Title of Invention

A NON-IMIDAZOLE ARYLOXYPIPERIDINES COMPOUND

Abstract Substituted non-imidazole aryloxypiperidine compounds, compositions containing them, and methods of making and using them to treat or prevent histamine-mediated conditions.
Full Text NON-IMIDAZOLE ARYLOXYPIPER1DINES
Field of the Invention
The present invention relates to aryloxypiperidines, their synthesis and their
use, for example, for the treatment of disorders and conditions mediated by the
histamine receptor.
Background of the Invention
Histamine [2-(imidazol-4-yI)ethylamine] is a transmitter substance. Histamine
exerts a physiological effect via multiple distinct G-protein coupled receptors. It plays
a role in immediate hypersensitivity reactions and is released from mast cells
following antigen IgE antibody interaction. The actions of released histamine on the
vasculature and smooth muscle system account for the symptoms of the allergic
response. These actions occur at the H, receptor (Ash, A.S.F. and Schiid, H.O., Br.
J. Pharmacol., 1966, 27,427) and are blocked by the classical antihistamines (e.g.
diphenhydramine). Histamine is also an important regulator of gastric acid secretion
through its action on parietal cells. These effects of hisiamine are mediated via the
H2 receptor (Black, J.W., Duncan, W.A.M,. Durant, C.J., Ganeltin, C.R. and Parsons,
E. M., Nature, 1972, 236, 385).and are blocked by H2 receptor antagonists (e.g.
cimetidine). The third histamine receptor —H3— was first described as a
presynaptic autoreceptor in the central nervous system (CMS) (Arrang, J.-M.,
Garbarg, M., and Schwartz, J.-C, Nature 1983, 302, 832) controlling the synthesis
and release of histamine. Recent evidence has emerged showing that the H3
receptors are also located presynaptically as heteroreceptors on serotonerglc,
noradrsnergic, dopaminergic, cholinergic, and GABAergic (gamma-aminobutyric
acid containing) neurons. These H3 receptors have aiso recently been identified in
peripheral tissues such as vascular smooth muscle Consequently there are many
potential therapeutic applications for histamine H3 agonists, antagonists, and inverse
agonists. (See: "The Histamine H3 Reuepior-A Target for New Drugs', Leurs, R., and
Timmerman, H., (Editors), Elsevier, 1998; Morisset et al., Nature, 2000, 408, 860-
864.) A fourth histamine receptor —H4— was recently described by Oda et al., (J.
Biol. Chem., 2000, 275, 36781-36786).
The potential use of histamine H3 agonists in sleep/wake and
arousal/vigilance disorders is suggested based on animal studies (Lin et al, Br. Res.,
1990, 523, 325; Monti et al Eur. J. Pharmacol., 1991, 205, 283). Their use in the
treatment of migraine has also been suggested (McLeod et al Abstr. Society
Neuroscience, 1996, 22, 2010) based on their ability to inhibit neurogenic
inflammation. Other applications could be a protective role in myocardial ischemia
and hypertension where blockade of norepinephrine release is beneficial (Imamura
et al J. Pharmacol. Expt. Ther., 1994, 271, 1259). It has been suggested that
histamine H3 agonists may be beneficial in asthma due to their ability to reduce non-
adrenergic non-cholinergic (NANC) neurotransmission in airways and to reduce
microvascular leakage (Ichinose et al Eur. J. Pharmacol., 1989, 174, 49).
Several indications for histamine H3 antagonists and inverse agonists have
similarly been proposed based on animal pharmacology experiments with known
histamine H3 antagonists (e.g. thioperamide). These include dementia, Alzheimer's
disease (Panula et al Abstr. Society Neuroscience, 1995, 21, 1977), epilepsy
(Yokoyama et al Eur. J. Pharmacol., 1993, 234, 129) narcolepsy, eating disorders
(Machidori et al Brain Research 1992, 590,180), motion sickness, vertigo, attention
deficit hyperactivity disorders (ADHD), learning and memory (Barnes et al Abstr.
Society Neuroscience, 1993, 19, 1813), schizophrenia (Schlicker et al Naunyn-
Schmiedeberg's Arch. Pharmacol., 1996, 353, 290-294); (also see; Stark et ai
Drugs Future, 1996, 21, 507 and Leurs et al Progress in Drug Research, 1995, 45,
107 and references cited therein). Histamine H3 antagonists, alone or in
combination with a histamine H1 antagonist, are reported to be useful for the
treatment of upper airway allergic response (U.S. Patent Nos. 5,217,986; 5,352,707
and 5,869,479). Recently, a histamine H3 antagonist (GT-2331) was identified and
is being developed by Gliatech Inc. (Gliatech Inc. Press Release Nov. 5, 1998;
Bioworld Today, March 2, 1999) for the treatment of CNS disorders.
As noted, the prior art related to histamine H3 ligands has been
comprehensively reviewed ('The Histamine H3 Receptor-A Target for New Drugs",
Leurs, R., and Timmerman, H., (Editors), Elsevier, 1998). Within this reference the
medicinal chemistry of histamine H3 agonists and antagonists was reviewed (see
Krause et al and Phillips et al respectively). The importance of an imidazole moiety
containing only a single substitution in the 4 position was noted together with the
deleterious effects of additional substitution on activity. Particularly methvlation of
the imidazole ring at any of the remaining unsubstituted positions was reported to
strongly decrease activity. Additional publications support the hypothesis that an
imidazole function is essential for high affinity histamine H3 receptor figands (See, Ali
et al J. Med. Chem., 1999, 42, 903 and Stark et al. Drugs Future, 1996, 21, 507 and
references cited therein). However many imidazole containing compounds are
. substrates for histamine methyl transferase, the major histamine metabolizing
enzyme in humans, which leads to shortened half lives and lower bioavailability
(See, Rouleau et al J. Pharmacol. Exp. Ther. 1997, 281, 1085). In addition,
imidazole containing drugs, via their interaction with the cytochrome P450
monooxygenase system, can result in unfavorable biotransformations due to
enzyme induction or enzyme inhibition. (Kapetanovic et al Drug Metab. Dispos.
1984, 12, 560; Sheets et al Drug Metab. Dispos. 1984, 12, 603; Back, et al Br. J.
Pharmacol. 1985, 85, 121; Lavrijsen et al Biochem. Pharmacol. 1986, 35, 1867;
Drug Saf., 1998, 18, 83). The poor blood brain barrier penetration of earlier
histamine H3 receptor ligands may also be associated with the imidazole fragment
(Ganellin et al Arch. Pharm. (Weinheim.Ger.) 1998, 331, 395).
More recently, several publications have described histamine H3 ligands that
do not contain an imidazole moiety. For example; Ganellin et al Arch. Pharm.
(Weinheim.Ger.) 1998, 331, 395; Walczynski et al Arch. Pharm. (Weinheim,Ger.)
1999, 332, 389; Walczynski et al Farmaco 1999, 684; Linney et al J. Med. Chem.
2000, 2362; Tozer and Kalindjian Exp. Opin. Ther. Patents 2000, 10, 1045-1055;
U.S. Patent 5,352, 707; PCT Application WO99/42458, Aug 26, 1999; and European
Patent Application 0978512, Feb 9, 2000.
The compounds of the present invention do not contain the imidazole moiety,
and its inherent liabilities, and maintain potency at the human H3 receptor. Thus in
the present invention receptor binding was determined using the human histamine
H3 receptor (See Lovenberg et al Mol. Pharmacol. 1999, 1107). Screening using the
human receptor is particularly important for the identification of new therapies for the
treatment of human diseas. Conventional binding assays for example are
determined using rat synaptosomes (Garbarg et al J. Pharmacol. Exp. Ther. 1992,
263, 304), rat cortical membranes (West et al Mol. Pharmacol. 1990,, 610), and
guinea pig brain (Korte et al Biochem. Biophys. Res. Commun. 1990, 978). Only
limited studies have been performed previously using human tissue but these allude
to significant differences in the pharmacology of rodent and primate receptors (West
et al Eur. J. Pharmacol. 1999, 233).
We now describe a series of aryloxypiperidines with the ability to modulate
the activity of the histamine receptor, specifically the H3 receptor, without the
(inherent problems associated with the presence of an imidazolyl moiety.
Summary of the Invention
The present invention is directed to a compound of formula (I):

wherein X is O;
n is an integer from 0 to 3;
R5 is C 1-10 alkyl, C 3-8 alkenyl, C 3-8 cycloalkyl, (C 3-8 cycloalkyl) C 1-6
alkyl, (phenyl)C 1-6 alkyl, (phenyl)C 3-6 alkenyl, or (C 1-8 alkylcarbonyl)C 1-8 alkyl;
one of R1, R2, and R3 is G or W, wherein one of the remaining two is
selected from H and halogen, and the third being hydrogen;
G is a nitrogen-containing group selected from one of the following:
–OL1Q, -L2Q. –N(L1Q)R5,-L3C(L1Q)R6R7, -C(L1Q)R6R7

wherein:
L1 is C 2-5 alkylene, C 3-8 cycloalkylene, C 4-6 alkenylene, C 4-6
alkynylene, C2-5 alkanoyl, (phenyl)C 1-6 alkylene, (naphthyl)C 1-6 alkylene, (C 2-5
heteroaryl)C 1-6 alkylene, (phenoxy)C1-6 alkylene, or (C 2-5 heteroaryioxy)C1-6
alkylene;
L2 is C1-6 alkylene, C 3-8 cycloalkylene, C 3-6 alkenylene, C 3-6 alkynylene,
C2-5 alkanoyl, (phenyl)C 1-6 alkylene, (naphthyl)C 1-6 alkylene, (C 1-5
heteroaryl)C 1-6 alkylene, (phenoxy)C1-6 alkylene, (C 1-6 heteroaryloxy)C1-6
alkylene, or (C 1-5 heteroarylthio)C1-6 alkylene;
L3 is C1-6 alkylene, C 2-6 alkenylene, C 2-6 alkynylene, C226 alkanoyl,
(phenyl)C 1-6 alkylene, phenyl, naphthyl, (naphthyl)C 1-6 alkylene, C 1-6
heteroaryl)C 1-6 alkylene, (phenoxy)C1-6 alkylene, (C 1-6 heteroaryloxy)C1-6
alkylene, or C 2-5 heteroaryl;
L4 is C1-5 alkylene;
L5 is C1-5 alkylene;
L6 is C1-5 alkylene;
L7 is C1-5 alkylene or absent;
Q is -NR8R9 or a non-aromatic C 2-15 heterocyclyl ring system containing
at least one nitrogen atom and optionally between 1 and 3 additional
heteroatoms selected from O, S, and N in each ring;
R6 is independently selected from hydrogen, C 1-8 alkyl,
C 1-6 alkoxy, C 2-8 alkenyl, C 3-7 cycloalkyl, (C3-7 cycloalkyl)C1-6 alkylene,
C2-15 heterocyclyl, and (C2-7 heterocyclyl)C1-6 alkylene;
R7 is H, hydroxyl, halo, C 2-6 alkoxy or absent where the carbon linking
L6 and L7 (or bonded to R6) participates in a double bond;
each of R6 and R9 is independently selected from hydrogen, C1-5
alkoxy, C 1-5 alkyl, C 3-8 alkenyl, C 3-7 cycloalkyl, (C3-7 cycloalkyl)C1-5 alkylene,
C2-15 heterocyclyl, phenyl, (C2-15heterocyclyl)C1-5 alkylene, and (phenyl) C1-6
alkylene;
R10 is H, C 1-5 alkyl, C 3-8 alkenyl, C 3-7 cycloalkyl, (C3-7 cycloalkyl)C1-6
alkylene, (C2-15 heterocyclyl)C1-6 alkylene, or (phenyl) C1-6 alkylene;
W is -CN, -CHO, halogen, C1-8 heterocyclyl, (C1-8 heterocyclyl)-O-,
phenoxy, phenyl, (phenyl)C1-5 alkylene-O-, -C(=O)RX, -C(OH)RxRy, C1-8 alkyl,
C1-8 cydoalkyl, or -NRxRy;
wherein each of Rx and Ry is independently selected from H, C1-6 alkyl,
C1-6 alkanoyl, C1-8 heterocyclyl, and phenyl;
wherein each of the above alkyl, alkylene, alkenyl, alkenylene, alkynyl,
alkynylene, heterocyclyl, cycloalkyl, and aryl groups may each be
independently and optionally substituted with between 1 and 3 substituents
selected from halo, amino, nitro, hydroxyl, and C 1-3 alkyl;
wherein substituents of Q can be further selected from carboxamide,
C2-6 alkyl, C1-8 heterocyclyl, N(C1-6 alkyl)C1-8 heterocyclyl), NH(C1-8
heterocyclyl), (C1-3 alkylene)(C1-8 heterocyclyl), O(C1-8 heterocyclyl), O(C1-6
alkyl), O(C3-6 cycloalkyl), phenyl, (C1-3 alkylene) phenyl, N(C1-6 alkyl)(C1-3
alkylene) phenyl, and 0(C1-3 alkylene) phenyl where each of above
heterocyclyl, phenyl, and alkyl groups may be optionally substituted with from
1 to 3 substituents independently selected from halogen, nitro, cyano, and C1
3 alkyl;
or a Dharmaceuticallv acceptable salt, ester, or amide thereof.
The invention also features a pharmaceutical composition comprising a
compound of the invention and a pharmaceutically acceptable carrier, and methods
of preparing or formulating such compositions. A composition of the invention may
further include more than one compound of the invention, or a combination therapy
(combination formulation or administering a combination of differently formulated
active agents).
The invention also provides methods of treating certain conditions and
diseases, each of which methods includes administering a therapeutically effective
(or jointly effective) amount of a comDOund or composition of the invention to a
subject in need of such treatment. The disclosed compounds are useful in methods
Tor treating or preventing neurologic disorders including sleep/wake and
arousal/vigilance disorders (e.g. insomnia and jet lag), attention deficit hyperactivity
disorders (ADHD), learning and memory disorders, cognitive dysfunction, migraine,
neurogenic inflammation, dementia, mild cognitive impairment (pre-dementia),
Alzheimer's disease, epilepsy, narcolepsy, eating disorders, obesity, motion
sickness, vertigo, schizophrenia, substance abuse, bipolar disorders, manic
disorders and depression, as well as other histamine H3 receptor mediated disorders
such as upper airway allergic response, asthma, itch, nasal congestion and allergic
rhinitis in a subject in need thereof. For example, the invention features methods for
preventing, inhibiting the progression of, or treating upper airway allergic response,
asthma, itch, nasal congestion and allergic rhinitis.
In yet another embodiment, the disclosed compounds may be used in a
combination therapy method including administering a jointly effective dose of an H3
antagonist and administering a jointly effective dose of a histamine H1 antagonist,
such as loratidine (CLARITIN™), desloratidine (CLARINEX™), fexofenadine
(ALLEGRA™) and cetirizine (ZYRTEC™), for the treatment of allergic rhinitis, nasal
congestion and allergic congestion.
In yet another embodiment, the disclosed compounds may be used in a
combination therapy method, including administering a jointly effective dose of an H3
antagonist and administering a jointly effective dose of a neurotransmitter re-uptake
blocker, such as a selective serotonin re-uptake inhibitor (SSRI) or a non-selective
seretemin,dopamine or norepinephrine re-uptake inhibitor, including fiuoxetine
(PROZAC™), Setraline (ZOLOFT™), paroxetine (PAXIL™) and amitryptyiine, for
the treatment of depression, mood disorders or schizophrenia.
Additional features and advantages of the invention will become apparent
from the detailed description and examples below, and the appended claims.
Detailed Description of the Invention
The present invention provides aryloxypiperidine derivatives useful for the
treatment of disorders and conditions modulated by a histamine receptor.
A. Terms
Certain terms are defined below and by their usage throughout this disclosure.
As used herein, "halogen" shall mean chlorine, bromine, fluorine and iodine, or
monovalent radicals thereof.
As used herein, the term "alkyl", whether used alone or as part of a
substituent group, shall include straight and branched carbon chains. For example,
alkyl radicals include methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, t-
butyl, pentyi and the like. Unless otherwise noted, "lower" when used with alkyl
means a carbon chain composition of 1-4 carbon atoms. "Alkylene" refers to a
bivalent hydrocarbyl group, such as methylene (CH2), ethylene (-CH2-CH2-) or
propylene (-CH2CH2CH2-).
As used herein, unless otherwise noted, "alkoxy" shall denote an oxygen ether
radical of the above described straight or branched chain alkyl groups. For example,
methoxy, ethoxy, n-propoxy, sec-butoxy, t-butoxy, n-hexyloxy and the like.
As used herein, unless otherwise noted, "cycloalkyl" shall denote a three- to
eight -membered, saturated monocyclic carbocyclic ring structure. Suitable examples
include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
As used herein, unless otherwise noted, "cycloalkenyl" shall denote a three- to
etght-membered, partially unsaturated, monocyclic, carbocyclic ring structure, wherein
the ring structure contains at least one double bond. Suitable examples include
cyclohexenyl, cyclopentenyl, cycloheptenyl, cyclooctenyl, cyclohex-1,3-dienyl and the
like.
As used herein, unless otherwise noted, "aryl" shall refer to carbocyclic
aromatic groups such as phenyl, naphthyl, and the like. Divalent radicals include
phenylene (-C6H4-) which is preferably phen-1,4-diyl, but may also be phen-1,3-diyl.
As used herein, unless otherwise noted, "aralkyl" shall mean any alkyl group
substituted with an aryl group such as phenyl, naphthyl and the like. Examples of
aralkyls include benzyl, phenethyl, and phenylpropyl.
As used herein, unless otherwise noted, the terms "heterocycle", "heterocyclyl"
and "heterocyclo" shall denote any five-, six-, or seven- membered monocyclic, nine or
ten membered brcyclic or thirteen or fourteen membered tricyclic ring structure
containing at least one heteroatom moiety selected from the group consisting of N, O,
SO, SO2, (C=O), and S, and preferably N, O, or S, optionally containing one to four
additional heteroatoms in each ring. In some embodiments, the heterocyclyl contains
between 1 and 3 or between 1 and 2 additional heteroatoms. Unless otherwise
specified, a heterocyclyl may be saturated, partially unsaturated, aromatic or partially
aromatic. The heterocyclyl group may be attached at any heteroatom or carbon
atom which results in the creation of a stable structure.
Exemplary monocyclic heterocyclic groups can include pyrrolidinyl, pyrrolyl,
indolyl, pyrazolyl, oxetanyl, pyrazolinyl, imidazolyl, imidazolinyl, imidazolidinyl,
oxazolyl, oxazolidinyl, isoxazolinyl, isoxazolyl, thiazaolyl, thiadiazolyl, thiazolidinyl,
isothiazolyl, isothiazolidinyl, furyl, tetrahydrofuryl, thienyl, oxadiazolyl, piperidinyl,
piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, 2-oxazepinyl,
11
azepinyl, hexahydroazepinyl, 4-piperidinyl, pyridyl, N-oxo-pyridyl, pyrazinyl,
pyrimidinyl, pyridazinyl, tetrahydropyranyl, tetrahydrothiopyranyl,
tetrahydrothiopyranyl sulfone, morpholinyl, thiomorpholinyl, thiomorpholinyl
sulfoxide, thiomorpholinyl sulfone, 1,3-dixolane and tetrahydro-1,1-dioxothienyl,
dioxanyl, isothiazolidinyl, thietanyl, thiiranyl, triazinyl, triazolyl, tetrazolyl, azetidinyl
and the like.
For example, where Q is a non-aromatic nitrogen-containing heterocyclyl,
preferred values for Q include piperidyl, piperazinyl, pyrrolinyl, pyrrolidinyl,
morpholinyl, and N-(C1-6 alkyl) piperazinyl. These may be linked to the rest of the
molecule by a nitrogen or a carbon atom; in general, N-linked heterocyclyls are
preferred. Q can be substituted with between 1 and 3 substituents selected from
pyridyl, pyrimidyl, furyl, thiofuryl, imidazolyl, (imidazolyOC,^ alkylene, oxazolyl,
thiazolyl, 2,3-dihydro-indolyl, benzimidazolyl, 2-oxobenzimidazolyl, (tetrazolyOC^
alkylene, tetrazolyl, (triazolyl)C1-6 alkylene, triazolyl, (pyrrolyOC^ alkylene, and
pyrrolyl. Examples of substituted Q, wherein the substituent comprises a
heterocyclyl, include: 4-(4-chloropyridin-2-yl)amino-piperidin-1-yl; 4-(4-
chloropyrimidin-2-yl)amino-piperidin-1-yl;2-([1,2,4]triazol-1-yl)methyl-morpholin-1-yl;
3-(pyrazin-2-yl)piperidin-1 -yl; 4-(pyrazol-1 -yi)piperidin-1 -yl; 4-(pyrimidin-2-
yl)piperazin-1-yl; 4-(furan-2-yl)methylpiperazin-1-yl; 4-(thiophen-2-
yl)methylpiperazin-1-yl; 4-(4-chloropyridin-2-yl)[1,4]diazepan-1-yl; and 5-(isoxazol-5-
yl)-2,5-diaza-bicyclo[2.2.1]heptah-2-yl.
Exemplary bicyclic heterocyclic groups include benzthiazolyl, benzoxazolyl,
benzoxazinyl, benzothienyl, quinuclidinyl, quinolinyl, quinolinyl-N-oxide,
tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl,
benzofuryl, chromonyl, coumarinyl, cinnolinyl, quinoxaliny!, indazolyl, pyrrolopridyl,
furopyridinyl (such as furo[2,3-c]pyridinyl, furo[3,1-b]pyridinyl), or furo[2,3-
b]pyridinyl), dihydroisoindolyl, dihydroquinazolinyl (such as 3,4-dihydro-4-oxo-

quinazolinyl), tetrahydroquinolinyl (such as 1,2,3,4-tetrahydroquinolinyl),
tetrahydroisoquinoliny!(such as 1,2,3,4-tetrahydroisoquiunolinyl), benzisothiazolyl,
benzisoxazolyl, benzodiazinyl, benzofurazanyl, benzothiopyranyl, benzotriazolyl,
benzpyrazolyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl,
dihydrobenzothiopyranyl sulfone, dihydrobenzopyranyl, indolinyl, isoindolyl,
tetrahydroindoazolyl (such as 4,5,6,7-tetrahydroindazolyl), isochromanyl,
isoindolinyl, naphthyridinyl, phthalazinyl, piperonyl, purinyl, pyridopyridyl,
quinazolinyl, tetrahydroquinolinyl, thienofuryl, thienopyridyl, thienothienyl,

Exemplary tricyclic heterocylclic groups include acridinyl, phenoxazinyl,
phenazinyl, phenothiazinyl, carbozolyl, perminidinyl, phenanthrolinyl, carbolinyl,
naphthothienyl, thianthrenyl, and the like.
Preferred heterocyclyl groups include morpholinyl, piperidinyl, piperazinyl,
pyrrolidinyl, pyrimidinyl, pyridyl, pyrrolyl, imidazolyl, oxazolyl, isoxazolyl, acridinyl,
azepinyl, hexahydroazepinyl, azetidinyi, indolyl, isoindolyl, thiazolyl, thiadiazolyl,
quinolinyl, isoquinolinyl, 1,2,3,4-tetrahydroquinolinyl, 1,3,4-trihydroisoquinolinyl,
4,5,6,7-tetrahydroindadolyl, benzoxazinyl, benzoaxzolyl, benzthiazolyl, benzimidazolyl,

