WO2014028322A1 - Modulateurs du récepteur h3 histaminique et traitement de troubles s'y rapportant - Google Patents

Modulateurs du récepteur h3 histaminique et traitement de troubles s'y rapportant Download PDF

Info

Publication number
WO2014028322A1
WO2014028322A1 PCT/US2013/054307 US2013054307W WO2014028322A1 WO 2014028322 A1 WO2014028322 A1 WO 2014028322A1 US 2013054307 W US2013054307 W US 2013054307W WO 2014028322 A1 WO2014028322 A1 WO 2014028322A1
Authority
WO
WIPO (PCT)
Prior art keywords
ethyl
methylpyrrolidin
phenyl
itch
group
Prior art date
Application number
PCT/US2013/054307
Other languages
English (en)
Inventor
Albert Ren
Graeme Semple
Thuy-Anh Tran
Zheng Wei
Andrew J. Grottick
David M. Mills
Brian M. Smith
Original Assignee
Arena Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Arena Pharmaceuticals, Inc. filed Critical Arena Pharmaceuticals, Inc.
Publication of WO2014028322A1 publication Critical patent/WO2014028322A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings

Definitions

  • the present invention relates to compounds of Formula (la) and pharmaceutical compositions thereof that modulate the activity of the histamine H3 receptor (H3R) and are useful in methods for the treatment of histamine H3 receptor-associated disorders, such as, cognitive disorders, epilepsy, brain trauma, depression, obesity, disorders of sleep and wakefulness such as excessive daytime sleepiness, narcolepsy, shift-work sleep disorder, drowsiness as a side effect from a medication, maintenance of vigilance to aid in the completion of tasks and the like, cataplexy, hypersomnia, somnolence syndrome, jet lag, sleep apnea and the like, attention deficit hyperactivity disorder (ADHD), schizophrenia, allergies, allergic responses in the upper airway, allergic rhinitis, nasal congestion, dementia, Alzheimer's disease, pain, pruritus, and the like.
  • ADHD attention deficit hyperactivity disorder
  • n, and p are each independently 0 or 1 ;
  • X is selected from the group: -S-, -0-, C3-C6 cycloalkylene, and CR C R C ;
  • Z is -CH 2 - or -(CO)-;
  • R 1 is H or C 1 -C4 alkyl
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl;
  • each R , R b , and R c is independently selected from the group: H, C 1 -C3 alkyl, amino, halogen, and hydroxyl.
  • Another aspect of the present invention relates to the unexpected discovery that inhibition of the H3R in an individual, such as by administration of a H3R antagonist, can reduce itching or pruritus.
  • the present invention further relates to compounds of the present invention that are useful in the treatment of a condition characterized by itching, for example, in an individual.
  • the individual is a human.
  • the inventors have discovered that the G-protein coupled histamine 3 receptor is a key effector of the itch sensation.
  • the present invention provides a method of treating pruritus in a subject in need thereof by administering a therapeutically effective amount of a compound of the present invention that modulates the H3R.
  • peripherally restricted antagonists of H3R are capable of mediating the inhibition of itch.
  • Peripherally restricted compounds may be advantageous to the extent that the peripheral restriction reduces the CNS effects of H3R inhibition. Such effects may include, for example, wakefulness.
  • compositions comprising a compound of the present invention.
  • One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods of inducing wakefulness in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating a histamine H3 receptor-associated disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for inducing wakefulness.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of a histamine H3 receptor-associated disorder.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of the human or animal body by therapy.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of inducing wakefulness.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method for the treatment of a histamine H3 receptor-associated disorder.
  • a process for preparing a composition comprising formulating a compound of the present invention and a pharmaceutically acceptable carrier.
  • Figure 1 shows a general synthesis of biaryl derivatives useful as intermediates in the preparation of compounds of the present invention.
  • a mesylate derivative is coupled with an R 1 substituted pyrrolidine and subsequently converted to an aryl boronic acid by treatment of an aryl lithium intermediate with triisopropylborate followed by hydrolysis.
  • the aryl boronic acid is coupled with an aryl bromide in the presence of a palladium catalyst to prepare the biaryl derivatives.
  • Figure 2 shows general methods for preparing compounds of the present invention.
  • Method A shows the use of carboxylic acids in the presence of a coupling agent to prepare compounds of the invention
  • Method B shows the use of acid chlorides to prepare compounds of the invention, optionally in the presence of a base, such as an amine base.
  • Figure 3 shows a general method for preparing compounds of the present invention.
  • Method C shows the use of acid anhydrides in the presence of a base to prepare compounds of the invention.
  • Figure 4 shows the data from the administration of Compound 1 in the chronic DNFB- mediated allergic pruritus mouse model. The data shows that Compound 1 effectively inhibits chronic DNFB-mediated allergic pruritus in mice.
  • Figure 5 shows the data from the administration of Compound 1 in the histamine- induced pruritus mouse model. The data shows that Compound 1 effectively inhibits histamine- induced pruritus in mice.
  • Figure 6 shows the data from the administration of Compound 3 in the histamine- induced pruritus mouse model. The data shows that Compound 3 effectively inhibits histamine- induced pruritus in mice.
  • Figure 7 shows that central administration of a H3R antagonist, Compound A, does not inhibit histamine-induced pruritus.
  • Figure 7A shows ICV administration of Compound A increases cognitive function in mice.
  • Figure 7B shows that ICV administration of Compound A is unable to inhibit histamine-induced pruritus. Suggesting that brain exposure of a H3R antagonist is not sufficient and that histamine induced pruritus is peripherally mediated.
  • agonists refers to moieties that interact and activate the receptor, such as the histamine H3 receptor and initiate a physiological or pharmacological response characteristic of that receptor. For example, when moieties activate the intracellular response upon binding to the receptor, or enhance GTP binding to membranes.
  • antagonists refers to moieties that competitively bind to the receptor at the same site as agonists (for example, the endogenous ligand), but which do not activate the intracellular response initiated by the active form of the receptor and can thereby inhibit the intracellular responses by agonists or partial agonists. Antagonists do not diminish the baseline intracellular response in the absence of an agonist or partial agonist.
  • contact or contacting refers to bringing the indicated moieties together, whether in an in vitro system or an in vivo system.
  • "contacting" a histamine H3 receptor with a compound of the invention includes the administration of a compound of the present invention to an individual, preferably a human, having a histamine H3 receptor, as well as, for example, introducing a compound of the invention into a sample containing a cellular or more purified preparation containing a histamine H3 receptor.
  • in need of treatment and the term “in need thereof” when referring to treatment are used interchangeably to mean a judgment made by a caregiver ⁇ e.g. physician, nurse, nurse practitioner, etc. in the case of humans; veterinarian in the case of animals, including non-human mammals) that an individual or animal requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, but that includes the knowledge that the individual or animal is ill, or will become ill, as the result of a disease, condition or disorder that is treatable by the compounds of the invention. Accordingly, the compounds of the invention can be used in a protective or preventive manner; or compounds of the invention can be used to alleviate, inhibit or ameliorate the disease, condition or disorder.
  • mice refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates and most preferably humans.
  • inverse agonists refers to moieties that bind to the endogenous form of the receptor or to the constitutively activated form of the receptor and which inhibit the baseline intracellular response initiated by the active form of the receptor below the normal base level of activity which is observed in the absence of agonists or partial agonists, or decrease GTP binding to membranes.
  • the baseline intracellular response is inhibited in the presence of the inverse agonist by at least 30%, more preferably by at least 50% and most preferably by at least 75%, as compared with the baseline response in the absence of the inverse agonist.
  • modulate or modulating refers to an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule.
  • composition refers to a composition comprising at least one active ingredient; including but not limited to, salts, solvates and hydrates of compounds of the present invention; whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human).
  • a mammal for example, without limitation, a human.
  • terapéuticaally effective amount refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician or caregiver; or by an individual, which includes one or more of the following:
  • Preventing the disease for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease,
  • Inhibiting the disease for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or
  • Ameliorating the disease for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
  • C 1 -C4 alkoxy is intended to mean a radical comprising a C 1 -C4 alkyl group attached directly to an oxygen atom, wherein C 1 -C4 alkyl has the same definition as found herein. Some embodiments contain 1 to 3 carbons. Some embodiments contain 1 or 2 carbons. Examples include, but are not limited to methoxy, ethoxy, w-propoxy, isopropoxy, w-butoxy, t- butoxy, isobutoxy, s-butoxy, and the like.
  • C 1 -C4 alkoxycarbonyl refers to a radical comprising a single C 1 -C4 alkoxy group attached to the carbon of a carbonyl group, wherein C 1 -C4 alkoxy has the same definition as found herein.
