water
(2 mL) for 1.5 hrs. The solution was extracted with EtOAc, the organic layer evaporated and
the residue chromatographed on silica to give the target compound. 1H NMR (300 MHz,
CDCI3) 6= 7.38 (s, 1H), 7.15 (d, J=8.0 Hz, 1H), 7.05 (br, 1H), 7.00 (d, 7.5 Hz, 1H), 4.47 (m,
1H), 3.95 (m, 2H), 1.8-1.2 (m, 6H), 0.95 (m, 3H). ESMS = 224.9 m/z (M+H)
Example 26
Synthesis of (994).
[0218] Methylaminopentanoate hydrochloride (720 mg, 4.3 mmol) and
benzophenoneimine (722 mg, 4.3 mmol) were stirred together in dichloromethane for 18 hrs.
At this point, the reaction was washed with aqueous sodium bicarbonate and the organic layer

WO 2006/045119 PCT/US2005/038282
67
dried with MgS04 and evaporated to leave 1.3 g of a thick oil. The product (530 mg, 1.8
mmol) was then dissolved in THF (15 mL) and cooled (under N2) to -78°C. DBAL ( 2.0 mL,
1 M solution) was added dropwise and the reaction stirred for lhr. When TLC indicated the
absence of the ester starting material and the presence of a aldehyde (via DNP stain), 1.3 eq
of butyl magnesium bromide was added to the reaction. This was allowed to warm to room
temperature over 2.5 hrs. The reaction was quenched with bicarbonate solution and the
organic layer was dried and evaporated to give the imino alcohol (350 mg) as an oil. The
imino alcohol (150 mg) was stirred with aqueous HC1 (1M, 1mL) and diethyl ether (5 mL)
overnight. The aqueous layer was evaporated and 1 equivalent of 1-adamantylisocyanate was
added as a solution in dichloromethane (5 ml) and triethylamine (0.5 mL). This was stirred
overnight. The crude reaction was chromatographed on silica gel (1:1 EtOAc rhexanes) to
give the product as an oil (35 mg). 1H NMR (300 MHz, CDC13) δ= 4.22 (br, 1H), 4.08 (br,
1H), 3.91 (br, 1H), 3.10 (m, 1H), 2.30 (br, 2H), 2.2 -1.0 (br m, 32 H)
Example 27
Synthesis of (996).
[0219] Dodecylamine (500 mg, 2.7 mmol) was added to a biphasic system of
dichloromethane (20 mL) and aqueous bicarbonate (20 mL, satd). The mixture was stirred
while triphosgene (264 mg, 0.9 mmol) was added. The reaction was stirred for 2 hrs. The
organic layer was removed and filtered through a plug of silica gel using EtOAc as the eluent.
Evaporation of the solvent lead to 564 mg of the corresponding isocyanate. The isocyanate
(leq) was combined with the amine (15 mg, 0.048 mmol) in dichloromethane. This was
stirred overnight. The reaction was loaded onto silica gel and chromatographed using EtOAc
to yield the product (22 mg, 89%) as an orange solid. 1H NMR (300 MHz, CDC13) δ= 7.69
(d, J=8.3 Hz, 1H), 5.81 (d, J = 8.3 Hz, 1H), 5.15 (d, J=6.72 Hz, 1H), 4.70 (m, 1H), 4.60 (m,
1H), 4.00 - 3.5 (br, 2H), 3.30 (t, J = 6.7 Hz, 2H), 3.17 (q, J=6.6 Hz, 2H), 2.4-2.0 (m, 2H), 1.9
- 0.9 (br m, 32H). ESMS = 523 m/z (M+H)
Example 28
Synthesis of (997).
[0220] 12-aminododecanaol (50 mg) was stirred in dichloromethane with 1-
adamantylisocyanate (44mg) overnight. The reaction was evaporated and

WO 2006/045119 PCT/US2005/038282
68
carbonyldiimidazole was added in 2 mL acetonitrile. This was refluxed 5 hrs. The solvent
was removed in vacuo and the solid was partitioned between dichloromethane and water. The
organic layer was washed repeatedly with water to yield the target compound (22mg). 1H
NMR (300 MHz, CDC13) δ = 8.17 (s, IE), 7.45 (s, 1H), 7.05 (s, 1H), 4.41 (t, J = 7.5 Hz, 2H),
4.32 (br, 1H), 4.21 (br, 1H), 3.08 (q, J = 6.7 Hz, 2 H), 2.0 - 1.0 (m, 35H).
Example 29
General procedure for the synthesis of trans-benzyloxy isomers: Synthesis of trans-1-(4-
Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7] decan-1-yl-urea 1032.
Synthesis oftrans-l-(4-Hydroxy-cyclohexyl)-3-tricyclo[3.3.L13,7]decan-1-yl-urea 1039
[0221] To a solution of 1-adamantyl isocyanate (10 g, 56.4 mmol) and trans-4-
aminocyclohexanol hydrochloride (10.3 g, 67.7 mmol) in DMF (300 mL) was added
triethylamine (6.9 g, 67.7 mmol) at 0 oC. The reaction mixture was stirred overnight. The
reaction mixture was poured into water, and the resulting precipitates were collected and
washed with water. The crude product was recrystallized from methanol/water. Yield: 15.5
g (94% of theory). M.P.: 254 °C. 1H NMR (300 MHz, DMSO-d6): δ 5.48 (d. J = 9 Hz, 1H),
5.38 (s, 1H), 4.48 (d, J = 4.7 Hz, 1H), 3.42-3.28 (m, 1H), 3.28-3.13 (m, 1H), 2.02-1.93 (m,
3H), 1.87-1.68 (m, 9H), 1.63-1.54 (m, 7H), 1.24-0.93 (4H).
Synthesis of trans-l-(4-Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-1-yl-urea 1032
[0222] To a solution of compound 1 in DMF (10 mL) was added 60% sodium hydride in
oil (60 mg, 1.5 mmol) at 0 °C. After 10 min, benzyl bromide (0.20 g, 1.2 mmol) was
introduced and then warmed up to room temperature and stirred overnight. The reaction was
quenched by adding water and the resulting white precipitates were collected and washed
with water. The solids were recrystallized from DCM/hexanes. Yield: 0.35 g (92% of
theory). M.P.:244°C. 1HNMR(300MHz,CDCl3): δ 7.40-7.23 (m, 5H), 4.52 (s, 2H),
4.10-3.92 (m, 2H), 3.58-3.41 (m, 1H), 3.37-3.24 (m, 1H), 2.11-1.81 (m, 13H), 1.50-1.33 (m,
6H), 1.50-1.33 (m, 2H), 1.17-0.99 (m, 2H).

WO 2006/045119 PCT/US2005/038282
69
Example 30
General procedure for the synthesis of cis-benzyloxy isomers: Synthesis ofcis-l-(4-
Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-l-yl-urea 1078
Synthesis of trans-4-Nitro-benzoic acid 4-(l,3-dioxo-l,3-dihydro-isoindol-2-yl)-cyclohexyl
ester
[0223] To a solution of trans-2-(4-Hydroxy-cyclohexyl)-isoindole-l,3-dione (38 g, 154.9
mmol), triphenylphosphine(65 g, 248 mmol), and 4-nitrobenzoic acid (41.4 g, 248 mmol) in
1500 mL of THF was added dropwise diisopropyl azodicarboxylate (50.1 g, 248 mmol) at
room temperature. The reaction mixture was stirred overnight. The solvent was evaporated,
and the resulting solid was recrystallized from methanol. Yield: 53 g (86.7% of theory). 1H
NMR (300 MHz, CDC13): δ 8.40-8.36 (m, 4H), 7.79 (ddd, J = 0.12, 0.02, and 0.02 Hz, 4H),
5.39 (s, 1H), 4.37-4.22 (m, 1H), 2.82-2.65 (m, 2H), 2.27-2.16 (m, 2H), 1.84-1.65 (m, 4H).
Synthesis of trans-4-Nitro-benzoic acid 4-amino-cyclohexyl ester
[0224] 35 wt% Hydrazine hydrate (0.93 g, 10.1 mmol) was added to a solution of the above
compound (2.0 g, 5.1 mmol) in DCM (50 mL) followed by MeOH (50 mL) at room
temperature. The reaction mixture was allowed to stir overnight. The resulting white
precipitates were filtered off and the solvent was removed in vacuo. The resulting white
solids were dissolved in aqueous 1N HCI solution and washed with DCM. Aqueous layer
was basified with excess IN NaOH solution and then extracted with DCM. After drying with
MgSO4, the solvent was evaporated affording crude trans-4-Nitro-benzoic acid 4-amino-
cyclohexyl ester as a solid, which was used in the next step without further purification.
Yield: 1.1 g (89% of theory). 1H NMR (DMSO-d6) δ 8.26 (dd, J = 43.5 and 8.8 Hz, 4H),
6.72 (d, J = 7.3 Hz, 2H), 5.08 (s, 1H), 2.00-1.36 (m, 9H).

WO 2006/045119 PCT/US2005/038282
70
Synthesis of trans-4-Nitro-benzoic acid 4-(3-tricyclo[3.3.L13,7]decan-l-yl-ureido)-
cyclohexyl ester 1076
[0225] To a solution the above compound (1.33 g, 5.1 mmol) in DMF was added 1-
adamantyl isocyanate (0.82 g, 4.6 mmol) followed by triethylamine (0.47 g, 4.6 mmol) at 0
°C. The reaction mixture was stirred overnight. The reaction mixture was poured into water,
and the resulting precipitates were collected and washed with water. The crude product was
recrystallized from DCM/hexanes. Yield: 1.83 g (90% of theory). M.P.:124°C. 1H NMR
(300 MHz, CDC13): δ 8.24 (dd, J = 28.7 and 9.1 Hz, 4H), 5.23 (s, 1H), 4.13 (d, J = 7.2 Hz,
1H), 4.05 (s, 1H), 3.75-3.61 (m, 1H), 2.17-1.41 (m, 23H).
Synthesis of cis-l-(4-Hydroxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-l-yl-urea 1077
[0226] To a solution of ester 1076 (1 g, 2.3 mmol) in THF (100 mL) was added IN NaOH
solution (4.6 mL, 4.6 mmol) at room temperature. The reaction mixture was stirred
overnight, at which time the reaction was quenched by addition of IN HC1 solution (5.5 mL).
The resulting white precipitate was collected by filtration and recrystallized from
methanol/water. Yield: 0.63 g (95% of theory). M.P.: 205 °C. 1H NMR (300 MHz, DMSO-
d6): δ 5.67 (d, J = 8.2 Hz, 1H), 5.45 (s, 1H), 4.41 (s, 1H), 3.63-3.51 (m, 1H), 3.46-3.36 (m,
1H), 2.00-1.92 (m, 3H), 1.87-1.72 (m, 6H), 1.66-1.28 (m, 14H).
Synthesis ofcis-l-(4-Benzyloxy-cyclohexyl)-3-tricyclo[3.3.1.13,7]decan-1-yl-wea 1078
[0227] Compound 1078 (2.22 g, 60%) was synthesized from compound 1077 (0.29 g, 1
mmol) by the same method) as that described for compound 1032 with benzyl bromide (0.20
g, 1.2 mmol) and 60% sodium hydride (0.06 g, 1.5 mmol). Yield: 0.35 g (92% of theory).
M.P.: 181 °C. 1H NMR (300 MHz, CDC13): δ 7.43-7.24 (m, 5H), 4.49 (s, 2H), 4.11 (d, J =
8.3 Hz, 1H), 4.02 (s, 1H), 3.66-3.51 (m, 2H), 2.23-1.07 (m, 23H).
Example 31