As used herein, unless otherwise noted, the term "heterocyclyl-alkyl" or
"heterocyclyl-alkylene" shall denote any alkyl group substituted with a heterocyclyl
group, wherein the heterocycly-alkyl group is bound through the alkyl portion to the
central part of the molecule. Suitable examples of heterocyclyl-alkyl groups include,
but are not limited to piperidinylmethyl, pyrrolidinylmethyl, piperidinylethyl,
piperazinylmethyl, pyrrolylbutyl, piperidinylisobutyl, pyridylmethyl, pyrimidylethyl, and
the like.
When a particular group is "substituted" (e.g., alkyl, alkylene, cycloalkyl, aryl,
heterocyclyl, heteroaryl), that group may have one or more substituents, preferably
from one to five substituents, more preferably from one to three substituents, most
preferably from one to two substituents, independently selected from the list of
substituents.
It is intended that the definition of any substituent or variable at a particular
location in a molecule be independent of its definitions elsewhere in that molecule.
It is understood that substituents and substitution patterns on the compounds of this
invention can be selected by one of ordinary skill in the art to provide compounds
that are chemically stable and that can be readily synthesized by techniques known
in the art as well as those methods set forth herein.
Under standard nomenclature used throughout this disclosure, the terminal
portion of the designated side chain is described first, followed by the adjacent
functionality toward the point of attachment. Thus, for example, a
"phenyl(alkyl)amido(alkyl)" substituent refers to a group of the formula
The term "subject" as used herein, refers to an animal, preferably a mammal,
most preferably a human, who has been the object of treatment, observation or
experiment.
The term "therapeutically effective amount" as used herein, means that amount
of active compound or pharmaceutical agent that elicits the biological or medicinal
response in a tissue system, animal or human that is being sought by a researcher,
veterinarian, medical doctor or other clinician, which includes prevention, inhibition of
onset, or alleviation of the symptoms of the disease or disorder being treated.
As used herein, the term "composition" 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 combinations of the specified ingredients in
the specified amounts.
Abbreviations used in the specification, particularly in the Schemes and
Examples, are as follows:
The next section describes the compounds provided by the invention in more
detail.
B. Compounds
The invention features compounds of formula (I) as described in the above
Summary section and, for example, in the claims. Preferred compounds include
those wherein:
(a) R5 is C 1-5 alkyl, C 3-4 alkenyl, C 3-6 cycloalkyl, (C 3-6 cycloalkyl) C 1aIkylene,
(phenyl)C 1-3 alkylene, or (phenyl)C 3-4 alkenylene;
(b) R5 is branched C 3-5 alkyl, C 3-6 cycloalkyl, and (C 3-6 cycloalkyl )C 1
alkylene;
(c) one of R2 and R3 is G;
(d) R2 is G;
(e) R3 is G;
(f) L1 is C 2-3 alkylene;
(g) L2 is C 1-6 alkylene, (C 1-5 heteroaryl)C 1-6 alkylene, or -phenyl-C 1-6
alkylene;
(h) L2 is methylene;
(I) L3 is ethylene, vinylene, ethynylene, and phenylene;
(j) Q is a non-aromaic nitrogen-containing C 2-5 heterocyclyl;
(k) Q is selected from piperidyl, N-(C ,.6 alkyl)piperazinyl, piperazinyl,
pyrrolinyl, pyrrolidinyl, and morpholinyl;
(I) Q is N-morpholinyl or N-piperidinyl, optionally substituted with between 1
and 3 substituents selected from hydroxyl, carboxamide, C1-6 alkyl, C1-8
heterocyclyl, N(C1-6 alkyl)(C1-8 heterocyclyl), NH(C1-8 heterocyclyl), (C1-3
alkylene)(C1-8 heterocyclyl), O(C1-8 heterocyclyl), O(C1-6 alkyl), O(C3-6
cycloalkyl), phenyl, (C1-3 alkylene) phenyl, N(C1-6 alkyl)(C1-3 alkylene)
phenyl, and O(C1-3 alkylene) phenyl where each of above heterocyclyl,
phenyl, and alkyl groups may be optionally substituted with from 1 to 3
substituents independently selected from halogen, nitro, cyano, and C1-3
alkyl;
(m) Q is substituted with a substituent comprising a C1-6 heterocyclyl group
selected from: pyridyl, pyrimidyl, furyl, thiofuryl, imidazolyl, (C1-6 alkyl)
imidazolyl, oxazolyl, thiazolyl, 2,3-dihydro-indolyl, benzimidazolyl, 2-
oxobenzimidazolyl, (C1-6 alkyl) tetrazolyl, tetrazolyl, (C1-6 alkyl) triazolyl,
triazolyl, (C1-6 alkyl) pyrrolyl, and pyrrolyl;
(n) Q is a substituted or unsubstituted N-morpholinyl;
(o) Q is NR8R9 wherein each of R8 or R9 is independently selected from
hydrogen, C 1-8 alkyl, C 3-8 alkenyl, C 3-7 cycloalkyl, (C3-7 cycloalkyl)C1-6
alkylene, C2-5 heterocyclyl, phenyl, (C2-5 heterocyclyl)C1-6 alkylene, and
(phenyl) C1-6 alkylene;
(p) one of R8 and R9 is hydrogen;
(q) R8 is H and R9 is phenyl or aromatic C 1-8 heterocyclyl optionally
substituted with 1-3 substituents selected from halo, nitro, cyano, and C1-3
alkyl;
(r) R9 is phenyl, pyridyl, pyrimidyl, furyl, thiofuryl, imidazolyl, (C1-6 alkyl)
imidazolyl, oxazolyl, thiazolyl, 2,3-dihydro-indolyl, benzimidazolyl, 2-
oxobenzimidazolyl, (C1-6 alkyl) tetrazolyl, tetrazolyl, (C1-6 alkyl) triazolyl,
triazolyl, (C1-6 alkyl) pyrrolyl, and pyrrolyl;
(s) R5 is C1-5 alkyl, C 3-4 alkenyl, C 3-6 cycloalkyl, (C 3-6 cycloalkyl) C 1 aIkylene,
(phenyl)C 1-3 alkylene, or (phenyl)C 3-4 alkenylene;
(t) n is 0 or 1;
(u) n is 0;
(v) G is selected from:
(1) formula (i) wherein L4 and L5 are independently selected from C 2-3
alkylene,
(2) formula (iii) wherein L6 is C2-3 alkylene and L7 is C 2-3 alkylene or
absent,
(3) L2Q wherein L2 is C 1-6 alkylene, phenyl C 1-4 alkylene, or
(aromatic C 1-5 heterocyclyl)C 1-4 alkylene, and
(4) OL1Q wherein L1 is C 2-3 alkylene;
(w) G is selected from:
(1) formula (i) wherein L4 and L5 are each C2 alkylene,
(2) formula (iii) wherein each of L6 and L7 is C2 alkylene, and
(3) L2Q wherein L2 is methylene;
(x) wherein G is L2Q;
(y) R10 is H, branched C 3-6 alkyl, or benzyl;
(2) R10 is isopropyl or benzyl;
(aa) Q is a non-aromatic C 2-5 heterocyclyl;
(bb) Q is selected from piperidyl, N-(C 1-6 alkyl)piperazinyl, piperazinyl,
pyrrolinyl, pyrrolidinyl, and morpholinyl;
(cc) R5 is C 1-5 alkyl, C 3-4 alkenyl, C 3-6 cycloalkyl, (C 3-6 cycloalkyl) C
1alkylene, (phenyl)C 1-3 alkylene, or (phenyl)C 3-4 alkenylene;
(dd) R7 is hydroxyl, halo, or absent where one of L6 and L7 provides a
double bond to the carbon atom to which R6 and R7 are attached;
(ee) one of R2 and R3 is G;
(ff) one of R2 and R3 is W, and W is a heterocyclyl selected from: pyridyl,
pyrimidyl, furyl, thiofuryl, imidazolyl, oxazolyl, thiazoiyl, 2,3-dihydro-indolyl,
benzimidazolyl, tetrazolyl, triazolyl, and pyrrolyl;
(gg) R5 is branched C 3-5 alkyl;
(hh) R5 is isopropyl or cyclopentyl; or
(ii) Combinations of the above.
Preferred compounds of the invention include: 4-(4-lmidazol-1-yl-phenoxy)-1-
isopropyl-piperidine, 4-(4-lmidazol-1-yl-phenoxy)-1-isobutyl-piperidine, 1-lsopropyl-4-
(4-pyrrol-1 -yl-phenoxy)-piperidine, 5-Chloro-2-[4-(1 -isopropyl-piperidin-4-yloxy)-
pheny)]-1 H-benzoimidazole, and more preferably, 4-(4-lmidazol-1-yl-phenoxy)-1-
isopropyl-piperidine, 4-(4-lmidazol-1-yl-phenoxy)-1-isobutyl-piperidine, and 5-Chloro-
2-[4-(1-isopropyl-piperidin-4-yloxy)-phenyl]-1 H-benzoimidazole.
The invention also features: [4-(1-lsopropyl-piperidin-4-yloxy)-phenyl]-phenyl-
methanone, 4-(Biphenyl-4-yloxy)-1 -isopropyl-piperidine, 4-(4-Benzyloxy-phenoxy)-1 -
isopropyl-piperidine, 1 -lsopropyl-4-(4-phenoxy-phenoxy)-piperidine, 4-(4-Benzyl-
phenoxy)-1 -isopropyl-piperidine, [4-(1 -lsopropyl-piperidin-4-yloxy)-phenyl]-phenyl-
methanol, N-[4-(1 -lsopropyl-piperidin-4-yloxy)-phenyl]-acetamide, 4-(4-Cyclopentyl-
phenoxy)-1 -isopropyl-piperidine, 4-(1 -Cyclopentyl-piperidin-4-yloxy)-benzonitrile, 4-
(1-Cyclobutyl-piperidin-4-yIoxy)-benzonitrile, 4-(1 -sec-Butyl-piperidin-4-yloxy)-
benzonitrile, 4-(1 -lsopropyl-piperidin-4-yloxy)-benzaldehyde, 4-(1 -Cyclohexyl-
piperidin-4-yloxy)-benzonitrile, 4-(1-lsopropyl-piperidin-4-yloxy)-benzonitrile1 4-(1-
Cyclopropylmethyl-piperidin-4-yloxy)-benzonitrile, 4-(1-lsobutyl-piperidin-4-yloxy)-
benzonitrile, and 4-(1-Propyl-piperidin-4-yloxy)-benzonitrile.
The invention also provides more preferred compounds such as: 4-(Biphenyl-
4-yloxy)-1 -isopropyl-piperidine, 4-(4-Benzyloxy-phenoxy)-1 -isopropyl-piperidine, 4-
(4-Benzyl-phenoxy)-1 -isopropyl-piperidine, 1 -lsopropyl-4-(4-phenoxy-phenoxy)-
piperidine, and N-[4-(1 -lsopropyl-piperidin-4-yloxy)-phenyl]-acetamide.
Preferred compounds also include those such as: (A) 1-lsopropyl-4-[4-(1-
isopropyl-piperidin-4-yloxy)-phenyl]-piperazine. and 1-[4-(1-lsopropyi-piperidin-4-
yloxy)-phenyl]-piperazine 1-[4-(1-lsopropyl-p:: ridin-4-yloxy)-phenyl]-piperazine; and
(B) 1 -[4-(1 -lsopropyl-piperidin-4-yloxy)-benzyl]-piperidine, 4-[4-(1 -sec-Butyl-piperidin-
4-yloxy)-benzyl]-morpholine, 1-[4-(1-Cyclopentyl-piperidin-4-yloxy)-benzyl]-
piperidine, 1-[4-(1-lsobutyl-piperidin-4-ylox) -oenzyl]-piperidine, 1-N-lsopropyl-4-{4-
[5-(1-isopropyl-piperidin-4-ylsulfanyl)-tetrazol-1-yl]-phenoxy}-piperidine, {1 -[4-( 1 -
lsopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-yl}-methanol, 1-[4-(1-lsopropyl-
piperidin-4-yloxy)-benzyl]-4-methyl-[1,4]diazepane, 1-[4-(1-lsopropyl-piperidin-4-
yloxy)-benzyl]-azepane, 1-[4-(1-lsobutyl-piperidin-4-yloxy)-benzyl]-piperidine, 1-[4-(1 -
lsopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-ol, [4-(1-lsopropyl-piperidin-4-yloxy)-
benzyl]-methyl-(1 -methyl-piperidin-4-yl)-amine, 1 -[4-(1 -isopropyl-piperidin-4-yloxy)-
benzyl]-4-benzyl-piperidine, N-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-N,N\N'-
trimethyl-ethane-1,2-diamine, 1 -[4-(1 -lsopropyl-piperidin-4-yloxy)-benzyl]-4-methyl-
piperazine, Cyclohexyl-[4-(1-isopropyl-piperidin-4-yloxy)-benzyl]-methyl-amine,
Butyl-[4-(1 -isopropyl-piperidin-4-yloxy)-benzyl]-methyl-amine, 4-[4-(1 -Cyclopentyl-
piperidin-4-yloxy)-benzyl]-morpholine, 1 -lsopropyl-4-(4-pyrrolidin-1 -ylmethyl-
phenoxy)-piperidine, Diethyl-[4-(1 -isopropyl-piperidin-4-yloxy)-benzyl]-amine, 4-[4-(1 -
sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine, 1 -[4-(1 -lsopropyl-piperidin-4-yloxy)-
benzyl]-4-phenyl-piperazine, 1 -Benzyl-4-[4-(1 -isopropyl-piperidin-4-yloxy)-benzyl]-
piperazine, 4-[4-(4-Benzylidene-piperidin-1 -ylmethyl)-phenoxy]-1 -isopropyl-
piperidine, 4-[4-(1 -lsopropyl-piperidin-4-yloxy)-benzyr|-morpholine, [4-(1 -Isopropyl-
piperidin-4-yloxy)-benzyl]-dimethyl-amine, 4-[4-(1-Cyclohexyl-piperidin-4-yloxy)-
benzyl]-morpholine, 4-[4-(1 -lsobutyl-piperidin-4-yloxy)-benzyl]-morpholine, 4-[4-(1 -
Propyl-piperidin-4-yloxy)-benzyl]-morpholine 1 -[4-(1 -Cyclohexyl-piperidin-4-yloxy )-
benzyl]-piperidine, 1-[4-(1-Benzyl-piperidin -yloxy)-benzyl]-piperidine, 1-[4-(1-
Cyclohexylmethyl-piperidin-4-yloxy)-benzyl]-piperidine, and 4-[4-(4-Piperidin-1 -
ylmethyl-phenoxy)-piperidin-1-yl]-butan-2-one.
Highly preferred compounds include: 1-[4-(1-lsopropyl-piperidin-4-yloxy)-
benzyl]-piperidine, 4-[4-{1-sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine, 1-[4-(1-
Cyclopentyl-piperidin-4-yloxy)-benzyl]-piperidine, 1 -[4-(1 -lsobutyl-piperidin-4-yloxy)-
benzylj-piperidine, 1-N-lsopropyi-4-{4-[5-(1-isopropyl-piperidin-4-ylsulfanyl)-tetrazol-
1-yl]-phenoxy}-piperidine, {1-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-yl}-
methanol, 1-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-4-methyl-[1,4]diazepane, 1-[4-
(1-lsopropyl-piperidin-4-yloxy)-benzyl]-azepane, 1-[4-(1-lsobutyl-piperidin-4-yloxy)-
benzylj-piperidine, 1-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-ol, [4-{1-
Isopropyl-piperidin-4-yloxy)-benzyl]-methyl-(1-methyl-piperidin-4-yl)-amine, 1-[4-(1-
isopropyl-piperidin-4-yloxy)-benzyl]-4-benzyl-piperidine, N-[4-(1-lsopropyl-piperidin-
4-yloxy)-benzyl]-N,Nl,N'-trimethyl-ethane-1,2-diamine, 1 -[4-(1 -lsopropyl-piperidin-4-
yloxy)-benzyl]-4-methyi-piperazine, Cyclohexyl-[4-(1-isopropyl-piperid/n-4-yloxy)-
benzyl]-methyl-amine, Butyl-[4-(1-isopropyl-piperidin-4-yloxy)-benzyl]-methyl-amine,
4-[4-(1 -Cyclopentyl-piperidin^-yloxyJ-benzyl]-morpholine, 1 -lsopropyl-4-(4-pyrrolidin-
1-ylmethyl-phenoxy)-piperidine, Diethyl-[4-(1-isopropyl-piperidin-4-yloxy)-benzyl]-
amine, 4-[4-(1 -sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine, 1-[4-(1 -Isopropyl-
piperidin-4-yloxy)-benzyl]-4-phenyl-piperazine, 1-Benzyl-4-[4-(1-isopropyl-piperidin-
4-yloxy)-benzyl]-piperazine, 4-[4-(4-Benzylidene-piperidin-1 -ylmethyl)-phenoxy]-1 -
isopropyl-piperidine, 4-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-morpholine, [4-(1-
lsopropyl-pjperidin-4-yloxy)-benzyl]-dimethyl-amine, 4-[4-(1-Cyclohexyl-piperidin-4-
yloxy)-benzyl]-morpholine, and 4-[4-(1 -lsobutyl-piperidin-4-yloxy)-benzyI]-
morpholine.
Examples of highly preferred compounds also include: Cyclopropyl-[4-(1-
isopropyl-piperidin-4-yioxy)-benzyl]-amine, [4-( 1 -lsopropyl-piperidin-4-yloxy)-benzyl]-
(5-methyl-pyridin-2-yl)-amine, [4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-pyridin-2-yi-
amine, [4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-phenyl-amine, and (5-Chloro-
pyridin-2-yl)-[4-(1-isopropyl-piperidin-4-yloxy)-benzyl]-amine.
The invention also provides compounds that are useful as synthetic
intermediates of the compounds of the invention. Such compounds, which
themselves may or may not have pharmaceutical activity, include those provided in
the Schemes and synthetic examples.
The invention also contemplates compounds isotopically-labelled to be
detectable by positron emission tomography (PET) or single-photon emission
computed tomography (SPECT) useful for studying H3-mediated disorders.
During any of the processes for preparation of the compounds of the present
invention, it may be necessary and/or desirabie to protect sensitive or reactive
groups on any of the molecules concerned. In addition, compounds of the invention
may be modified by using protecting groups; such compounds, precursors, or
prodrugs are also within the scope of the invention. This may be achieved by means
of conventional protecting groups, such as those described in "Protective Groups in
Organic Chemistry", ed. J.F.W. McOmie, Plenum Press, 1973; and T.W. Greene &
P.G.M. Wuts, "Protective Groups in Organic Synthesis", 3rd ed., John Wiley & Sons,
1999. The protecting groups may be removed at a convenient subsequent stage
using methods known from the art.
HYDROXYL PROTECTING GROUPS
Protection for the hydroxyl group includes methyl ethers, substituted methyl
ethers, substituted ethyl ethers, substitute benzyl ethers, and silyl ethers.
Substituted Methyl Ethers
Examples of substituted methyl ethers include methyoxymethyl,
methylthiomethyl, t-butylthiomethyl, (phenyldimethylsilyl)methoxymethyl,
benzyloxymethyl, p-methoxybenzyloxymethyl, (4-methoxyphenoxy)methyl,
guaiacolmethyl, t-butoxymethyl, 4-pentenyloxymethyl, siloxymethyl, 2-
methoxyethoxymethyl, 2,2,2-trichloroethoxymethyl, bis(2-chloroethoxy)methyl, 2-
(trimethylsilyl)ethoxymethyl, tetrahydropyranyl, 3-bromotetrahydropyranyl,
tetrahydrothiopyranyl, 1-methoxycyclohexyl, 4-methoxytetrahydropyranyl, 4-
methoxytetrahydrothiopyranyl, 4-methoxytetrahydrothiopyranyl S,S-dioxido, 1-[(2-
chloro-4-methyl)phenyl]-4-methoxypiperidin-4-yl, 1,4-dioxan-2-yl, tetrahydrofuranyl,
tetrahydrothiofuranyl and 2,3,3a,4,5,6,7,7a-octahydro-7,8,8-trimethyl-4,7-
methanobenzofuran-2-yl.
Substituted Ethyl Ethers
Examples of substituted ethyl ethers include 1-ethoxyethyl, 1-(2-
chloroethoxy)ethyl, 1-methyl-1-methoxyethyl, 1-methyl-1-benzyloxyethy[, 1-methyl-1-
benzyloxy-2-fluoroethyl, 2,2,2-trichloroethyl, 2-trimethylsiiylethyl, 2-
(phenylselenyl)ethyl, t-butyl, allyl, p-chlorophenyl, p-methoxyphenyl, 2,4-
dinitrophenyl, and benzyl.
Substituted Benzyl Ethers
Examples of substituted benzyl ethers include p-methoxybenzyl, 3,4-
dimethoxybenzyl, o-nitrobenzyl, p-nitrobenzyl, p-halobenzyl, 2,6-dichlorobenzyl, p-
cyanobenzyl, p-phenylbenzyl, 2- and 4-picolyl, 3-methyl-2-picolyl N-oxido,
diphenylmethyl, p, p'-dinitrobenzhydryl, 5-dibenzosuberyl, triphenylmethyl, a-
naphthyldiphenylmethyl, p-methoxyphenyldiphenylmethyl, di(p-
methoxyphenyl)phenylmethyl, tri(p-methoxyphenyl)methyl, 4-(4'-
bromophenacyloxy)phenyldiphenylmethyl, 4I4',4"-tris(4,5-
dichlorophthalimidophenyl)methyl,4,4',4"-tris(levulinoyloxyphenyl)methyl, 4,4',4"-
tris(benzoyloxyphenyl)methyl, 3-(lmidazol-1-ylmethyl)bis(4 ',4"-
dimethoxyphenyl)methyl, 1,1-bis(4-methoxyphenyl)-1'-pyrenylmethyll 9-anthryl, 9-(9-
phenyl)xanthenyl, 9-(9-phenyl-10-oxo)anthryl, 1,3-benzodithiolan-2-yl, and
benzisothiazolyl S,S-dioxido.
Silyl Ethers
Examples of silyl ethers include trimethylsilyl, triethylsilyl, triisopropylsilyl,
dimethylisopropylsilyl, diethylisopropylsilyl, dimethylthexylsilyl, f-butyldimethylsilyl, t-
butyldiphenylsilyl, tribenzylsilyl, tri-p-xylylsilyl, triphenylsilyl, diphenylmethylsilyl, and
f-butylmethoxyphenylsilyl.
Esters
In addition to ethers, a hydroxyl group may be protected as an ester.
Examples of esters include formate, benzoylformate, acetate, chloroacetate,
dichloroacetate, trichloroacetate, trifluoroacetate, methoxyacetate,
triphenylmethoxyacetate, phenoxyacetate, p-chlorophenoxyacetate, p-P-
phenylacetate, 3-phenylpropionate, 4-oxopentanoate(levulinate), 4,4-
(ethylenedithio)pentanoate, pivaloate, adamantoate, crotonate, 4-methoxycrotonate,
benzoate, p-phenylbenzoate, 2,4,6-trimethylbenzoate(mesitoate)
Carbonates
Examples of carbonates include methyl, 9-fluorenylmethyl, ethyl, 2,2,2-
trichloroethyl, 2-(trimethylsilyl)ethyl, 2-(phenylsulfonyl)ethyl, 2-
(triphenylphosphonio)ethyl, isobutyl, vinyl, allyl, p-nitrophenyl, benzyl, p-
methoxybenzyl, 3,4-dimethoxybenzyl, o-nitrobenzyl, p-nitrobenzyl, S-benzyl
thiocarbonate, 4-ethoxy-1-naphthyl, and methyl dithiocarbonate.
Assisted Cleavage
Examples of assisted cleavage include 2-iodobenzoate, 4-azidobutyrate, 4-
nitro-4-methylpentanoate, o-(dibromomethyl)benzoate, 2-formylbenzenesulfonate, 2-
(methylthiomethoxy)ethyl carbonate, 4-(methylthiomethoxy)butyrate, and 2-
(methylthiomethoxymethyl)benzoate.
Miscellaneous Esters
Examples of miscellaneous esters include 2,6-dichloro-4-
methylphenoxyacetate, 2,6-dichloro-4-(1,1,3,3-tetramethylbutyl)phenoxyacetate,
2,4-bis(1,1-dimethylpropyl)phenoxyacetate, chlorodiphenylacetate, isobutyrate,
monosuccinoate, (E)-2-methyl-2-butenoate(tigloate), o-(methoxycarbonyl)benzoate,
p-P-benzoate, a-naphthoate, nitrate, alkyl N,N,N',N'-
tetramethylphosphorodiamidate, N-phenylcarbamate, borate,
dimethylphosphinothioyl, and 2,4-dinitrophenylsulfenate
Sulfonates
Examples of sulfonates include sulfate, methanesulfonate(mesylate),
benzylsulfonate, and tosylate.
PROTECTION FOR 1,2-AND 1,3-DIOLS
Cyclic Acetals and Ketals
Examples of cyclic acetals and ketals include methylene, ethylidene, 1-f-
butylethylidene, 1-phenylethylidene, (4-methoxyphenyl)ethylidene, 2,2,2-
trichloroethylidene, acetonide (isopropylidene), cyclopentylidene, cyclohexylidene,
cycloheptylidene, benzylidene, p-methoxybenzylidene, 2,4-dimethoxybenzylidene,
3,4-dimethoxybenzylidene, and 2-nitrobenzylidene.
Cyclic Ortho Esters
Examples of cyclic ortho esters include methoxymethylene, ethoxymethylene,
dimethoxymethylene, 1-methoxyethylidene, 1-ethoxyethylidine, 1,2-
dimethoxyethylidene, a-methoxybenzylidene, 1 -(N,N-dimethylamino)ethylidene
25
derivative, a-(N,N-dimethylamino)benzylidene derivative, and 2-
oxacyclopentylidene.
Silyl Derivatives
Examples of silyl derivatives include di- f-butylsilylene group, and 1,3-(1,1,3,3-
tetraisopropyldisiloxanylidene) derivative.
AMINO PROTECTING GROUPS
Protection for the amino group includes carbamates, amides, and special -
NH protective groups.
Examples of carbamates include methyl and ethyl carbamates, substituted
ethyl carbamates, assisted cleavage carbamates, photolytic cleavage carbamates,
urea-type derivatives, and miscellaneous carbamates.
Carbamates
Examples of methyl and ethyl carbamates include methyl and ethyl, 9-
fluorenylmethyl, 9-(2-sulfo)fluorenylmethyl, 9-(2,7-dibromo)fiuorenylmethyl, 2,7-di-f-
butyl-[9-(10,10-dioxo-10,10,10,10-tetrahydrothioxanthyl)]methyl, and 4-
methoxyphenacyl.
Substituted Ethyl
Examples of substituted ethyl carbamates include 2,2,2-trichloroethyl, 2-
trimethylsilylethyl, 2-phenylethyl, 1-(1-adamantyl)-1-methylethyl, 1,1-dimethyl-2-
haloethyl, 1,1-dimethyI-2,2-dibromoethyl, 1,1-dimethyl-2,2,2-trichloroethyl, 1-methyl
1-(4-biphenylyl)ethyl, 1-(3,5-di-f-butylphenyl)-1-methylethyl, 2-(2'- and 4'-
pyridyl)ethyl, 2-(N,N-dicyclohexylcarboxamido)ethyl, f-butyl, 1-adamantyl, vinyl, allyl,
1-isopropylallyl, cinnamyl, 4-nitrocinnamyl, 8-quinolyl, N-hydroxypiperidinyl,
26
alkyldithio, benzyl, p-methoxybenzyl, p-nitrobenzyl, p-bromobenzyl, p-chlorobenzyl,
2,4-dichlorobenzyl, 4-methylsulfinylbenzyl, 9-anthrylmethyl and diphenylmethyl.
Assisted Cleavage
Examples of assisted cleavage include 2-methylthioethyl, 2-
methyisulfonylethyl, 2-(p-toluenesulfonyl)ethyl, [2-(1,3-dithianyl)]methyl, 4-
methylthiophenyl, 2,4-dimethylthiophenyl, 2-phosphonioethyl, 2-
triphenylphosphonioisopropyl, 1,1-dimethyl-2-cyanoethyl, m-chloro-p-acyloxybenzyl,
p-(dihydroxyboryl)benzyl, 5-benzisoxazolylmethyl, and 2-(trifluoromethyl)-6-
chromonylmethyl.
Photolytic Cleavage
Examples of photolytic cleavage include m-nitrophenyl, 3,5-dimethoxybenzyl,
o-nitrobenzyl, 3,4-dimethoxy-6-nitrobenzyl, and phenyl(o-nitrophenyl)methyl.
Urea-Type Derivatives
Examples of urea-type derivatives include phenothiazinyl-(10)-carbonyl
derivative, N'-p-toluenesulfonylaminocarbonyl, and N'-phenylaminothiocarbonyl.
Miscellaneous Carbamates
Examples of miscellaneous carbamates include f-amyl, S-benzyl
thiocarbamate, p-cyanobenzyl, cyclobutyl, cyclohexyl, cyclopentyl,
cyclopropylmethyl, p-decyloxybenzyl, diisopropytmethyl, 2,2-dimethoxycarbonylvinyl,
o-(N,N-dimethylcarboxamido)benzyl, 1,1-dimethyl-3-(N,N-
dimethylcarboxamido)propyl, 1,1-dimethylpropynyl, di(2-pyridyl)methyl, 2-
furanylmethyl, 2-iodoethyl, isobornyl, isobutyl, isonicotinyl, p-(p'-
methoxyphenylazo)benzyl, 1-methylcyclobutyl, 1-methylcyclohexyl, 1-methyM-
cyclopropylmethyl, 1-methyl-1-(3,5-dimethoxyphenyl)ethyl, 1-methyl-1-(p-
phenylazophenyl)ethyl, 1-methyl-1-phenylethyl, 1-methyl-1-(4-pyridyI)ethyl, phenyi,
27
p-(phenylazo)benzyl, 2,4,6-tri-f-butylphenyl, 4-(trimethyiammonium)benzyl, and
2,4,6-trimethyl benzyl.
Examples of amides include:
Amides
N-formyl, N-acetyl, N-chloroacetyl, N-trichloroacetyl, N-trifluoroacetyl, N-
phenylacetyl, N-3-phenylpropionyl, N-picolinoyl, N-3-pyridylcarboxamide, N-
benzoylphenylalanyl derivative, N-benzoyl, N-p-phenylbenzoyl.
Assisted Cleavage
N-o-nitrophenylacetyl, N-o-nitrophenoxyacetyl, N-acetoacetyl, (N'-
dithiobenzyloxycarbonylamino)acetyl, N-3-(p-hydroxyphenyl)propionyl, N-3-(o-
nitrophenyl)propionyl, N-2-methyl-2-(o-nitrophenoxy)propionyl, N-2-methyl-2-(o-
phenylazophenoxy)propionyl, N-4-chlorobutyryl, N-3-methyl-3-nitrobutyryl, N-o-
nitrocinnamoyl, N-acetylmethionine derivative, N-o-nitrobenzoyl, N-o-
(benzoyloxymethyl)benzoyl, and 4,5-diphenyl-3-oxazolin-2-one.
Cyclic Imide Derivatives
N-phthalimide, N-dithiasuccinoyl, N-2,3-diphenylmaleoyl, N-2,5-
dimethylpyrrolyl, N-1,1,4,4-tetramethyldisilylazacyclopentane adduct, 5-substituted
1,3-dimethyl-1,3,5-triazacyclohexan-2-one, 5-substituted 1,3-dibenzyl-1,3,5-
triazacyclohexan-2-one, and 1-substituted 3,5-dinitro-4-pyridonyl.
SPECIAL - NH PROTECTIVE GROUPS
Examples of special NH protective groups include:
N-Alkyl and N-Aryl Amines
28
N-methyl, N-allyl, N-[2-(trimethylsilyi)ethoxy]methyl, N-3-acetoxypropyl, N-(1-
isopropyl-4-nitro-2-oxo-3-pyrrolin-3-yl), quaternary ammonium salts, N-benzyl, N-4-
methoxybenzyl, N-di(4-methoxyphenyl)methyl, N-5-dibenzosuberyl, N-
triphenylmethyl, N-(4-methoxyphenyl)diphenylmethyl, N-9-phenylfluorenyl, N-2,7-
dichloro-9-fluorenylmethylene, N-ferrocenylmethyl, and N-2-picolylamine N'-oxide.
Imine Derivatives
N-1,1-dimethylthiomethyiene, N-benzyiidene, N-p-methoxybenzylidene, N-
diphenylmethylene, N-[(2-pyridyl)mesityl]methylene, and N-(N' ,N'-
dimethylaminomethylene).
PROTECTION FOR THE CARBONYL GROUP
Acyclic Acetals and Ketals
Examples of acyclic acetals and ketals include dimethyl, bis(2,2,2-
trichloroethyl), dibenzyl, bis(2-nitrobenzyl) and diacetyl.
Cyclic Acetals and Ketals
Examples of cyclic acetals and ketals include 1,3-dioxanes, 5-methylene-1,3-
dioxane, 5,5-dibromo-1,3-dioxane, 5-(2-pyridyl)-1,3-dioxane, 1,3-dioxolanes, 4-
bromomethyl-1,3-dioxolane, 4-(3-butenyl)-1,3-dioxolane, 4-phenyl-1,3-dioxolane, 4-
(2-nitrophenyl)-1,3-dioxolane, 4,5-dimethoxymethyl-1,3-dioxolane, 0,0'-
phenylenedioxy and 1,5-dihydro-3H-2,4-benzodioxepin.
Acyclic Dithio Acetals and Ketals
Examples of acyclic dithio acetals and ketals include S.S'-dimethyl, S,S'-
diethyl, S,S'-dipropyl, S,S'-dibutyl, S.S'-dipentyl, S,S'-diphenyl, S,S'-dibenzyi and
S,S'-diacetyl.
Cyclic Dithio Acetals and Ketals
Examples of cyclic dithio acetals and ketals include 1,3-dithiane, 1,3-
dithiolane and 1,5-dihydro-3H-2,4-benzodithiepin.
Acyclic Monothio Acetals and Ketals
Examples of acyclic monothio acetals and ketals include O-trimethylsilyl-S-
alkyl, O-methyl-S-alkyl or -S-phenyl and O-methyl-S-2-(methylthio)ethyl.
Cyclic Monothio Acetals and Ketals
Examples of cyclic monothio acetals and ketals include 1,3-oxathiolanes.
MISCELLANEOUS DERIVATIVES
O-Substituted Cyanohydrins
Examples of O-substituted cyanohydrins include O-acetyl, O-trimethylsilyl, O-
1-ethoxyethyl and O-tetrahydropyranyl.
Substituted Hydrazones
Examples of substituted hydrazones include N.N-dimethyl and 2,4-
dinitrophenyl.
Oxime Derivatives
Examples of oxime derivatives include O-methyl, O-benzyl and O-
phenylthiomethyl.
I mines
Substituted Methylene Derivatives, Cyclic Derivatives
Examples of substituted methylene and cyclic derivatives include
axazolidines, 1 -methyl-2-(1 '-hydroxyalkyl)imidazoles, N.N'-dimethylimidazolidines,
2,3-dihydro-1,3-benzothiazoles, diethylamine adducts, and methylaluminum bis(2,6-
di-f-butyl-4-methylphenoxide)(MAD)complex.
MONOPROTECTION OF DICARBONYL COMPOUNDS
Selective Protection Of a-and b-Diketones
Examples of selective protection of a-and b-diketones include enamines, enol
acetates, enol ethers, methyl, ethyl, /-butyl, piperidinyl, morpholinyl, 4-methyl-1,3-
dioxolanyl, pyrrolidinyl, benzyl, S-butyl, and trimethylsilyl.
Cyclic Ketals, Monothio and Dithio Ketals
Examples of cyclic ketals, monothio and dithio ketals include
bismethylenedioxy derivatives and tetramethylbismethylenedioxy derivatives.
PROTECTION FOR THE CARBOXYL GROUP
Esters
Substituted Methyl Esters
Examples of substituted methyl esters include 9-fiuorenylmethyl,
methoxymethyl, methylthiomethyl, tetrahydropyranyl, tetrahydrofuranyl,
methoxyethoxymethyl, 2-(trimethylsilyl)ethoxymethyl, benzyloxymethyi, phenacyl, p-
bromophenacyl, a-methylphenacyl, p-methoxyphenacyl, carboxamidomethyl, and N-
phthalimidomethyl.
2-Substituted Ethyl Esters
Examoles of 2-substituted ethyl esters include 2,2,2-trichloroethyl,
2-haloethyl, ?-chloroalkyl, 2-(trimethylsilyl)ethyl, 2-methylthioethyl, 1,3-
dithianyl-2-methyl, 2-(p-nitrophenylsulfenyl)ethyi, 2-(p-toluenesulfonyl)ethyl,
2-(2'-pyridyl)ethyl, 2-(diphenylphosphino)ethyl, 1-methyl-1-phenylethyl, f-butyl,
cyclopentyl, cyclohexyl, allyl, 3-buten-1-yl, 4-(trimethylsilyl)-2-buten-1-yl, cinnamyl, a-
methylcinnamyl, phenyl, p-(methyimercapto)phenyl and benzyl.
Substituted Benzyl Esters
Examples of substituted benzyl esters include triphenylmethyl,
diphenylmethyl, bis(o-nitropheny!)methyi, 9-anthrylmethyl, 2-(9,10-
dioxo)anthrylmethyl, 5-dibenzosuberyI, 1-pyrenylmethyl, 2-(trifluoromethyl)-6-
chromylmethyl, 2,4,6-trimethylbenzyl, p-bromobenzyl, o-nitrobenzyl, p-nitrobenzyl, p-
methoxybenzyl, 2,6-dimethoxybenzyl, 4-(methylsulfinyl)benzyl, 4-sulfobenzyl,
piperonyl, 4-picolyl and p-P-benzyl.
Silyl Esters
Examples of silyl esters include trimethylsilyl, triethylsilyl, f-butyldimethylsilyl,
/-propyldimethyisilyl, phenyldimethylsilyl and di-f-butylmethylsilyl.
Activated Esters
Examples of activated esters include thiols.
Miscellaneous Derivatives
Examples of miscellaneous derivatives include oxazoles, 2-alkyl-1,3-
oxazolines, 4-alkyl-5-oxo-1,3-oxazolidines, 5-alkyl-4-oxo-1,3-dioxoianes, ortho
esters, phenyl group and pentaaminocobalt(lll) complex.
Stannyl Esters
Examples of stannyl esters include triethylstannyl and tri-n-butylstannyl.
AMIDES AND HYDRAZIDES
Amides
Examples of amides include N,N-dimethyl, pyrrolidinyl, piperidinyl, 5,6-
dihydrophenanthridinyl, o-nitroanilides, N-7-nitroindolyl, N-8-Nitro-1,2,3,4-
tetrahydroquinolyl, and p-P-benzenesulfonamides.
Hydrazides
Examples of hydrazides include N-phenyl and N,N'-diisopropyl.
The compounds of the invention can be prepared according to the methods
described in the next section.
C. Synthesis
The compounds of the invention can be prepared according to traditional
synthetic organic methods and matrix or combinatorial chemistry methods, as shown
in Schemes 1-10 below and in Examples 1-86. A person of ordinary skill will be
aware of variations and adaptations of the schemes and examples provided to
achieve the compounds of the invention.
Throughout the schemes when the reacting functionality is located at R3, one
skilled in the art will recognize that the choice of R3 is illustrative only and that the
reacting functionality could also be located at R1 and R2 also.
Compounds of formula (I) may be prepared according to the processes
outlined in Scheme 1.
Scheme 1
A compound of formula (I) is prepared as outlined in Scheme 1 by reacting a
compound of formula (II) or a compound of formula (III) with a compound of formula
(IV). A compound of formula (II) is reacted with a compound of formula (IV) in the
presence of a base such as Cs2CO3, K2CO3, Na2CO3, NaOH, KOH, LDA, LHMDS, or
the like, in a solvent, for example DMF, DMA, THF, DME, DCM, and DMSO at a
temperature from ambient to about 200 °C. In a preferred embodiment the base is
selected from Cs2CO3, K,CO3, and Na2CO3 in a solvent selected from DMF, DMA. In
a particularly preferred embodiment the base is Cs2CO3 and the solvent DMF at a
temperature from 80 to 150 °C. One skilled in the art will recognize that the
compound of formula (II) should contain both an X, substituent which is a leaving
group, and a R1 or R3 group which is electron-withdrawing in order that the reaction