  • the alkoxycarbonyl group may be represented by the following:
  • C 1 -C 4 alkyl refers to a straight or branched carbon radical containing 1 to 4 carbons. Some embodiments are 1 to 3 carbons. Some embodiments are 1 to 2 carbons. Some embodiments are 1 carbon. Examples of an alkyl include methyl, ethyl, w-propyl, wo-propyl, n- butyl, and s-butyl, isobutyl.
  • amino refers to the group -NH 2 .
  • C 3 -C6 cycloalkylene refers to a saturated ring di-radical containing 3 to 6 carbons. Some embodiments contain 3 to 5 carbons. Some embodiments contain 5 to 6 carbons. Some embodiments contain 3 to 4 carbons. Examples include cyclopropanediyl,
  • C 3 -C 6 cycloalkylene is selected from cyclopropane- 1, 1 -diyl, cyclobutane- 1, 1 -diyl, cyclopentane- 1 ,1- diyl, cyclohexane- 1,1 -diyl, cyclopropane- 1,2-diyl, cyclobutane- 1,2-diyl, cyclopentane- 1,2-diyl, cyclohexane- 1,2 -diyl, cyclopentane-l,3-diyl, cyclohexane- 1,3-diyl, cyclohexane- 1,4-diyl, and the :
  • halogen refers to a fluoro, chloro, bromo or iodo group.
  • hydroxyl refers to the group -OH.
  • One aspect of the present invention pertains to certain compounds as shown in Formula (la):
  • R 1 , R 2 , R , R b , X, Z, m, n, and p have the same definitions as described herein, supra and infra.
  • One aspect of the present invention pertains to certain compounds as shown in Formula (Ic):
  • R 1 , R 2 , R , R b , X, Z, m, n, and p have the same definitions as described herein, supra and infra.
  • One aspect of the present invention pertains to certain compounds as shown in Formula
  • R 1 , R 2 , R , R b , X, Z, m, n, and p have the same definitions as described herein, supra and infra.
  • substituted indicates that at least one hydrogen atom of the chemical group is replaced by a non-hydrogen substituent or group, the non-hydrogen substituent or group can be monovalent or divalent. When the substituent or group is divalent, then it is understood that this group is further substituted with another substituent or group.
  • a chemical group herein when a chemical group herein is "substituted" it may have up to the full valance of substitution; for example, a methyl group can be substituted by 1, 2, or 3 substituents, a methylene group can be substituted by 1 or 2 substituents, a phenyl group can be substituted by 1, 2, 3, 4, or 5 substituents, a naphthyl group can be substituted by 1, 2, 3, 4, 5, 6, or 7 substituents and the like.
  • substituted with one or more substituents refers to the substitution of a group with one substituent up to the total number of substituents physically allowed by the group. Further, when a group is substituted with more than one group they can be identical or they can be different.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. It is understood that the various tautomeric forms are within the scope of the compounds of the present invention.
  • meso isomers Such meso isomers may be referred to as cis and trans.
  • the trans meso isomers of compounds of Formula (la) are named herein using the prefix (lr,4r) and the cis meso isomers of compounds of Formula (la) are named herein using the prefix (li,4i), as illustrated with Compound 25 shown below:
  • Z is -CH 2 -.
  • One aspect of the present invention pertains to certain compounds as shown in Formula
  • R 1 , R 2 , R , R b , X, m, n, and p have the same definitions as described herein, supra and infra.
  • One aspect of the present invention pertains to certain compounds as shown in Formula
  • R 1 , R 2 , R , R b , X, m, n, and p have the same definitions as described herein, supra and infra.
  • R 1 is H or C 1 -C4 alkyl.
  • R 1 is H.
  • R 1 is C 1 -C4 alkyl.
  • R 1 is methyl.
  • R 2 is a group consisting of benzyl ether sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurethane, N-(2-aminoethyl)-2-a group consisting of benzyl sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylurea sulfonylure
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl.
  • R 2 is C 1 -C4 alkoxycarbonyl.
  • R 2 is selected from the group: methoxycarbonyl and
  • R 2 is carboxyl
  • R 2 is l/f-tetrazol-5-yl.
  • each R is selected independently from the group: H, C 1 -C3 alkyl, amino, halogen, and hydroxyl.
  • R is H.
  • R is halogen
  • R is hydroxyl
  • each R is selected independently from the group: H, C 1 -C3 alkyl, and amino.
  • R is C 1 -C3 alkyl.
  • each R is selected independently from the group: H, methyl, and amino.
  • R is methyl
  • R is amino.
  • the Group R b is amino.
  • each R b is selected independently from the group: H, C 1 -C3 alkyl, amino, halogen, and hydroxyl.
  • R b is H.
  • R b is halogen
  • R b is hydroxyl
  • each R b is selected independently from the group: H, C 1 -C3 alkyl, and halogen.
  • each R b is selected independently from the group: H, methyl, and fluoro.
  • R b is C 1 -C3 alkyl.
  • R b is methyl
  • R b is fluoro.
  • R c is fluoro.
  • each R c is selected independently from the group: H, C 1 -C3 alkyl, amino, halogen, and hydroxyl.
  • each R c is selected independently from the group: H, C 1 -C3 alkyl, halogen, and hydroxyl.
  • R c is independently selected from the group: H, methyl, amino, fluoro, and hydroxyl.
  • R c is H.
  • R c is C 1 -C3 alkyl.
  • R c is methyl
  • R c is amino
  • R c is halogen
  • R c is fluoro
  • R c is hydroxyl
  • X is -S-.
  • X is -0-.
  • X is C3-C6 cycloalkylene.
  • X is selected from the group: cyclopropane- 1,1-diyl, cyclopentane-l,l-diyl, cyclohexane- 1,1-diyl, cyclohexane-l,2-diyl, cyclohexane-l,3-diyl, and cyclohexane- 1 ,4 -diyl.
  • X is cyclopropane- 1,1 -diyl.
  • X is cyclopentane- 1 , 1 -diyl.
  • X is cyclohexane- 1,1 -diyl.
  • X is cyclohexane-l,2-diyl.
  • X is cyclohexane- 1,3-diyl.
  • X is cyclohexane-l,4-diyl.
  • X is CR C R C .
  • X is CR C R C , and each R c is selected independently from the group: H, C 1 -C3 alkyl, halogen, and hydroxyl.
  • X is CR C R C , and each R c is selected independently from the group: H, methyl, fluoro, and hydroxyl.
  • each R , R , and R c is independently selected from the group: H, methyl, amino, fluoro, and hydroxyl.
  • each R , R b , and R c is independently selected from the group: H, methyl, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (lie) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • n, and p are each independently 0 or 1, provided that when m is 1 then both n and p are both 1 ;
  • X is selected from the group: -S-, -0-, C3-C6 cycloalkylene, and CR C R C ;
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl;
  • each R , R , and R c is independently selected from the group: H, C 1 -C3 alkyl, amino, halogen, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (He) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • n, and p are each independently 0 or 1, provided that when m is 1 then n and p are both 1 ;
  • X is selected from the group: -S-, -0-, cyclopropane- 1,1-diyl, cyclopentane-l,l -diyl, cyclohexane-l,l -diyl, cyclohexane-l ,2-diyl, cyclohexane-l ,3-diyl, cyclohexane-l ,4-diyl, and CR C R C ;
  • R 2 is selected from the group: methoxycarbonyl, ethoxycarbonyl, carboxyl, and IH- tetrazol-5-yl;
  • each R , R b , and R c is independently selected from the group: H, methyl, amino, fluoro, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (He) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • n, and p are each independently 0 or 1, provided that when m is 1 then both n and p are both 1 ;
  • X is selected from the group: -S-, -0-, C 3 -C 6 cycloalkylene, and CR C R C ;
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl;
  • each R , R , and R c is independently selected from the group: H, C 1 -C3 alkyl, amino, halogen, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (He) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • n, and p are each independently 0 or 1, provided that when m is 1 then n and p are both 1;
  • X is selected from the group: -S-, -0-, cyclopropane- 1,1-diyl, cyclopentane-l,l-diyl, cyclohexane-l,l-diyl, cyclohexane-l,2-diyl, cyclohexane-l,3-diyl, cyclohexane-l,4-diyl, and CR C R C ;
  • R 2 is selected from the group: methoxycarbonyl, ethoxycarbonyl, carboxyl, and IH- tetrazol-5-yl;
  • each R , R b , and R c is independently selected from the group: H, methyl, amino, fluoro, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Ilg) and pharmaceutically accharides
  • X is selected from the group: -S-, -0-, C3-C6 cycloalkylene, and CR C R C ;
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl and carboxyl; and each R , R b , and R c is independently selected from the group: H, C 1 -C3 alkyl, amino, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Ilg) and pharmaceutically accharides
  • X is selected from the group: -S-, -0-, cyclopentane- l,l-diyl, cyclohexane-l,l-diyl, and CR C R C ;
  • R 2 is selected from the group: methoxycarbonyl, ethoxycarbonyl, and carboxyl; and each R , R b , and R c is independently selected from the group: H, methyl, amino, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Hi) and pharmaceutically accharides
  • X is selected from the group: -S-, -0-, C 3 -C 6 cycloalkylene, and CR C R C ;
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl and carboxyl;
  • each R , R b , and R c is independently selected from the group: H, C 1 -C3 alkyl, amino, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Hi) and pharmaceutically accharides
  • X is selected from the group: -S-, -0-, cyclopentane- l,l-diyl, cyclohexane-l,l-diyl, and CR C R C ;
  • R 2 is selected from the group: methoxycarbonyl, ethoxycarbonyl, and carboxyl; and each R , R b , and R c is independently selected from the group: H, methyl, amino, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Hi) and pharmaceutically acceptable salts, solvates and hydrates thereof, wherein X is CR C R C ; and each R , R b , and R c is independently selected from the group: H, methyl, and hydroxyl.