WO 2006/045119 PCT/US2005/038282
71
General procedure for the synthesis of cis-phenoxy isomers: Synthesis ofcis-l-[4-(4-
Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7] decan-1-yl-urea 1135
Synthesis of cis-2-[4-(4-Fluoro-phenoxy)-cyclohexyl]-isoindole-l,3-dione
[0228] To a solution of trans-2-(4-Hydroxy-cyclohexyl)-isoindole-l,3-dione 3 (1.0 g, 4.1
mmol), triphenylphosphine (1.3 g, 4.9 mmol), and 4-fluorophenol (0.55g, 4.9 mmol) in 40
mL of THF was added dropwise at room temperature diisopropyl azodicarboxylate (0.99 g,
4.9 mmol). The reaction mixture was stirred overnight. The solvent was evaporated after 12
h, and the resulting solid was purified by recrystallization from methanol. Yield: 1.1 g (80%
of theory). 1H NMR (300 MHz, CDC13): δ 7.84-7.80 (m, 2H), 7.71-7.67 (m, 2H), 6.98-6.94
(m, 4H), 4.51 (s, 1H), 4.26-4.12 (m, 1H), 2.76 - 2.60 (m, 2H), 2.18 (d, J = 13 Hz, 2H), 1.79-
1.49 (m,4H).
Synthesis ofcis-l-[4-(4-Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7]decan-1-yl-urea
1135
[0229] 35 wt% Hydrazine hydrate (0.27 g, 2.9 mmol) was added to a solution of the above
compound (0.5 g, 1.5 mmol) in DCM (15 mL) followed by MeOH (15 mL) at room
temperature. The reaction mixture was allowed to stir overnight The resulting white
precipitates were filtered off and the solvent was removed in vacuo. The resulting white
solids were dissolved in aqueous 1N HC1 solution and washed with DCM. Aqueous layer
was basified with excess 1N NaOH solution and then extracted with DCM. After drying with
MgSO4, the solvent was evaporated affording crude cis-4-(4-Fluoro-phenoxy)-
cyclohexylamine 10 as a solid, which was used in the next step without further purification.
[0230] To a solution compound 10 in DMF was added 1-adamantyl isocyanate (0.16 g,
0.91 mmol) followed by triethylamine (0.10 g, 1.0 mmol) at 0 °C. The reaction mixture was
stirred overnight. The reaction mixture was poured into water, and the resulting precipitates
were collected and washed with water. The crude product was recrystallized from
DCM/hexanes. Yield: 0.31 g (88% of theory). M.P.: 207 °C. 1H NMR (300 MHz, CDCl3):
δ 6.98-6.91 (m, 2H), 6.84-6.78 (m, 2H), 4.34 (s, 1H), 4.30 (d, J = 9.8 Hz, 1H), 4.20 (s, 1H),
3.71-3.56 (m, 1H), 2.13 -1.44 (m, 23H).

WO 2006/045119 PCT/US2005/038282
72
Example 32
General procedure for the synthesis of trans-phenoxy isomers: Synthesis of trans-1-[4-(4-
Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7]decan-l-yl-urealVi6.
Synthesis of cis-2-(4-Hydroxy-cyclohexyl)-isoindole-l,3-dione
[0231] IN NaOH solution (19 mL, 19 mmol) was added at room temperature to a solution
of ester 4 (5 g, 12.7 mmol) in THF (100 mL). The mixture was stirred overnight at room
temperature, at which time the reaction was quenched by addition of IN HC1 solution (40
mL). The solvent was removed under reduced pressure, and the resulting white precipitate
formed was collected by filtration and dissolved in DMF. After adding triethylamine (6.5 g,
64 mmol) at room temperature, the reaction mixture heated at 150 °C for 30 min in the
microwave. After cooling to r.t., the reaction mixture was poured into water and then
extracted with ether. The organic layer was washed with water thoroughly. After drying
with MgSO4, the solvent was removed in vacuo. The resulting white solids were
recrystallized from DCM/hexanes. Yield: 1.9 g (60% of theory). 1H NMR (300 MHz,
CDC13): δ 7.76 (ddd, J = 37.6, 5.4 and 3.1 Hz, 4H), 4.21-4.07 (m, 2H), 2.72-2.55 (m, 2H),
1.96 (d, J = 13.9 Hz, 2H), 1.73-1.50 (m, 4H).
Synthesis of trans-2-[4-(4-Fluoro-phenoxy)-cyclohexyl]-isoindole-1,3-dione
[0232] Synthesis of trans-2-[4-(4-Fluoro-phenoxy)-cyclohexyl]-isoindole-l,3-dione was
synthesized from the above compound (1 g, 4.1 mmol) by the same method as that described
with DIAD (0.99g, 4.9 mmol), PPh3 (1.3 g, 4.9 mmol), and 4-fluorophenol (0.55g, 4.9
mmol). Yield: 0.56 g (40% of theory). 1H NMR (300 MHz, CDCl3): δ 7.77 (ddd, J = 37.6,
5.4 and 3.1 Hz, 4H), 7.00-6.84 (m, 4H), 4.30-4.15 (m, 2H), 2.48-2.31 (m, 2H), 2.26 (d, J =
11.4 Hz, 2H), 1.89-1.77 (m, 2H), 1.65-1.49 (m, 4H).
Synthesis oftrans-l-[4-(4-Fluoro-phenoxy)-cyclohexyl]-3-tricyclo[3.3.1.13,7]decan-l-yl-
urea 1136

WO 2006/045119 PCT/US2005/038282
73
[0233] Compound 1136was synthesized (0.5 g, 1.5 mmol) by the same method as that
described above with 35 wt% hydrazine (0.27g, 2.95 mmol) in 15 mL of DCM and 15 mL of
MeOH followed by the reaction with 1-adamantyl isocyanate (0.12g, 0.67 mmol) and
triethylamine (0.07g, 0.74 mmol) in DMF (5 mL). Yield: 0.24 g (93% of theory). M.P.:
243 °C. 1HNMR (300 MHz, CDC13): δ 6.98-6.91 (m, 2H), 6.85-6.79 (m, 2H), 4.12-3.94 (m,
3H), 3.66-3.51 (m, 1H), 2.17-1.88 (m, 12H), 1.73-1.45 (m, 9H), 1.28-1.11 (m, 2H).
Example 33
Combinatorial synthesis of urea inhibitors
[0234] A mixture of PS-Indole-CHO resin (0.5 g, 0.46 mmol), THF (3 mL), TEOF (3 mL)
and the primary amine (1.0 mmol) was agitated at ambient temperature for 4 h. Then, a
solution of NaBH3CN (1.0 mL, 1 M) in THF and acetic acid (O.lmL) were added. The
resulting mixture was stirred for 2h. The supernatant liquid was drained off and the resin
washed with THF (8 mL*2), MeOH (8 mL*3) and DCM (8 mLx2).
[0235] To a suspension of the resin-bound secondary amine (0.1 g) in DCM (2 mL) was
added an isocyanate (0.5 mmol). The resulting mixture was agitated overnight at ambient
temperature. The supernatant liquid is drained off and the resin washed with DMF, MeOH
and DCM.
[0236] The resin-bound product was suspended hi 1 % (v/v) TFA (3 mL, ca. 4 mol equiv.)
and the mixture agitated at ambient temperature for 4 h. The color of the resin became deep
purple. The supernatant liquid was collected and the resin was washed with DCM (2x2 mL).
The combined solution was concentrated to afford pure products in excellent yields.
Example 34
[0237] This example provides assays and illustrates the inhibition of mouse and human
soluble epoxide hydrolases by compounds of the invention having a secondary
pharmacophore that is a carboxylic acid or carboxylic methyl ester functional group.
Enzyme preparation
[0238] Recombinant mouse sEH and human sEH were produced in a baculovirus
expression system and purified by affinity chromatography. The preparations were at least
97% pure as judged by SDS-PAGE and scanning densitometry. No detectable esterase or
glutathione transferase activity, which can interfere with this sEH assay, was observed.

WO 2006/045119 PCT/US2005/038282
74
Protein concentration was quantified by using the Pierce BCA assay using Fraction V bovine
serum albumin as the calibrating standard.
IC50 Assay conditions
[0239] IC50 values were determined in one of two method. One method uses racemic 4-
mtophenyl-trans-2,3-epoxy-3-phenylpropyl carbonate as substrate. Enzymes (0.12 fiM
mouse sEH or 0.24 μM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer, 0.1 M pH 7.4, at 30°C before substrate introduction([S] = 40 μM). Activity
was assessed by measuring the appearance of the 4-nitrophenolate anion at 405 nm at 30°C
during 1 min (Spectramax 200; Molecular Devices). Assays were performed in triplicate.
IC50 is a concentration of inhibitor, which reduces enzyme activity by 50%, and was
determined by regression of at least five datum points with a minimum of two points in the
linear region of the curve on either side of the IC50. The curve was generated from at least
three separate runs, each in triplicate, to obtain the standard deviation (SD) given in Table 1
thru Table 4.
[0240] Other IC50 values were determined using the procedure described in Analytical
Biochemistry 343 66-75 (2005) using cyano(6-methoxy-naphthalen-2-yl)methyl trans-[(3-
phenyloxiran-2-yl)methyl] carbonate as a substrate (see Table 18b). Enzymes (0.88 nM for
murine and 0.96 nM for human sEH) were incubated with inhibitors ([T] = 0.5-10,000 nM)
for 5 min in BisTris-HCl buffer (25 mM, pH 7.0, containing 0.1 mg/ml of BSA) at 30 °C
prior to substrate introduction ([S] = 51M). Enzyme activity was measured by monitoring the
appearance of 6-methoxy-2-naphthaldehyde. Assays were performed in triplicate. By
definition, IC50 values are concentrations of inhibitor that reduce enzyme activity by 50%.
IC50 values were determined by regression of at least five datum points, with a minimum of
two datum points in the linear region of the curve on either side of the IC50 values. The curve
was generated from at least three separate runs, each in triplicate.
[0241] Assays were conducted with the compounds indicated in Table 1, as described
above.