with a compound of formula (IV) proceed. Preferred R1 and R3 substituents include -
NO,, -CHO, -CN, CO2R15, COR16, -CONR17R18l or the like. Preferred X1 substituents
include -F and -Cl, and -Br. Particularly preferred R., or R3 substituents include -NO2,
-CHO, -CN, and COR16. Particularly preferred X1 substituents include -F and -Cl. A
compound of formula (!) may be prepared by reacting a compound of formula (III)
with a compound of formula (IV) according to the Mitsunobu procedure (in the
presence of triphenylphosphine or polymer supported triphenyl phosphine and
DBAD or DEAD, in an organic solvent such as DCM, THF, and the like), to yield the
corresponding compound of formula (I).
A compound of formula (VI) is prepared from a compound of formula (V) as
outlined in Scheme 2.
A compound of formula (VI) is prepared by reacting a compound of formula
(V) with a compound of formula (II), or a compound of formula (III) according to the
procedures of Step A and Step B of Scheme 1, respectively. The group Y,
represents a protecting group. One skilled in the art will select the appropriate
protecting group compatible with the desired reactions. Examples of preferred
protecting groups include; carbamates, benzyl and substituted benzyl groups.
Especially preferred protecting groups are; tert-butyloxycarbonyl, 2,2,2-
trichloroethoxycarbonyl, alpha-chloroethoxycarbonyl, benzyl, 4-n'rtrobenzyl and
diphenylmethyl.
A compound of formula (XI) is prepared from a compound of formula (VI) as
outlined in Scheme 3.
Thus in Step A, a compound of formula (VI) in which Y2 is a nitrogen
protecting group is reacted under the appropriate conditions to obtain a compound
of formula (VII). In a preferred embodiment Y2 is a benzyl or tert-butoxycarbonyl
group which may be removed via hydrogenolysis or acidic hydrolysis respectively.
In a more preferred embodiment the protecting group is tert-butoxycarbonyl and is
removed with HCI in dioxane. A compound of formula (VIII) is obtained in Step B by
reacting a compound of formula (VII) and a compound of formula (XXa) in the
presence of a reducing agent such as sodium borohydride, sodium
cyanoborohydride, sodium triacetoxyborohydride, or hydrogen gas or phenylsilane in
the presence of a catalyst, and the like, in a solvent such as tetrahydrofuran,
methanol, ethanol, 1,2-dichloroethane, trifluoroethanol, and the like, to yield the
compound of formula (VIII). One skilled in the art will recognize that addition of acid
to decrease the pH of the reaction mixture to a pH of less than about 7 may be
necessary to effect reaction, wherein the acid is added as needed and is such as
acetic acid, hydrochloric acid, and the like. Preferred reducing agents are sodium
cyanoborohydride or sodium triacetoxyborohydride. A compound of formula (IX) is
prepared from a compound of formula (VIM) in Step C by reacting a compound of
formula (VIII), wherein R3 is -CN, with a reducing agent. A preferred reducing agent
is DIBAL in toluene at a temperature form -78 °C to ambient temperature, preferably
at 0 °C. A compound of formula (XI) is prepared from a compound of formula (IX) in
Step D, by reacting a compound of formula (IX) with a compound of formula (X) in
the presence of a reducing agent such as sodium borohydride, sodium
cyanoborohydride, sodium triacetoxyborohydride, hydrogen gas in the presence of a
catalyst, and the like, in a solvent such as methanol, ethanol, 1,2-dichloroethane,
trifluoroethanol, and the like. One skilled in the art will recognize that addition of
acid to decrease the pH of the reaction mixture to a pH of less than about 7 may be
necessary to effect reaction, wherein the acid is added as needed and is such as
acetic acid, hydrochloric acid, and the like. One skilled in the art will furthermore
recognize that a substituted or unsubstituted nonaromatic heterocycle containing
secondary amine functionality may be used in place of the compound of formula (X).
Preferred reducing agents are sodium cyanoborohydride or sodium
triacetoxyborohydride. In an alternative embodiment a compound of formula (XI) is
obtained from a compound of formula (VI). Thus a compound of formula (XII) is
obtained from a compound of formula (VI) using the conditions of Step C. A
compound of formula (XIII) is obtained from a compound of formula (XII) by reacting
38
a compound of formula (XII) with a compound of formula (X) according to the
conditions of Step D. A compound of formula (XIV) is obtained from a compound of
formula (XIII) according to the conditions of Step A. A compound of formula (XI) is
obtained from a compound of formula (XIV) by reacting a compound of formula (XIV)
with a compound of formula (XXa) according to the procedure of Step B.
A compound of formula (XVI) is prepared from a compound of formula (IX),
and a compound of formula (XVII) is prepared from a compound of formula (XII) as
outlined in Scheme 4.
A compound of formula (XVI) is prepared from a compound of formula (IX) by
reacting a compound of formula (IX) with a compound of formula (XV). Thus a
compound of formula (XVI) is reacted with a compound of formula (XV) in the
presence of sodium metabisulfite in DMA at 80 to 120 °C to give a compound of
formula (XVI). In a further embodiment a compound of formula (XII), wherein Y2 is a
protecting group, is reacted with a compound of formula (XV) under the same
conditions to give a compound of formula (XVII).
A compound of formula (XXI), formula (XXIII) and formula (XIX) is prepared
as outlined in Scheme 5.
Scheme 5
A compound of formula (XXI) is prepared from a compound of formula (I) in
which R3 is halogen by reacting a compound of formula (XXII) according to the
Sonogashira coupling or Heck coupling procedures. In both cases the preferred
halogen is Br and I with I being especially preferred. In a preferred embodiment a
compound of formula (I) is reacted with a compound of formula (XXII) in the
presence of a palladium catalyst and in the presence or absence of a copper
catalyst. In a more preferred embodiment the catalyst is selected from; Pd2(dba)3,
PdCI2 and Pd(OAc)2 with or without a phosphine additive. Preferred additives are
triphenylphosphine and tri-(tert-butyl)phosphine. When the compound of formula
(XXII) contains a triple bond (that is, when XXII is a terminal acetylene) an additional
copper catalyst may be desirable. A preferred catalyst is a Cu(l) halide with Cul
being especially preferred. A further preferred embodiment includes catalysts,
together with a base selected from an amine, Na2CO3, K2CO3, or the like. A
particularly preferred base is aqueous K2CO3. These reactions are carried out in a
solvent selected from, THF, DMF, DME, DMA, benzene, toluene, DCM, or the like.
Preferred solvents are THF and DME at from ambient temperature to the boiling
point of the solvent. A compound of formula (XXIII) is prepared from a compound of
formula (1) by reacting a compound of formula (I) where R3 is halogen, preferably I,
with a compound of formula (XXIV), according to the Suzuki procedure, in the
presence of a catalyst such as tetrakis(triphenylphosphine) palladium (0), and the
like, in the presence of a base such as sodium carbonate, potassium carbonate. A
compound of formula (XIX) is prepared from a compound of formula (I). Thus a
compound of formula (I), in which R3 is halogen, preferably Br and I, is reacted with
an organolithium reagent and subsequently with a compound of formula (XXb), in a
solvent to give a compound of formula (XVIII). In a preferred embodiment the
organolithium reagent is n-BuLi in THF at a temperature from -100 to 0 °C,
preferably at -78 °C. A compound of formula (XIX) is prepared from a compound of
formula (XVIII) by reacting a compound of formula (XVIII) with a reducing agent,
42
such as sodium borohydride, or sodium cyanoborohydride in the presence of an acid
such as HCI, AcOH, TFA, or the like, in a solvent such as THF and ether. In an
alternative embodiment the compound of formula (XVIII) may be reacted with either
hydrogen in the presence of a catalyst such as palladium on carbon, or triethylsilane
in the presence of TFA. In preferred embodiment a compound of formula (XVIII) is
reacted with triethylsilane in DCM at ambient temperature in the presence of TFA.
A compound of formula (XXV) is prepared from a compound of formula
(XXIV) according to the procedure outlined in Scheme 6.
A compound of formula (XXIV), where Y, represents an oxygen protecting
group, is reacted with a compound of formula (XXVII), where Z2H represents a
heterocycle in which a nitrogen-hydrogen bond is present, to give a compound of
formula (XXVI) where the group Z2 is attached via a nitrogen atom, in a preferred
embodiment a compound of formula (XXIV) is reacted with a compound of formula
(XXVII) in the presence of Cu(OAc)2, in the presence of a base such as pyridine, a
dehydrating agent such as molecular sieves 4A in DCM at ambient temperature to
give a compound of formula (XXVI). Removal of the protecting group Y1 is
accomplished under the appropriate conditions to give a compound of formula
(XXVIII). One skilled in the art would be capable of selecting an appropriate
protecting group. In one embodiment Y1 is a substituted benzyl ether, which may be
removed upon treatment with hydrogen in the presence of palladium on carbon. A
compound of formula (XXV) is obtained by reacting a compound of formula (XXVIII)
with a compound of formula (IV) according to the procedures outlined in Step B of
Scheme 1.
A compound of formula (XXX) is obtained from a compound of formula (XXIX)
as outlined in Scheme 7.
A compound of formula (XXXI) is obtained from a compound of formula
(XXIX) by reacting a compound of formula (XXIX) with a reducing agent in a solvent.
Reducing agents include hydrogen gas over a catalyst, for example palladium,
platinum, Raney nickel or the like. In a preferred embodiment the reducing agent is
SnCI2 in ethanol in the presence or absence of water at ambient temperature to the
boiling point of the solvent. A compound of formula (XXXII) is prepared from a
compound of formula (XXXI) by reacting a compound of formula (XXXI) with a
compound of formula (XXXIII) in which X2 is a leaving group. In a preferred
embodiment X2 is CI and OH. Thus when X2 is Cl the compound of formula (XXXI)
is reacted with a compound of formula (XXXIII) in a solvent such as DCM or THF in
the presence of a base such as triethylamine or pyridine. When X2 is OH a
compound of formula (XXXI) is reacted with a compound of formula (XXXIII) via
peptide coupling, for example in the presence of DCC in a solvent such as DCM or
THF. A compound of formula (XXX) is prepared from a compound of formula
(XXXII) using a reducing agent. Reducing agents include; BH3:THF, and BH3:Me2S.
A preferred reducing agent is BH3:Me2S, in THF at elevated temperature, preferably
at the boiling point of the solvent.
A compound of formula (XXXV) may be prepared according to the processes
outlined in Scheme 8.
A compound of formula (XXXIV) is reacted with a compound of formula (X) in
the presence of a reducing agent such as sodium borohydride, sodium
cyanoborohydride, sodium triacetoxyborohydride, hydrogen gas in the presence of a
catalyst, and the like, in a solvent such as methanol, ethanol, 1,2-dichloroethane,
trifluoroethanol, and the like, to yield the compound of formula (XXXVI). One skilled
in the art will recognize that addition of acid to decrease the pH of the reaction
mixture to a pH of less than about 7 may be necessary to effect reaction, wherein
the acid is added as needed and is such as acetic acid, hydrochloric acid, and the
like. One skilled in the art will furthermore recognize that a substituted or
unsubstituted nonaromatic heterocycle containing secondary amine functionality
may be used in place of the compound of formula (X). A compound of formula
(XXXVI) is reacted with a compound of formula (IV) according to the procedure of
Step B, Scheme 1, to give a compound of formula (XXXV).
Compounds of formula (XXXVIII) may be prepared according to the
processes outlined in Scheme 9.
A compound of formula (XXXVII) where R3 is selected from Br and I,
preferably I, is reacted with a compound of formula (XXXIX) in the presence of a
catalyst such as tris(dibenzylidineacetone)dipalladium(0), and the like, in the
presence of a base such as sodium t-butoxide, cesium carbonate, triethylamine,
potassium carbonate, and the like, in an organic solvent such as THF or dioxane,
and the like, preferably in the presence of BINAP (2,2'-bis(diphenylphosphino)-1,1'-
dinaphthyl) and 18-Crown-6 (a crown ether), to yield the corresponding compound of
formula (XL). A compound of formula (XLI) may be obtained from a compound of
formula (XL) via the removal of the protecting group Y1 followed by reaction with the
compound of formula (IV) according to the procedures of Scheme 1, Step B. A
compound of formula (XLII) is prepared from a compound of formula (XLI) via
removal of the protecting group Y2. One skilled in the art will further recognize and
understand the concept of orthogonal protection such that the groups Y^ and Y2 may
be removed separately and at the appropriate points in the Scheme. A compound of
formula (XXXVIII) is prepared from a compound of formula (XLII) by reacting a
compound of formula (XLII) with a compound of formula (XXc) in the presence of a
reducing agent such as sodium borohydride, sodium cyanoborohydride, sodium
triacetoxyborohydride, hydrogen gas in the presence of a catalyst, and the like, in a
solvent such as methanol, ethanol, 1,2-dichloroethane, trifluoroethanol, and the like,
to yield the compound of formula (XXXVII). One skilled in the art will recognize that
addition of acid to decrease the pH of the reaction mixture to a pH of less than about
7 may be necessary to effect reaction, wherein the acid is added as needed and is
such as acetic acid, hydrochloric acid, and the like.
A compound of formula (XLIV) is prepared from a compound of formula
(XLIII) according to the procedure outlined in Scheme 10.
Compounds of formula (XLIII) are reacted with compounds of formula (IV)
according to the procedure of Step B, Scheme 1 to give compounds of formula
(XLV). Removal of the protecting group Y1 affords compound of formula (XLVI). In
a preferred embodiment the group Y1 is a benzyl group, thus the compound of
formula (XLV) is reacted with hydrogen or ammonium formate in the presence of a
catalyst such as palladium on carbon, or the like, in a solvent such as methanol,
ethanol and the like (i.e. catalytic hydrogenolysis) to yield the corresponding
compound of formula (XLVI). The compound of formula (XLVI) is reacted with a
compound of formula (XXXVII) where X3 is selected from the group consisting Cl, Br,
I, tosylate, mesylate, and the like, in the presence of a base such as sodium
hydroxide. TEA, sodium hydride, potassium carbonate, and the like, in an organic
solvent such as DCM, THF, DMF, DMA, and the like, to yield the corresponding
compound of formula (XLIV). In an alternative embodiment the compound of
formula (XXXVII) where X3 is OH is reacted with a compound of formula (XLVI)
according to the procedures of Step B, Scheme 1.
D. Formulation, Administration, and Therapy
The disclosed compounds, alone or in combination (with, for example, a
histamine H1 receptor antagonist), are useful for treating or preventing neurologic
disorders including sleep/wake and arousal/vigilance disorders (e.g. insomnia and
jet lag), attention deficit hyperactivity disorders (ADHD), learning and memory
disorders, cognitive dysfunction, migraine, neurogenic inflammation, dementia, mild
cognitive impairment (pre-dementia), Alzheimer's disease, epilepsy, narcolepsy,
eating disorders, obesity, motion sickness, vertigo, schizophrenia, substance abuse,
bipolar disorders, manic disorders and depression, as well as other histamine H3
receptor mediated disorders such as upper airway allergic response, asthma, itch,
nasal congestion and allergic rhinitis in a subject in need thereof.
1. Formulation and Administration
The compounds or compositions of the invention may be formulated and
administered to a subject by any conventional route of administration, including, but
not limited to, intravenous, oral, subcutaneous, intramuscular, intradermal and
parenteral administration. The quantity of the compound which is effective for
treating each condition may vary, and can be determined by one of ordinary skill in
the art.
For use in medicine, the salts of the compounds of this invention refer to non-
toxic "pharmaceutically acceptable salts." Other salts may, however, be useful in the
preparation of compounds according to this invention or of their pharmaceutically
acceptable salts. Suitable pharmaceutically acceptable salts of the compounds
include acid addition salts which may, for example, be formed by mixing a solution of
the compound with a solution of a pharmaceutically acceptable acid such as
hydrochloric acid, sulfuric acid, fumaric acid, maleic acid, succinic acid, acetic acid,
benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid.
Furthermore, where the compounds of the invention carry an acidic moiety, suitable
pharmaceutically acceptable salts thereof may include alkali ml salts, e.g., sodium or
potassium salts; alkaline earth ml salts, e.g., calcium or magnesium salts; and salts
formed with suitable organic ligands, e.g., quaternary ammonium salts.
Thus, representative pharmaceutically acceptable salts include the following:
acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate,
bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate,
dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate,
gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine,
hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isothionate, lactate,
lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide,
methylnitrate, methylsulfate, mucate, napsylate, nitrate, N-methylglucamine
ammonium salt, oleate, pamoate (embonate), palmitate, pantothenate,
phosphate/diphosphate, polygalacturonate, salicylate, stearate, sulfate, subacetate,
succinate, tannate, tartrate, teoclate, tosylate, triethiodide and valerate.
The present invention includes within its scope prodrugs of the compounds of
this invention. In general, such prodrugs will be functional derivatives of the
compounds which are readily convertible in vivo into the required compound. Thus,
irrthe methods of treatment of the present invention, the term "administering" shall
encompass the treatment of the various disorders described with the compound
specifically disclosed or with a compound which may not be specifically disclosed,
but which converts to the specified compound in vivo after administration to the
patient. Conventional procedures for the selection and preption of suitable prodrug
derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard,
Elsevier, 1985. In addition to salts, the invention provides the esters, amides, and
other protected or derivatized forms of the described compounds.
Where the compounds according to this invention have at least one chiral
center, they may accordingly exist as enantiomers. Where the compounds possess
two or more chiral centers, they may additionally exist as diastereomers. It is to be
understood that all such isomers and mixtures thereof are encompassed within the
scope of the present invention. Furthermore, some of the crystalline forms for the
compounds may exist as polymorphs and as such are intended to be included in the
present invention. In addition, some of the compounds may form solvates with
water (i.e., hydrates) or common organic solvents, and such solvates are also
intended to be encompassed within the scope of this invention.
The present invention also provides pharmaceutical compositions comprising
one or more compounds of this invention in association with a pharmaceutically
acceptable carrier and optionally additional pharmaceutical agents such as H1
antagonists or SSRIs. Preferably these compositions are in unit dosage forms such
as pills, tablets, caplets, capsules (each including immediate release, timed release
and sustained release formulations), powders, granules, sterile parenteral solutions
or suspensions (including syrups and emulsions), metered aerosol or liquid sprays,
drops, ampoules, autoinjector devices or suppositories; for oral parenteral,
intranasal, sublingual or rectal administration, or for administration by inhalation or
insufflation. Alternatively, the composition may be presented in a form suitable for
once-weekly or once-monthly administration; for example, an insoluble salt of the
active compound, such as the decanoate salt, may be adapted to provide a depot
preparation for intramuscular injection. For preparing solid compositions such as
tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g.
conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol,
talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other
pharmaceutical diluents, e.g. water, to form a solid preformulation composition
containing a homogeneous mixture of a compound of the present invention, or a
pharmaceutically acceptable salt thereof. When referring to these preformulation
compositions as homogeneous, it is meant that the active ingredient is dispersed
evenly throughout the composition so that the composition may be readily
subdivided into equally effective dosage forms such as tablets, pills and capsules.
This solid preformulation composition is then subdivided into unit dosage forms of
the type described above containing from 5 to about 1000 mg of the active
ingredient of the present invention. Examples include 5 mg, 7 mg, 10 mg, 15 mg, 20
mg, 35 mg, 50 mg, 75 mg, 100 mg, 120 mg, 150 mg, and so on. The tablets or pills
of the disclosed compositions can be coated or otherwise compounded to provide a
dosage form affording the advantage of prolonged action. For example, the tablet or
pill can comprise an inner dosage and an outer dosage component, the latter being
in the form of an envelope over the former. The two components can be septed by
an enteric layer which serves to resist disintegration in the stomach and permits the
inner component to pass intact into the duodenum or to be delayed in release. A
variety of material can be used for such enteric layers or coatings, such materials
including a number of polymeric acids with such materials as shellac, cetyl alcohol
and cellulose acetate.
The liquid forms in which the compounds and compositions of the present
invention may be incorporated for administration orally or by injection include,
aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions, and
flavoured emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil
or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Suitable
dispersing or suspending agents for aqueous suspensions, include synthetic and
natural gums such as tragacanth, acacia, alginate, dextran, sodium
carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone or gelatin.
Where the processes for the preparation of the compounds according to the
invention give rise to mixture of stereoisomers, these isomers may be separated by
conventional techniques such as preparative chromatography. The compounds may
be prepared in racemic form, or individual enantiomers may be prepared either by
enantiospecific synthesis or by resolution. The compounds may, for example, be
resolved into their component enantiomers by standard techniques, such as the
formation of diastereomeric pairs by salt formation with an optically active acid, such
as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by
fractional crystallization and regeneration of the free base. The compounds may
also be resolved by formation of diastereomeric esters or amides, followed by
chromatographic separation and removal of the chiral auxiliary. Alternatively, the
compounds may be resolved using a chiral HPLC column.
Advantageously, compounds of the present invention may be administered in a
single daily dose, or the total daily dosage may be administered in divided doses of
two, three or four times daily. Furthermore, compounds for the present invention can
be administered in intranasal form via topical use of suitable intranasai vehicles, or via
transdermal skin patches well known to those of ordinary skill in that art. To be
administered in the form of a transdermal delivery system, the dosage administration
will, of course, be continuous rather than intermittent throughout the dosage regimen.
For instance, for oral administration in the form of a tablet or capsule, the active
drug component can be combined with an oral, non-toxic pharmaceutically acceptable
inert carrier such as ethanol, glycerol, water and the like. Moreover, when desired or
necessary, suitable binders, lubricants, disintegrating agents and coloring agents can
also be incorporated into the mixture. Suitable binders include, without limitation,
starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners,
natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium
stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and
the like. Disintegrators include, without limitation, starch, methyl cellulose, agar,
bentonite, xanthan gum and the like.
The compound of the present invention can also be administered in the form of
liposome delivery systems, such as small unilamellar vesicles, large unilamellar
vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of
phospholipids, such as cholesterol, stearylamine or phophanaylcholines.
Compounds of the present invention may also be delivered by the use of
monoclonal antibodies as individual carriers to which the compound molecules are
coupled. The compounds of the present invention may also be coupled with soluble
polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone,
pyran copolymer, polyhydroxypropylmethacrylamidephenol,
polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysine substituted with
palmitoyl residue. Furthermore, the compounds of the present invention may be
coupled to a class of biodegradable polymers useful in achieving controlled release of
a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric
acid, polyoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-
linked or amphipathic block copolymers of hydrogels.
Compounds of this invention may be administered in any of the foregoing
compositions and according to dosage regimens established in the art whenever
treatment of ADHD is required.
The daily dosage of the products may be varied over a wide range from 1 to
1,000 mg per adult human per day. For oral administration, the compositions are
preferably provided in the form of tablets containing 1.0,5.0,10.0,15.0, 25.0, 50.0,
100, 250 and 500 milligrams of the active ingredient for the symptomatic adjustment of
the dosage to the subject to be treated. An effective amount of the drug is ordinarily
supplied at a dosage level of from about 0.01 mg/kg to about 20 mg/kg of body weight
per day. Preferably, the range is from about 0.02 mg/kg to about 10 mg/kg of body
weight per day, and especially from about 0.05 mg/kg to about 10 mg/kg of body
weight per day. The compounds may be administered on a regimen of 1 to 4 times
per day.
Optimal dosages to be administered may be readily determined by those skilled
in the art, and will vary with the particular compound used, the mode of administration,
the strength of the preparation, the mode of administration, and the advancement of
the disease condition. In addition, factors associated with the particular patient being
treated, including patient age, weight, diet and time of administration, will result in the
need to adjust dosages.
2. Combination Therapy
The disclosed compounds are useful in combination with other therapeutic
agents, including H1 receptor antagonists, H2 receptor antagonists, and
neurotransmitter modulators such as SSRIs and non-selective serotonin re-uptake
inhibitors (NSSRIs).
Methods are known in the art for determining effective doses for therapeutic
and prophylactic purposes for the disclosed pharmaceutical compositions or the
disclosed drug combinations, whether or not formulated in the same composition.
For therapeutic purposes, the term "jointly effective amount" as used herein, means
that amount of each active compound or pharmaceutical agent, alone or in
combination, that elicits the biological or medicinal response in a tissue system,
animal or human that is being sought by a researcher, veterinarian, medical doctor
or other clinician, which includes alleviation of the symptoms of the disease or
disorder being treated. For prophylactic purposes (i.e., inhibiting the onset or
progression of a disorder), the term "jointly effective amount" refers to that amount of
each active compound or pharmaceutical agent, alone or in combination, that
inhibits in a subject the onset or progression of a disorder as being sought by a
researcher, veterinarian, medical doctor or other clinician, the delaying of which
disorder is mediated, at least in part, by the modulation of one or more histamine
receptors. Thus, the present invention provides combinations of two or more drugs
wherein, for example, (a) each drug is administered in an independently
therapeutically or prophylactically effective amount; (b) at least one drug in the
combination is administered in an amount that is sub-therapeutic or sub-prophylactic
if administered alone, but is therapeutic or prophylactic when administered in
combination with the second or additional drugs according to the invention; or
(c) both drugs are administered in an amount that is sub-therapeutic or sub-
prophylactic if administered alone, but are therapeutic or prophylactic when
administered together. Combinations of three or more drugs are analogously
possible. Methods of combination therapy include co-administration of a single
formulation containing all active agents; essentially contemporaneous administration
of more than one formulation; and administration of two or more active agents
separately formulated.
E. Examples