  • X is CR C R C ; and each R , R b , and R c is independently selected from the group: H, methyl, and hydroxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Ilk) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • R 2 is selected from the group: C1-C 4 alkoxycarbonyl and carboxyl;
  • each R b , and R c is independently selected from the group: H, C1-C3 alkyl, and halogen.
  • R is selected from the group: methoxycarbonyl, ethoxycarbonyl, and carboxyl; and each R b , and R c is independently selected from the group: H, methyl, and fluoro.
  • R 2 is selected from the group: C1-C 4 alkoxycarbonyl and carboxyl;
  • each R b , and R c is independently selected from the group: H, C1-C3 alkyl, and halogen.
  • R j 2 is selected from the group: methoxycarbonyl, ethoxycarbonyl, and carboxyl; and each R b , and R c is independently selected from the group: H, methyl, and fluoro.
  • X is C 3 -C6 cycloalkylene
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, and carboxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (IIo) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • X is selected from the group: cyclopropane- 1,1-diyl, cyclohexane-l,2-diyl, cyclohexane-l,3-diyl, and cyclohexane-l,4-diyl; and
  • R 2 is selected from the group: methoxycarbonyl, ethoxycarbonyl, and carboxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Ilq) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • X is C3-C6 cycloalkylene
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, and carboxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (Ilq) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • X is selected from the group: cyclopropane- 1,1-diyl, cyclohexane-l,2-diyl, cyclohexane-l,3-diyl, and cyclohexane-l,4-diyl; and
  • R 2 is selected from the group: methoxycarbonyl, ethoxycarbonyl, and carboxyl.
  • Some embodiments of the present invention pertain to compounds of Formula (lis) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl;
  • each R b is independently selected from the group: H, and C 1 -C3 alkyl.
  • Some embodiments of the present invention pertain to compounds of Formula (lis) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • R j 2 is selected from the group: methoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl; and each R b is independently selected from the group: H, and methyl.
  • Some embodiments of the present invention pertain to compounds of Formula (IIu) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl;
  • each R is independently selected from the group: H, and C 1 -C3 alkyl.
  • R is selected from the group: methoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl; and each R b is independently selected from the group: H, and methyl.
  • Some embodiments of the present invention pertain to compounds of Formula (IIw) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl.
  • R is selected from the group: methoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl.
  • Some embodiments of the present invention pertain to compounds of Formula (Ily) and pharmaceutically acceptable salts, solvates and hydrates thereof:
  • R 2 is selected from the group: C 1 -C4 alkoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl.
  • R 2 is selected from the group: methoxycarbonyl, carboxyl, and l/f-tetrazol-5-yl.
  • One aspect of the present invention encompasses every combination of one or more compounds selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One aspect of the present invention encompasses every combination of one or more compounds selected from the compounds in Table A and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • Table A
  • individual compounds and chemical genera of the present invention encompass all pharmaceutically acceptable salts, solvates and particularly hydrates, thereof.
  • the compounds of the Formula (la) of the present invention may be prepared according to relevant published literature procedures that are used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter in the working Examples.
  • Protection and deprotection may be carried out by procedures generally known in the art (see, for example, Greene, T. W. and Wuts, P. G. M., Protecting Groups in Organic Synthesis, 3 rd Edition, 1999 [Wiley]). It is understood that the present invention embraces each diastereoisomer, each enantiomer and mixtures thereof of each compound and generic formulae disclosed herein just as if they were each individually disclosed with the specific stereochemical designation for each chiral carbon.
  • Histamine [2-(imidazol-4-yl)ethylamine] exerts its physiological effects through four distinct G-protein coupled receptors (GPCRs), termed HI, H2, H3 and H4.
  • GPCRs G-protein coupled receptors
  • the histamine H3 receptor was first identified in 1983, when it was determined that the H3 receptor acted as an autoreceptor controlling both the synthesis and release of histamine (see: Arrang et al. Nature 1983, 302, 832-7).
  • At least four human and three rat splice variants have proven functional activity in pharmacological assays (Passani et al., Trends in Pharmacol. Sci. 2004, 25, 618-625).
  • Rat and human histamine H3 receptors also show constitutive activity which means that they can transduce a signal even in the absence of a ligand. Histamine H3 receptors also function as heteroceptors, modulating the release of a number of other transmitter substances including serotonin, acetylcholine, dopamine and noradrenaline (see: Brown et al. Prog. Neurobiol. 2001, 63, 637-672).
  • ligands which target the histamine H3 receptor, where the ligand functions as either an antagonist or inverse agonist (for reviews see: Leurs et al. Nat. Rev. Drug. Discov. 2005, 4, 107-120; Passani et al. Trends Pharmacol. Sci. 2004, 25, 618-625).
  • H3 receptor antagonists have been shown to increase wakefulness (e.g. Lin J. S. et al. Brain Research 1990, 523, 325-330). This effect demonstrates that H3 receptor antagonists can be useful for disorders of sleep and wakefulness (Parmentier et al. J. Neurosci. 2002, 22, 7695-7711; Ligneau et al. J. Pharmacol. Exp. Ther. 1998, 287, 658-666).
  • histamine H3 receptor antagonists and inverse agonists can be used to treat the somnolence syndrome associated with different pathological conditions, for example, sleep apnea and Parkinson's disease or circumstances associated with lifestyle, for example, daytime somnolence from sleep deprivation as a result of nocturnal jobs, overwork, or jet-lag (see Passani et al., Trends Pharmacol. Sci. 2004, 25, 618-625). Somnolence is one of the major problems of public health because of its high prevalence (19-37% of the general population) and risk for causing work and traffic accidents.
  • Sleep apnea is a common sleep disorder characterized by brief interruptions of breathing during sleep. These episodes, called apneas, last 10 seconds or more and occur repeatedly throughout the night. People with sleep apnea partially awaken as they struggle to breathe, but in the morning they may not be aware of the disturbances in their sleep.
  • the most common type of sleep apnea is obstructive sleep apnea (OSA), caused by relaxation of soft tissue in the back of the throat that blocks the passage of air.
  • OSA obstructive sleep apnea
  • CSA Central sleep apnea
  • the hallmark symptom of the disorder is excessive daytime sleepiness.
  • sleep apnea Additional symptoms of sleep apnea include restless sleep, loud snoring (with periods of silence followed by gasps), falling asleep during the day, morning headaches, trouble concentrating, irritability, forgetfulness, mood or behaviour changes, weight gain, increased heart rate, anxiety, and depression.
  • histamine H3 receptor antagonists and inverse agonists can be used to treat narcolepsy (Tedford et al. Soc. Neurosci. Abstr. 1999, 25, 460.3).
  • Narcolepsy is a neurological condition most often characterized by Excessive Daytime Sleepiness (EDS), episodes of sleep and disorder of REM or rapid eye movement sleep.
  • EDS Excessive Daytime Sleepiness
  • the main characteristic of narcolepsy is overwhelming Excessive Daytime Sleepiness (EDS), even after adequate nighttime sleep.
  • a person with narcolepsy is likely to become drowsy or to fall asleep, often at inappropriate times and places.
  • nighttime sleep may be fragmented with frequent wakenings.
  • Classic symptoms of narcolepsy include, for example, cataplexy which is sudden episodes of loss of muscle function, ranging from slight weakness (such as limpness at the neck or knees, sagging facial muscles, or inability to speak clearly) to complete body collapse. Episodes may be triggered by sudden emotional reactions such as laughter, anger, surprise, or fear, and may last from a few seconds to several minutes.
  • Another symptom of narcolepsy is sleep paralysis, which is the temporary inability to talk or move when waking up.