WO 2006/045119 PCTYUS2005/038282
75
Table 1: Inhibition of mouse and human sBH by l-cyclohexyl-3-n-(substituted)alkylureas

a Enzymes (0.12 uM mouse sEH and 0.24 uM human sEH) were incubated with
inhibitors for 5 min in sodium phosphate buffer (pH 7.4) at 30°C before substrate
introduction ([S] = 40μM). Results are means ± SD of three separate experiments.
[0242] As can be seen from the above table, the conversion of a carboxylic acid function to
its methyl ester (549, 635, and 774) increased inhibition potency for both mouse and human
i
sEHs. Moreover, the methyl ester of butanoic acid (774) showed 8-100 fold higher activity
than the esters of acetic and propanoic acids (549 and 635) for both enzymes, indicating that
a polar functional group located three carbon units (carbonyl on the fourth carbon, about 7.5
angstroms from the urea carbonyl) from the carbonyl of the primary urea pharmacophore can
be effective for making potent sEH inhibitors of improved water solubility, hi addition, the

WO 2006/045119 PCT/US2005/038282
76
distance from the carbonyl of the primary urea pharmacophore to the secondary ester
pharmacophore in compound 854 is about 8.9 A showing that the secondary pharmacophore
may be located about 7 A to about 9 A from the carbonyl of the primary urea pharmacophore
group.
Example 35
[0243] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention having a secondary pharmacophore, with
comparison to compounds having only a primary pharmacophore. As can be seen from the
results in Table 2, the activity is relatively consistent.
[0244] Assays were conducted with the compounds indicated in Table 2, according to
established protocols (see, above).
Table 2: Inhibition of mouse and human sEH by l-cycloalkyl-3-alkylureasa


WO 2006/045119 PCT/US2005/038282
77

Enzymes (0.12 μM mouse sbn and 0.24 uM human sEH) were incubated with inhibitors for 5 mm in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40μM). Results are means ± SD of three
separate experiments.
[0245] As shown in the above table, the substitution at R with a cyclohexyl (772) or
adamantyl (789) increased inhibitor potency 10-fold over the 3-chlorophenyl analog (767, see
Table 3 below). Furthermore, these compounds functionalized with a polar group were as
active and potent as non-functionalized lipophilic inhibitors (for example, 791, 790,297, and
686) for both murine and human enzymes. Adding polar groups to compounds generally
increases their water solubility, and this was the case when one compares compounds 772 or
789 to 791 and 790. In addition, stripping water of hydration out of the enzyme catalytic site
requires about the same amount of energy that is gained by forming a new hydrogen bond
between the inhibitor and the enzyme. Thus addition of polar groups which hydrogen bond
to a target enzyme does not dramatically increase potency if the inhibitor is already potent.
However, the presence of an additional polar group can be expected to dramatically increase
specificity by decreasing hydrophobic binding to biological molecules other than the primary
target (sEH). In this way combining several active pharmacophores into a single molecule
often has a massive increase in specificity and biological activity in complex biological
systems.
Example 36
[0246] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention having a secondary pharmacophore that is a
ketone, amide, alcohol, carbonate, carbamate, urea, carboxylate ester functional group.
[0247] Based on the initial activity shown in Table 1, urea compounds were prepared
having a polar carbonyl group located approximately 7.5 angstroms from the carbonyl of the
primary urea pharmacophore to improve water solubility of lipophilic sEH inhibitors (192

WO 2006/045119 PCT/US2005/038282
78
and 686). The table below shows various functionalities such as ketone, ester, amide,
carbonate, carbamate, and urea which contribute a carbonyl group, and are termed as the
secondary pharmacophores. To determine the effect for each of the secondary
pharmacophores, a 3-chlorophenyl group was held constant as one of substituents of the urea
pharmacophore. The 3-chlorophenyl group is also particularly useful for monitoring
chemical reactions quickly via chromatography. After optimizing the secondary
pharmacophore, the aryl substituent can be replaced by a cyclohexyl, adamantyl or other
group leading to more potent inhibitors.
[0248] Assays were conducted with the compounds indicated in Table 3, according to
established protocols (see, above).
Table 3: Inhibition of mouse and human sEH by l-(3-chlorophenyl)-3-(2-alkylated
ethyl)ureasa


WO 2006/045119 PCT/US2005/038282
79

a Enzymes (0.12 μM mouse sEH and 0.24 uM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40μM). Results are means ± SD of three
separate experiments.
[0249] When the left of the carbonyl (X) is a methylene carbon, the best inhibition was
obtained if a methylene carbon (ketone, 794) or oxygen (ester, 767) is present in the right
position (Y). The ester bond can be stabilized by stearic hindrance of the alcohol or acid
moiety or both (805). The presence of nitrogen (amide, 768) reduced the activity. In
compounds with an oxygen in the left of the carbonyl group, a > 10-fold drop in activity was
observed and there was not any change in tide activity even if the right position, Y, was
modified with a methylene carbon (ester, 761), oxygen (carbonate, 760), or nitrogen
(carbamate, 762), respectively. All compounds (765,777, and 766) with nitrogen in the left
position had lower activities than 794 or 767. Comparing compounds 767 and 761, the
presence of a methylene carbon around the carbonyl showed a very different effect on the
inhibition activity. The compound with a methylene carbon in the left of the carbonyl (767)
showed a 20-fold better inhibition than that in the right (761). While the rank-order potency
of this inhibitor series was equivalent with mouse and human sEH, a 3-5-fold higher
inhibition potency was observed for the murine enzyme.
Example 37
[0250] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention having no secondary pharmacophore, but having a
tertiary pharmacophore that is an amide or a carboxylate ester functional group (with alkyl,
alkenyl, alkynyl, cycloalkyl and arylalkyl ester groups).
[0251] Compound 687, having a carboxylic acid group at the end of twelve carbon chain,
was found to be an excellent inhibitor of both the mouse and human enzymes. Additionally,
an ester found to be a suitable secondary pharmacophore. As a result, a variety of ester
derivatives having a carbonyl group located eleven carbon units from the urea
pharmacophore were synthesized and evaluated to examine contributions of a tertiary
pharmacophore.

WO 2006/045119 PCT/US2005/038282
80
[0252] Assays were conducted with the compounds indicated in Table 4, according to
established protocols (see, above).
Table 4: Inhibition of mouse and human sEH by l-(l-adamantyl)-3-(l 1-alkylated undecyl)-
ureasa


WO 2006/045119 PCT/US2005/038282
81
a Enzymes (0.12 μM mouse sEH and 0.24 μM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40μM). Results are means ± SD of three
separate experiments.
[0253] While the presence of a polar group at the end of a shorter chain reduced inhibition
potency for both enzymes (see Table 1), when the carboxylic acid was modified to esters with
various aliphatic groups (780,784,783,781,788,800,785,801,802, and 803) inhibition
potencies were as high as that of the acid (687) for both enzymes. Ethyl (792) and isopropyl
(793) amide derivatives were also potent inhibitors. Compounds with methyl-branched
aliphatic chains were also potent (785, 801,802, 803, and 793). Still further, larger bulky
group such as 1-adamantylmethyl (786), benzyl (804), 2-chlorobenzyl (782) or 2-
naphthylmethyl (787) provided good levels of activity, although slightly reduced (1.5-3-fold)
for both enzymes. These results identified an additional site within the sEH inhibitor
structure which allows the inclusion of a third polar function, i.e. a tertiary pharmacophore.
Example 38
[0254] In order to further explore the effects of functional groups which improve water
solubility on the bioavailability and potency of the inhibitors, seven amide derivatives of
compound 687 with various functionalities were synthesized. As shown in Table 5, alkyl,
sulfonyl, lipoamino acid, and glucosylamide derivatives were prepared, and their inhibition
potency on sEHs, melting point, and solubility in water and oil were examined. For mouse
sEH, there was no change in the inhibition activity when the acid function of compound 687
was substituted by an alkyl (792 and 793), sulfonyl (848 and 914), lipoamino acid (1001), or
glucosyl (1002 and 798) groups. Interestingly, for human sEH, significant differences in
inhibition potencies, which were measured by the spectrometric and fluorescent based assays,
were observed when replacing the acid of compound 687 by the substituents. While
introduction of the ethyl amide group (792) resulted in a 1.5-fold decrease in potency, the
isopropyl analogue (793) exhibited very similar potency to ADUA (687). The corresponding
ester derivatives 784 and 785 in Table 5 exhibited a 2-3-fold improvement in inhibition of the
human enzyme. Two sulfonylamides with a methyl (848) or phenyl (914) group improved the
inhibition potency of compound 4 about 1.5-fold. On the other hand, a 25-fold and 3-fold
decrease in potency was observed when the sulfonylamide was replaced by a lipoamino acid
(1001) or glucosyl groups (1002 and 798), respectively. This suggests that such bulky, and in
the case of glucose, highly polar functional groups are not effective in retaining the inhibition

WO 2006/045119 PCT/US2005/038282
82
potency on human sEH. Comparing compounds 1002 and 798, similar inhibition on the
human enzyme was shown in these two compounds although an octyl group is present
between the ADUA and sugar moieties of compound 1002, suggesting that relatively
lipophilic alkyl groups located around the amide function do not alter the binding activity of
inhibitors to the enzyme. Melting points of most of the amides in Table 5 were measured in a
range of 100-140°C, which was similar to 114°C of the acid compound 687, while the ester
derivates of compound 687 in Table 5 showed 23-66°C lower melting points than that of
compound 687. An exception was compound 1001 for which we were unable to obtain a
crystalline solid at room temperature. These results imply that the amide function is not as
useful in reducing the melting point of AUDA derives as the esters. As with the esters
prepared for this study, the amides failed to result in a large increase in potency on the target
enzyme. Thus any advantage of these amides would result from improvements in ease of
formulation, oral availability and pharmacokinetics. When water solubility of three
compounds (848,1001, and 1002) was compared to that of compound 4, a 3-fold lower or
similar solubility was shown in compounds with a rnethanesulfonyl (848) or glucosyl (1002)
group, respectively. Compound 1001, the lipoamino acid conjugate, surprisingly had a 2.5-
fold better water solubility than compound 687. The 25-fold decrease in the inhibitory
potency of compound 1001 compared to 687 indicates that with compound 1001 the
biological activity will be attributed largely to the production of AUDA rather than being due
to both AUDA and its ester or amide. In addition, no valuable improvement in oil solubility
was obtained in the amides. Although significant improvements in the properties of the
compounds were not observed in the seven amide derivatives, their relative stability and
inhibition potency is sufficient to encourage the further exploration of other amide
compounds to develop bioavailable inhibitors with improved physical properties. We have
reported that modification of the urea pharmacophore of potent sEH inhibitors to an amide
functionality does not dramatically alter the inhibition potency and that at least a 10-fold
improvement hi water solubility and a decrease in the melting points of these amide inhibitors
is observed.( Kim, et al. J. Med. Chem. 2005, 48, 3621-3629). This suggests that the
inhibition potency and physical properties of the amide derivatives in Table 5 might be
improved with the modification of the urea function to the corresponding amide
pharmacophore.