4-Piperidin-1-ylmethyl-phenol
A solution of 4-hydroxybenzaldehyde (10 g), piperidine (8.9 mL), and acetic
acid (4.7 mL) in DCE (200 mL) was treated with sodium triacetoxyborohydride
(24 g). After 16 h, the resulting mixture was treated with saturated aqueous sodium
bicarbonate (100 mL) and extracted with DCM (5x100 mL). The combined organic
phases were dried (magnesium sulfate) and evaporated. Trituration of the residue
with ethyl acetate gave the title compound as a white crystalline solid (5.5 g).

4-(1-lsopropyl-piperidin-4-yloxy)-benzaldehyde
A suspension of the product of Example 3 (5./ g). 4-fluorobenzaldehyde
(1.7 mL), and cesium carbonate (13 mg) in DMF (40 mL) was heated to 100 °C for
22 h, and allowed to cool to RT. Water (100 mL) was added, and the resulting
mixture was extracted with ether (3x100 mL). The combined organic phases were
dried (magnesium sulfate) and evaporated. Chromatography of the residue (0-15%
2M methanolic ammonia) gave the title compound as a colorless oil (2.0 g). 1H NMR
(400 MHz, CDCI3): 9.86 (s, 1H), 7.81 (d, J=8.6 Hz, 2H), 6.99 (d, J=8.6 Hz, 2H), 4.43
(m, 1H), 2.82-2.72 (m, 3H), 2.42 (m, 4H), 2.08-2.00 (m, 2H), 1.89-1.80 (m, 2H), 1.06
(d, J=6.6 Hz, 6H).

A solution of 1-isopropyl-piperidin-4-one (51.2 g) in absolute ethanol (350 mL)
was treated with sodium borohydride (7.30 g) at a rate not to exceed an internal
temperature of 50°C. After 48h, the solvent was evaporated and the resulting yellow
paste was partitioned between DCM (300 mL) and 5% NaOH (300 mL). This mixture
was stirred for 6h and then extracted with DCM (4x100 mL). The combined organic
phases were dried (sodium sulfate) and evaporated. Distillation of the yellow oil (bp
68 °C, 1.5 mm Hg) gave the title compound as an off-white waxy solid (35.3 g).
4-[4-(1-tert-Butoxycarbonyl-piperidin-4-yloxy)-phenyi]-pipera2ine-1-carboxylic
acid tert-butyl ester
A suspension of 4-(4-Hydroxy-phenyl)-piperazine-1 -carboxylic acid tert-butyl
ester (3.11 g), 4-Hydroxy-piperidine-1-carboxylic acid tert-butyl ester (3.51g), and
polymer supported triphenylphosphine (6.25 g; loading: 3 mmol/g) in DCM (100 mL)
was treated with di-tert-butylazodicarboxylate (3.78 g). After 24 h, the resulting
mixture was filtered, and the filtrate was evaporated. Chromatography of the residue
(15-30% ethyl acetate/hexanes) gave the title compound as a white solid (3.20 g).
A solution of 4-hydroxy-piperidine-1-carboxylic acid tert-butyl ester (14.99 g)
and 4-chlorobenzonitrile (10.35 g) in DMF (100 mL) was treated with sodium hydride
(60%, 3.8g). The resulting dark mixture was then heated to 65 C for 16 h, and
allowed to cool to RT. The mixture was poured into water (1 L) and extracted with
ether (3x400 mL). The combined organic phases were evaporated and the brown oil
dissolved in methanol (500 mL) and treated with concentrated hydrochloric acid (20
mL). After 24h the methanol was removed, 5% aqueous sodium hydroxide (300 mL)
and water (300 mL) were added, and the mixture was extracted with DCM
(3x300 mL). The combined organic phases were dried (sodium sulfate) and
evaporated. Bulb to bulb distillation of the residue gave the title compound as an off
white waxy solid (12.23 g).

4-(4-l midazol-1 -y!-phenoxy)-piperidine
A solution of 4-hydroxy-piperidine-1-carboxylic acid tert-butyl ester (1.6 g), 4-
imidazol-1-yl-phenol (1.2 g), and triphenylphosphine (2.4 g) in THF (10 mL) was
treated with a solution of di-tert-butylazodicarboxylate (2.1 g) in THF (5 mL). After
24 h, the resulting mixture was evaporated. Chromatography of the residue (0-10%
2 M methanolic ammonia/DCM) gave a colorless glassy solid (1.9 g). This material
was dissolved in methanol (10 mL) and treated with 2 M ethereal hydrogen chloride
(14 mL). After 24 h, a white solid formed, which was filtered and washed with 1:1
methanol-ether and ether, yielding a white powder (1.2 g). To this material (1.1 g)
was added 10% aqueous sodium hydroxide (20 mL) and DCM (20 mL). After 10 min
vigorous stirring, the resulting mixture was extracted with DCM (2x15 mL). The
combined organic phases were dried (magnesium sulfate) and evaporated, giving
the title compound as a light pink solid (781 mg).

4-(4-Moroholin-4-ylmethyl-phenoxy)-piperidine-1 -carboxylic acid tert-butyl
ester
A suspension of tert-butyl 4-hydroxy-1-piperidinecarboxylate (32.4 g), 4-
flourobenzaldehyde (20.0 g), and cesium carbonate (52.5 g) in DMF (320 mL) was
heated to 110 C for 48 h, and allowed to cool to RT. Water (400 mL) was added,
and the resulting mixture was extracted with ether (3x500 mL). The combined
organic phases were washed with water (3x200 mL), brine (200 mL), dried
(magnesium sulfate) and evaporated. Toluene (2x200 mL) was added, and then
evaporated, giving a brown-yellow oil (47.2 g). A solution of this oil (7.0 g),
morpholine (2.4 mL), and acetic acid (1.3 mL) in DCM (300 mL) was treated with
sodium triacetoxyborohydride (5.9 g). After 16 h, the resulting mixture was treated
with 10% aqueous sodium hydroxide (50 mL), and extracted with DCM (2x500 mL).
The combined organic phases were washed with water (100 mL), brine (100 mL),
and then dried (magnesium sulfate) and evaporated. Chromatography of the residue
(0.5 to 5.5% 2 M methanolic ammonia/DCM) gave the title compound as a yellow
solid (6.6 g).

4-[4-(Piperidin-4-yloxy)-benzyl]-morpholine
A solution of the product of Example 11 (6.6 g) in dioxane (40 mL) was
treated with 4 N hydrogen chloride in dioxane (30 mL). The resulting mixture was
stirred at RT for 16 h. Solvent was evaporated and the residue was treated with 10%
aqueous sodium hydroxide (50 mL). The resulting mixture was extracted with DCM
(2x500 mL). The combined organic phases were washed with water (200 mL), brine
(200 mL), dried (magnesium sulfate), and evaporated to give the title compound as
a yellow oil (5.4 g). 1H NMR (400 MHz, CDCI3): 7.21 (d, J = 8.6 Hz, 2H), 6.86 (d, J =
8.6 Hz, 2H), 4.38-4.34 (m, 1H), 3.70 (t, J = 4.6 Hz, 4H), 3.43 (s, 2H), 3.19-3.13 (m,
2H), 2.42 (t, J = 4.3 Hz, 2H), 2.10-2.00 (m, 2H), 1.73-1.65 (m, 2H).
1-[4-(Piperidin-4-yloxy)-benzyl]-piperidine
A suspension of tert-butyl 4-hydroxy-1-piperidinecarboxylate (32.4 g), 4-
fluorobenzaldehyde (20.0 g), and cesium carbonate (52.5 g) in DMF (320 mL) was
heated to 110 C for 48 h and allowed to cool to RT. Water (400 mL) was added,
and the resulting mixture was extracted with ether (3x500 mL). The combined
organic phases were washed with water (3x200 mL), brine (200 mL), dried
(magnesium sulfate) and evaporated. The residue was twice treated with toluene
(2x200 mL) and evaporated, giving a brown-yellow oil (47.2 g). A solution of this oil
(7.0 g), piperidine (2.8 mL), and acetic acid (1.3 mL) in DCM (300 mL) was treated
with sodium triacetoxyborohydride (5.9 g). After 16 h, the resulting mixture was
treated with 10% aqueous sodium hydroxide (50 mL), and extracted with DCM
(2x500 mL). The combined organic phases were washed with water (100 mL), brine
(100 mL), and then dried (magnesium sulfate) and evaporated. Chromatography of
the residue (0.5 to 5.5% 2 M methanolic ammonia/DCM) gave the title compound as
a yellow oil (8.6 g).
A solution of this oil (8.6 g) in dioxane (30 mL) was added 4 N hydrogen
chloride in dioxane (40 mL). The resulting mixture was stirred at RT for 16 h and
evaporated. The residue was treated with 10% aqueous sodium hydroxide (50 mL)
and extracted with DCM (2x500 mL). The combined organic phases were washed
with water (200 mL), brine (200 mL), dried (magnesium sulfate), and evaporated.
Chromatography of the residue (0.5 to 5.5% 2 M methanolic ammonia/DCM) gave
the title compound as an ivory solid (3.4 g). 1H NMR (400 MHz, CDCI3): 7.20 (d, J =
8.5 Hz, 2H), 6.85 (d, J = 8.6 Hz, 2H), 4.37-4.30 (m, 1H), 3.40 (s, 2H), 3.17-3.11 (m,
2H), 2.75-2.68 (m, 2H), 2.35 (brs, 4H), 2.03-1.99 (m, 2H), 1.69-1.61 (m, 2H), 1.59-
1.53 (m, 2H), 1.43-1.39 (m, 2H).