  • hypnagogic hallucinations which are vivid, often frightening, dream-like experiences that occur while dozing, falling asleep and/or while awakening, and automatic behaviour which occurs when a person continues to function (talking, putting things away, etc.) during sleep episodes, but awakens with no memory of performing such activities.
  • Daytime sleepiness, sleep paralysis, and hypnagogic hallucinations also occur in people who do not have narcolepsy, such as in people who are suffering from extreme lack of sleep. Cataplexy is generally considered unique to narcolepsy.
  • narcolepsy treat the symptoms, but not the underlying cause.
  • antidepressant medications and other drugs that suppress REM sleep are prescribed.
  • the drowsiness is normally treated using stimulants such as methylphenidate (Ritalin), amphetamines (Adderall), dextroamphetamine (Dexedrine), methamphetamine (Desoxyn), modafinil (Provigil), etc.
  • Other medications used are codeine and selegiline.
  • the cataplexy is treated using clomipramine, imipramine, or pro trip tyline but this need only be done in severe cases.
  • the drug gamma-hydro xybutyrate (GHB) (Xyrem) is approved in the USA by the Food and Drug Administration to treat both the cataplexy and excessive daytime sleepiness associated with narcolepsy.
  • histamine H3 receptor antagonists and inverse agonists can be used for the treatment and/or prevention of conditions associated with excessive daytime sleepiness such as hypersomnia, narcolepsy, sleep apnea, time zone change disorder, and other disorders which are associated with excessive daytime sleepiness such as fibromyalgia, and multiple sclerosis (Parmentier et al., J. Neurosci. 2002, 22, 7695-7711 ; Ligneau et al. J. Pharmacol. Exp. Ther. 1998, 287, 658-666).
  • Other conditions include excessive sleepiness due to shift-work, medical disorders, psychiatric disorders, narcolepsy, primary hypersomnia, and the like.
  • Histamine H3 receptor antagonists and inverse agonists can also be used occasionally to promote wakefulness or vigilance in shift workers, sleep deprivation, post anesthesia grogginess, drowsiness as a side effect from a medication, military use and the like.
  • histamine H3 receptor antagonists and inverse agonists have been shown to improve cognitive performance in various animal models (Hancock and Fox in Milestones in Drug Therapy, ed. Buccafusco, 2003). These compounds can be used as pro-cognitive agents and can increase vigilance. Therefore, histamine H3 receptor antagonists and inverse agonists can be used in aging or degenerative disorders in which vigilance, attention and memory are impaired, for example, as in Alzheimer's disease or other dementias.
  • AD Alzheimer's disease
  • NMDA NMDA antagonists
  • Histamine H3 receptor antagonists and inverse agonists can be used to treat or prevent cognitive disorders (Passani et al. Trends Pharmacol. Sci. 2004, 25, 618-625), epilepsy (Vohora et al. Pharmacol. Biochem. Behav. 2001, 68, 735-741), depression (Perez-Garcia et al.
  • Histamine H3 receptor antagonists or inverse agonists can also be used as a novel therapeutic approach to restore cortical activation in comatose or brain-traumatized patients (Passani et al., Trends in Pharmacol. Sci. 2004, 25, 618-625).
  • epilepsy is a chronic neurological condition characterized by recurrent unprovoked seizures. In terms of their pattern of activity, seizures may be described as either partial (focal) or generalized. Partial seizures only involve a localized part of the brain, whereas generalized seizures involve the entire cortex. There are many different epilepsy syndromes, each presenting with its own unique combination of seizure type, typical age of onset, EEG findings, treatment, and prognosis.
  • Some common seizure syndromes include, for example, infantile spasms (West syndrome), childhood absence epilepsy, and benign focal epilepsy of childhood (Benign Rolandic epilepsy), juvenile myoclonic epilepsy, temporal lobe epilepsy, frontal lobe epilepsy and Lennox-Gastaut syndrome.
  • Compounds of the present invention can be used in combination with various known drugs.
  • compounds of the present invention can be used with one or more drugs that prevent seizures or reduce seizure frequency: these include carbamazepine (common brand name Tegretol), clobazam (Frisium), clonazepam (Klonopin), ethosuximide (Zarontin), felbamate (Felbatol), fosphenytoin (Cerebyx), flurazepam (Dalmane), gabapentin (Neurontin), lamotrigine (Lamictal), levetiracetam (Keppra), oxcarbazepine (Trileptal), mephenytoin (Mesantoin), phenobarbital (Luminal), phenytoin (Dilantin), pregabalin (Lyrica), primidone (Mysoline), sodium valproate (Epilim), tiagabine (Gabitril), topiramate (Topamax), valproate semisodium (Depakot
  • Drugs used only in the treatment of refractory status epilepticus include paraldehyde (Paral) and pentobarbital (Nembutal).
  • a histamine H3 receptor antagonist or inverse agonist can be used as the sole agent of treatment or can be used in combination with other agents.
  • Vohora et al. show that a histamine H3 receptor antagonist can work as an anti-epilepsy, antiseizure drug and also showed effect with sub-effective doses of the H3 receptor antagonist in combination with sub-effective doses of known anti-epileptic drugs (Vohora et al. Pharmacol. Biochem. Behav. 2001, 68, 735-741).
  • Perez-Garcia et al. tested the ability of a histamine H3 receptor agonist and antagonist on experimental mouse models of anxiety (elevated plus-maze) and depression (forced swimming test). They found that while the compounds did not have a significant effect on the model of anxiety, an H3 receptor antagonist did have a significant dose-dependent effect in the model of depression. Thus, histamine H3 receptor antagonists or inverse agonists can have antidepressant effects.
  • Clinical depression is a state of sadness or melancholia that has advanced to the point of being disruptive to an individual's social functioning and/or activities of daily living. Clinical depression affects about 16% of the population on at least one occasion in their lives. Clinical depression is currently the leading cause of disability in the U.S. as well as other countries, and is expected to become the second leading cause of disability worldwide (after heart disease) by the year 2020, according to the World Health Organization.
  • compounds of the present invention can be used in combination with various known drugs.
  • compounds of the present invention can be used with one or more of the drugs currently available that can relieve the symptoms of depression.
  • They include, for example, monoamine oxidase inhibitors (MAOIs) such as Nardil or Moclobemide (Manerix), tricyclic antidepressants, selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine (Prozac), paroxetine (Paxil), escitalopram (Lexapro), and sertraline (Zoloft), norepinephrine reuptake inhibitors such as reboxetine (Edronax), and serotonin-norepinephrine reuptake inhibitors (SNRIs) such as venlafaxine (Effexor) and duloxetine (Cymbalta).
  • MAOIs monoamine oxidase inhibitors
  • SSRIs selective serotonin reuptake inhibitor
  • histamine H3 receptor antagonists and inverse agonists can be used to treat or prevent attention deficit hyperactivity disorder (ADHD).
  • ADHD attention deficit hyperactivity disorder
  • Diagnostic and Statistical Manual of Mental Disorders-IV-TR ADHD is a developmental disorder that arises in childhood, in most cases before the age of 7 years, is characterized by developmentally inappropriate levels of inattention and/or hyperactive-impulsive behavior, and results in impairment in one or more major life activities, such as family, peer, educational, occupational, social, or adaptive functioning. ADHD can also be diagnosed in adulthood.
  • the first-line medications used to treat ADHD are mostly stimulants, which work by stimulating the areas of the brain responsible for focus, attention, and impulse control.
  • the use of stimulants to treat a syndrome often characterized by hyperactivity is sometimes referred to as a paradoxical effect, but there is no real paradox in that stimulants activate brain inhibitory and self-organizing mechanisms permitting the individual to have greater self -regulation.
  • the stimulants used include, for example, methylphenidate (sold as Ritalin, Ritalin SR and Ritalin LA), Metadate, Metadate ER, Metadate CD, Concerta, Focalin, Focalin XR or Methylin.
  • the stimulants also include, for example, amphetamines such dextroamphetamine, sold as
  • Dexedrine, Dexedrine Spansules, Adderall, and Adderall XR a trade name for a mixture of dextroamphetamine and laevoamphetamine salts, methamphetamine sold as Desoxyn, bupropion, a dopamine and norepinephrine reuptake inhibitor, marketed under the brand name Wellbutrin.
  • a non-stimulant medication to treat ADHD is Atomoxetine (sold as Strattera) a norepinephrine reuptake inhibitor.
  • Other drugs sometimes used for ADHD include, for example, benzphetamine, Provigil/Alertec/modafinil and clonidine.
  • a histamine H3 receptor antagonist was at least as effective as methylphenidate (Ritalin) (Hancock and Fox in Milestones in Drug Therapy, ed. Buccafusco, 2003).
  • Compounds of the present invention can be used in combination with various known drugs.