WO 2006/045119 PCT/US2005/038282
83
Table 5. Inhibition of mouse and human sEH by 12-(3-adamantan-l-yl-ureido)dodecanoic
acid alkyl or sulfonylamide derivatives

a Spectrometric-based assay: enzymes (0.12 μM mouse sEH or 0.24 uM human sEH) were
incubated with inhibitors for 5 min in sodium phosphate buffer (200 uL; pH 7.4) at 30°C
before substrate introduction ([S] = 40 uM), results are means ± SD of three separate
experiments.
b Fluorescent-based assay: enzymes (0.88 nM mouse sEH or 0.96 nM human sEH) were
incubated with inhibitors for 5 min in Bis-Tris/HCl buffer (25mM; pH 7.0) at 30°C before
substrate introduction ([S] = 5 uM), results are means ± SD of three separate experiments
c Melting point

WO 2006/045119 PCT/US2005/038282
84
Example 39
[0255] This example provides assays and illustrates the inhibition of mouse and human
soluble epoxide hydrolases by compounds of the invention having a both a secondary and
tertiary pharmacophore that is a carboxylic ester functional group.
[0256] Assays were conducted with the compounds indicated in Table 6, according to
established protocols (see, above).
Table 6. Inhibition of mouse and human sEH by 4-(3-adamantan-l-yl-ureido)butyryloxy
comoounds

No. n TAa Mouse sEHb Human sEHb MP(°C) cLogP0
lC50 (μM) IC90 (μM) IC50 (μM) IC90 (μM)
857 1 8 0.05±0.01 0.11±0.01 0.39±0.01 9±2 123 0.98±0.47
876 2 9 0.05±0.01 0.63±0.02 0.54±0.05 9±2 95-97 1.27±0.47
858 3 10 0.05±0.01 0.16±0.01 0.12±0.01 5.0±0.1 89-91 1.55±0.47
877 4 11 0.05±0.01 0.10±0.01 0.13±0.01 1.5±0.1 84-86 1.97±0.47
878 6 13 0.05±0.01 0.13±0.01 0.12+0.01 0.81±0.01 65-67 2.81±0.47
879 7 14 0.05±0.01 0.16±0.02 0.11±0.01 0.72±0.01 58-59 3.22± .47
880 9 16 0.05±0.01 0.26±0.03 0.10±0.01 0.68±0.01 60-61 4.06±0.47
881 10 17 0.05±0.01 0.35±0.05 0.10±0.01 1.2±0.1 54-55 4.48±0.47
882 11 18 0.05±0.01 0.63+0.04 0.10±0.01 1.8±0.2 64-65 4.89±0.47
a The total number of atoms extending from the carbonyl group of the primary urea pharmacophore, TA = n + 7
b Enzymes (0.12 μM mouse sEH and 0.24 uM human sEH) were incubated with inhibitors for 5 min in sodium
phosphate buffer (pH 7.4) at 30°C before substrate introduction ([S] = 40 μM). Results are means ± SD of three
separate experiments.
c cLog P: calculated log P by Crippen's method by using CS ChemDraw 6.0 version

WO 2006/045119 PCT/US2005/038282
85
[0257] As can be seen from the above table, in increasing the distance between the
secondary ester phannacphore and the tertiary ester pharmacaphore (549, 635, and 774)
increased inhibition potency for human sEHs but mouse EH activity remained relatively
consistent.
Example 40
[0258] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having a secondary ether
pharmacophore.
[0259] Adamantyl-urea compounds were prepared having a polar ether group located
various distances from the carbonyl of the primary urea pharmacophore. These compounds
were prepared to improve water solubility of lipophilic sEH inhibitors (192 and 686). As can
be seen from the results in Table 7, the activity is relatively consistent.
[0260] Assays were conducted with the compounds indicated in Table 7, according to
established protocols (see, above).
Table 7. Inhibition of mouse and human sEH by alkyl ether derivatives


WO 2006/045119 PCT/US2005/038282
86

[0261] As shown in the above table, these compounds fiinctionalized with a single ether
group could be as active and potent as non-functionalized lipophilic inhibitors (790, see Table
2 above) for both murine and human enzymes. Adding a polar ether group to these
compounds increased their water solubility (compare compound 866-870 with 790). The
distance from the carbonyl of the primary urea pharmacophore to the secondary ether
. pharmacophore in compound 869 is about 8.9 A showing that the secondary pharmacophore
may be located about 7 A to about 9 A from the carbonyl of the primary urea pharmacophore
group.
Example 41
[0262] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having a secondary ether or
polyether pharmacophore, with comparison to compounds further including a tertiary
pharmacophore.
[0263] Because compounds having a ether secondary pharmacophore were found to be
suitable inhibitors of both the mouse and human enzymes, a variety of polyether derivatives
were synthesized and evaluated along with contributions of a tertiary pharmacophore. As can
be seen from the results in Table 8, the activity is relatively consistent.
[0264] Assays were conducted with the compounds indicated in Table 8, according to
established protocols (see, above).

WO 2006/045119 PCT/US2005/038282
87
Table 8. Inhibition of mouse and human sEH by substituted ether derivatives


WO 2006/045119 PCT7US2005/038282
88

[0265] Compounds with from two to four ether groups (908, 950, and 952) had inhibition
potencies that were as high as non-functionalized lipophilic inhibitors (790, see Table 2
above) for both murine and human enzymes, as well as increased water solubility and
improved pharmacokinetics (See Figures 14-24). Including a tertiary pharmacophore were
also potent inhibitors but did not further increase their activity (compare compounds 913 and
940 with 908 and compound 951 with 950).
Example 42
[0266] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having a primary amide
pharmacophore.
[0267] Adamantyl-amide compounds were prepared having a polar secondary
pharmacophore group located various distances from the carbonyl of the primary amide
pharmacophore.
[0268] Assays were conducted with the compounds indicated in Table 9, according to
established protocols (see, above).

WO 2006/045119 PCT7US2005/038282
89
Table 9. Inhibition of mouse and human sEH by adamantyl-amide derivatives

[0269] As shown in the above table, these compounds functionalized with a amide group
could be as active and potent as urea inhibitors for both murine and human enzymes. The
nitrogen to the right of the amide carbonyl group is important for activity.

WO 2006/045119 PCTYUS2005/038282
90
Example 43
[0270] This example illustrates the inhibition of mouse and human soluble epoxide
hydrolases by compounds of the invention (formula (I)) having an arylene or cycloalkylene
linker.
[0271] Because compounds having an alkylene linker between the primary and secondary
pharmacophore were found to be excellent inhibitors of both the mouse and human enzymes,
a variety of admantyl-urea derivatives having a phenyl or cyclohexyl spacer between a
primary urea and secondary pharmacophore were synthesized and evaluated to examine the
contributions of the linker.
[0272] Assays were conducted with the compounds indicated in Table 10, according to
established protocols (see, above).
Table 10. Inhibition of mouse and human sEH by substituted phenyl and cyclohexyl
derivatives


WO 2006/045119 PCT7US2005/038282
91


WO 2006/045119 PCT/US2005/038282
92

[0273] Compounds with alkylene and arylene linker groups (859 and 861) had inhibition
potencies that were higher than compounds with alkylene linkers (789, see Table 2 above,
and 868, see Table 7 above) for both murine and human enzymes, independent of the
topography {compare compound 859 with 860 and compound 861 with 863) or type of the
secondary phannacophore {compare compounds 860 and 863 with 909).
Example 44
[0274] This example illustrates the inhibition of mouse soluble epoxide hydrolases by
compounds of the invention (formula (T)) having a secondary phannacophore, and further
including a mono amino acid moiety. This example further illustrates the use of a
combinatorial approach toward compound preparation and evaluation.
[0275] The utility of a combinatorial approach is illustrated by using the butanoic acid
derivatives from Table 11 and Table 12 to form amide bonds with one or more natural or
synthetic amino acids. This approach rapidly leads to a large number of compounds that are
highly active and can be recognized by the intestinal peptide uptake system. As shown
above, polar groups could be incorporated into one of the alkyl groups of the dialkyl-urea
sEH inhibitors without loss of activity, when placed at an appropriate distance from the urea
function. These modifications give the new inhibitors better solubility and availability. To
expand this assessment of inhibitor structure refinement a semi-combinatorial approach was
used with amino acids. Because amino acids are simple bifunctional synthons with a wide
variety of side chains, mono and di-peptidic derivatives of 4-(3-cyclohexyl-ureido)-butyric
acid 625 were synthesized. This parent compound (acid 625) was selected due to its low
inhibition of sEH. Furthermore, to make the peptidic bond, reactants were used, such as 1-
ethyl-3-(3-(dimethylamino)-propyl) carbodiimide, that themselves or their reaction product,
such as l-efhyl-3-(3-dimethylamino)-propyl urea, are not inhibitors of sEH. Therefore, any
inhibition observed was derived from the targeted peptidic derivatives. This approach allows
the preparation of compounds on an analytical scale (10 μmol) without purification of the

WO 2006/045119 PCT/US2005/038282
93
products. The presence of the desired products was confirmed by LC-MS and the ratio of the
LC-MS peak of the desire compounds with the starting material was used to estimate the
reaction yield. Because each inhibitor presents a single carboxyl group for negative mode
ionization, the estimation of yield is reasonably quantitative.
[0276] Syntheses of amino acid derivatives of 4-(3-cyclohexyl-ureido)-butyric acid (632)
were performed at analytical scale. Reactions were performed in 2mL glass vials for each
amino acid. To 100 μL of a solution of 632 in DMF at 100 mM (10 μmol), 200 μL of a
solution of l-ethyl-3-(3-(dimethylamino)-propyl) carbodiimide in DMF at 100 mM (20
μmol) was added. After 15 minutes reaction at room temperature, 400 μL of amino acid
methyl ester solution at 100 mM (40 umol) in 90:10 DMF:1 N NaOH was added. The
reaction was strongly mixed at 40°C overnight. Three hundred microliters of 1 N NaOH was
then added and allowed to react overnight at 40°C. Product formation was confirmed for
each amino acid using electrospray-ionization mass spectrometry (ESI-MS). Reaction
solutions were used directly for inhibitor potency measurement with a theoretical
concentration of 10 mM.
[0277] Assays were conducted with the compounds indicated in Table 11, according to
established protocols (see, above).
Table 11: Inhibition of mouse sEH by mono-amino acid derivatives of 4-(3-cyclohexyl-
ureido)- butyric acid (632).