3-(1-lsopropyl-piperidin-4-yloxy)-benzaldehyde
A solution of the product of Example 3 (716 mg), 3-hydroxybenzaldehyde
(672 mg), and triphenylphosphine (1.6 g) in THF (5 mL) was treated with a solution
of di-terf-butylazodicarboxylate (1.4 g) in THF (5 mL). After 16 h, the resulting
mixture was evaporated. The residue was treated with ether (20 mL) and 20%
aqueous hydrochloric acid (20 mL). The aqueous phase was washed with ether
(20 mL), neutralized with solid sodium carbonate, and extracted with ether
(3x20 mL). The combined organic phases were dried (magnesium sulfate), and
evaporated. Chromatography of the residue (0-10% 2M methanolic ammonia/DCM)
gave the title compound as a colorless oil (50 mg).
4-[4-(1-Benzyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), benzaldehyde (0.051 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
ceiite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a white solid (94 mg). 1H NMR (400 MHz, CDCI3): 7.33-7.23 (m, 5H),
7.20 (d, J = 8.6 Hz, 2H), 6.85 (d, J = 8.6 Hz, 2H), 4.32-4.23 (m, 1H), 3.69 (t, J = 4.6
Hz, 4H), 3.53 (s, 2H), 3.42 (s, 2H), 2.77-2.73 (m, 2H), 2.42 (t, J = 4.3 Hz, 4H), 2.32-
2.26 (m, 2H), 2.02-1.96 (m, 2H), 1.85-1.77 (m, 2H).
4-[4-(1-Cydohexyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), cyclohexanone (0.052 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
celite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (73 mg). 1H NMR (400 MHz, CDCI3): 7.21 (d, J = 8.6 Hz,
2H), 6.85 (d, J = 8.6 Hz, 2H), 4.33-4.29 (m, 1H), 3.70 (t, J = 4.6 Hz, 4H), 3.42 (s,
2H), 2.92-2.86 (m, 2 H), 2.57-2.51 (m, 2H), 2.43-2.28 (m, 7 H), 2.09-2.02 (m, 2H),
1.92-1.81 (m, 6H), 1.31-1.20 (m, 4H).
4-[4-(4-Morpholin-4-ylmethyl-phenoxy)-piperidin-1-yl]-butan-2-one
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), 4-hydroxy-2-butanone (44 mg) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsiiane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
celite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (33 mg). 1H NMR (400 MHz, CDCl3): 7.04 (d, J = 8.5 Hz,
2H), 6.84 (d, J = 8.6 Hz, 2H), 4.32-4.27 (m, 1H), 3.70 (t, J = 4.6 Hz, 4 H), 3.42 (s,
2H), 2.74-2.62 (m, 4H), 2.43 (t, J = 4.2 Hz, 4H), 2.33-2.27 (m, 2H), 2.18 (s, 3H),
2.02-1.76 (m,6H).
4-[4-(1-Cyclohexylmethyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), cyclohexanecarboxaldehyde (0.061 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
ceiite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (100 mg). 1H NMR (400 MHz, CDCI3): 7.20 (d, J = 8.6
Hz, 2H), 6.85 (d, J = 8.6 Hz, 2H), 4.29-4.23 (m, 1H), 3.70 (t, J = 4.6 Hz, 4H), 3.42 (s,
2H), 2.70 (t, J = 4.6 Hz, 2H), 2.43 (t, J = 4.2 Hz, 4H), 2.20 (t, J = 9.1 Hz, 1H), 2.13
(d, J = 7.1 Hz, 1H), 2.00-1.64 (m, 9H), 1.52-1.43 (m, 1H), 1.28-1.13 (m, 3H), 0.91-
0.82 (m, 2H).
4-[4-( 1 -lsobutyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), isobutyraldehyde (0.091 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
celite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (198 mg). 1H NMR (400 MHz, CDCI3): 7.20 (d, J = 8.6
Hz, 2H), 6.85 (d, J = 8.6 Hz, 2H), 4.29-4.23 (m, 1H), 3.70 (t, J = 4.7 Hz, 4H), 3.42 (s,
2H), 2.72-2.69 (m, 2H), 2.42 (t, J = 4.4 Hz, 4H), 2.19 (t, J = 9.0 Hz, 1H), 2.09 (d, J =
7.3 Hz, 1H), 2.00-1.95 (m, 2H), 1.83-1.72 (m, 3H), 0.90 (d, J = 6.6 Hz, 6H).
4-[4-(1-Propyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), propionaldehyde (0.072 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
celite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (85 mg). 1H NMR (400 MHz, CDCI3): 7.21 (d, J = 8.6 Hz,
2H), 6.85 (d, J = 8.6 Hz, 2H), 4.34-4.28 (m, 1H), 3.70 (t, J = 4.7 Hz, 4H), 3.42 (s,
2H), 2.79-2.75 (m, 2H), 2.42 (t, J = 4.3 Hz, 4H), 2.37-2.33 (m, 4H), 2.06-1.99 (m,
2H), 1.88-1.80 (m, 2H), 1.59-1.50 (m, 2H), 0.91 (t, J = 7.4 Hz, 6H).
4-{4-[1 -(1 -Methyl-heptyl)-piperidin-4-yloxy]-benzyl}-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), 2-octanone (0.156 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
celite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (108 mg). 1H NMR (400 MHz, CDCI3): 7.20 (d, J = 8.5
Hz, 2H), 6.85 (d, J = 8.6 Hz, 2H), 4.29-4.24 (m, 1H), 3.70 (t, J = 4.7 Hz, 4H), 3.42 (s,
2H), 2.80-2.73 (m, 2H), 2.60-2.57 (m, 1H), 2.48-2.33 (m, 6H), 1.99 (m, 2H), 1.84-
1.74 (m, 1H), 1.57-1.54 (m, 9H), 0.94 (d, J = 6.5 Hz, 3H), 0.90-0.87 (m, 3H).
4-[4-(1-sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), 2-butanone (0.089 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
celite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (75 mg). 1H NMR (400 MHz, CDCI3): 7.20 (d, J = 8.6 Hz,
2H), 6.85 (d, J = 8.6 Hz, 2H), 4.29-4.24 (m, 1H), 3.70 (t, J = 4.7 Hz, 4H), 3.42 (s,
2H), 2.81-2.73 (m, 2H), 2.51-2.32 (m, 6H), 2.05-1.95 (m, 2H), 1.83-1.75 (m, 2H),
1.62-1.56 (m, 1H), 1.34-1.26 (m, 2H), 0.99 (d, J = 6.6 Hz, 3H), 0.90 (t, J = 7.4 Hz,
3H).
4-[4-( 1 -Cyclopentyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 10 (137 mg), cyclopentanone (0.088 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, additional phenylsilane
(0.036 mL) was added. After 4 h, the resulting mixture was filtered through a pad of
celite, and the pad was washed with DCM (3x3 mL). The combined filtrates were
chromatographed (0.5-5.5% 2 M methanolic ammonia/DCM), giving the title
compound as a colorless oil (168 mg). 1H NMR (400 MHz, CDCI3): 7.22 (d, J = 8.6
Hz, 2H). 6.85 (d, J = 8.6 Hz, 2H), 4.44-4.35 (m, 1H), 3.70 (t, J = 4.7 Hz, 4H), 3.43 (s,
2H), 2.95-2.88 (m, 1H), 2.76-2.54 (m, 2H), 2.42 (t, J = 4.4 Hz, 4H), 2.16-2.08 (m,
2H), 1.94-1.91 (m, 4H), 1.78-1.72 (m, 2H), 1.62-1.57 ( m, 4H).
4-[4-(4-Piperidin-1 -ylmethyl-phenoxy)-piperidin-1 -yl]-butan-2-one
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 9 (137 mg), 4-hydroxy-2-butanone (44 mg) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celrte, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (42 mg). 'H NMR (400
MHz, CDCI3): 7.19 (d, J = 8.6 Hz, 2H), 6.83 (d, J = 8.6 Hz, 2H), 4.31-4.26 (m, 1H),
3.40 (s, 2H), 2.75-2.61 (m, 4H), 2.36-2.22 (m, 6H), 2.18 (s, 3H), 2.21 A.12 (m, 6H),
1.59-1.53 (m, 4H), 1.47-1.35 (m, 2H).
[4-(1 -lsopropyl-piperidin-4-yloxy)-benzyI]-(5-methyl-pyndln-2-yl)-amine
A solution of the product of Example 2 (175 mg), 2-amino5-methyipyridine
(87 mg), and acetic add (0.05 mL) in DCE (3 mL) was treated with sodium
triacetoxyborohydride (257 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (1 mL), and the mixture was extracted with DCM (3x3 mL).
The combined organic phases were dried (magnesium sulfate) and evaporated.
Chromatography of the residue (0-8% 2M methanolic ammonia/DCM) gave the title
compound as a yellow waxy solid (103 mg). 1H NMR (400 MHz, COCy: 7.93 (s, 1H),
7.26-7.21 (m, 3H), 6.86 (d, J = 8.6 Hz, 2 H), 6.32 (d, J = 8.41,1 H), 4.66-4.61 (br m,
1 H). 4.39 (d. J = 5.7,2H). 4.31-424 (m. 1H). 2.81-2.70 (m. 3H), 2.42-2.34 (m, 2H).
2.17 (s, 3H), 2.04-1.96 (m, 2H), 1.86-1.76 (m, 2H). 1.07 (d, J = 6.7,6H).
(5-Chloro-pyrldin-2-yl)-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-amine
A solution of the product of Example 2 (200 mg), 2-amino-5-chloropyridine
(104 mg), and acetic acid (0.05 mL) in DCE (3 ml_) was treated with sodium
triaoetoxyborohydride (257 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (1 mL), and the mixture was extracted with DCM (3x3 mL).
The combined organic phases were dried (magnesium suHate) and evaporated.
Chromatography of the residue (0-8% 2M methanolic ammonia/DCM) gave the title
compound as a yelow amorphous solid (137 mg). 1H NMR (400 MHz. CDCW: 8.05
(s, 1H), 7.35 (dd, J = 8.8, 2.5 Hz, 1H), 7.24 (d, J =» 8.6 Hz, 2H), 6.87 (d, J = 8.8 Hz,
2H), 6.32 (d. J * 9.0 Hz, 1H), 4.84-4.78 (m, 1H), 4.39 (d, J = 5.7 Hz, 2H). 4.31-4.25
(m, 1H), 2.81-2.70 (m. 3H), 2.41-2.35 (m, 2H), 2.05-1.96 (m, 2H). 1.84-1.76 (m, 2H),
1.05(d,J = 6.1Hz,6H).
[4-(1-isopropyl-piperidin-4-yloxy)-benzyl]-phenyl-amine
A solution of the product of Example 2 (200 mg), aniline (104 mg), and acetic
acid (0.05 mL) in DCE (3 mL) was treated with sodium triacetoxyborohydride
(257 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(1 mL), and the mixture was extracted with DCM (3x3 mL). The combined organic
phases were dried (magnesium sulfate) and evaporated. Chromatography of the
residue (0-6% 2M methanolic ammonia/DCM) gave the title compound as a yellow
amorphous solid (136 mg). 1H NMR (400 MHz, CDCl3: 7.27 (d, J = 8.2 Hz, 1H),
7.19-7.15 (m, 2H), 6.89 (d. J = 8.6 Hz, 2H), 6.73-6.69 (m. 1H). 6.65-6.62 (m, 2H).
4.31-4.23 (m, 3H), 3.96-3.90 (brs, 1H), 2.83-2.70 (m, 3H), 2.43-2.34 (m, 2H). 2.05-
1.96 (m, 2H), 1.86-1.76 (m, 2H). 1.06 (d. J = 6.7,6H).
1 -[4-{1 -Benzyl-piperidin-4-yloxy)-benzyl]-piperidine
Phenyteilane (0.068 mL) was added dropwise to a solution of the product of
Example 9 (137 mg), benzaldehyde (0.051 mL) and scandium
trifluoromethanesuffonate (13 mg) in THF (1 mL). After 16 h. the resulting mixture
was filtered through a pad of cellte, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM). giving the title compound as a white solid (145 mg). 1H NMR (400
MHz, COO,): 7.34-7.23 (m, 5H), 7.21 (d, J = 8.6 Hz, 2H), 6.85 (d, J = 8.6 Hz, 2H),
4.32-4.26 (m, 1H), 3.53 (s, 2H), 3.48 (s, 2H), 2.90 (bs. 1H). 2.76-2.72 (m, 2H), 2.43
(bs, 3H), 2.39 (t J = 8.7 Hz, 2H), 2.01-1.96 (m. 2H), 1.85-1.77 (m, 2H), 1.63-1.57
(m,4H), 1.47-1.38 (m,2H).
1 -{4-(1-Cycohexyl-piperidin-4-yloxy)-benzyl]-piperidina
Phenyfsliane (0.068 mL) was added dropwtee to a solution of the product of
Example 9 (137 mg), cydohexanone (0.052 mL) and scandium
trrfluorornethanesulfonate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celite, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (141 mg). 1H NMR (400
MHz. CDCI,): 7.10 (d, J = 8.6 Hz, 2H), 6.84 (d. J = 8.6 Hz, 2H), 4.30-4.25 (m, 1H),
3.40 (s, 2H). 2.88-2.83 (m, 2H), 2.50-2.45 (m, 2H), 2.40-2.29 (m, 5H), 2.07-1.74 (m,
8H), 1.65-1.62 (m, 1H). 1.59-1.54 (m. 4H), 1.45-1.42 (m, 4H), 1.30-1.19 (m. 4H),
1.14-1.07 (m,1H).
1-4-1(1-Cyclohexylmethyl-piperdin-4-yloxy)-benzyl]-piperidine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 9 (137 mg), cyclohexanecarbozaldehyde (0.061 mL) and scandium
trifuoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celite, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (163 mg). 1H NMR (400
MHz, COCy: 7.19 (d, J = 8.4 Hz. 2H), 6.84 (d, J = 8.5 Hz, 2H), 4.27-4.23 (m, 1H),
3.40 (s. 2H). 2.76-2.64 (m, 2H), 2.43-2.28 (bs, 4H), 2.24-2.08 (m, 4H), 2.03-1.92 (m,
2H), 1.65-1.62 (m, 1H). 1.85-1.36 (m, 13H), 1.30-1.09 (m, 4H), 0.93-0.79 (m, 2H).
4-[3-{1-Isopropyl-piperidin-4-yloxy)-benzyl]-morpholine
A solution of the product of Example 10 (50 mg) and morpholine (21 mg) in
DCE (2 mL) was treated with sodium triaoatoxybofohydride (64 mg). After 16 h. the
resultina mixture was treated with 10% sodium hydroxide (1 mL), and the mixture
was extracted with DCM (3x2 mL). The combined organic phases were dried
(magnesium suffate)andevaporatod.- Chromatography^of the-residue (0-15% 2M
methanoiic ammonia/DCM) gave the title compound as a colorless glassy OH
(46 mg) 1H NMR (400MHz,-CDCl2) 7 20(t, J = 7.6 Hz, 1H), 6.91-6.87 (m, 2H),
6.82-6.78 (m. 1H), 4.34-4.27 (m. 1H), 3.71 (t J = 3.71.4H), 3.46 (s. 2H). 2.83-2.71
(m, 3H), 2.47-2.35 (m. 6H). 2.05-1.97 (m. 2H), 1,86-1,77 (m, 2H), 1.06 (d. J = 6.5
Hz, 6H).
1-[4-(1 -Propyl-piperidin-4-yloxy)-benzyi]-piperidine
Phenyteitene (0.068 mL) was added dropwise to a solution of the product of
Example 9 (137 mg), propbnaldehyde (0.072 mL) and scandium
trifluoromethanesutfbnate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celite, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanofic
ammonia/DCM), giving the title compound as a colorless oil (105 mg). 1H NMR (400
MHz. CDCg: 7.19 (d, J = 8.4 Hz, 2H). 6.84 (d, J = 8.5 Hz, 2H), 4.30-4.26 (m. 1H),
3.40 (s, 2H), 2.75-2.70 (m. 2H). 2.40-2.20 (m, 7H), 2.06-1.93 (m, 2H). 1.86-1.68 (m,
5H), 1.59-1.37 (m, 6H). 0.92-0.86 (m, 3H).
1-[4-(1-lsobutyl-piperidin-4-yioxy)-benzyq-piperidine
PhenylsBane (0.068 mL) was added dropwise to a solution of the product of
Example 9 (137 mg), teotxrtyraldehyde (0.091 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celite, and the pad was washed with DCM (3x3 mL).
The combined fitrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (91 mg). 'H NMR (400
MHz, CDCI,): 7.19 (d, J = 8.6 Hz, 2H), 6.84 (d, J = 8.6 Hz, 2H). 4.28-4.23 (m, 1H),
3.39 (s. 2H), 2.72-2.69 (m. 2H), 2.35 (bs, 4H), 2.21-2.16 (m, 2H), 2.09 (d. 2H), 2.01-
1.95 (m, 3H), 1.59-1.53 (m, 3H), 1.44-1.38 (m, 2H). 0.90 (d, J = 6.6 Hz, 6H).
K, = 16 nM
1 -[4-[1 -(1 -Methyl-hepty|)-piperidin-4-yloxy]-benzyl}-piperidine
Phenytsilane (0.068 rnL) was added dropwise to a solution of the product of
Example 9 (137 mg), 2-octanone (0.156 mL) and scandium
trifluoromethanesutfonate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celite, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (228 mg). 1H NMR (400
MHz. CDCI3): 7.19 (d, J = 8.5 Hz, 2H), 6.84 (d, J = 8.6 Hz, 2H), 4.28-4.22 (m, 1H),
3.40 (s, 2H), 2.79-2.72 (m, 2H), 2.59-2.55 (m, 1H), 2.46-2.31 (m, 6H), 1.83-1.73 (m,
2H), 1.57-1.53 (m, 4H), 1.43-1.38 (m, 8H), 1.32-1.21 (m, 10H), 0.98 (d, J = 6.5 Hz,
3H), 0.89 (t, J =* 6.8 Hz, 3H).
4-[4-(1-sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 9 (137 mg), 2-butanone (0.089 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celite, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (164 mg). 1H NMR (400
MHz, CDCI3): 7.19 (d, J = 8.6 Hz, 2H), 6.84 (d, J = 8.6 Hz, 2H), 4.28-4.22 (m, 1H),
3.39 (s, 2H), 2.89-2.72 (m, 2H), 2.50-2.31 (m, 6H), 1.98-1.95 (m, 2H), 1.83-1.72 (m,
2H), 1.59-1.53 (m, 6H), 1.43-1.37 (m, 2H), 1.34-1.23 (m, 1H), 0.98 (d, J a 6.5 Hz,
3H), 0.90 (t, J = 7.4 Hz, 3H).

1 -[4-( 1 -Cyclopentyl-piperidin-4-yloxy)-benzyf]-pfperidine
Phenylsilane (0.068 mL) was added dropwise to a solution of the product of
Example 9 (137 mg), cyclopentanone (0.088 mL) and scandium
trifluoromethanesulfonate (13 mg) in THF (1 mL). After 16 h, the resulting mixture
was filtered through a pad of celite, and the pad was washed with DCM (3x3 mL).
The combined filtrates were chromatographed (0.5-5.5% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (250 mg). 1H NMR (400
MHz, CDCI3): 7.19 (d, J = 8.5 Hz. 2H), 6.84 (d, J = 8.6 Hz. 2H). 4.34-4.23 (m. 1H).
3.39 (s. 2H), 2.87-2.73 (m, 2H), 2.55-2.47 (m, 1H), 2.40-2.26 (m. 6H), 2.05-1.95 (m,
2H), 1.91-1.79 (m, 4H), 1.73-1.49 (m, 8H). 1.46-1.37 (m, 4H).
4-(4-l midazol-1-yl-phenoxy)-1 -isobutyl-piperidine
A solution of the product of Example 6 (130 mg),.cydohexanone (0.06 mL),
and dibutyttin dichloride (3 mg) in THF (0.1 mL) was treated with phenylsMane
(0.07 mL). After 16 h, the resulting mixture was chromatographed (0-8% 2M
methanolic ammonia/DCM), giving the title compound as a waxy solid (18 mg). 1H
NMR (400 MHz, CDCI3): 7.75 (t, J = 1.2 Hz, 1H), 7.28 (d, J = 9.0 Hz. 2H). 7.20 (t, J =
1.4 Hz, 1H), 7.18 (t, J = 1.2 Hz, 1H), 6.99 (d. J = 8.8 Hz, 2H), 4.35-4.27 (m, 1H),
2.76-2.67 (m, 2H), 2.26-2.18 (m, 2H), 2.10 (d, J = 7.24 Hz. 2H). 2.05-1.96 (m. 2H).
1.87-1.73 (m, 3 H), 0.90 (d. J = 6.5 Hz. 6H).
1 -Cyclopentyl-4-(4-imidazol-1 -yl-phenoxy)-piperidine
A solution of the product of Example 6 (130 mg), isobutyraldehyde (0.06 mL).
and dibutyttin dichloride (3 mg) in THF (0.1 mL) was treated with phenylsilane
(0.07 mL). After 16 h, the resulting mixture was chromatographed (0-8% 2M
methanolic ammonia/DCM), giving the title compound as a waxy solid (57 mg). 'H
NMR (400 MHz, CDCI3): 7.75 (t, J = 1.2 Hz, 1H). 7.28 (d, J = 9.0 Hz, 2H), 7.20 (t, J =
1.2 Hz, 1H), 7.17 (t, J = 1.2 Hz. 1H), 6.99 (d, J = 9.0 Hz, 2H), 4.39-4.29 (m, 1H),
2.87-2.77 (br m, 1H), 2.57-2.48 (m, 1H), 2.40-2.30 (m. 2H), 2.08-1.99 (m, 2H). 1.94-
1.82 (m, 5H), 1.75-1.64 (m, 2H), 1.61-1.61 (m, 2H), 1.47-1.37 (m, 2H).

[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-pyridin-2-yl-amine
A solution of the product of Example 2 (522 mg), 2-aminopyridine (230 mg),
and acetic acid (0.013 mL) in DCM (7 mL) was treated with sodium
triacetoxyborohydride (720 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (8 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a yellow solid (417 mg). 1H NMR (400 MHz. CDCI3): 8.10 (m, 1H). 7.40
(m, 1H), 7.28-7.24 (m, 2H). 6.9-6.85 (m, 2H), 6.58 (m, 1H), 6.37 (m, 1H), 4.77 (m.
1H), 4.41 (d, J=5.8 Hz, 2H), 4.28 (m, 1H), 2.82-2.71 (m, 4H), 2.39 (m, 3H). 2.05-1.97
(m, 3H), 1.85-1.76 (m, 3H), 1.06 (d, J=6.6 Hz, 6H).