  • compounds of the present invention can be used with one or more of the drugs used to treat ADHD and related disorders.
  • histamine H3 receptor antagonists and inverse agonists can be used to treat or prevent schizophrenia.
  • Schizophrenia is a psychiatric diagnosis that describes a mental disorder characterized by impairments in the perception or expression of reality and by significant social or occupational dysfunction.
  • a person experiencing untreated schizophrenia is typically characterized as demonstrating disorganized thinking, and as experiencing delusions or auditory hallucinations.
  • the disorder is primarily thought to affect cognition, it can also contribute to chronic problems with behavior and emotion.
  • Schizophrenia is often described in terms of "positive” and "negative” symptoms. Positive symptoms include delusions, auditory hallucinations and thought disorder, and are typically regarded as manifestations of psychosis.
  • Negative symptoms are so named because they are considered to be the loss or absence of normal traits or abilities, and include features such as flat, blunted or constricted affect and emotion, poverty of speech and lack of motivation.
  • Some models of schizophrenia include formal thought disorder and planning difficulties in a third group, a "disorganization syndrome.”
  • the first line pharmacological therapy for schizophrenia is usually the use of antipsychotic medication.
  • Antipsychotic drugs are only thought to provide symptomatic relief from the positive symptoms of psychosis.
  • the newer atypical antipsychotic medications are usually preferred over older typical antipsychotic medications (such as chlorpromazine and haloperidol) due to their favorable side-effect profile. While the atypical antipsychotics are associated with less extra pyramidal side-effects and tardive dyskinesia than the conventional antipsychotics, some of the agents in this class (especially olanzapine and clozapine) appear to be associated with metabolic side effects such as weight gain, hyperglycemia and hypertriglyceridemia that must be considered when choosing appropriate pharmacotherapy.
  • Histamine H3 receptor antagonists or inverse agonists can be used to treat obesity
  • histamine H3 receptor antagonists have been investigated in various preclinical models of obesity and have shown to be effective in reducing food intake, reducing weight, and decreasing total body fat in mice (Hancock, et al. Eur. J. Pharmacol. 2004, 487, 183-197).
  • the most common drugs used for the treatment of obesity are sibutramine (Meridia) and orlistat (Xenical), both of which have limited effectiveness and significant side effects. Therefore, novel anti-obesity agents, such as histamine H3 receptor antagonists or inverse agonists, are needed.
  • Histamine H3 receptor antagonists or inverse agonists can also be used to treat upper airway allergic responses (U.S. Pat. Nos. 5,217,986; 5,352,707 and 5,869,479) including allergic rhinitis and nasal congestion. Allergic rhinitis is a frequently occurring chronic disease that affects a large number of people. Recent analysis of histamine H3 receptor expression in the periphery by quantitative PCR revealed that H3 receptor mRNA is abundantly expressed in human nasal mucosa (Varty et al. Eur. J. Pharmacol. 2004, 484, 83-89).
  • histamine H3 receptor antagonists in a cat model of nasal decongestion, a combination of histamine H3 receptor antagonists with the HI receptor antagonist chlorpheniramine resulted in significant nasal decongestion without the hypertensive effect seen with adrenergic agonists.
  • histamine H3 receptor antagonists or inverse agonists can be used alone or in combination with HI receptor blockage for the treatment of allergic rhinitis and nasal congestion.
  • Histamine H3 receptor antagonists or inverse agonists have therapeutic potential for the treatment of pain (Medhurst et al. Biochemical Pharmacology (2007), 73(8), 1182-1194).
  • Itch or pruritus, is the unpleasant sensation that leads to a desire to scratch (for reviews, see Journal of Investigative Dermatology (2006) 126: 1705-1718; and Lancet (2003) 361 : 690- 94). It is a common and distressing symptom in a variety of conditions and diseases. Itch typically occurs in peripheral diseases such as allergic conjunctivitis, allergic rhinitis, hemorrhoids, atopic dermatitis, allergic dermatitis, acute and chronic urticaria (hives), psoriasis and dermatoses of fungal, allergic and non-allergic origin.
  • peripheral diseases such as allergic conjunctivitis, allergic rhinitis, hemorrhoids, atopic dermatitis, allergic dermatitis, acute and chronic urticaria (hives), psoriasis and dermatoses of fungal, allergic and non-allergic origin.
  • Itching can also be a major symptom of many systemic diseases such as, Hodgkin's disease, chronic renal failure, polycythemia vera, hyperthyroidism, malignancy, infection, chronic cholestatic liver disease and end-stage renal disease, and cholestasis.
  • systemic diseases such as, Hodgkin's disease, chronic renal failure, polycythemia vera, hyperthyroidism, malignancy, infection, chronic cholestatic liver disease and end-stage renal disease, and cholestasis.
  • senile itch without an obvious cause, except perhaps xerosis, occurs in more than half of the population aged 70 years. In all cases chronic severe generalized itch can be disabling.
  • diseases or conditions associated with itch further include primary biliary cirrhosis (PBC), primary sclerosis cholangitis (PSC), chronic renal disease, epidural morphine, pregnancy, diabetes mellitus, thyroid illness, hyperparathyroidism, iron deficiency anemia, viral infection, aquagenic pruritus, and psychogenic itch. Itch causes sufferers to scratch, leading to skin damage, increased risk of skin infection, and worsening of inflammation. However, despite the prevalence of this clinically important symptom, the pathogenesis of itch is not well understood, and treatment options are limited (Paus, R., et al., J. Clin. Invest. 2006, 116: 1174-1185).
  • H3R histamine 3 receptor
  • the present invention provides a method of preventing and/or treating itch in an individual in need thereof by administering a therapeutically effective amount of a compound or agent that modulates the H3R.
  • peripherally restricted inhibitors of H3R are capable of mediating the inhibition of itch. Accordingly, screening for peripherally restricted inhibitors (i.e. antagonists), and the application of peripherally restricted inhibitors in the various embodiments of the invention is contemplated. Peripherally restricted compounds may be advantageous to the extent that the peripheral restriction reduces the CNS effects of H3R inhibition. Such effects may include, for example, wakefulness.
  • H3R inhibitors for example antagonists and inverse agonists, are peripherally restricted and may be assayed or screened based on their inability or reduced ability to inhibit H3R in the CNS (for example, in the brain).
  • peripherally restricted compounds of the present invention can be advantageous in the treatment of pruritus to the extent that the peripheral restriction reduces the CNS effects associated with H3R inhibition.
  • an individual suffering from pruritus can benefit from a treatment with a peripherally restricted compound of the present invention and yet not experience or experience to a less extent the CNS effects associated with H3R inhibition, such as, wakefulness.
  • the compound of the present invention is restricted in some manner from crossing the blood brain barrier. In another embodiment, the compound of the present invention is less than 100% restricted to the periphery. In one embodiment, the compound of the present invention has a brain:plasma ratio (B P) which is less than 1 in at least one species. In some embodiments, the species is a mouse. In some embodiments, the species is a C57BL6 mouse.
  • the H3R antagonist has a brain to plasma ratio of about 0.5 or less. In another embodiment, the H3R antagonist has a brain to plasma ratio of about 0.4 or less. In another embodiment, the H3R antagonist has a brain to plasma ratio of about 0.3 or less. In another embodiment, the H3R antagonist has a brain to plasma ratio of about 0.2 or less. In another embodiment, the H3R antagonist has a brain to plasma ratio of about 0.15 or less. In another embodiment, the H3R antagonist has a brain to plasma ratio of about 0.1 or less. In another embodiment, the H3R antagonist has a brain to plasma ratio of about 0.01 or less. In another embodiment, the H3R antagonist has a brain to plasma ratio of about 0.1 to about 0.001.
  • the invention comprises a method for treating or preventing itching or the symptoms thereof in an individual wherein an inhibitor of H3R is administered to the individual.
  • the invention disclosed herein is suitable for the prevention and/or treatment of itch that is associated with a disease or disorder selected from eczema, atopic eczematous dermatitis, seborrheic dermatitis, atopic dermatitis, contact dermatitis, irritant dermatitis, xerosis (dry skin), psoriasis, a fungal infection, athlete's foot, a yeast infection, diaper rash, vaginal itch, parasitic infections, parasitic infestations including scabies and lice, lichen planus, lichen simplex, lichen simplex chronicus, lichen sclerosis, itch secondary to medications, senile itch, uremia, idiopathic itch, itch associated with liver cirrhosis, itch associated with inflammation, itch associated with allergies, itch associated with cancer, itch associated with chemotherapy, itch associated with kidney disease, itch associated with haemodialysis, burns, scalds, sunburn
  • Itching can be associated with various types of cancers such as Hodgkin's disease, Lymphoma, Leukemia, Kaposi's sarcoma, AIDs, liver metastases, and renal failure, and may be associated with some antibiotics. Acute itching, during the infusion of chemotherapy could be an early sign of a hypersensitivity reaction.