WO 2006/045119 PCT/US2005/038282
94
Glycine 285.2 286.6 >50
Histidine 365.2 366.6 1.9 ±0.2
Isoleucine 341.2 342.7 18±3
Leucine 341.2 342.7 >50
Lysine 356.3 357.7 2.2 ±0.5
Methionine 359.2 360.7 >50
Phenylalanine 375.2 376.7 5.6 ± 0.4
Proline 325.2 326.7 >50
Serine 315.2 316.7 >50
Threonine 329.2 330.7 >50
Tryptophane 414.2 415.8 1.6 ±0.2
Tyrosine 391.2 392.8 0.59 ± 0.03
Valine 327.2 328.7 >50
Results are means ± SD of three separate experiments.
[0278] Significant improvement of the inhibition potency was observed for the aromatic
derivatives (phenylalanine, tryptophane and tyrosine), histidine and lysine. Again, without
intending to be bound by theory, it is believed that the specificity of the interaction of the
enzyme with the five peptidic inhibitors listed results from specific pi-pi stacking between
tryptophane 334 (Trp334) located in close proximity to the secondary pharmacophore, and the
aromatic moieties with four of the five amino acids above. This interaction should alter the
fluorescence spectrum of the enzyme. For the lysine derivative, because reaction can occur
with the side chain amino group, the resulting product could resemble the alkyl derivatives
synthesized above with the acid function playing the role of the third pharmacophore.
Example 45
[0279] This example illustrates the inhibition of mouse soluble epoxide hydrolases by
compounds of the invention (formula (I)) having a secondary pharmacophore, and further
including a dipeptide moiety.

WO 2006/045119 PCT/US2005/038282
95
[0280] Compounds in the amino acid derivative series, 625-Tyr, showed an inhibition
potency in the hundreds of nanomolar range, prompting the evaluation of the effect of adding
a second amino acid.
[0281] In a manner similar to that described above, syntheses of amino acid derivatives of
2-[4-(3-Cyclohexyl-ureido)-butyrylamino}-3- (4-hydroxy-phenyl)-propionic acid (632-Tyr)
that are examples of dipetide derivatives of 632 were done on an analytical scale. Synthesis
was performed as described above for the derivatives of 632, simply substituting this
compound by 632-Tyr. Product formation was confirmed by ESI-MS.
[0282] Assays were conducted with the compounds indicated in Table 12, according to
established protocols (see, above).
Table 12: Inhibition of mouse sEH by mono-amino acid derivatives of 4-(3-cyclohexyl-
ureido)- butyryl-tyrosine.


WO 2006/045119 PCT/US2005/038282
96
Methionine 522.8 521.2 2 0.05 2.0
Phenylalanine 538.7 537.5 1 0.05 1.6
Proline 488.6 487.4 1 0.06 6.3
Serine 478.6 477.3 1 0.07 3.3
Threonine 492.6 491.3 4 0.12 12.5
Tryptophane 577.7 576.4 1 0.05 1.0
Tyrosine 554.7 553.4 5 0.05 2.5
Valine 490.6 489.4 2 0.05 3.1
Results are means ± SD of three separate experiments.
[0283] Significant improvement of inhibition potency was observed for almost all the
derivatives tested except for alanine, isoleucine, leucine and threonine. These results indicate
that the enzyme has a narrower specificity close to the catalytic center than toward the end of
the active site tunnel. The inhibition potency found for the best dipeptidic derivatives are
similar to those found for the corresponding alkyl inhibitors (see, C. Morisseau, et al.,
Biochem. Pharm. 63:1599-1608 (2002)), indicating that such peptide-mimics are excellent
inhibitors of sEH. Because of the presence of the amino acid derivatives in their structure,
these compounds have excellent water solubility. Furthermore, because of the presence of
active small peptide transport system in the gut, the dipeptidic urea derivatives will be
absorbed in the gut by such systems as observed for several peptide derivative drugs (see, E.
Walter, et al., Pharm. Res. 12: 360-365 (1995) and K. Watanabe, et al., Biol. Pharm. Bull 25:
1345-1350 (2002)), giving these compounds excellent bioavailability.
Example 46
[0284] This example provides studies directed to the metabolic stability of certain
inhibitors of sEH.
[0285] To evaluate the metabolic stability of these inhibitors, the microsomal and NADPH
dependent metabolism of a number of potent sEH inhibitors was evaluated. The rates of
metabolism among the compounds varied dramatically, however the appearance of an
omega-terminal acid was observed for all inhibitors containing n-alkane substitutions. When

WO 2006/045119 PCT/US2005/038282
97
tested, the potent alkyl derivatives (e.g. 686) are rapidly metabolized in microsomal
preparations by P450 dependents processes (see Figure 6), while the omega acid analogs (e.g.
687) were stable (see Figure 7). The first step in the metabolic transformation of the n-alkyl
to n-alkanoic acid derivatives is an NAPDH dependent process carried out by cytochrome
P450 dependent omega hydroxylation in rodent and human hepatic tissue preparations (see
Figure 8). The metabolites identified along this metabolic route are provided in Table 13.
When in vivo metabolism was evaluated, evidence for the beta-oxidation of the alkanoic acid
derivatives was also found (see Figure 9). Together, these data indicate that P450 omega
hydroxylation can result in the rapid in vivo metabolic inactivation and excretion of these
inhibitors.
Table 13: Structure of metabolites formed from compound 686.

Example 47
[0286] This example provides the structures of compounds of the invention designed to
slow esterase dependent inactivation, block beta-oxidation, block cytochrome P450
dependent omega hydroxylation, or inhibit cytochrome P450 omega hydrolase.
[0287] Beta-oxidation can be blocked in a variety of ways, for example with an alpha
halogen or alpha branched alkyl group (806), cyclopropane (807) or aromatic groups (808),
or by replacing the acid or ester functional groups with alternate functionalities, such as
sulfonamides (809 and 810), which mimic ester and acid functional groups yet provide
metabolic stability in vivo. Similarly in pharmacology heterocyclic groups are used for
hydrogen bond donors and acceptors to mimic carboxylic acids and esters (811). In addition,
P450 omega hydroxylation can be blocked by including acetylene (812), trifluoromethyl
(813), or aryl (814) groups at the terminus of the alkyl chain. This series of inhibitors also

WO 2006/045119 PCT/US2005/038282
98
illustrates that with both the secondary and tertiary phannacophore, replacement can be made
for the carbonyl with other functionalities as hydrogen bond donors and acceptors.
Table 14: Structures of sEH inhibitors designed to prevent beta-oxidation and P450 omega
5 hydroxylation.

Example 48
[0288] This example illustrates a comparison of cyclohexyl and adamantyl groups in
stability and solubility.
[0289] Another consistent observation during the metabolism studies was that the
adamantyl substituent (both 192 and 686 substituted) provided compounds having improved
stability (see Figure 6). Surprisingly the adamantyl compounds were approximately 2x more
soluble than the corresponding cyclohexyl derivatives (772 vs. 789, 791 vs. 790, and 297 vs.
686 see Table 2 for structures). Surprisingly, the LC-MS/MS analyses producing collision
induced dissociation of compounds containing the adamantyl substituent provided extremely

WO 2006/045119 PCT/US2005/038282
99
high abundance ions, which dramatically enhanced the analytical sensitivity for these
inhibitors (see Table 15 below). This enhanced sensitivity is a distinct advantage for drug
metabolism studies using either in vivo or in vitro systems. Moreover, adamantane represents
the smallest diamond nucleus and the adamantyl substituents not only yield compounds of
improved metabolic stability and pharmacokinetic parameters, but also compounds that are
very easy to detect.
Table 15: Calibration curves and detections limit (DL) of inhibitors analyzed by HPLC-
MS/MS.

Example 49
[0290] This example provides the pharmacokinetic studies carried out using compounds of
the present invention.
[0291] The pharmacokinetic properties of some of the most potent sEH inhibitors was
evaluated following oral gavage in mice. As noted above, the use of 1-adamantyl urea
inhibitors afforded exquisite sensitivity, allowing the determination of the determined
pharmacokinetic parameters from serial blood samples collected from individual mice (see
Table 17).

WO 2006/045119 PCT/US2005/038282
100
[0292] Animals. Male Swiss Webster mice, 6 weeks-old, were obtained from Charles
River (CA, USA). After 1-2 week acclimation period, healthy animals were assigned to
study groups based on body-weight stratified randomization procedure. The body weight of
animals used in all the experiments ranged from 28 g to 38 g. Mice were maintained on a 12
h light /12 h dark cycle under controlled temperature and humidity conditions, and food and
water available ad libid ran.
[0293] Administration and measurement. Pharmacokinetic studies in mice used a 5
mg/kg dose of sEH inhibitors dissolved in corn oil and 4% DMSO administered orally.
Serial tail bled blood samples (5-10 aL) were collected in heparinized 1.5 mL tubes at
various time points (0.5,1, 2, 3, 4, 5, 6, and 24 hr) after the administration for measuring
parent compounds and their metabolites by using LC-MS/MS: a Waters 2790 liquid
chromatograph equipped with a 30 X 2.1 mm 3 um C18 Xterra™ column (Waters) and a
Micromass Quattro Ultima triple quadrupole tandem mass spectrometer (Micromass,
Manchester, UK). To the collected samples were added l00μL of distilled water, 25 uL of
internal standard (500 ng/mL; l-cyclohexyl-3-tetradecylurea, CTU), and 500uL of ethyl
acetate. Then the samples were centrifuged at 6000 rpm for 5 min, and the ethyl acetate layer
was dried under nitrogen. The residue was reconstituted in 25 μL of methanol, and aliquots
(5μL) were injected onto the LC-MS/MS system.
[0294] Pharmacokinetic studies using a human subject employed doses of 0.1-1.0 mg/kg of
sEH inhibitors (800) or a 0.3 mg/kg dose of 687 dissolved in olive oil administered orally.
Serial bled blood samples (3-50 uL) were collected from finger tips into 50 uL heparinized
capillary tube at various time points (0.5,1, 2,4,6,12 and 24 hr) after administration. These
samples were used to measure parent compounds and their metabolites using LC-MS/MS as
described above for experiments with mice. Blood samples were added 400 μL of distilled
water and 25 μL of internal standard (500 ng/mL CTU), and vortexed. The blood samples
were then extracted with 500 μL of ethyl acetate twice and the ethyl acetate layer was dried
under nitrogen. The residue was reconstituted in 25 μL of methanol, and aliquots (10 μL)
were injected onto the LC-MS/MS system as described above. Biological end points came
from clinical chemistry samples run at The University of California Davis Clinical
Laboratory and a series of 6 inflammatory markers including C reactive protein were run
blind at the University of California Davis Department of Nephrology.