4-(1-Ethyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (226 mg), acetaldehyde (0.5 mL), and
acetic acid (0.09 mL) in DCM (3 mL) was treated with sodium triacetoxyborohydride
(360 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(5 mL), and the mixture was extracted with DCM (3x10 mL). The combined organic
phases were dried (sodium sulfate) and evaporated. Chromatography of the residue
(1-7% 2M methanolic ammonia/DCM) gave the title compound as a white solid
(67 mg). 1H NMR (400 MHz. CDCI3): 7.53 (d, J=8.8 Hz, 2H). 6.91 (d, J=8.8 Hz. 2H).
4.37 (m, 1H). 2.71 (m, 2H), 2.41 (q, J=7.1, 2H), 2.28 (m, 2H), 1.99 (m, 2H), 1.82 (m,
2H). 1.07(t,J=7.1.3H).
4-(1-Phenethyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (203 mg), phenylacetaldehyde
(0.14 mL), and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (320 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a white solid (145 mg). 1H NMR (400 MHz, CDCI3): 7.58 (d, J=8.8 Hz,
2H), 7.32-7.28 (m, 2H), 7.21 (m, 3H), 6.94 (d, J=8.8 Hz, 2H). 4.45 (br,1H), 2.84 (br,
4H), 2.66 (br. 2H). 2.42 (br, 1.5H), 2.04 (br, 1.5H), 1.91 (br, 2H), 1.55 (br, 1H).
4-(1-Cydobutyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (206 mg), cyclobutanone (0.1 mL), and
acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium triacetoxyborohydride
(320 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(5 mL), and the mixture was extracted with DCM (3x10 mL). The combined organic
phases were dried (sodium sulfate) and evaporated. Chromatography of the residue
(1-7% 2M methanolic ammonia/DCM) gave the title compound as a white solid
(66 mg). 1H NMR (400 MHz, CDCI3): 7.58 (d, J=8.8 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H).
4.41 (br, 1H), 2.81-2.64 (br, m, 3H), 2.23-1.46 (br, m, 12H).

4-( 1 -Methyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (213 mg), paraformaldehyde (0.52 g),
and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (340 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a white solid (91 mg). 1H NMR (400 MHz, CDCI3): 7.57 (d, J=9.1Hz,
2H), 6.93 (d, J=9.1 Hz, 2H), 4.40 (m, 1H), 2.69 (br, 2H), 2.37-2.29 (br, 2H), 2.32 (s,
3H), 2.03 (m. 2H), 1.87 (m, 2H).

4-(1-sec-Butyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (211 mg), 2-butanone (0.13 mL), and
acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium triacetoxyborohydride
(330 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(5 mL), and the mixture was extracted with DCM (3x10 mL). The combined organic
phases were dried (sodium sulfate) and evaporated. Chromatography of the residue
(1-7% 2M methanolic ammonia/DCM) gave the title compound as a white solid
(91 mg). 1H NMR (400 MHz, CDCI3): 7.56 (d, J=9.1 Hz, 2H), 6.93 (d, J=9.1 Hz, 2H),
4.36 (br, 1H), 2.77 (br, 2H). 2.50 (br, 2H), 2.37 (br, 1H), 2.02 (br, 2H), 1.81 (br, 2H),
1.58 (br, 1H), 1.30 (m, 1H). 1.0 (br, d, J=6.1 Hz, 3H), 0.91 (t, J=7.3 Hz, 3H).
4-(1-BenzyI-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (203 mg), benzaldehyde (0.13 mL),
and acetic add (0.07 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (330 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanoiic ammonia/DCM) gave the title
compound as a white solid (112 mg). 'H NMR (400 MHz, CDCI3): 7.56 (d, J=8.8 Hz.
2H), 7.33 (m, 3H), 7.27 (m, 2H), 6.93 (d, J=8.8 Hz, 2H), 4.40 (br, 1H), 3.54 (br, 2H),
2.73 (br, 2H), 2.32 (br, 2H), 1.99 (br, 2H), 1.84 (br, 2H), 1.56 (br, 1H).
4-[1-(2-Hydroxy-1-methyl-ethyl)-piperidin-4-yloxy]-benzonitrite
A solution of the product of Example 5 (205 mg), 1-hydroxy-2-propanone
(0.14 mL), and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (330 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the tide
compound as a white solid (161 mg). 1H NMR (400 MHz, CDCJ,): 7.57 (d, J=8.8 Hz,
2H), 6.93 (d, J=8.8 Hz, 2H), 4.42 (m, 1H), 3.42 (m, 1H), 3.32 (m, 1H), 2.88 (m, 2H),
2.64 (m, 2H), 2.33 (m, 1H), 2.03 (m, 2H), 1.84 (m, 2H), 0.92 (d, J=6.6 Hz, 3H).

4-(1 -Cydohexylmethyl-piperidin-4-yk>xy>-ben2x>nitrile
A solution of the product of Example 5 (221 mg), cyclohexanecarboxaldehyde
(0.2 mL), and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (340 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a white solid (205 mg). 1H NMR (400 MHz. CDCI,): 7.57 (d, J=9.1 Hz,
2H), 6.93 (d, J=9.1 Hz, 2H), 4.37 (br, 1H), 2.69 (br, 2H), 2.22 (br, 2H), 2.14 (br, 2H),
1.98 (br, 2H), 1.86-1.63 (m, 7H). 1.47 (br, 1H), 1.29-1.10 (m, 3H), 0.88 (m, 2H).

4-(1-Cyclohexyl-piperidin-4-ytoxy)-benzonltrile
A solution of the product of Example 5 (202 mg), cydohexanone (0.17 mL),
and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (340 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a white solid (241 mg). 1H NMR (400 MHz, CDCI3): 7.56 (d, J=8.8 Hz,
2H), 6.93 (d, J=8.8 Hz, 2H), 4.37 (m, 1H), 2.84 (m, 2H), 2.47 (m, 2H), 2.32 (br, 1H),
2.01 (br, 2H). 1.89-1.77 (m, 6H), 1.63 (m, 1H), 1.30-1.17 (m, 4H), 1.10 (m, 1H).
4-(1-Cyclopentyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (210 mg), cyclopentanone (0.14 mL),
and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (330 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a white solid (237 mg). 1H NMR (400 MHz, CDCL,): 7.57 (d, J=8.8 Hz,
2H), 6.94 (d, J=8.9 Hz, 2H), 4.40 (br, 1H), 2.91-2.23 (brr, m, 5H), 2.13-1.35 (m,
12H).
4-(1-Propyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (211 mg), propanaldehyde (0.15 ml_),
and acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (330 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a white solid (64 mg). 1H NMR (400 MHz, CDCI3): 7.57 (d, J=9.1 Hz,
2H), 6.93 (d, J=9.1 Hz, 2H), 4.42 (br, 1H), 2.76 (br, 2H), 2.35 (br, 3H), 2.05 (br, 2H),
1.87 (br, 2H), 1.56 (br, 2H), 0.92 (t J=7.3 Hz, 3H).

4-(1-lsobutyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (202 mg), isobutyraldehyde (0.21 mL),
and acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (360 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a white solid (211 mg). 1H NMR (400 MHz, CDCI3): 7.56 (d. J=9.1 Hz,
2H), 6.93 (d, J=9.1 Hz, 2H), 4.37 (br, 1H), 2.69 (br, 2H), 2.22 (br, 2H), 2.09 (br, 2H),
1.98 (br, 2H), 187-1.73 (m, 3H), 0.90 (d, br, J=7.3 Hz, 6H).

4-(1-Cydopropylmethy1-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 5 (215 mg),
cyclopropanecarboxaldehyde (0.16 mL). and acetic acid (0.07 mL) in DCM (3 mL)
was treated with sodium triacetoxyborohydride (340 mg). After 16 h, the resulting
mixture was treated with 10% sodium hydroxide (5 mL), and the mixture was
extracted with DCM (3x10 mL). The combined organic phases were dried (sodium
sulfate) and evaporated. Chromatography of the residue (1-7% 2M methanolic
ammonia/DCM) gave the title compound as a white solid (173 mg). 'H NMR (400
MHz. CDCI3): 7.56 (d, J=9.1 Hz, 2H), 6.93 (d, J=9.1 Hz, 2H), 4.40 (m, 1H), 2.83 (m,
2H), 2.40 (m, 2H), 2.29 (d, J=6.6 Hz, 2H), 2.03 (m, 2H), 1.87 (m, 2H), 0.88 (m, 1H),
0.52 (m,2H), 0.11 (m, 2H).
5-Chloro-2-[4-(1-isopropyl-piperidin-4-yloxy)-phenyl]-1H-benzoimidazole
A solution of the product of Example 2 (260 mg), 4-chloro-benzene-1,2-
diamine (156 mg), and sodium metabisutftte (280 mg) in DMA (2 mL) was heated to
100 °C for 12h. The resulting mixture was chromatographed (1-7% 2M methanolic
ammonia/DCM), giving the title compound as a pink solid (191 mg). 1H NMR (400
MHz, CDCI3): 7.99 (d, J=9.1 Hz, 2H), 7.54 (m, 1H), 7.51 (m, 1H), 7.21 (m, 1H), 7.09
(d, J=9.1 Hz, 2H), 4.51 (m, 1H), 2.84 (m, 2H), 2.77 (m, 1H), 2.51 (m, 2H), 2.07 (m,
2H), 1.82 (m, 2H), 1.10 (d, J=6.6 Hz. 6H).
1 -[4-(1 -lsopropyl-piperidin-4-yloxy)-benzyl]-4-methyl-[1,4]diazepane
A solution of the product of Example 2 (171 mg), N-methyl homopiperazine
(0.09 mL), and acetic acid (0.07 ml_) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (121 mg). 'H NMR (400 MHz, CDCI3): 7.22 (d, J=8.6 Hz,
2H), 6.84 (d, J=8.6 Hz, 2H), 4.26 (m, 1H). 3.55 (s, 2H), 2.82-2.72 (m. 3H), 2.71-2.63
(m, 6H), 2.61-2.57 (m, 2H), 2.41-2.36 (m, 2H). 2.35 (s, 3H), 2.00 (m, 2H). 1.85-1.76
(m, 4H), 1.06 (d. J=6.6 Hz. 6H).
Butyt-[4-(1-isopropyl-piperidin-4-yloxy)-benzyl]-methy1-amine
A solution of the product of Example 2 (167 mg), butyl-methyl-amine
(0.08 mL), and acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sutfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (157 mg). 1H NMR (400 MHz, CDCI3): 7.19 (d, J=8.6 Hz,
2H), 6.85 (d, J=8.6 Hz, 2H), 4.27 (m, 1H), 3.40 (s, 2H), 2.82-2.71 (m, 3H), 2.39 (m,
2H), 2.34 (m, 2H), 2.16 (s, 3H), 2.00 (m, 2H), 1.85-1.76 (m, 2H), 1.52-1.45 (m, 2H),
1.38-1.27 (m, 2H) 1.06 (d, J=6.6 Hz, 6H), 0.90 (t, J=7.3 Hz, 3H).
N-[4-(1 -lsopropyl-piperidin-4-yloxy)-ben2yl]-N,N',N'-trimethyl-ethane-1,2-
diamine
A solution of the product of Example 2 (171 mg), N,N,N'-trimethy1-ethane-1,2-
diamine (0.09 mL), and acetic acid (0.07 mL) in DCM (3 mL) was treated with
sodium triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated
with 10% sodium hydroxide (5 mL), and the mixture was extracted with DCM
(3x10 mL). The combined organic phases were dried (sodium sulfate) and
evaporated. Chromatography of the residue (1-7% 2M methanolic ammonia/DCM)
gave the title compound as a clear oil (126 mg). 1H NMR (400 MHz, CDCI3): 7.19 (d,
J=8.6 Hz, 2H), 6.84 (d, J=8.6 Hz. 2H), 4.26 (m, 1H), 3.44 (s, 2H), 2.82-2.71 (m, 3H),
2.48-2.34 (m, 6H), 2.22 (s. 3H), 2.20 (s. 6H), 2.04-1.96 (m, 2H), 1.85-1.75 (m, 2H),
1.06 (d, J=6.6Hz, 6H).
Cyclohexyl-[4-(1-isopropyl-pjperidin-4-yloxy)-benzyI]-methyl-amine
A solution of the product of Example 2 (169 mg), cyclohexyl-methyl-amine
(0.09 mL), and acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium suifate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (165 mg). *H NMR (400 MHz, CDCI3): 7.19 (d, J=8.6 Hz.
2H), 6.84 (d, J=8.6 Hz, 2H), 4.26 (m, 1H), 3.48 (s, 2H), 2.82-2.70 (m. 3H), 2.46-2.33
(m, 3H), 2.17 (s, 3H), 2.04-1.96 (m, 2H), 1.89-1.76 (m, 6H), 1.62 (m, 1H), 1.34-1.15
(m, 4H) 1.05 (d, J=6.6 Hz, 6H).
1-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-azepane
A solution of the product of Example 2 (167 mg), hexamethyleneimine
(0.08 mL), and acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (163 mg). 'H NMR (400 MHz. CDCI3): 7.22 (d, J=8.6 Hz,
2H), 6.84 (d, J=8.6 Hz, 2H), 4.26 (m, 1H), 3.56 (s, 2H), 2.85-2.71 (m, 3H), 2.60 (m,
4H), 2.38 (m, 2H), 2.04-1.96 (m, 2H), 1.85-1.76 (m, 2H), 1.60 (m, 9H), 1.06 (d, J=6.6
Hz, 6H).
Diethyl-[4-(1 -isopropyl-piperidin-4-yloxy)-benzyl]-amine
A solution of the product of Example 2 (170 mg), diethylamine (0.08 mL), and
acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium triacetoxyborohydride
(220 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(5 mL), and the mixture was extracted with DCM (3x10 mL). The combined organic
phases were dried (sodium suifate) and evaporated. Chromatography of the residue
(1-7% 2M methanolic ammonia/DCM) gave the title compound as a clear oil
(78 mg). 1H NMR (400 MHz, CDCU,): 7.21 (d, J=8.6 Hz, 2H), 6.84 (d, J=8.6 Hz, 2H),
4.26 (m, 1H), 3.49 (s, 2H), 2.82-2.70 (m, 4H), 2.50 (q, J=7.1 Hz, 4H), 2.37 (m, 2H),
2.04-1.96 (m, 2H), 1.85-1.76 (m, 2H), 1.60 (m, 9H), 1.07-1.00 (m, 12H).
1-lsopropyl-4-(4-pyrrolidin-1-ylmethy(-phenoxy)-pericline
A solution of the product of Example 2 (169 mg), pyrrolidine (0.06 mL), and
acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium triacetoxyborohydride
(220 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(5 mL), and the mixture was extracted with DCM (3x10 mL). The combined organic
phases were dried (sodium sulfate) and evaporated. Chromatography of the residue
(1-7% 2M methanolic ammonia/DCM) gave the title compound as a clear oil
(144 mg). 1H NMR (400 MHz, CDCI3): 7.21 (d, J=8.8 Hz, 2H), 6.84 (d, J=8.8 Hz, 2H),
4.26 (m, 1H). 3.53 (s, 2H), 2.82-2.71 (m, 3H), 2.48 (m, 4H), 2.37 (m, 2H), 2.03-1.96
(m, 2H), 1.85-1.74 (m. 6H), 1.05 (d, J=6.6 Hz, 6H).
N-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-O,N-dimethyl-hydrDxylamine
A solution of the product of Example 2 (170 mg) and O,N-dimethyl-
hydroxylamine hydrochloride (0.15 g) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (179 mg). 'H NMR (400 MHz, CDCI3): 7.24 (d, J=8.8 Hz,
2H), 6.85 (d, J=8.8 Hz, 2H), 4.27 (m, 1H), 3.70 (s, 2H), 3.37 (s, 3H), 2.81-2.70 (m,
3H), 2.58 (s, 3H), 2.48 (m, 4H), 2.37 (m, 2H), 2.03-1.96 (m, 2H), 1.85-1.76 (m, 2H),
1.05(d,J=6.6Hz,6H).
[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-dimethyl-amine
A solution of the product of Example 2 (170 mg) and dimethylamine
hydrochloride (0.12 g) in DCM (3 mL) was treated with sodium triacetoxyborohydride
(220 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(5 mlL), and the mixture was extracted with DCM (3x10 mL). The combined organic
phases were dried (sodium sulfate) and evaporated. Chromatography of the residue
(1-7% 2M methanolic ammonia/DCM) gave the title compound as a clear oil
(163 mg). 1H NMR (400 MHz, CDCI3): 7.18 (d, J=8.6 Hz, 2H), 6.85 (d, J=8.6 Hz, 2H),
4.27 (m, 1H), 3.34 (s, 2H), 3.37 (s, 3H). 2.81-2.70 (m, 3H). 2.38 (m, 2H), 2.21 (s,
6H), 2.05-1.96 (m, 2H), 1.85-1.75 (m, 2H), 1.05 (d, J=6.6 Hz, 6H).
1-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-4-methyl-piperazine
A solution of the product of Example 2 (173 mg), N-methylpiperazine
(0.08 mL), and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (217 mg). 1H NMR (400 MHz, CDCI3): 7.19 (d, J=8.6 Hz.
2H), 6.83 (d, J=8.6 Hz, 2H), 4.26 (m, 1H), 3.42 (s, 2H), 2.81-2.70 (m, 3H), 2.55-2.33
(m, 811), 2.26 (s, 311), 2.03-1.97 (m, 2H), 1.84-1.74 (m, 211), 1.04 (d, J=6.6 Hz, 6H).
4-[4-(4-Benzylidene-piperidin-1-yl methyl )-phenoxy]-1-isopropyl-piperidine
A solution of the product of Example 2 (136 mg), 4-benzylidene-piperidine
(94 mg), and acetic acid (0.05 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (190 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium suffate) and evaporated.
Chromatography of the residue (1-7% 2M methanoiic ammonia/DCM) gave the title
compound as a clear oil (54 mg). 1H NMR (400 MHz, CDCI3): 7.32-7.27 (m, 2H),
7.23-7.16 (m, 5H), 6.86 (d, J=8.6 Hz, 2H), 6.27 (s, 1H), 4.28 (m, 1H), 3.46 (s, 2H),
2,83-2.71 (m, 3H), 2.54-2.49 (m, 4H), 2.43-2.35 (m, 6H), 2.05-1.97 (m, 2H), 1.86-
1.76 (m, 2H), 1.06 (d, J=6.6 Hz, 6H).
1 -Benzyl-4-[4-(1-isopropyl-piperidin-4-yloxy)-benzyl]-piperazine
A solution of the product of Example 2 (188 mg), N-benzylpiperazine
(0.13 mL), and acetic acid (0.06 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (260 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (239 mg). 'H NMR (400 MHz, CDO,): 7.31-7.29 (m, 4H),
7.25-7.23 (m, 1H), 7.18 (d, J=8.6 Hz, 2H) 6.84 (d, J=8.6 Hz. 2H), 4.26 (m, 1H), 3.50
(S. 2H), 3.43 (s, 2H), 2.82-2.70 (m, 3H). 2.54-2.34 (m, 9H). 2.03-1.96 (m, 2H), 1.84-
1.75 (m, 2H), 1.06 (d, J=6.6 Hz. 6H).
1 -[4-(1-(sopropyl-piperidin-4-yloxy)-benzyl]-4-phenyl-piperazine
A solution of the product of Example 2 (140 mg), N-phenvipiperazine
(0.09 mL), and acetic acid (0.05 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (190 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sutfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (78 mg). 1H NMR (400 MHz. CDCI,): 7.30-7.22 (m, 4H),
6.65-6.82 (m, 5H), 4.29 (m, 1H), 3.50 (s, 2H), 3.19 (m, 2H), 3.16-3.12 (m, 2H), 2.83-
2.71 (m, 3H), 2.59 (m, 2H), 2.39 (m, 2H). 2.06-1.99 (m. 2H). 1.87-1.78 (m, 2H), 1.07
(d. J=6.6 Hz, 6H).
1 -[4-(1-isoprDpyl-piperidin-4-yloxy)-benzyl]-4-benzyl-piperidine
A solution of the product of Example 2 (188 mg), N-benzylpiperidine
(0.13 mL), and acetic acid (0.05 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (250 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium suffate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (160 mg). 1H NMR (400 MHz, CDCI3): 7.29-7.23 (m, 3H),
7.19-7.10 (m, 4H), 6.83 (m, 2H), 4.26 (m, 1H), 3.39 (s, 2H), 2.87-2.70 (m, 5H), 2.52
(d, J=7.1 Hz, 2H). 2.38 (m, 2H), 2.03-1.96 (m, 2H), 1.59 (m, 2H), 1.50 (m, 1H), 1.29
(m, 2H), 1.06 (d, J=6.6 Hz, 6H).
Cydopropyl-[4-(1-isopropyl-plpericlin-4-y1oxy)-benzyl]-amine
A solution of the product of Example 2 (250 mg), cydopropyiamine (0.10 mL),
and acetic acid (0.07 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (340 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a clear oil (88 mg). 1H NMR (400 MHz. CDCI,): 7.19 (d, J=8.6 Hz, 2H),
6.84 (d, J=8.6 Hz, 2H), 4.25 (m, 1H), 3.74 (s, 2H)f 2.81-2.69 (m, 3H), 2.37 (m, 2H),
2.12 (m. 1H). 2.03-1.95 (m, 2H), 1.83-1.74 (m. 2H). 1.04 (d, J=6.6 Hz, 6H), 0.44-
0.33 (m, 4H).
Example 65
K,= 1.5 nM
[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-methyl-(1-methyl-piperidin-4-yl)-
amine
A solution of the product of Example 2 (146 mg), 1-methyl-4-
(methylamino)pfperidine (0.09 mL), and acetic acid (0.09 mL) in DCM (3 mL) was
treated with sodium triacetoxyborohydride (200 mg). After 16 h, the resulting mixture
was treated with 10% sodium hydroxide (5 mL), and the mixture was extracted with
DCM (3x10 mL). The combined organic phases were dried (sodium sulfate) and
evaporated. Chromatography of the residue (1-7% 2M methanolic ammonia/DCM)
gave the title compound as a clear oil (137 mg). *H NMR (400 MHz, CDCI,): 7.16 (d,
J=8.6 Hz, 2H), 6.82 (d, J=8.6 Hz, 2H), 4.24 (m, 1H). 3.47 (s, 2H), 2.88 (m, 2H),
2.79-2.68 (m. 3H), 2.43-2.32 (m, 3H), 2.23 (s, 3H), 2.15 (s. 3H), 2.01-1.94 (m, 2H),
1.89 (m, 2H), 1.82-1.73 (m. 4H), 1.69-1.59 (m, 2H) 1.03 (d, J=6.6 Hz, 6H).
{1 -[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-yl]-methanol
A solution of the product of Example 2 (158 mg), 4-piperidinemethanol
(78 mg). and acetic add (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium sulfate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a dear oil (166 mg). 'H NMR (400 MHz. CDCI3): 7.17 (d. J=8.6 Hz.
2H). 6.82 (d. J=8.6 Hz. 2H). 4.25 (m. 1H). 3.24 (d, J=6.6 Hz. 2H). 3.39 (s, 2H). 2.87
(m, 2H). 2.80-2.69 (m, 3H), 2.37 (m. 2H), 2.02-1.94 (m. 2H), 1.90 (m, 2H). 1.83-1.74
(m. 2H), 1.69 (m, 2H) 1.45 (m. 1H). 1.23 (m. 2H). 1.04 (d. J=6.6 Hz, 6H).
1-[4-(1-lsopropyl-piperidin-4-yloxy)4-benzyl]-piperidin-4-ol
A solution of the product of Example 2 (167 mg), 4-hydroxypiperidine (73 mg),
and acetic acid (0.08 mL) in DCM (3 mL) was treated with sodium
triacetoxyborohydride (220 mg). After 16 h, the resulting mixture was treated with
10% sodium hydroxide (5 mL), and the mixture was extracted with DCM (3x10 mL).
The combined organic phases were dried (sodium suffate) and evaporated.
Chromatography of the residue (1-7% 2M methanolic ammonia/DCM) gave the title
compound as a dear oil (168 mg). 1H NMR (400 MHz, CDCI3): 7.17 (d, J=8.6 Hz,
2H), 6.82 (d, J=8.6 Hz, 2H), 4.25 (m. 1H), 3.64 (m. 1H). 3.42 (s, 1H). 3.40 (s. 2H),
2.80-2.69 (m, 5H), 2.36 (m, 2H), 2.08 (m, 2H). 2.02-1.94 (m, 2H). 1.87-1.74 (m, 2H),
1.55 (m, 2H), 1.04 (d. J=6.6 Hz, 6H).
4-[4-(1-lsopropyl-piperidin-4-yloxy)-benzyl]-morpholine
A solution of the product of Example 2 (360 mg), morphofine (0.13 mL), and
acetic add (0.09 mL) in DCM (5 mL) was treated with sodium triacetoxyborohydride
(450 mg). After 16 h, the resulting mixture was treated with 10% sodium hydroxide
(10 mL), and the mixture was extracted with DCM (3x20 mL). The combined organic
phases were dried (sodium sulfate) and evaporated. Chromatography of the residue
(1-7% 2M methanolic ammonia/DCM) gave the title compound as a dear oil (366
mg). 1H NMR (400 MHz, CDCI3): 7.20 (d. J=8.6 Hz, 2H), 6.82 (d, J=8.6 Hz, 2H), 4.27
(m, 1H), 3.69 (m, 4H), 3.42 (s, 2H), 2.82-2.70 (m, 3H), 2.41 (m, 4H), 2.36 (m, 2H),
2.04-1.96 (m, 2H). 1.84-1.75 (m, 2H). 1.05 (d, J=6.6 Hz, 6H).
1 -[4-(1-Isopropyl-piperidin-4-ylmethoxy)-bonzyl]-piperidine
A solution of 4-hydroxymethyipiperidine (23.1 g), acetone (40 mL), acetic acid
(12 mL) and sodium triacetoxyborohydride (62 g) in DCM (3 mL) was stirred under
nitrogen overnight at room temperature. The reaction mixture was basified with 10 %
aqueous sodium hydroxide to a pH of 12-13. The resulting mixture was extracted
with dichloromethane (3x150 mL). The combined extracts were dried (sodium
sulfate), filtered and evaporated, yielding an oil (31.47 g). A portion of this oH (595
mg) was added to a suspension of sodium hydride (151 mg) in dimethyl fbrmamide
(4 mL) under nitrogen. After 30 min, a solution of 4-cyanochiorobenzene (521 mg) in
dimethyl fbrmamide (2 mL) was added. The reaction mixture was heated to 65-68 °C
for 18 hours, cooled to RT, and poured into water (200 mL). The resulting mixture
was extracted with ethyl acetate (3x30 mL). The combined extracts were washed
with water, brine and water and then dried (sodium sulfate). The residue was
chromatographed (5% 2 M methanolic ammonia/DCM). This product (230 mg) was
then dissolved in toluene, cooled to 0 °C, and treated with di-isobutyl aluminum
hydride (2.2 mL of a 1 M solution in hexane). The reaction mixture was warmed to
RT, stirred for 16 hours, and quenched with ethyl acetate (1 mL). Sodium hydroxide
(20 mL of a 1 M aqueous solution) was added, and the resulting mixture was stirred
for five minutes and then extracted with DCM (2x30 mL). The combined organic
phases were evaporated, and the residue was chromatographed (5% 2 M
methanolic ammonia/DCM). A suspension of this product (100 mg). piperidine (38
µL), acetic acid (22 µL) and sodium triacetoxyborohydride (122 mg) in DCE (3 mL)
was stirred overnight. The reaction mixture was quenched with 1 N aqueous sodium
hydroxide (1 mL) and extracted with DCM (3x20 mL). The combined extracts were
evaporated to dryness. Chromatography of the residue (5% 2 M methanolic
ammonia/DCM) yielded the title compound (13 mg). 1H NMR (400 MHz, CDCI3): 7.20
(d, J = 8.59 Hz, 2 H), 6.83 (d. J = 8.6 Hz, 2 H), 3.78 (d. J = 6.3 Hz, 2 H). 2.95 (d. J =
12 Hz, 2 H), 2.75 (m, 1 H), 2.37 (brs, 4 H), 2.19 (m, 3 H), 1.83 (m, 4 H), 1.57 (m, 4
H), 1.42 (m, 4 H). 1.08 (d, J = 6.6 Hz, 6 H).
4-(1-lsopropyl-piperidin-4-yloxy)-benzonitrile
A solution of the product of Example 3 (10.74 g) and 4-Chloro-benzonitriIe
(11.45 g) in DMF (100 mL) was treated with NaH (60%, 3.7g). The resulting dark
mixture was then heated to 65 °C for 16 h, and allowed to cool to RT. The mixture
was poured into water (1.5 L) and extracted with ether (3x400 mL). The combined
organic phases were dried (sodium sulfate) and evaporated which gave the title
compound as a light brown solid (16.6 g). 1H NMR (400 MHz. CDCI,): 7.56 (d, J=8.6
Hz, 2H), 6.93 (d, J=8.6 Hz, 2H),4.37 (m, 1H), 2.81-2.72 (m, 3H), 2.41 (m, 4H). 2.06-
1.98 (m, 2H), 1.87-1.77 (m, 2H), 1.06 (d. J=6.6 Hz, 6H).
1 -lsopropyl-4-[4-(1-isopropyl-pipeiidin-4-yloxy)-phenyi]-piperazine
A solution of the product of Example 74 (0.061 mg), acetone (2 mL), and
acetic acid (0.012 mL) in DCM (10 mL) was treated with sodium
triacetoxyborohydride (59 mg). After 16 h, the resulting mixture was treated with
10% aqueous sodium hydroxide (5 mL). The aqueous phase was extracted with
DCM (2x50 mL). The combined organic phases were washed with water (10 mL),
brine (10 mL), dried (magnesium sulfate), and evaporated. Chromatography of the
residue (4-10% 2M methanolic ammonia) gave the title compound as a colorless
solid (40 mg). 1H NMR (400 MHz, CDCI3): 6.90-6.83 (m, 4H). 4.19-4.15 (m, 1H), 3.11
(t, J = 5.0 Hz, 4H), 2.81-2.67 (m, 8H), 2.38-2.33 (m. 2H), 2.02-1.95 (m, 2H), 1.82-
1.74 (m. 2H), 1.09 (d, J = 6.5 Hz, 6H), 1.05 (d, J = 6.6 Hz, 6H).
1 -[4-(1 -lsopropyl-piperidin-4-yloxy)-benzyl]-piperidine
A suspension of the product of Example 3 (129 mg), the product of Example
1 (172 mg), and polymer-supported triphenylphosphine (600 mg) in DCM (5 mL) was
treated with di-tert-butyl azodicarboxylate (311 mg). The resulting mixture was
shaken for 16 h and filtered through a pad of celite. The pad was washed with DCM
(3x1 mL) and the combined filtrates were chromatographed (1-6% 2 M methanolic
ammonia/DCM), giving the title compound as a colorless oil (75 mg). 1H NMR (400
MHz, CDCI3): 7.19 (d. J = 8.5 Hz, 2H), 6.84 (d. J = 8.6 Hz, 2H), 4.29-4.25 (m, 1H),
3.40 (s, 2H), 2.82-2.71 (m, 3H), 2.41-2.35 (m, 6H). 2.40-2.26 (m, 6H). 2.03-1.99 (m,
2H), 1.85-1.77 (m, 2H), 1.59-1.53 (m, 4H), 1.43-1.38 (m, 2H), 1.06 (d, J = 6.6 Hz,
6H),
1-lsopropyl-4-(4-pyrrol-1-yl-phenoxy)-piperidine
A suspension of the product of Example 3 (129 mg), 4-(1 H-pyrrol)-1-yl)phenol
(143 mg), and polymer-supported triphenylphosphine (600 mg) in DCM (5 mL) was
treated with di-tert-butyl azodicarboxylate (311 mg). The resulting mixture was
shaken for 16 h and filtered through a pad of celite. The pad was washed with DCM
(3x1 mL) and the combined filtrates were chromatographed (1-6% 2 M methanolic
ammonia/DCM), giving the title compound as a light yellow oil (92 mg). 1H NMR (400
MHz, CDCI,): 7.27 (d, J = 8.9 Hz, 2H), 6.99-6.98 (m, 2H), 6.93 (d, J = 8.9 Hz, 2H),
6.31-6.30 (m, 2H), 4.30-4.26 (m, 1H), 2.82-2.71 (m, 3H), 2.42-2.36 (m, 2H), 2.04-
2.00 (m, 2H), 1.86-1.78 (m, 2H), 1.06 (d, J = 6.6 Hz, 6H).
1-[4-(1-lsopropyl-piperidin-4-yloxy)-phenyl]piperazine
A solution of 1-(4-hydroxyphenyl)-plperazine (12.0 g) in THF (50 mL) was
treated with dkert butyl dicarbonate (72 mL of a 1 M solution in THF). After 10 min,
saturated aqueous sodium bicarbonate was added. After 16 h, the resulting mixture
was extracted with ethyl acetate (1.5 L). The organic phase was washed with water
(2x200 mL), brine (200 mL), and then dried (magnesium sulfate) and evaporated.
The resulting brown oil was triturated with hexane, giving a brown solid (16.3 g). A
suspension of this brown solid (501 mg), the product of Example 66 (258 mg). and
polymer-supported triphenytphosphine (1.2 g) in DCM (12 mL) was treated with di-
tert-butyl azodicarboxytate (622 mg). The resulting mixture was shaken for 16 h and
filtered through a pad of celite. The pad was washed with DCM (3x2 mL) and the
combined filtrates were chromatographed (1-6% 2 M methanolic ammonia/DCM)
giving the title compound as a brown oil (275 mg). A solution of this brown oil (259
mg) in dioxane (5 mL) was treated with hydrogen chloride (5 mL of a 4 N solution in
dioxane). After 16 h solvent was evaporated and the residue was treated with 10%
aqueous sodium hydroxide (20 mL) and extracted with DCM (2x100 mL). The
combined organic phases were washed with water (50 mL), brine (50 mL), and dried
(magnesium sulfate) and evaporated. Chromatography of the residue (6-20% 2 M
methanolic ammonia/DCM) gave the title compound as a colorless oil (61 mg). 1H
NMR (400 MHz, CDCI3): 6.89-6.83 (m, 4H), 4.21-4.15 (m, 1H), 3.03 (s, 8H), 2.82-
2.72 (m, 3H), 2.41-2.35 (m, 2H), 2.03-1.97 (m, 2H), 1.83-1.75 (m, 2H), 1.06 (d, J =
6.6 Hz, 6H).