  • Chemotherapy medications commonly associated with risk of allergic reactions include: L-asparaginase, paclitaxel, docetaxel, teniposide, procarbazine, and cytarabine.
  • One aspect of the present invention pertains to methods of inducing wakefulness in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating a histamine H3 receptor-associated disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating a histamine H3 receptor-associated disorder selected from the group: a cognitive disorder, epilepsy, brain trauma, depression, obesity, disorders of sleep and wakefulness, narcolepsy, shift-work sleep disorder, cataplexy, hypersomnia, somnolence syndrome, jet lag, sleep apnea, excessive daytime sleepiness, attention deficit hyperactivity disorder (ADHD), schizophrenia, allergies, allergic responses in the upper airway, allergic rhinitis, nasal congestion, dementia, Alzheimer's disease, pain, and pruritus in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • a cognitive disorder selected from the group: a cognitive disorder, epilepsy, brain trauma, depression, obesity, disorders of sleep and wakefulness, narcolepsy, shift-work sleep disorder, cataplexy, hypersomnia, somnolence syndrome, jet lag, sleep apnea, excessive day
  • One aspect of the present invention pertains to methods for treating a cognitive disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating epilepsy in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating a disorder of sleep and wakefulness in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating narcolepsy in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating shift-work sleep disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating cataplexy in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating jet lag in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating sleep apnea in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating excessive daytime sleepiness in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating attention deficit hyperactivity disorder in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating schizophrenia in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating pain in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating pruritus in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof.
  • One aspect of the present invention pertains to methods for treating pruritus in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition thereof, wherein pruritus is associated with a disorder selected from eczema, atopic eczematous dermatitis, seborrheic dermatitis, atopic dermatitis, contact dermatitis, irritant dermatitis, xerosis (dry skin), psoriasis, a fungal infection, athlete's foot, a yeast infection, diaper rash, vaginal itch, parasitic infections, parasitic infestations including scabies and lice, lichen planus, lichen simplex, lichen simplex chronicus, lichen sclerosis, itch secondary to medications, senile itch, uremia, idiopathic itch, itch associated with liver cirrhosis, itch associated with inflammation, itch associated with allergies, itch associated with cancer,
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for inducing wakefulness.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of a histamine H3 receptor-associated disorder.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of a disorder selected from the group: a cognitive disorder, epilepsy, brain trauma, depression, obesity, disorders of sleep and wakefulness, narcolepsy, shift- work sleep disorder, cataplexy, hypersomnia, somnolence syndrome, jet lag, sleep apnea, excessive daytime sleepiness, attention deficit hyperactivity disorder (ADHD), schizophrenia, allergies, allergic responses in the upper airway, allergic rhinitis, nasal congestion, dementia, Alzheimer's disease, pain, and pruritus.
  • a cognitive disorder selected from the group: a cognitive disorder, epilepsy, brain trauma, depression, obesity, disorders of sleep and wakefulness, narcolepsy, shift- work sleep disorder, cataplexy, hypersomnia, somnolence syndrome, jet lag, sleep apnea, excessive daytime sleepiness, attention deficit hyperactivity disorder (ADHD), schizophrenia, allergies,
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of a cognitive disorder.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of epilepsy.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of a disorder of sleep and wakefulness.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of narcolepsy.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of shift-work sleep disorder.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of cataplexy.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of jet lag.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of sleep apnea.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of excessive daytime sleepiness.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of attention deficit hyperactivity disorder.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of schizophrenia.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of pain.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of pruritus.
  • One aspect of the present invention pertains to the use of a compound of the present invention or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment of pruritus, wherein pruritus is associated with a disorder selected from eczema, atopic eczematous dermatitis, seborrheic dermatitis, atopic dermatitis, contact dermatitis, irritant dermatitis, xerosis (dry skin), psoriasis, a fungal infection, athlete's foot, a yeast infection, diaper rash, vaginal itch, parasitic infections, parasitic infestations including scabies and lice, lichen planus, lichen simplex, lichen simplex chronicus, lichen sclerosis, itch secondary to medications, senile itch, uremia, idiopathic itch, itch associated with liver cirrhosis, itch associated with inflammation, itch associated with allergies, itch associated with cancer, itch associated with chemotherapy, itch associated with
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of inducing wakefulness.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of a histamine H3 receptor- associated disorder.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of a histamine H3 receptor- associated disorder selected from the group: a cognitive disorder, epilepsy, brain trauma, depression, obesity, disorders of sleep and wakefulness, narcolepsy, shift-work sleep disorder, cataplexy, hypersomnia, somnolence syndrome, jet lag, sleep apnea, excessive daytime sleepiness, attention deficit hyperactivity disorder (ADHD), schizophrenia, allergies, allergic responses in the upper airway, allergic rhinitis, nasal congestion, dementia, Alzheimer's disease, pain, and pruritus.
  • a cognitive disorder selected from the group: a cognitive disorder, epilepsy, brain trauma, depression, obesity, disorders of sleep and wakefulness, narcolepsy, shift-work sleep disorder, cataplexy, hypersomnia, somnolence syndrome, jet lag, sleep apnea, excessive daytime sleepiness, attention deficit hyperactivity disorder (ADHD), schizophrenia
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of a cognitive disorder.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of epilepsy.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of a disorder of sleep and wakefulness.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of narcolepsy.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of shift-work sleep disorder.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of cataplexy.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of jet lag.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of sleep apnea.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of excessive daytime sleepiness.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of attention deficit hyperactivity disorder.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of schizophrenia.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of pain.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of pruritus.
  • One aspect of the present invention pertains to a compound of the present invention or a pharmaceutical composition thereof for use in a method of treatment of pruritus, wherein the pruritus is associated with a disorder selected from eczema, atopic eczematous dermatitis, seborrheic dermatitis, atopic dermatitis, contact dermatitis, irritant dermatitis, xerosis (dry skin), psoriasis, a fungal infection, athlete's foot, a yeast infection, diaper rash, vaginal itch, parasitic infections, parasitic infestations including scabies and lice, lichen planus, lichen simplex, lichen simplex chronicus, lichen sclerosis, itch secondary to medications, senile itch, uremia, idiopathic itch, itch associated with liver cirrhosis, itch associated with inflammation, itch associated with allergies, itch associated with cancer, itch associated with chemotherapy, itch associated with kidney disease,
  • a further aspect of the present invention pertains to pharmaceutical compositions comprising one or more compounds as described herein and one or more pharmaceutically acceptable carriers. Some embodiments pertain to pharmaceutical compositions comprising a compound of the present invention and a pharmaceutically acceptable carrier.
  • Some embodiments of the present invention include a method of producing a pharmaceutical composition comprising formulating at least one compound according to any of the compound embodiments disclosed herein and a pharmaceutically acceptable carrier.
  • Some embodiments of the present invention include a method of producing a pharmaceutical composition
  • a method of producing a pharmaceutical composition comprising admixing at least one compound according to any of the compound embodiments disclosed herein and a pharmaceutically acceptable carrier.
  • Formulations may be prepared by any suitable method, typically by uniformly mixing the active compound(s) with liquids or finely divided solid carriers, or both, in the required proportions and then, if necessary, forming the resulting mixture into a desired shape.
  • excipients such as binding agents, fillers, acceptable wetting agents, tabletting lubricants and disintegrants may be used in tablets and capsules for oral
  • Liquid preparations for oral administration may be in the form of solutions, emulsions, aqueous or oily suspensions and syrups.
  • the oral preparations may be in the form of dry powder that can be reconstituted with water or another suitable liquid vehicle before use. Additional additives such as suspending or emulsifying agents, non-aqueous vehicles (including edible oils), preservatives and flavorings and colorants may be added to the liquid preparations.
  • Parenteral dosage forms may be prepared by dissolving the compound of the invention in a suitable liquid vehicle and filter sterilizing the solution before filling and sealing an appropriate vial or ampule. These are just a few examples of the many appropriate methods well known in the art for preparing dosage forms.
  • a compound of the present invention can be formulated into pharmaceutical compositions using techniques well known to those in the art. Suitable pharmaceutically- acceptable carriers, outside those mentioned herein, are known in the art; for example, see Remington, The Science and Practice of Pharmacy, 20* Edition, 2000, Lippincott Williams & Wilkins, (Editors: Gennaro et al.).
  • a compound of the invention may, in an alternative use, be administered as a raw or pure chemical, it is preferable however to present the compound or active ingredient as a pharmaceutical formulation or composition further comprising a pharmaceutically acceptable carrier.