WO 2006/045119 PCT/US2005/038282
101
[0295] Analysis. Pharmacokinetics analysis was performed using SigmaPlot software
system (SPSS science, Chicago, IL). A one-compartment model was used for blood
concentration-time profiles for the oral gavage dosing and fits to the following equation (see,
Gibson, G.G. and Skett, P.: INTRODUCTION TO DRUG METABOLISM, SECOND ED.,
Chapman and Hall, New York 1994,199-210):
C = ae-bt
The half-life (t1/2) for the elimination phase was calculated by the following equation:
t1/2 = 0.693/b
The area under the concentration (AUC) was calculated by the following equation:
AUC = alb
Where:
- C = the total blood concentration at time t
- a = the extrapolated zero intercept
- b = the apparent first-order elimination rate constant
Table 17: Pharmacokinetic parameters of l-(l-adamantyl)-3-(l 1 -alkylated undecyl)ureasa


WO 2006/045119 PCT/US2005/038282
102

5 mg/kg dosing of compounds were administered orally to male Swill Webster mice, maximum
concentration,c time of maximum concentration,d area under concentration,c half-life.
[0296] The ester compounds were generally hydrolyzed to the acid compound (687) when
administered orally. An example of the time course of free acid appearance is shown in
Figure 10. When compound 687 was administered orally, it reached the maximum
concentration (2-fold higher than 686) in 30 min, while compound 686 reached its maximum
concentration in 2 h. Furthermore, the area under the curve (AUC) for 687 was 2-fold higher,
indicating an improvement in oral bioavailability. The maximum concentrations of primary
esters (780, 784, 783,781, 788, 800, 803 and 804) esters were 1.5-5-fold higher than 687, and
the AUC increased 1.2-2.3-fold for the ester compounds indicating higher bioavailabilities.
On the other hand, secondary esters (785 and 802) showed similar maximum concentrations
and bioavailabilities to those of 687 in mice, while the tertiary ester (801) displayed a 4-8-
fold decrease in maximum concentration and bioavailability. Accordingly, the alkylation of a
potent acid inhibitor (687) to form primary esters improves the oral availability of these
inhibitors.
Example 50
[0297] This example provides a table of structures for compounds of the invention having
all three pharmacophores present.

WO 2006/045119 PCT/US2005/038282
103
Tables 18a and b: Structures and inhibition of mouse and human sEH by other sEH
inhibitors containing the primary, secondary, and tertiary pharmacophores.
Table 18a


WO 2006/045119 PCT7US2005/038282
104


WO 2006/045119 PCTVUS2005/038282
105


WO 2006/045119 PCTYUS2005/038282
106


WO 2006/045119 PCT/US2005/038282
107


WO 2006/045119 PCT/US2005/038282
108


WO 2006/045119 PCT/US2005/038282
109


WO 2006/045119 PCT/US2005/038282
110


WO 2006/045119 PCT/US2005/038282
111


WO 2006/045119 PCT/US2005/038282
112
Table 18b._____________________________________________________________


WO 2006/045119 PCT/US2005/038282
113


WO 2006/045119 PCT7US2005/038282
114


WO 2006/045119 PCT/US2005/038282
115


WO 2006/045119 PCT/US2005/038282
116


WO 2006/045119 PCT/US2005/038282
117


WO 2006/045119 PCT/US2005/038282
118


WO 2006/045119 PCT/US2005/038282
119


WO 2006/045119 PCT/US2005/038282
120


WO 2006/045119 PCT/US2005/038282
121


WO 2006/045119 PCT/US2005/038282
122


WO 2006/045119 PCT/US2005/038282
123


WO 2006/045119 PCT/US2005/038282
124


WO 2006/045119 PCT/US2005/038282
125


WO 2006/045119 PCT/US2005/038282
126


WO 2006/045119 PCTYUS2005/038282
127


WO 2006/045119 PCT/US2005/038282
128


WO 2006/045119 PCT/US2005/038282
129


WO 2006/045119 PCT/US2005/038282
130


WO 2006/045119 PCT7US2005/038282
131


WO 2006/045119 PCT/US2005/038282
132


WO 2006/045119 PCT/US2005/038282
133


WO 2006/045119 PCT/US2005/038282
134


WO 2006/045119 PCT/US2005/038282
135


WO 2006/045119 PCT/US2005/038282
136


WO 2006/045119 PCT/US2005/038282
137


WO 2006/045119 PCT/US2005/038282
138


WO 2006/045119 PCT/US2005/038282
139


WO 2006/045119 PCT/US2005/038282
140


WO 2006/045119 PCT/US2005/038282
141


WO 2006/045119 PCT/US2005/038282
142


WO 2006/045119 PCT/US2005/038282
143


WO 2006/045119 PCT/US2005/038282
144


WO 2006/045119 PCT/US2005/038282
145


WO 2006/045119 PCT/US2005/038282
146


WO 2006/045119 PCT/US2005/038282
147


WO 2006/045119 PCT/US2005/038282
148


WO 2006/045119 PCT/US2005/038282
149


WO 2006/045119 PCT/US2005/038282
150

* Inhibition potencies were determined using a fluorescent based high-throughput assay. Inhibitors in solution at
10 mM in DMSO were serially diluted by 10-fold increment in Bis/Tris HC1 buffer (25 mM PH 7.0) containing
0.1 mg/mL of BSA (Buffer A). In black 96-well plates, 20pL of the inhibitor dilution or buffer were delivered in
every well, and then 130μL of Human sEH at ~0.4 μg/mL in solution in Buffer A were added to each well The
plate was then mixed and incubated at room temperature for 5 minutes. Fifty microliters of substrate ((3-Phenyl-
oxiranyl)-acetic acid cyano-(6-methoxy-naphthalen-2-yl)-methyl ester; PHOME) at 200μM in solution in 96:4
Buffer A:DMSO was then added to each well to give [S]final = 50μM and [E]final= ~4nM. The plate was then

WO 2006/045119 PCT/US2005/038282
151
mixed and incubated in the dark at room temperature (~25°C) for 90 min. Activity was measured by determining
the relative quantity of 6-methoxy-2-naphthaldehyde formed with an excitation wavelength of 316 nm and an
emission wavelength of 460 mn measured with a SpectraMax M-2 fluorometer (molecular Devices, Sunnyvale
CA). Results are not reported.
[0298] The primary urea pharmacophore can be varied (compound #) with amide or
carbamate functionality to improve physical properties of sEH inhibitors as well. The
carbonyls can be replaced by heterocyclic or acyclic hydrogen bond acceptors and donators
as shown in Table 14.
Example 51
[0299] This example shows the effect of sEH inhibitors on serum and urinary oxylipin
profiles in rodents.
[0300] The described soluble epoxide inhibitors have been shown to modulate the relative
abundance and amounts of epoxy and dihydroxy fatty acids formed in treated animals. One
such example of this alteration is provided in Figure 13. In this example, hypertension was
induced in one group of Sprague-Dawley rats by the infusion of angiotensin II (ANGII). A
second group of rats received both ANGII and a subcutaneous injection of the model sEH
inhibitor l-adamantyl-3-(dodecanoic acid) urea (i.e. compound 687). Urine samples were
collected for 24hr post exposure to compound 687 and analyzed for linoleate (Panel A) and
arachidonate (Panel B) derived epoxides and diols using LC/MS/MS. As shown in Figure 13,
ANGII exposure decreased the concentration of both linoleate (EpOMEs) and arachidonate
(EETs) derived epoxides and increased arachidonate derived diols (DHETs) but not linoleate
derived diols (DHOMEs). hi the case of both lipid classes, treating animals with compound
687 resulted in an increase in urinary epoxides, as well as a decrease in diol concentrations.
Example 52
[0301] This example illustrates the effect of certain compounds of the invention on
members of the arachidonic acid cascade.
[0302] For epoxy fatty acid hydrolysis, the soluble epoxide hydrolase prefers substrates
with epoxide moieties that are more distant from the carboxyl terminal. Specifically the
substrate preference decreases in the order of 14,15-EET > 11,12-EET > 8,9-EET >>> 5,6-
EET for the epoxides of arachidonic acid. Independently, the relative substrate turnover of

WO 2006/045119 PCT7US2005/038282
152
the epoxy arachidonates were calculated at 0.1:8.1:14.3 when a 1:1:2 mixture of 8,9-, 11,12-,
and 14,15-EET fatty acid was hydrolyzed to 30% by rat renal cortex cytosol. By considering
the primary pharmacophore of the urea to be a transition-state analog of epoxide hydrolysis,
inhibitors have now been developed which incorporate long aliphatic acids. These
compounds are better substrate and transition state mimics than those incorporating shorter
aliphatic acids. Accordingly, optimal soluble epoxide hydrolase inhibitors can be obtained by
producing compounds with aliphatic acid substituents (i.e. a tertiary pharmacophore) which
are separated from the primary pharmacophore by an equivalent distance as the terminal acid
is separated from the epoxide in optimal substrates. Within the enzyme active site, epoxy
fatty acids have been predicted to exist in an extended or pseudo-linear confirmation.
Therefore, both the epoxy fatty acids and the aliphatic acid containing urea structures were
approximated as two dimensional linear representations and measurements were made on
each species. The critical measurements taken were distances (in angstroms) from the
carboxylate hydroxyl to the urea carbonyl and the urea nitrogens.
[0303] The distance of the carboxylate to the urea function of l-cyclohexyl-3-octanoic acid
is similar to the distance of the epoxide to the carboxylate in 8,9-EET. Therefore, the
calculated inhibitor potencies were normalized to this compound, resulting in a ranked
inhibitor potency. We then correlated epoxide to carbonyl distance with respect to relative
substrate turnover rate to establish a correlative regression. By plotting the relative inhibitor
potency on this graph we find that the distances of the carboxyl to the N' -nitrogen correlate
best with the carboxyl to epoxide oxygen distance. These data further highlight the similarity
between inhibitor and substrate interaction with the soluble epoxide hydrolase.
Programs:
[0304] All structures were drawn and exported as MDL MOL files using
ACD/ChemSketch v 4.55 (5/06/2000) Advanced Chemistry Development Inc., Toronto,
Ontario, Canada). Distance measurements were made on the corresponding MOL file image
using ACD/3D v 4.52 (4/10/2000). Structural optimizations were not used.
Table 19 provides results for this analysis (see also, Figure 12).
Table 19: Linear distances between the primary and secondary pharmacophores of a series of sEH
inhibitors and their rank order potencies with the mouse (MsEH) and human sEHs (HsEH) are
shown in comparison with the epoxide to free acid distances and relative turnover rate of the four
arachidonic acid epoxides with the rat sEH.