[4-(1-lsopropyl-piperidin-4-yloxy)-phenyl]-phenyl-methanol
A solution of the product of Example 75 (65 mg) in ethanol (10 mL) was
treated with sodium borohydride (1 g). After 16 h, the resulting mixture was treated
with saturated aqueous sodium bicarbonate (20 mL), and extracted with DCM
(2x50 mL). The combined organic phases were dried (magnesium sulfate) and
evaporated. Chromatography of the residue (1-10% 2 M methanolic ammonia/DCM)
gave the title compound as a colorless oil (52 mg). 1H NMR (400 MHz, CDCI3): 7.38-
7.21 (m, 7H), 6.86-6.82 (m, 2H), 5.76 (s, 1H), 4.27-4.23 (m, 1H), 2.77-2.68 (m, 3H),
2.39-2.34 (m, 2H). 2.01-1.96 (m, 2H), 1.81-1.72 (m, 2H), 1.04 (d. J = 6.6 Hz. 6H).
[4-(1-lsopropyl-piperidin-4-yloxy)-phenyl]-pheny(-methanone
A suspension of the product of Example 66 (258 mg), 4-
hydroxybenzophenone (357 mg), and polymer-supported tripnenyiphosphine (1.2 g)
in DCM (6 mL) was treated with di-tert-butyl azodicarboxylate (622 mg). The
resulting mixture was shaken for 16 h and filtered through a pad of celite. The pad
was washed with OCM (3x1 mL) and the combined filtrates were chromatographed
(1-6% 2 M methanolic ammonia/DCM) giving the title compound as a colorless oil
(151 mg). 1H NMR (400 MHz, CDCI3): 7.81 (d, J = 8.9 Hz, 2H), 7.76-7.74 (m, 2H),
7.59-7.54 (m, 1H), 6.96 (d, J = 8.8 Hz, 2H). 4.46-4.40 (m, 1H). 2.83-2.73 (m, 3H).
2.46-2.40 (m, 2H). 2.08-2.03 (m, 2H). 1.90-1.82 (m, 2H), 1.07 (d. J = 6.5 Hz, 6H).
N-lsopropyl-4-{4-[5-(1-isopropyl-piperidin-4-ylsulfanyl)-tetrazol-1-yI}-phenoxy}-
piperidine
A suspension of the product of Example 66 (258 mg), 1-(4-hydroxyphenyl)-
1H-tetrazole-5-thiol (175 mg), and polymer-supported triphenylphosphine (1.2 g) in
DCM (6 mL) was treated with di-tert-butyl azodicarboxylate (622 mg). The resulting
mixture was shaken for 16 h and filtered through a pad of celite. The pad was
washed with DCM (3x1 mL) and the combined filtrates were chromatographed (1-
6% 2 M methanolic ammonia/DCM) giving the title compound as a colorless oil
(54 mg). 1H NMR (400 MHz, CDCI3): 7.44-7.41 (m, 2H), 7.05-7.02 (m, 2H), 4.39-4.36
(m, 1H), 3.97-3.92(m, 1H), 2.86-2.70 (m, 6H), 2.46-2.40 (m, 4H), 2.38-2.24 (m, 2H),
2.07-2.03 (m, 2H), 1.89-1.74 (m, 4H), 1.07 (d, J = 6.5 Hz, 6H), 1.04 (d, J = 6.6 Hz,
6H).
4-{4-lmidazol-1-yl-phenoxy)-1-Isopropyl-piperidine
A suspension of the product of Example 66 (258 mg), 4-(imidazol-yl)phenol
(144 mg), and polymer-supported triphenylphosphine (1.2 g) in DCM (6 mL) was
treated with di-tert-butyl azodicarboxytate (622 mg). The resulting mixture was
shaken for 16 h and filtered through a pad of cetite. The pad was washed with DCM
(3x1 mL) and the combined filtrates were chromatographed (1-6% 2 M methanolic
ammonia/DCM) giving the title compound as a colorless oil (67 mg). 1H NMR (400
MHz, CDCI,): 7.76 (s, 1H), 7.30-7.26 (m, 2H), 7.21-7.18 (m, 2H), 7.01-6.97 (m, 2H),
4.36-4.30 (m. 1H), 2.86-2.75 (m. 3H), 2.44-2.39 (m, 2H), 2.07-2.01 (m, 2H), 1.88-
1.80 (m, 2H), 1.07 (d, J = 6.6 Hz, 6H).
N-[4-(1-lsopropyl-piperidin-4-yloxy)-phenyl]-acetamide
A suspension of the product of Example 66 (258 mg), acetaminophen
(136 mg), and polymer-supported triphenylphosphine (1.2 g) in DCM (6 mL) was
treated with di-tert-butyl azodicarisoxytate (622 mg). The resulting mixture was
shaken for 16 h and filtered through a pad of celite. The pad was washed with DCM
(3x1 mL) and the combined filtrates were chromatographed (1-6% 2 M methanolic
ammonia/DCM) giving the title compound as a colorless oil (82 mg). 1H NMR (400
MHz, CDCl3): 8.14 (s, 1H). 7.39-7.35 (m, 2H), 6.84-6.80 (m, 2H), 4.24-4.18 (m, 1H),
2.80-2.72 (m, 3H), 2.39-2.33 (m, 2H), 2.10 (s, 3H), 2.00-1.95 (m. 2H). 1.81-1.73 (m,
2H),1.05(d,J = 6.6Hz,6H).
4-(4-Cyclopentyl-phenoxy)-1-isopropyl-piperidine
A suspension of the product of Example 66 (258 mg), 4-cyclopentylphenol
(146 mg), and polymer-supported triphenylphosphine (1.2 g) in DCM (6 mL) was
treated with di-tert-butyl azodicarboxylate (622 mg). The resulting mixture was
shaken for 16 h and filtered through a pad of celite. The pad was washed with DCM
(3x1 mL) and the combined filtrates were chromatographed (1-6% 2 M methanolic
ammonia/DCM) giving the title compound as a colorless oil (19 mg). 1H NMR (400
MHz, CDCL3): 7.15-7.12 (m, 2H), 6.85-6.81 (m, 2H), 4.28-4.22 (m, 1H), 2.97-2.88 (m,
1H), 2.42-2.36 (m, 2H), 2.07-1.99 (m, 4H), 1.85-1.49 (m, 8H), 1.06 (d, J = 6.6 Hz,
6H).
[4-(1-lsopropyl-piperldin-4-yloxy)-phenyl]-phenyl-amine
A suspension of the product of Example 66 (258 mg), 4-
hydroxydiphenylamine (136 mg), and polymer-supported triphenyiphosphine (1.2 g)
in DCM (6 mL) was treated with di-tert-butyl azodicarboxyiate (622 mg). The
resulting mixture was shaken for 16 h and filtered through a pad of celite. The pad
was washed with DCM (3x1 mL) and the combined filtrates were chromatographed
(1-6% 2 M methanolic ammonia/DCM) giving the title compound as a brown oil
(25 mg). 1H NMR (400 MHz, CDCI3): 7.23-7.19 (m, 2H), 7.06-7.03 (m, 2H), 6.93-6.91
(m, 2H), 6.88-6.81 (m, 3H), 4.26-4.20 (m, 1H), 2.84-2.74 (m, 3H), 2.44-2.38 (m, 2H),
2.06-2.00 (m, 2H), 1.07 (d, J = 6.6 Hz, 6H).
K, = 6 nM
4-(4-Benzyloxy-phenoxy)-1-isopropyl-piperidine
A suspension of the product of Example 66 (258 mg), 4-(benzyloxy)phenol
(180 mg), and polymer-supported triphenylphosphine (1.2 g) in DCM (6 mL) was
treated with di-tert-butyl azodicarboxylate (622 mg). The resulting mixture was
shaken for 16 h and filtered through a pad of celite. The pad was washed with DCM
(3x1 mL) and the combined filtrates were chromatographed (1-6% 2 M methanolic
ammonia/DCM) giving the title compound as a light pink solid (102 mg). 1H NMR
(400 MHz, CDCI3): 7.43-7.28 (m, 5H), 6.91-6.82 (m, 4H). 5.00 (s, 2H), 4.18-4.13 (m,
1H), 2.81-2.69 (m, 3H), 2.38-2.32 (m, 2H), 2.01-1.95 (m, 2H). 1.82-1.74 (m, 2H),
1.05(d,J = 6.6Hz, 6H).

1-lsopropyl-4-(4-phenoxy-phenoxy)-piperidine
A suspension of the product of Example 66 (258 mg), 4-phenoxyphenol
(168 mg), and polymer-supported triphenylphosphine (1.2 g) in DCM (6 mL) was
treated with di-tert-butyl azodicarboxylate (622 mg). The resulting mixture was
shaken for 16 h and filtered through a pad of celite. The pad was washed with DCM
(3x1 mL) and the combined filtrates were chromatographed (1-6% 2 M methanolic
ammonia/DCM) giving the title compound as a colorless oil (77 mg). 1H NMR (400
MHz, CDCL3): 7.31-7.26 (m, 2H), 7.06-7.01 (m. 1H), 6.97-6.92 (m, 4H), 6.90-6.86 (m,
2H), 4.25-4.20 (m, 1H). 2.83-2.72 (m. 3H). 2.41-2.35 (m. 2H). 2.05-1.99 (m, 2H).
1.86-1.77 (m. 2H). 1.06 (d, J =6.6 Hz. 6H).