  • the invention thus further provides pharmaceutical formulations comprising a compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate or derivative thereof together with one or more pharmaceutically acceptable carriers thereof and/or prophylactic ingredients.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not overly deleterious to the recipient thereof.
  • Transdermal patches dispense a drug at a controlled rate by presenting the drug for absorption in an efficient manner with a minimum of degradation of the drug.
  • transdermal patches comprise an impermeable backing layer, a single pressure sensitive adhesive and a removable protective layer with a release liner.
  • the compounds of the invention may thus be placed into the form of pharmaceutical formulations and unit dosages thereof and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, gels or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use.
  • Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
  • the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
  • dosage units are capsules, tablets, powders, granules or a suspension, with conventional additives such as lactose, mannitol, corn starch or potato starch; with binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators such as corn starch, potato starch or sodium carboxymethyl-cellulose; and with lubricants such as talc or magnesium stearate.
  • the active ingredient may also be administered by injection as a composition wherein, for example, saline, dextrose or water may be used as a suitable pharmaceutically acceptable carrier.
  • active ingredient is defined in the context of a "pharmaceutical composition” and refers to a component of a pharmaceutical composition that provides the primary pharmacological effect, as opposed to an "inactive ingredient” which would generally be recognized as providing no pharmaceutical benefit.
  • the dose when using the compounds of the present invention can vary within wide limits and as is customary and is known to the physician, it is to be tailored to the individual conditions in each individual case. It depends, for example, on the nature and severity of the illness to be treated, on the condition of the patient, on the compound employed or on whether an acute or chronic disease state is treated or prophylaxis is conducted or on whether further active compounds are administered in addition to the compounds of the present invention.
  • Representative doses of the present invention include, but not limited to, about 0.001 mg to about 5000 mg, about 0.001 mg to about 2500 mg, about 0.001 mg to about 1000 mg, 0.001 mg to about 500 mg, 0.001 mg to about 250 mg, about 0.001 mg to 100 mg, about 0.001 mg to about 50 mg and about 0.001 mg to about 25 mg.
  • Multiple doses may be administered during the day, especially when relatively large amounts are deemed to be needed, for example 2, 3 or 4 doses. Depending on the individual and as deemed appropriate from the patient' s physician or caregiver it may be necessary to deviate upward or downward from the doses described herein.
  • the amount of active ingredient, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will ultimately be at the discretion of the attendant physician or clinician.
  • a model system typically an animal model
  • these extrapolations may merely be based on the weight of the animal model in comparison to another, such as a mammal, preferably a human, however, more often, these extrapolations are not simply based on weights, but rather incorporate a variety of factors.
  • compositions of this invention are selected in accordance with a variety factors as cited above.
  • the actual dosage regimen employed may vary widely and therefore may deviate from a preferred dosage regimen and one skilled in the art will recognize that dosage and dosage regimen outside these typical ranges can be tested and, where appropriate, may be used in the methods of this invention.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations.
  • the daily dose can be divided, especially when relatively large amounts are administered as deemed appropriate, into several, for example 2, 3 or 4 part administrations. If appropriate, depending on individual behavior, it may be necessary to deviate upward or downward from the daily dose indicated.
  • the compounds of the present invention can be administrated in a wide variety of oral and parenteral dosage forms. It will be obvious to those skilled in the art that the following dosage forms may comprise, as the active component, either a compound of the invention or a pharmaceutically acceptable salt, solvate or hydrate of a compound of the invention.
  • a suitable pharmaceutically acceptable carrier can be either solid, liquid or a mixture of both.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories and dispersible granules.
  • a solid carrier can be one or more substances which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • the carrier is a finely divided solid which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted to the desire shape and size.
  • the powders and tablets may contain varying percentage amounts of the active compound.
  • a representative amount in a powder or tablet may contain from 0.5 to about 90 percent of the active compound; however, an artisan would know when amounts outside of this range are necessary.
  • Suitable carriers for powders and tablets are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter and the like.
  • the term "preparation" is intended to include the formulation of the active compound with encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it.
  • cachets and lozenges are included. Tablets, powders, capsules, pills, cachets and lozenges can be used as solid forms suitable for oral administration.
  • a low melting wax such as an admixture of fatty acid glycerides or cocoa butter
  • the active component is dispersed homogeneously therein, as by stirring.
  • the molten homogenous mixture is then poured into convenient sized molds, allowed to cool and thereby to solidify.
  • Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Liquid form preparations include solutions, suspensions and emulsions, for example, water or water-propylene glycol solutions.
  • parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the compounds according to the present invention may thus be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi-dose containers with an added preservative.
  • the pharmaceutical compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • a suitable vehicle e.g. sterile, pyrogen-free water
  • Aqueous formulations suitable for oral use can be prepared by dissolving or suspending the active component in water and adding suitable colorants, flavors, stabilizing and thickening agents, as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • viscous material such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well-known suspending agents.
  • solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • liquid forms include solutions, suspensions and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents and the like.
  • the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • Formulations suitable for topical administration in the mouth include lozenges comprising active agent in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the formulations may be provided in single or multi-dose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray, this may be achieved for example by means of a metering atomizing spray pump.
  • Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the active ingredient is provided in a pressurized pack with a suitable propellant.
  • aerosol formulation in which the active ingredient is provided in a pressurized pack with a suitable propellant.
  • the compounds of the present invention or pharmaceutical compositions comprising them are administered as aerosols, for example as nasal aerosols or by inhalation, this can be carried out, for example, using a spray, a nebulizer, a pump nebulizer, an inhalation apparatus, a metered inhaler or a dry powder inhaler.
  • Pharmaceutical forms for administration of the compounds of the present invention as an aerosol can be prepared by processes well known to the person skilled in the art.
  • solutions or dispersions of the compounds of the present invention in water, water/alcohol mixtures or suitable saline solutions can be employed using customary additives, for example benzyl alcohol or other suitable preservatives, absorption enhancers for increasing the bioavailability, solubilizers, dispersants and others and, if appropriate, customary propellants, for example include carbon dioxide,
  • CFCs such as, dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane; and the like.
  • the aerosol may conveniently also contain a surfactant such as lecithin.
  • the dose of drug may be controlled by provision of a metered valve.
  • the compound In formulations intended for administration to the respiratory tract, including intranasal formulations, the compound will generally have a small particle size for example of the order of 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization. When desired, formulations adapted to give sustained release of the active ingredient may be employed.
  • the active ingredients may be provided in the form of a dry powder, for example, a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form for example in capsules or cartridges of, e.g., gelatin, or blister packs from which the powder may be administered by means of an inhaler.
  • the pharmaceutical preparations are preferably in unit dosage forms.
  • the preparation is subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • Tablets or capsules for oral administration and liquids for intravenous administration are preferred compositions.
  • the compounds according to the invention may optionally exist as pharmaceutically acceptable salts including pharmaceutically acceptable acid addition salts prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids.
  • Representative acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, dichloroacetic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic,
  • the acid addition salts may be obtained as the direct products of compound synthesis.
  • the free base may be dissolved in a suitable solvent containing the appropriate acid and the salt isolated by evaporating the solvent or otherwise separating the salt and solvent.
  • the compounds of this invention may form solvates with standard low molecular weight solvents using methods known to the skilled artisan.
  • Pro-drugs refers to compounds that have been modified with specific chemical groups known in the art and when administered into an individual these groups undergo biotransformation to give the parent compound. Pro-drugs can thus be viewed as compounds of the invention containing one or more specialized non-toxic protective groups used in a transient manner to alter or to eliminate a property of the compound. In one general aspect, the "pro-drug” approach is utilized to facilitate oral absorption.
  • T. Higuchi and V. Stella Prodrugs as Novel Delivery Systems Vol. 14 of the A.C.S. Symposium Series; and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
  • Some embodiments of the present invention include a method of producing a pharmaceutical composition for "combination-therapy" comprising admixing at least one compound according to any of the compound embodiments disclosed herein, together with at least one known pharmaceutical agent as described herein and a pharmaceutically acceptable carrier.
  • histamine H3 receptor modulators are utilized as active ingredients in a pharmaceutical composition, these are not intended for use only in humans, but in other non-human mammals as well. Indeed, recent advances in the area of animal health-care mandate that consideration be given for the use of active agents, such as histamine H3 receptor modulators, for the treatment of an H3-associated disease or disorder in companionship animals (e.g., cats, dogs, etc.) and in livestock animals (e.g., cows, chickens, fish, etc.) Those of ordinary skill in the art are readily credited with understanding the utility of such compounds in such settings.
  • active agents such as histamine H3 receptor modulators
  • the dosage forms described herein may comprise, as the active component, either a compound described herein or a pharmaceutically acceptable salt or as a solvate or hydrate thereof.