WO 2006/045119 PCT/US2005/038282
153


WO 2006/045119 PCT/US2005/038282
154
WHAT IS CLAIMED IS:
1. A compound having a formula:

and their pharmaceutically acceptable salts, wherein
R1 is a member selected from the group consisting of substituted or unsubstituted
alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted
cycloalkylalkyl, substituted or unsubstituted cycloalkylheteroalkyl, substituted
or unsubstituted arylalkyl, substituted or unsubstituted arylheteroalkyl,
substituted or unsubstituted C5-C12 cycloalkyl, substituted or unsubstituted
aryl, substituted or unsubstituted heteroaryl and combinations thereof, wherein
said cycloalkyl portions are monocyclic or polycyclic;
P1 is a primary pharmacophore selected from the group consisting of -C(O)O-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-
,-NHC(O)NH-,--OC(O)NH-, -NHC(O)O-, -NHC(S)NH-, -CH2C(O)NH-,
-NHC(O)CH2-, -C(O)NH-, -NHC(O)-, and

P2 is a secondary pharmacophore selected from the group consisting of -NH-, -
-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -NHC(NH)NH-, -
NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)- and -NHC(S)NH-;
P3 is a tertiary pharmacophore selected from the group consisting of C2-C6 alkenyl,
C2-C6 alkynyl,C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl,
-O(CH2CH2O)q-R2, OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-
C4alkyl-C(O)OR2, -C(O)R2, -C(0)OR2 and carboxylic acid analogs, wherein
R2 is a member selected from the group consisting of hydrogen, substituted or
unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl,
substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl;

WO 2006/045119 PCT/US2005/038282
155
the subscripts n and m are each independently 0 or 1, and at least one of n or m is 1,
and the subscript q is 0 to 6;
L1 is a first linker selected from the group consisting of substituted or unsubstituted
C1-C6 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, substituted
or unsubstituted arylene and substituted or unsubstituted heteroarylene;
L2 is a second linker selected from the group consisting of substituted or unsubstituted
C1-C12 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, substituted
or unsubstituted arylene, substituted or unsubstituted heteroarylene; an amino
acid, a dipeptide and a dipeptide analog; and combinations thereof; or is H
when m is 0.
2. The compound in accordance with claim 1, wherein wherein R1 is
selected from the group consisting of substituted or unsubstituted alkyl, substituted or
unsubstituted heteroalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or
unsubstituted cycloalkylheteroalkyl, substituted or unsubstituted arylalkyl and substituted or
unsubstituted arylheteroalkyl.
3. The compound in accordance with claim 2, wherein R1 is a member
selected from the group consisting of alkyl, heteroalkyl, cycloalkylalkyl, arylalkyl and
arylheteroalkyl, each of which is optionally substituted with from 1 to 2 substitutuents
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, niiro,
haloalkoxy, thioalkyl, phenyl..
4. The compound in accordance with claim 3, wherein R1 is a member
selected from the group consisting of cycloalkyl and aryl, each of which is optionally
substituted with from 1 to 2 substituents each independently selected from the group
consisting of alkyl, halo, haloalkyl, alkoxy, nitro, haloalkoxy, thioalkyl, phenyl.
5. The compound in accordance with claim 4, wherein R1 is selected from
the group consisting of C5-C12 cycloalkyl, phenyl and naphthyl.
6. The compound in accordance with any of the preceeding claims,
wherein P1 is selected from the group consisting of -NHC(O)NH-NHC(NH)NH-, -
NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(NH)-, -C(NH)NH-,-NHC(S)NH-,-NHC(O)CH2-,
CH2C(O)NH-, NHC(O)CH2, NHC(O)- and -C(O)NH-.

WO 2006/045119 PCT/US2005/038282
156
7. The compound in accordance with claim 6, wherein P1 is selected from
the group consisting of-NHC(O)NH-, -C(O)NH- and -NHC(O) -.
8. The compound in accordance any one of the proceeding claims,
wherein P2 is selected from the group consisting of -NB-, -OC(O)O-, -O(CH2CH2O)q-, -
NHC(NH)NH-5 -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(S)NH-, -NHC(S)CH2-,
CH2C(S)NH-, -SC(O)CH2-, - CH2C(O)S-, -SC(NH)CH2-, - CH2C(NH)S-, -NON-,

9. The compound in accordance with claim 8, wherein P2 is selected from
the group consisting of -NH-, -C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)-; and -NHC(S)NH~.
10. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of-NHC(O)NH-, -C(O)NH- and -NHC(O) -; P2 is selected from the
group consisting of-NH-,-C(O)-, -CH(OH)-, -O(CH2CH2O)q-, -C(O)O-, -OC(O)-, -
NHC(NH)NH-, -NHC(NH)CH2-, -CH2C(NH)NH-, -NHC(O)NH-, -OC(O)NH-, -NHC(O)O-,
-C(O)NH-, -NHC(O)-, and -NHC(S)NH-; m is 0, n is 1 and L1 is selected from the group
consisting of unsubstituted C1-C6 alkylene, substituted or unsubstituted C3-C6 cycloalkylene,

WO 2006/045119 PCT/US2005/038282
157
and substituted or unsubstituted arylene; and L2 is selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C12 alkylene, substituted or unsubstituted C3-C12
cycloalkylene, substituted or unsubstituted arylene and substituted or unsubstituted
heteroarylene.
11. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -C(O)NH- and -NHC(O) -; P2 is selected from the
group consisting of-CH(OH)-, -C(O)O-5 -O(CH2CH2O)q-! -OC(O)-, -C(O)NH- and
-NHC(O)-; n and m are each 1; L1 is selected from the group consisting of unsubstituted C1-
C6 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, and substituted or
unsubstituted arylene; L2 is selected from the group consisting of substituted or unsubstituted
C1-C12 alkylene, substituted or unsubstituted C3-C6 cycloalkylene, and substituted or
unsubstituted arylene; and P3 is selected from the group consisting of C2-C6 alkenyl, C2-C6
alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, OR2, -C(O)NHR2, -C(O)NHS(O)2R2,
-NHS(O)2R2, -OC2-C4alkyl-C(O)OR2, -C(O)R2, -C(O)OR2 and carboxylic acid analogs,
wherein R2 is a member selected from the group consisting of hydrogen, substituted or
unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or
unsubstituted heterocyclyl; substituted or unsubstituted aryl and substituted or unsubstituted
aryl C1-C4 alkyl.
12. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; n is 0; m is 1; L1 is
selected from the group consisting of unsubstituted C1-C6 alkylene, substituted or
unsubstituted C3-C6 cycloalkylene, and substituted or unsubstituted arylene; L2 is selected
from the group consisting of substituted or unsubstituted C1-C6 alkylene, substituted or
unsubstituted C3-C12 cycloalkylene, and substituted or unsubstituted arylene and substituted
or unsubstituted heteroarylene; and P3 is selected from the group consisting of C2-C6 alkenyl,
C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl, heterocyclyl, OR2, -C(O)NHR2,
-C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(O)OR2, -C(O)R2, -C(O)OR2 and
carboxylic acid analogs, wherein R2 is a member selected from the group consisting of
hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8
cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or unsubstituted aryl and
substituted or unsubstituted aryl C1-C4 alkyl.

WO 2006/045119 PCT/US2005/038282
158
13. The compound in accordance with claim 1, wherein P3 is selected from
the group consisting of C2-C6 alkenyl, heterocyclyl, OR2, -OC2-C4alkyl-C(O)OR2 and
-C(O)R2, wherein R2 is a member selected from the group consisting of hydrogen, substituted
or unsubstituted C1-C4 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or
unsubstituted heterocyclyl; substituted or unsubstituted aryl and substituted or unsubstituted
aryl C1-C4 alkyl.
14. The compound in accordance with claim 1, wherein P3 is selected from
the group consisting of haloalkoxy, morpholino, dioxothiomorpholino,.
15. The compound in accordance with claim 1, wherein L2 is selected from
the group consisting of substituted or unsubstituted C3-C6 cycloalkylene, substituted or
unsubstituted arylene and substituted or unsubstituted heteroarylene or is H when m is 0.
16. The compound in accordance with claim 15, wherein L2 is selected
from the group consisting of substituted or unsubstituted C3-C6 cycloalkylene and substituted
or unsubstituted heteroarylene or is H when m is 0.
17. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; P2 is selected from the
group consisting of -C(O)O-, -CH(OH)-, -O(CH2CH2O)q-, -OC(O)-, -C(O)NH- and
-NHC(O)-; n and m are each 1; L1 is selected from the group consisting of unsubstituted C1-
C6 alkylene, substituted or unsubstituted C3-C6cycloalkylene, and substituted or
unsubstituted arylene; L2 is selected from the group consisting of substituted or unsubstituted
C1-C6 alkylene; and P3 is selected from the group consisting of -C(O)NHR2,
-C(O)NHS(O)2R2, -NHS(O)2R2, and -C(O)OR2, wherein R2 is a member selected from the
group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl.
18. The compound in accordance with claim 1, wherein P1 is selected from
the group consisting of -NHC(O)NH-, -OC(O)NH- and -NHC(O)O-; n is 0; m is 1; L1 is
selected from the group consisting of unsubstituted C1-C6 alkylene, substituted or
unsubstituted C3-C6cycloalkylene, and substituted or unsubstituted arylene; L2 is selected
from the group consisting of substituted or unsubstituted C1-C6 alkylene; and P3 is selected

WO 2006/045119 PCT/US2005/038282
159
from the group consisting of C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl,
heterocyclyl, OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(0)OR2,
-C(O)R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is a member selected from the
group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl.
19. The compound in accordance with claim 1, wherein R1 is a member
selected from the group consisting of C5-C12 cycloalkyl, wherein said cycloalkyl portions are
monocyclic or polycyclic; P1 is selected from the group consisting of -NHC(O)NH-; P2 is
selected from the group consisting of -O(CH2CH2O)q- and -C(O)O-; P3 is selected from the
group consisting of C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 haloalkyl, aryl, heteroaryl,
heterocyclyl, OR2, -C(O)NHR2, -C(O)NHS(O)2R2, -NHS(O)2R2, -OC2-C4alkyl-C(O)OR2,
-C(O)R2, -C(O)OR2 and carboxylic acid analogs, wherein R2 is a member selected from the
group consisting of hydrogen, substituted or unsubstituted C1-C4 alkyl, substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl; m is 1 and q is 0 to 6; L1
is selected from the group consisting of substituted or unsubstituted C1-C6 alkylene,
substituted or unsubstituted C3-C6 cycloalkylene, and substituted or unsubstituted arylene;
and L2 is selected from the group consisting of substituted or unsubstituted C1-C12 alkylene.
20. The compound in accordance with claim 1, wherein L2 is a dipeptide or
dipeptide analog.
21. The compound in accordance with claim 20, wherein L2 is a dipeptide
having an N-terminal residue selected from the group consisting of Tyr, His, Lys, Phe and
Trp, and a C-terminal residue selected from the group consisting of Ala, Arg, Asp, Gly, He,
Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr and Val.
22. The compound in accordance with claim 1, having the formula:

wherein R1 is a member selected from the group consisting of alkyl, aryl,
ilkylaryl, cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each