4-(4-Benzyl-phenoxy)-1-leopropyl-piperidine
A suspension of the product of Example 66 (258 mg), 4-
hydroxydiphenyimethane (166 mg), and polymer-supported triphenylphosphine (1.2
g) In OCM (6 mL) was treated with di-tert-butyl azodicarboxylate (622 mg). The
resulting mixture was shaken for 16 h and filtered through a pad of celite. The pad
was washed with DCM (3x1 mL) and the combined filtrates were chromatographed
(1-6% 2 M methanolic ammonia/DCM) giving the title compound as a light yellow ofl
(108 mg). 1H NMR (400 MHz, CDCI,): 7.30-7.26 (m, 2H), 7.20-7.16 (m, 3H). 7.09-
7.06 (m, 2H), 6.84-6.81 (m, 2H), 4.27-4.21 (m, 1H), 3.91 (s, 2H), 2.81-2,67 (m, 3H),
2.40-2.34 (m, 2H), 2.02-1.92 (m, 2H), 1.83-1.75 (m. 2H), 1.05 (d, J = 6.6 Hz, 6H).
4-(Biphenyl-4-yloxy)-1-isopropyl-piperidine
A suspension of the product of Example 66 (258 mg), acetone (0.039 mL),
and polymer-supported triphenylphosphine (1.2 g) in OCM (6 mL) was treated with
di-tert-butyl azodicarboxylate (622 mg). The resulting mixture was shaken for 16 h
and filtered through a pad of ceirte. The pad was washed with DCM (3x1 mL) and
the combined filtrates were chromatographed (1-6% 2 M methanolic ammonia/DCM)
giving the title compound as a white solid (91 mg). 1H NMR (400 MHz. CDCI3): 7.55-
7.49 (m, 4H), 7.42-7.38 (m, 2H), 7.30-7.26 (m, 1H), 6.98-6.95 (m, 2H), 4.36-4.31 (m,
1H), 2.83-2.73 (m, 3H), 2.46-2.41 (m, 2H), 2.09-2.05 (m, 2H), 1.90-1.82 (m, 2H),
1.08 (d. J = 6.6 Hz,6H).
4-(Biphenyl-4-yloxy)-piperidine
A suspension of tert-butyl 4-hydroxy-1-piperidine carboxyiate (497 mg), 4-
phenylphenol (300 mg), and polymer-supported triphenyiphosphine (1.2 g) in DCM
(10 mL) was treated with di-tert-butyl azodicarboxylate (608 mg). The resulting
mixture was shaken for 16 h, and filtered through a pad of celite. The pad was
washed with DCM (3x5 mL) and the combined filtrates were evaporated. The
residue was dissolved in dioxane (5 mL) and treated with hydrogen chloride (5 mL of
a 4 N solution in dioxane). After 16 h, solvent was removed and the residue was
treated with 10% aqueous sodium hydroxide (20 mL). The resulting mixture was
extracted with DCM (2x500 mL), and the combined organic phases were dried
(magnesium sulfate) and evaporated. Chromatography of the residue (1-6% 2M
methanolic ammonia/DCM) gave the title compound as a white solid (107 mg). 1H
NMR (400 MHz, CDCI3): 7.56-7.49 (m, 4H), 7.42-7.39 (m. 2H), 7.31-7.27 (m, 1H),
6.99-6.96 (m, 2H), 4.43-4.37 (m, 1H), 3.18-3.13 (m, 3H), 2.77-2.70 (m, 2H), 2.07-
2.01 (m, 2H), 1.73-1.64 (m, 2H).
Example 87
BIOLOGICAL METHODS
In Vitro
Transfectlon of cells with human histamine receptor
A 10 cm tissue culture dish with a confluent monolayer of SK-N-MC cells was
split two days prior to transfection. Using sterile technique the media was removed
and the cells were detached from the dish by the addition of trypsin. One fifth of the
cells were then placed onto a new 10 cm dish. Cells were grown in a 37°C
incubator with 5% CO2 in Minimal Essential Media Eagle with 10% Fetal Bovine
Serum. After two days cells were approximately 80% confluent. These were
removed from the dish with trypsin and pelleted in a clinical centrifuge. The pellet
was then re-suspended in 400 µL complete media and transferred to an
electroporation cuvette with a 0.4 cm gap between the electrodes (Bio-Rad #165-
2088). One microgram of supercoiled H, receptor cDNA was added to the cells and
mixed. The voltage for the electroporation was set at 0.25 kV, the capacitance was
set at 960 µF. After electroporation the ceils were diluted into 10 mL complete
media and plated onto four 10 cm dishes. Because of the variability in the efficiency
of electroporation, four different concentrations of cells were plated. The ratios used
were; 1:20,1:10,1:5, with the remainder of the cells being added to the fourth dish.
The cells were allowed to recover for 24 hours before adding the selection media
(complete media with 600 µL G418). After 10 days dishes were analyzed for
surviving colonies of cells. Dishes with well isolated colonies were used. Cells from
individual colonies were isolated and tested. SK-N-MC cells were used because they
give efficient coupling for inhibition of adenylate cyclase. The clones that gave the
most robust inhibition of adenylate cyclase in response to histamine were used for
further study.
[3H]-N-methylhistamine binding
Cell pellets from histamine H3 receptor-expressing SK-N-MC cells were
homogenized in 20 mM TrisHCL/0.5 mM EOTA. Supernatants from a 800 g spin
were collected, recentrifuged at 30.000 g for 30 minutes. Pellets were re-
homogenized in 50 mM Tris/5 mM EDTA (pH 7.4). Membranes were incubated with
0.8 nM [3H]-N-methylhistamine plus/minus test compounds for 45 min at 25°C and
harvested by rapid filtration over GF/C glass fiber filters (pretreated with 0.3 %
polyetnylenimine) followed by four washes with ice cold buffer. Filters were dried,
added to 4 mL scintillation cocktail and then counted on a liquid scintillation counter.
Non-specific binding was defined with 10 µM histamine. The pK, values were
calculated based on a Kd of 800 pM and a ligand concentration ([L]) of 800 pM
according to the formula:
In Vivo
Elucidation of oral absorption and blood-brain barrier penetration profiles of H3
receptor antagonists in the rat
A rat in vivo system was used to determine the blood-brain barrier penetration
profiles and kinetics of various H, receptor antagonists after single bolus oral
administration.
Female Sprague Oawley Rats (~300 gram body weight) were housed in
accordance with institutional standards and allowed to acclimate for at least 7 days
prior to the study. Each H3 antagonist was formulated in 0.5% hydroxypropylmethyl
cellulose at a concentration of 1 mg/mL for oral dosing. The test compound was
administered to each of eight animals as a single oral dose of 10 mL/kg (10 mg/kg).
Remaining dosing solution was retained for analysis. Two animals from each
original group of eight were euthanized via CO2 asphyxiation at t = 1, 6, 24, and 48
hours. After each animal was euthanized, 0.1 mL of its blood was sampled via
cardiac puncture, and its brain was removed via dissection of the cranial bones and
placed in a pre-weighed 50 mL conical tube on dry ice.
The blood was added to 0.3 mL of 6% trichloroacetic acid, and the acidified
sample was vortexed and then centrifuged (5 minutes at 14,000 rpm in a
microcentrifuge). The clear supernatant was retained for analysis. The frozen brain
was weighed, homogenized in 6% trichloroacetic acid (3 mL/g wet weight of tissue),
and then centrifuged. The clear supernatant was retained for analysis. The
supematants from the blood and brain samples were analyzed by liquid
chromatography with mass spectral detection utilizing selective reaction monitoring
(LC-MS/MS). The LC method used a Phenomonex Polar RP column (2 x 50 mm)
and a linear solvent gradient of water and acetonitrite (both 1 % in acetic acid).
Graphs of H3 receptor antagonist concentration versus time for blood and
brain were generated from the LC-MS/MS results. The mean residency time (MRT)
of the H3 receptor antagonist in blood or in the brain, was calculated from the ratio
of the area under the first moment curve (AUMC) to the area under the
concentration time curve (AUC): AUMC/AUC. The Blood Bran Barrier index was
calculated from the log of AUCtu/AUCblood.
F. Other Embodiments
The features and advantages of the invention will be apparent to one of
ordinary skill in view of the discussion, examples, embodiments, and claims relating
to the invention. The invention also contemplates variations and adaptations, based
on the disclosure herein concerning the key features and advantages of the
invention, and within the abilities of one of ordinary skill.
What is claimed is:
1. A compound of formula (I):
wherein X is O;
n is an integer from 0 to 3;
R6 is C 1-10 alkyl, C 3-4 alkenyl, C 3-8 cycloalkyl, (C 3-8 cycloalkyl) C 1-6
alkyl, (pheny()C 1-6 alkyl, (phenyl)C 3-8 alkenyl, or (C 1-8 alkylcarbonyl)C 1-5 alkyl;
one of R1, R2, and R3 is G or W, wherein one of the remaining two is
selected from H and halogen, and the third being hydrogen;
wherein:
L1 is C 2-8 alkylene, C 3-8 cycloalkylerie, C 4-6 alkenylene, C 4-6
alkynyiene, C2-6 alkanoyl, (phenyl)C 1-6 alkylene, (naphithyl)C 1-6 alkylene, (C 2-5
heteroaryl )C 1-5 alkylene, (phenoxy)C1-6 alkylene, or (C 2-5 heteroaryloxy}C1-6
alkylene;
L2 Is C1-6 alkytene, C 3-8 cycloalkylene, C 3-6 alkenylene, C 3-6 alkynylene,
C3-6 alkanoyl, (phenyl)C 1-6 alkylene, (naphthyl)C 1-6 alkylene, (C 1-6
heteroaryl)C 1-6 alkylene, (phenoxy)C1-6 alkylene, (C1-5 heteroaryioxy)C1-6
alkylene, or (C1-6 heteroarythio)C1-6 alkylene;
L3 is C1-6 alkylene, C 2-6 alkenylene, C 2-6 alkynyiene, C2-6 alkanoyl,
(phenyl)C 1-6 alkylene, pnenyl, naphthyl, (naphthyl)C 1-6 alkylene, C1-6
heteroaryl)C 1-6 alkylene, (phenoxy)C1-6 alkylene, (C 1-6 heteroaryloxy)C1-6
alkylene, or C 2-6 heteroaryl;
L4 is C1-5 alkylene;
L5 is C1-6 alkylene;
L6 is C1-5 alkylene;
L7 is C1-5 alkylene or absent;
Q is -NR8R9 or a non-aromatic C 2-15 heterocyclyl ring system containing
at least one nitrogen atom and optionally between 1 and 3 additional
heteroatoms selected from O, S, and N in each ring;
R6 is independently selected from hydrogen, C1-6 alkyl,
C1-6 alkoxy, C 2-8 alkenyl, C 3-7 cycloalkyl, (C3-7 cycloalkyl)C1-6 alkylene,
C2-16 heterocyclyl, and (C2-7 hetenocycyl)C1-6 alkylene;
R7 is H, hydroxyl, halo, C2-8 alkoxy or absent where the carbon linking
L6 and L7 (or bonded to R6) participates in a double bond;
each of R8 and R9 is independently selected from hydrogen, C1-6
alkoxy, C1-8 alkyl, C 3-8 alkenyl, C 3-7 cycloalkyl. (C3-7 cycloalkyl)C1-6, alkylene,
C2.15 heterocyclyl, phenyl, (C2-15heterocyclyl)C1-6 alkylene, and (phenyl) C1-6
alkylene;
R10 is H, C 1-6 alkyl, C 3-8 alkenyl, C 3-7 cycloalkyl, (C3-7, cycloalkyl)C1-8
aikylene, (C3-15heterocyclyl)C1-6, alkylene, or (phenyl) C1-4 alkylene;
W is -CN, -CHO, halogen, C1-8 heterocyclyl, (C1-8 heterocyclyl)-O-,
phenoxy, phenyl, (phenyl)C1-6 alkylene-O-, -C(O)R, -C(OH)RxRy, C1-6 alkyl.
C1-6 cycloalkyl, or -NRxRy;
wherein each of Rx and Ry is independently selected from H, C1-6 alkyl,
C1-6 alkanoyl, C1-6 heterocyclyl, and phenyl;
wherein each of the above alkyl. alkylene, alkenyl, alkenylene, alkynyl,
alkynylene, heterocydyl, cydoalkyl. and aryl groups may each be
independently and optionally substituted with between 1 and 3 substituents
selected from halo, amino. nitro, hydroxyl. and C 1-3 alkyl;
wherein substituents of Q can be further selected from carboxamlde,
C2-6 alkyl, C1-6 heterocyclyl, N(C1-6 alkyl)(C1-6 heterocydyl), NH(C1-6
heterocydyl), (C1-8 alkylene)(C1-6 heterocyclyl), O(C1-4 heterocyclyl), O(C1-4
alkyl), O(C1-6 cycloalkyl), phenyl, (C1-3 alkylene) phenyl, N(C1-4 alkyl)(C1-6
alkylene) phenyl, and O(C1-3 alkylene) phenyl where each of above
hetarocydyt, phenyl, and alkyl groups may be optionally substituted with from
1 to 3 substituents independently selected from halogen, nitro, cyano, and C1-
3 alkyl;
or a pharmaceuticaly acceptable salt, ester, or omide thereof.
2. A compound of claim 1, wherein R6 is C1-6 alkyl, C3-4 alkenyl, C2-6
cydoalkyl, (C 3-6 cydoalkyl) C 1aIkylene, (phenyl)C 1-3 alkylene, or
(pnonyl)C 3-4 alkenylene.
3. A compound of claim 2, wherein R, is branched C 3-5 alkyl, C 3-5
cydoalkyl, and (C 3-6 cydoalkyl )C 1 alkylene.
4. A compound of claim 1, wherein one of R2 and R3 is G.
5. A compound of claim 4, wherein R, is G.

6. A compound of claim 4, wherein R3 is G.
7. A compound of claim 1, wherein L1 is C2-3 alkylene.
8. A compound of claim 1, wherein L2 is C1-6 alkylene, (C1-5
heteroaryl) C1-6 alkylene, or-phenyl-C1-6 alkylene.
9. A compound of claim 8, wherein L2 is methylene.
10. A compound of claim 1, wherein L3 is ethylene, vinylene,
ethynylene, and phenylene.
11. A compound of claim 1, wherein Q is NR8R9 wherein each of R8
or R9 is independently hydrogen, C1-8alkyl, C3-8alkenyl, C3-
7cycloalkyl, (C3-7Cycloalkyl) C1-6alkylene, C2-5heterocyclyl,
phenyl, (C2-5heteroxcyclyl)C1-6 alkylene or (phenyl) C1-6alkylene.
12. A compound of claim 11, wherein one of R8 or R9 is hydrogen.
13. A compound of claim 12, wherein R8 is H and R9 is phenyl or
aromatic C1-8 heterocyclyl optionally substituted with 1-3 halo,
nitro, cyano or C1-3 alkyl substituents.
14. A compound of claim 13, wherein R9 is phenyl, pyridyl, pyrimidyl,
furyl, thiofuryl, imidazolyl, (C1-6 alkyl) imidazolyl, oxazolyl,
thiazolyl, 2,3dihydro-indolyl, benzimidazolyl, 2-
oxobenzimidazolyl, (C1-6alkyl) tetrazolyl, tetrazolyl, (C1-6alkyl)
triazolyl, triazolyl, (C1-6alkyl)pyrrolyl or pyrrolyl.
15. A compound of claim 13, wherein R5 is C1-5alkyl, C3-4alkenyl, C3-
6cycloalkyl, (C3-6cycloalkyl)C1alkylene, (phenyl) C1-3alkylene, or
(phenyl)C3-4alkylene.
16. A compound of claim 1, wherein n is 0 or 1.
17. A compound of claim 16, wherein n is 0.
18. A compound of claim 1, wherein G is:
(4) formula (i) wherein L4 and L5 are independently C2-3alkylene,
(5) formula (iii) wherein L6 is C2-3alkylene and L7 is C2-3alkylene
or absent,
(6) L2Q wherein L2 is C1-6alkylene, phenylC1-4alkylene, or
(aromatic C1-5heterocyclyl)C1-4alkylene, or
(7) OL1Q wherein L1, is C2-3 alkylene.
19. A compound of claim 18, wherein G is selected from:
(8) formula (i) wherein L4 and L5 are each C2alkylene,
(9) formula (iii) wherein each of L6 and L7 is C2alkylene, or
(10) L2Q wherein L2 is methylene.
20. A compound of claim 19, wherein G is L2Q.
21. A compound of claim 18, wherein R10 is H, branched C3-6alkyl, or
benzyl.
22. A compound of claim 21, wherein R]0 is isopropyl or benzyl.
23. A compound of any one of claim 1 to 22, wherein Q is a non-
aromatic C2-5 heterocyclyl.
24. A compound of claim 23, wherein Q is piperidyl, N-(C1-6alkyl)
piperazinyl, piperazinyl, pyrrolinyl, pyrrolidinyl or morpholinyl.
25. A compound of claim 23, wherein Q is N-morpholinyl or N-
piperidinyl, optionally substituted with between 1 and 3
substituents hydroxyl, carboxamide, (C1-6alkyl, C1-8heterocyclyl,
N(C1-6alkyl)(C1-8heterocyclyl), NH (C1-8heterocyclyl),
(C13alkylene) (C1-8heterocyclyl), (C1-3alkylene) (C1-8heterocyclyl),
O(C1-8heterocyclyl), O(C1-6alkyl), O(C3-6cycloalkyl), phenyl
(C13alkylene)phenyl, N(C1alkyl)(C1-3alkylene)phenyl, and
O(C13alkylene) phenyl where each of above heterocyclyl, phenyl,
and alkyl groups may be optionally independently substituted with
from 1 to 3 halogen, nitro, cyano or C1-3alkyl substituents.
26. A compound of claim 25, wherein Q is substituted with pyridyl,
pyrimidyl, furyl, thiofuryl, imidazolyl, (C1-6alkyl) imidazolyl,
oxazolyl, thiazolyl, 2,3dihydro-indolyl, benzimidazolyl, 2-
oxobenzimidazolyl, (C1-6alklyl) tetrazolyl, tetrazolyl,
(C16alkyl)triazolyl, triazolyl, (C1-6alkyl) pyrrolyl orpyrrolyl.
27. . A compound of claim 26, wherein Q is a substituted or
unsubstituted N-morpholinyl.
28. A compound of claim 18, wherein R5 is C1-5alkyl, C3-4alkenyl,
C36cycloalkyl, (C3-6Cycloalkyl) C1alkylene, (phenyl) C1-3alkylene,
or (phenyl)C3-4alkenylene.
29. A compound of claim 18, wherein R7 is hydroxyl, halo or absent
where one of L6 and R7 provides a double bond to the carbon atom
to which R6 and R7 are attached.
30. A compound of claim 1, wherein one of R2 and R3 is W and W is
pyridyl, pyrimidyl, furyl, thiofuryl, imidazolyl, oxazolyl, thiazolyl,
2,3-dihydro-indolyl, benzimidazolyl, tetrazolyl, triazolyl or
pyrrolyl.
31. A compound of claim 16, wherein R5 is branched C3-5alkyl.
32. A compound of claim 16, wherein R5 is isopropyl or cyclopentyl.
33. Any one of the following compounds:
4-(4-Imidazol-1 -yl-phenoxy)-1 -isopropyl-piperidine,
4-(4-Imidazol-1 -yl-phenoxy)-1 -isobuty 1-piperidine,
1 -Isopropyl-4-(4-pyrrol-1 -yl-phenoxy )-piperidine,
5-Chloro-2-[4-( 1 -isopropyl-piperidin»4-yloxy)-phenyl]-1H-
benzoimidazole,
4-(4-Imidazol-1 -yl-phenoxy)-1 -isopropyl-piperidine,
4-(4-Imidazol-1 -yl-phenoxy)-1 -isobutyl-piperidine,
5-Chloro-2-[4-( 1 -isopropyl-piperidin-4-yloxy)-phenyl]-1H-
benzoimidazole,
[4-( 1 -Isopropyl-piperidin-4-yloxy)-phenyl]-phenyl-methanone,
4-(Biphenyl-4-yloxy)-1 -isopropyl piperidine,
4-(4-Benzyloxy-phenoxy)-1 -isopropyl-piperidine,
l-Isopropyl-4-(4-phenoxy-phenoxy)-piperidine,
4-(4-Benzyl-phenoxy)-1 -isopropyl-piperidine,
[4-( 1 -Isopropyl-piperidin-4-yloxy)-phenyl]-phenyl methanone,
N-[4-( 1 -Isopropyl-piperidin-4-yloxy)-phenyl]-acetamide,
4-(4-Cyclophenyl-phenoxy)-1 -isopropyl-piperidine,
4-( 1 -Cyclopentyl-piperidin-4-yloxy)-benzonitrile,
4-( 1 -Cyclobutyl-piperidin-4-yloxy)-benzonitrile,
4-( 1 -sec-Butyl-piperidin-4-yloxy)-benzonitrile,
4-( 1 -Isopropyl-piperidin-4-yloxy)-benzaldehyde,
4-( 1 -Cyclohexyl-piperidin-4-yloxy)-benzonitrile,
4-( 1 -Isopropyl-piperidin-4-yloxy)-benzonitrile,
4-( 1 -Cyclopropylmethyl-piperidin-4-yloxy)-benzonitrile,
4-Isobutyl-piperidin-4-yloxy)-benzonitrile,
4-( 1 -Propyl-piperidin-4-yloxy)-benzonitrile,
4-(Biphenyl-4-yloxy)-1 -isopropyl-piperidine,
4-(4-Benzyloxy-phenoxy)-1 -isopropyl-piperidine,
4-(4-Benzyl-phenoxy)-1 -isopropyl-piperidine,
1-Isopropyl-4-(4-phenoxy-phenoxy )-piperidine,
N-[4-( 1 -Isopropyl-piperidin-4-yloxy)-phenyl]-acetamide,
1 -Isopropyl-4-[4-( 1 -isopropyl-piperidin-4-yloxy)-phenyl]-
piperazine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-phenyl]-piperazine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy )-phenyl]-piperazine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-piperazine,
4-[4-( 1 -sec-Butyl-piperidin-4-yloxy)-benzyl]morpholine,
1 -[4-( 1 -Cyclopentyl-piperidin-4-yloxy)-benzyl]-piperidine,
1 -[4-( 1 -Isobutyl-piperidin-4-yloxy)-benzyl]-piperidine,
1 -N-Isopropyl-4-4-[5-( 1 -isopropyl-piperidin-4-ylsulfanyl)-tetrazol-
1 -yl]-phenoxy} -piperixine,
{l-[4-(l-Isopropyl-piperidin-4-yloxy )-benzyl]-piperidin-4-yl}-
methanol,
1 -[4-(1 -Isopropyl-piperidin-4-yloxy)-benzyl]-4-methyl-[1,
4]diazepane,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-azepane,
1 -[4-( 1 -Isobutyl-piperidin-4-yloxy)-benzyl]-piperazine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-ol,
[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-methyl-( 1 -methyl-
piperidin-4-yl)-amine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy )-benzyl]-4-benzyl-piperidine,
N-[4-(l-Isopropyl-piperidin-4-yloxy)-benzyl]-N,N'N'-trimethyl-
ethane-1,2-diamine,
1 -[4-( 1 -Isopropyl piperidin-4-yloxy)-benzyl]-4-methyl-piperazine,
Cyclohexyl-[4-(l -isopropyl-piperidin-4-yloxy )-benzyl]-methyl-
amine,
Butyl-[4-(l-isopropyl-piperidin-4-yloxy)-benzyl]-methyl-amine,
4-[4-( 1-Cyclopentyl piperidin-4-yloxy)-benzyl]-morpholine,
1 -Isopropyl-4-(4-pyrrolidin-1 -ylmethyl-phenoxy)-piperidine,
Diethyl-[4-(l-isopropyl-piperidin-4-yloxy)-benzyl]-amine,
4-[4-( 1 -sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine,
l-[4-(l-Isoropyl-piperidin-4-yloxy)-benzyl]-4-phenyl-piperazine,
1 -Benzyl-4-[4-( 1 -isopropyl-piperidin-4-yloxy)-benzyl]-piperazine,
4-[4-(4-Benzylidene-piperidin-1 -ylmethyl)-phenoxy]-1 -isopropyl-
piperidine,
4-[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-morpholine,
[4-( 1 -Isopropyl piperidin-4-yloxy)-bezyl]-dimethyl-amine,
4-[4-(l-Cyclohexyl-piperidin-4-yloxy)-benzyl]-morpholine,
4-[4-( 1 -Isobutyl-piperidin-4-yloxy)-benzyl] morpholine,
4-[4-( 1 -Propyl-piperidin-4-yloxy)-benzyl]-morpholine,
1 -[4-( 1 -Cyclohexyl-piperidin-4-yloxy)-benzyl]-piperidine,
l-[4-(l-Benzyl-piperidin-4-yloxy)-benzyl]-piperidine,
1 -[4-( 1 -Cyclohexylmethyl-piperidin-4-y loxy)-benzyl]-piperidine,
4-[4-(4-Piperidin-1 -ylmethyl-phenoxy)-piperidin-1 -yl]-butan-2-
one,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-piperidine,
4-[4-( 1 -sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine,
1 -[4-( 1 -Cyclopentyl-piperidin-4-yloxy)-benzyl]-piperidine,
1 -[4-( 1 -Isobutyl-piperidin-4-yloxy)-benzyl]-piperidine,
l-N-Isopropyl-4-4-[5-(l-isopropyl-piperidin-4-ylsulfanyl)-tetrazol-
1 -yl]-phenoxy }-piperixine,
{l-[4-(l-Isopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-yl}-
methanol,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-4-methyl-[ 1,
4]diazepane,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-azepane,
1 -[4-( 1 -Isobutyl-piperidin-4-yloxy)-benzyl]-piperidine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-piperidin-4-ol,
[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-methyl-( 1 -methyl-
piperidin-4-yl)-amine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-4-benzyl-piperidine,
N-[4-(l-Isopropyl-piperidin-4-yloxy)-benzyl]-N,N',N'-trimethyl-
ethane-1,2-diamine,
1 -[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-4-methyl-piperazine,
Cyclohexyl-[4-(l-isopropyl-piperidin-4-yloxy)-benzyl]-methyl-
amine,
Butyl-[4-(l-isopropyl-piperidin-4-yloxy)-benzyl]-methyl-amine,
4-[4-( 1 -Cyclopentyl piperidin-4-yloxy)-benzyl]-morpholine,
1-Isopropyl-4-(4-pyrrolidin-l-ylmethyl-phenoxy)-piperidine,
Diethy l-[4-( 1 -isopropyl-piperidin-4-yloxy)-benzyl]-amine,
4-[4-( 1 -sec-Butyl-piperidin-4-yloxy)-benzyl]-morpholine,
1 -[4-( 1 -Isoropyl-piperidin-4-yloxy)-benzyl]-4-phenyl-piperazine,
1 -Benzyl-4-[4-( 1 -isopropyl-piperidin-4-yloxy)-benzyl]-piperazine,
4-[4-(4-Benzylidene-piperidin-1 -ylmethy l)-phenoxy]-1 -isopropyl-
piperidine,
4-[4-( 1 -Isopropyl-piperidin-4-yloxy)-benzyl]-morpholine,
[4-( 1 -Isopropyl-piperidin-4-yloxy)-bezyl]-dimethyl-amine,
4-[4-( 1 -Cyclohexyl-piperidin-4-yloxy)-benzyl]-morpholine,
4-[4-( 1 -Isobutyl-piperidin-4-yloxy) benzyl]-morpholine,
Cyclopropyl-[4-(l-isopropyl-piperidin-4-yloxy)-benzyl]-amine,
[4-(l-Isopropyl-piperidin-4-yloxy)-benzyl]-(4-methyl-pyridin-2-
yl)-amine,
[4-(l-Isopropyl-piperidin-4-yloxy)-benzyl]-pyridin-2-yl-amine,
[4-(l -Isopropyt-piperidin-4-yloxy)-benzyl]-phenyl-amine, or
(5-Chloro-pyridin-2-y l)-[4-( 1 -isopropy l-piperidin-4-yloxy)-
benzyl]-amine.
Substituted non-imidazole aryloxypiperidine compounds,
compositions containing them, and methods of making and using
them to treat or prevent histamine-mediated conditions.

Documents:

156-kolnp-2003-granted-abstract.pdf

156-kolnp-2003-granted-assignment.pdf

156-kolnp-2003-granted-claims.pdf

156-kolnp-2003-granted-correspondence.pdf

156-kolnp-2003-granted-description (complete).pdf

156-kolnp-2003-granted-examination report.pdf

156-kolnp-2003-granted-form 1.pdf

156-kolnp-2003-granted-form 18.pdf

156-kolnp-2003-granted-form 2.pdf

156-kolnp-2003-granted-form 26.pdf

156-kolnp-2003-granted-form 3.pdf

156-kolnp-2003-granted-form 5.pdf

156-kolnp-2003-granted-reply to examination report.pdf

156-kolnp-2003-granted-specification.pdf


Patent Number 223388
Indian Patent Application Number 156/KOLNP/2003
PG Journal Number 37/2008
Publication Date 12-Sep-2008
Grant Date 10-Sep-2008
Date of Filing 10-Feb-2003
Name of Patentee ORTHO MCNEIL PHARMACEUTICAL, INC.
Applicant Address U.S. ROUTE 202, RARITAN, NJ 08869 A DELAWARE CORPORATION
Inventors:
# Inventor's Name Inventor's Address
1 APODACA, RICHARD 8248 STATION VILLAGE LANE NO. 2216 SAN DIEGO, CA 92108
2 CARRUTHERS, NICHOLAS, I. 14370 SILVER HEIGHTS ROAD, POWAY, CA 92064
3 DVORAK, CURT, A. 7657 ANGELENO ROAD, SAN DIEGO, CA 92126
4 SHAH, CHANDRAVADAN, R. 14213 DALHOUSIE ROAD, SAN DIEGO, CA 92129
5 XIAO, W EI. 4043 CARMEL SPRINGS WAY, SAN DIEGO, CA 92130
PCT International Classification Number C07D 211/46, 211/44
PCT International Application Number PCT/US01/24660
PCT International Filing date 2001-08-06
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/223,768 2000-08-08 U.S.A.
2 09/922,619 2001-08-06 U.S.A.