WO 2006/045119 PCT/US2005/038282
160
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl;
L2 is selected from the group consisting of phenylene or methylenephenylene,
heteroarylene, optionally substituted with from 1 to 2 substituents each independently
selected from the group consisting of halo and haloalkyl.
23. The compound in accordance with claim 22, having the formula:

24. The compound in accordance with claim 22, having the formula:

25. The compound in accordance with claim 1, having the formula:

wherein R2 is selected from the group consisting of substituted or
unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted heterocyclyl; substituted or
unsubstituted aryl and substituted or unsubstituted aryl C1-C4 alkyl.
26. The compound in accordance with claim 25, having the formula:

wherein R2 is substituted or unsubstituted aryl.
27. The compound in accordance with claim 26, having the formula:


WO 2006/045119 PCT/US2005/038282
161
wherein R1 is a member selected from the group consisting of alkyl, aryl,
alkylaryl, cycloalkyl, cycloalkylaryl, optionally substituted with from 1 to 2 substituents each
independently selected from the group consisting of alkyl, halo, haloalkyl, alkoxy, nitro,
haloalkoxy, thioalkyl and phenyl.
28. The compound having the formula 972, 973, 975,1003,1004,1005,
1006,1011 described in Table 8, the formula 960, 961,981,982,983,984,985,1009,1014
described in Table 10, and the compounds in Tables 9 and 18 and their pharmaceutically
acceptable salts.
29. A method for inhibiting a soluble epoxide hydrolase, comprising
contacting said soluble epoxide hydrolase with an inhibiting amount of a compound of any of
claims 1 to 28.
30. A method of treating diseases modulated by soluble epoxide
hydrolases, said method comprising administering to a subject in need of such treatment an
effective amount of a compound of any of claims 1 to 28.
31. The method in accordance with claim 30, wherein said disease is
selected from the group consisting of hypertension, inflammation, adult respiratory distress
syndrome; diabetic complications; end stage renal disease; Raynaud syndrome and arthritis.
32. The method in accordance with claim 31, wherein said treatment
increases sodium excretion, reduces vascular and renal inflammation, and reduces male
erectile dysfunction.
- 33. The method in accordance with claim 31, wherein said hypertension is
selected from the group consisting of renal hypertension, pulmonary hypertension and hepatic
hypertension.
34. The method in accordance with claim 31, wherein said inflammation is
selected from the group consisting of renal inflammation, vascular inflammation, and lung
inflammation.
35. A method for reducing renal deterioration in a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28.

WO 2006/045119 PCT/US2005/038282
162
36. The method in accordance with claim 35, wherein said renal
deterioration is present in said subject afflicted with diabetes, hypertension or an
inflammatory disorder.
37. A method for inhibiting progression of nephropathy in a subject, said
method comprising administering to said subject an effective amount of a compound of any
of claims 1 to 28.
38. The method in accordance with claim 37 wherein the subject is (a) a
person with diabetes mellitus whose blood pressure is 130/85 or less, (b) a person with
metabolic syndrome whose blood pressure is 130/85 or less, (c) a person with a triglyceride
level over 215 mg/dL, or (d) a person with a cholesterol level over 200 mg/dL.
39. A method for reducing blood pressure in a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28. .
40. The method in accordance with claim 39, said method further
comprising administering to said subject an effective amount of a cis-epoxyeicosantrienoic
acid.
41. The method in accordance with claim 40, wherein said cis-
epoxyeicosantrienoic acid is administered with said compound having formula (I).
42. A method of increasing vasodilation hi a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28.
43. A method of inhibiting the progression of obstructive pulmonary
disease, an interstitial lung disease, or asthma in a subject, said method comprising
administering to said subject an effective amount of a compound of any of claims 1 to 28.
44. The method in accordance with claim 43, wherein said obstructive
pulmonary disease is selected from the group consisting of chronic obstructive pulmonary
disease, emphysema, and chronic bronchitis.

WO 2006/045119 PCT/US2005/038282
163
45. The method in accordance with claim 43, wherein said interstitial lung
disease is idiopathic pulmonary fibrosis or is one associated with exposure to dust.
46. The method in accordance with claim 43, said method further
comprising administering to said subject an effective amount of a cis-epoxyeicosantrienoic
acid.
47. The method in accordance with claim 46, wherein said cis-
epoxyeicosantrienoic acid is administered with said compound having formula (I).
48. A method of reducing vascular inflammation in a subject, said method
comprising administering to said subject an effective amount of a compound of any of claims
1 to 28.
49. A method of reducing renal inflammation in a subject, said method
comprising administering to a subject an effective amount of a compound of any of claims 1
to 28.
50. A method of regulating endothelial cell function in a subject, said
method comprising administering to said subject an effective amount of a compound of any
of claims 1 to 28.
51. A method of decreasing endothelial cell inflammation in a subject, said
method comprising administering to said subject an effective amount of a compound of any
of claims 1 to 28.
52. A pharmaceutical composition comprising a pharmaceutically
acceptable excipient and a compound of any of claims 1 to 28.
53. A method for stabilizing biologically active epoxides in the presence of
a soluble epoxide hydrolase, said method comprising contacting said soluble epoxide
hydrolase with an amount of a compound of any of claims 1 to 28, sufficient to inhibit the
activity of said soluble epoxide hydrolase and stabilize said biologically active epoxide.
54. The method in accordance with claim 53, wherein said contacting is
conducted in an in vitro assay.

WO 2006/045119 PCT/US2005/038282
164
55. The method in accordance with claim 53, wherein said contacting is
conducted in vivo.
56. The method for reducing the formation of a biologically active diol
produced by the action of a soluble epoxide hydrolase, said method comprising contacting
said soluble epoxide hydrolase with an amount of a compound of any of claims 1 to 28,
sufficient to inhibit the activity of said soluble epoxide hydrolase and reduce the formation of
said biologically active diol.
57. The method in accordance with claim 56, wherein said contacting is
conducted in an in vitro assay.
58. The method in accordance with claim 56, wherein said contacting is
conducted in vivo.
59. A method for monitoring the activity of a soluble epoxide hydrolase,
said method comprising contacting said soluble epoxide hydrolase with an amount of a
compound of any of claims 1 to 28 sufficient to produce a detectable change in fluorescence
of said soluble epoxide hydrolase by interacting with one or more tryptophan residues present
in the catalytic site of said sEH.
60. The method in accordance with claim 59, wherein said compound has
an aryl group present in one or more components selected from the group consisting of R1,
L1,P3 and L2.
61. A method of increasing ease of formulation, oral availability, or serum
half-life of a compound comprising covalently attaching a polyether substituent to said
compound.

Inhibitors of the soluble epoxide hydrolase (sEH) are provided that incorporate multiple pharmacophores and are
useful in the treatment of diseases.

Documents:

01641-kolnp-2007-abstract.pdf

01641-kolnp-2007-claims.pdf

01641-kolnp-2007-correspondence others 1.1.pdf

01641-kolnp-2007-correspondence others 1.2.pdf

01641-kolnp-2007-correspondence others.pdf

01641-kolnp-2007-description complete.pdf

01641-kolnp-2007-drawings.pdf

01641-kolnp-2007-form 1.pdf

01641-kolnp-2007-form 3 1.1.pdf

01641-kolnp-2007-form 3.pdf

01641-kolnp-2007-form 5.pdf

01641-kolnp-2007-gpa.pdf

01641-kolnp-2007-international publication.pdf

01641-kolnp-2007-international search report.pdf

01641-kolnp-2007-pct request form 1.1.pdf

01641-kolnp-2007-pct request form.pdf

01641-kolnp-2007-priority document.pdf

1641-KOLNP-2007-(18-04-2012)-CORRESPONDENCE.pdf

1641-KOLNP-2007-(29-02-2012)-ABSTRACT.pdf

1641-KOLNP-2007-(29-02-2012)-AMANDED CLAIMS.pdf

1641-KOLNP-2007-(29-02-2012)-DESCRIPTION (COMPLETE).pdf

1641-KOLNP-2007-(29-02-2012)-DRAWINGS.pdf

1641-KOLNP-2007-(29-02-2012)-EXAMINATION REPORT REPLY RECEIVED.pdf

1641-KOLNP-2007-(29-02-2012)-FORM-1.pdf

1641-KOLNP-2007-(29-02-2012)-FORM-13.pdf

1641-KOLNP-2007-(29-02-2012)-FORM-2.pdf

1641-KOLNP-2007-(29-02-2012)-FORM-3.pdf

1641-KOLNP-2007-(29-02-2012)-FORM-5.pdf

1641-KOLNP-2007-(29-02-2012)-OTHERS.pdf

1641-KOLNP-2007-(29-02-2012)-PETITION UNDER RULE 137-1.pdf

1641-KOLNP-2007-(29-02-2012)-PETITION UNDER RULE 137.pdf

1641-KOLNP-2007-ASSIGNMENT.pdf


Patent Number 256840
Indian Patent Application Number 1641/KOLNP/2007
PG Journal Number 32/2013
Publication Date 09-Aug-2013
Grant Date 01-Aug-2013
Date of Filing 08-May-2007
Name of Patentee THE REGENTS OF THE UNIVERSITY OF CALIFORNIA
Applicant Address 1111 FRANKLIN STREET, 12TH FLOOR, OAKLAND CALIFORNIA
Inventors:
# Inventor's Name Inventor's Address
1 HAMMOCK, BRUCE D. 3134 CHESAPEAKE BAY AVE., DAVIS, CALIFORNIA 95616
2 MORISSEAU, CHRISTOPHE 3376 BONAIRE ROAD, WEST SACRAMENTO, CALIFORNIA 95691
3 WATANABE TAKAHO 4-28-3 TSURUMAKIMINAMI HADANO, KANAGAWA-KEN 257-0002
4 NEWMAN, JOHN, W 1621 PACIFIC DRIVE, DAVIS, CALIFORNIA 95616
5 JONES, PAUL 120 RAVINE DRIVE, APT 63B, MATAWAN, NJ 07747
6 HWANG, SUNG HEE 2730 PORTAGE BAY E. #1801, DAVIS, CA 95616
7 WHETSTONE, PAUL 800 DWIGHT WAY, PO BOX 1986, BERKELEY, CA 94701-1986
8 KIM, IN-HAE 33-7 HANGGUK APARTMENT 203-1003, YEONJE-DONG, BUK-GU, GWANGJU 500-834, SOUTH KOREA
PCT International Classification Number C07D 405/12
PCT International Application Number PCT/US2005/038282
PCT International Filing date 2005-10-20
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/651,487 2004-10-20 U.S.A.