  • various hydrates and solvates of the compounds described herein and their salts will find use as intermediates in the manufacture of pharmaceutical compositions. Typical procedures for making and identifying suitable hydrates and solvates, outside those mentioned herein, are well known to those in the art; see for example, pages 202-209 of K.J. Guillory, "Generation of Polymorphs, Hydrates, Solvates, and Amorphous Solids," in: Polymorphism in Pharmaceutical Solids, ed. Harry G. England, Vol.
  • one aspect of the present invention pertains to methods of administering hydrates and solvates of compounds described herein and/or their pharmaceutical acceptable salts, that can be isolated and characterized by methods known in the art, such as, thermogravimetric analysis (TGA), TGA-mass spectroscopy, TGA-Infrared spectroscopy, powder X-ray diffraction (XRPD), Karl Fisher titration, high resolution X-ray diffraction, and the like.
  • TGA thermogravimetric analysis
  • TGA-mass spectroscopy TGA-Infrared spectroscopy
  • powder X-ray diffraction (XRPD) powder X-ray diffraction
  • Karl Fisher titration high resolution X-ray diffraction
  • the present disclosure includes all isotopes of atoms occurring in the present compounds, intermediates, salts and crystalline forms thereof.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • One aspect of the present invention includes every combination of one or more atoms in the present compounds, intermediates, salts, and crystalline forms thereof that is replaced with an atom having the same atomic number but a different mass number.
  • One such example is the replacement of an atom that is the most naturally abundant isotope, such as H or 12 C, found in one the present compounds, intermediates, salts, and crystalline forms thereof, with a different atom that is not the most
  • isotopes of hydrogen include 2 H (deuterium) and 3 H (tritium).
  • isotopes of carbon include n C, 13 C, and 14 C.
  • Isotopes of oxygen include O, O, and C.
  • An isotope of fluorine includes 18 F.
  • An isotope of sulfur includes 35 S.
  • An isotope of chlorine includes 36 C1.
  • Isotopes of bromine include 75 Br, 76 Br, 77 Br, and 82 Br.
  • Isotopes of iodine include 123 I, 124 1, 125 I, and 131 I.
  • compositions such as, those prepared during synthesis, preformulation, and the like, and pharmaceutical compositions, such as, those prepared with the intent of using in a mammal for the treatment of one or more of the disorders described herein, comprising one or more of the present compounds, intermediates, salts, and crystalline forms thereof, wherein the naturally occurring distribution of the isotopes in the composition is perturbed.
  • compositions and pharmaceutical compositions comprising compounds as described herein wherein the compound is enriched at one or more positions with an isotope other than the most naturally abundant isotope.
  • Another object of the present invention relates to radio-labeled compounds of the present invention that would be useful not only in radio-imaging but also in assays, both in vitro and in vivo, for localizing and quantitating the histamine H3 receptor in tissue samples, including human and for identifying histamine H3 receptor ligands by inhibition binding of a radio-labeled compound. It is a further object of this invention to develop novel H3 receptor assays of which comprise such radio-labeled compounds.
  • the present invention embraces isotopically-labeled compounds of the present invention.
  • Isotopically or radio-labeled compounds are those which are identical to compounds disclosed herein, but for the fact that one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number most commonly found in nature.
  • Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 2 H (also written as D for deuterium), 3 H (also written as T for tritium), n C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 18 F, 35 S, 36 C1, 75 Br, 76 Br, 77 Br, 82 Br,
  • radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro histamine H3 receptor labeling and competition assays, compounds that 3 14 82 125 131 35
  • a “radio-labeled " or “labeled compound” is a compound of Formula (la), (Ic), (Ie), (Ig), (Ii), (lie), (He), (Ilg), (Hi), (Ilk), (Urn), (IIo), (Ilq), (lis), (IIu), (II w), and (Hy) that has incorporated at least one radionuclide; in some embodiments the
  • radionuclide is selected from the group consisting of H, C, I , S and Br.
  • isotopically-labeled compounds of the present invention are useful in compound and/or substrate tissue distribution assays.
  • the radionuclide 3 H and/or 14 C isotopes are useful in these studies.
  • substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability
  • Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Drawings and Examples infra, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent. Other synthetic methods that are useful are discussed infra. Moreover, it should be understood that all of the atoms represented in the compounds of the invention can be either the most commonly occurring isotope of such atoms or the scarcer radio-isotope or nonradioactive isotope.
  • Synthetic methods for incorporating radio-isotopes into organic compounds are applicable to compounds of the invention and are well known in the art. These synthetic methods, for example, incorporating activity levels of tritium into target molecules, are as follows:
  • Tritium Gas Exposure Labeling This procedure involves exposing precursors containing exchangeable protons to tritium gas in the presence of a suitable catalyst.
  • Synthetic methods for incorporating activity levels of 125 I into target molecules include: A. Sandmeyer and like reactions: This procedure transforms an aryl amine or a heteroaryl amine into a diazonium salt, such as a diazonium tetrafluoroborate salt and subsequently to 125 I labeled compound using Na 125 I. A represented procedure was reported by Zhu, G-D. and co-workers in J. Org. Chem., 2002, 67, 943-948.
  • Aryl and heteroaryl bromide exchange with 125 I This method is generally a two step process.
  • the first step is the conversion of the aryl or heteroaryl bromide to the corresponding tri-alkyltin intermediate using for example, a Pd catalyzed reaction [i.e. Pd(Ph 3 P) 4 ] or through an aryl or heteroaryl lithium, in the presence of a tri-alkyltinhalide or hexaalkylditin [e.g. ,
  • a radiolabeled histamine H3 receptor compound of Formula (la) can be used in a screening assay to identify/evaluate compounds.
  • a newly synthesized or identified compound ⁇ i.e. , test compound) can be evaluated for its ability to reduce binding of the "radio-labeled compound of Formula (la)" to the H3 receptor. Accordingly, the ability of a test compound to compete with the "radio-labeled compound of Formula (la)" for the binding to the histamine H3 receptor directly correlates to its binding affinity.
  • the labeled compounds of the present invention bind to the histamine H3 receptor.
  • the labeled compound has an IC 50 less than about 500 ⁇
  • the labeled compound has an IC 50 less than about 100 ⁇
  • the labeled compound has an IC 50 less than about 10 ⁇
  • the labeled compound has an IC 50 less than about 1 ⁇
  • the labeled inhibitor has an IC 50 less than about 0.1 ⁇ .
  • Example 1 Syntheses of compounds of the present invention. Illustrated syntheses for compounds of the present invention are shown in Figures 1 through 3 where the symbols have the same definitions as used throughout this disclosure.
  • Microwave irradiations were carried out using a Smith SynthesizerTM or an Emrys OptimizerTM (Biotage). Thin-layer chromatography (TLC) was performed on silica gel 60 F 2 5 4 (Merck), preparatory thin-layer chromatography (prep TLC) was preformed on PK6F silica gel 60 A 1 mm plates (Whatman) and column chromatography was carried out on a silica gel column using Kieselgel 60, 0.063-0.200 mm (Merck). Evaporation was done under reduced pressure on a Biichi rotary evaporator.
  • LCMS spec HPLC -pumps: LC-10AD VP, Shimadzu Inc.; HPLC system controller: SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-10A VP, Shimadzu Inc; Autosampler: CTC HTS, PAL, Leap Scientific; Mass spectrometer: API 150EX with Turbo Ion Spray source, AB/MDS Sciex; Software: Analyst 1.2.
  • Example 1.1 Preparation of Intermediate (/f)-6-(4-(2-(2-Methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline.
  • Step A Preparation of (/f)-l-(4-Bromophenethyl)-2-methylpyrrolidine.
  • Step B Preparation of (/f)-4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenylboronic Acid.
  • Step C Preparation of Intermediate (/f)-6-(4-(2-(2-Methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline.
  • 6-bromo-l,2,3,4-tetrahydroisoquinoline hydrochloride (2.00 g, 8.05 mmol)
  • (R)-4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenylboronic acid 2.063 g, 8.85 mmol
  • tetrakis(triphenylphosphine)palladium (0) 0.279 g, 0.241 mmol
  • benzene 30.00 mL
  • ethanol (10.00 mL)
  • 2.0 M aqueous solution of sodium bicarbonate (8.05 mL, 16.09 mmol).
  • the reaction mixture was refluxed for 6 h. Upon completion, water was added and the mixture was extracted with ethyl acetate. The organic layer was washed with brine, dried over Na 2 S0 4 , and concentrated. The residue was taken up in 1 M HC1 solution and washed with ethyl acetate. The aqueous layer was basified with 10% aqueous NaOH to pH ⁇ l 1, extracted with ethyl acetate, and concentrated. The residue was purified by silica gel column, eluting with 5-10% 2.0 M ammonia in methanol DCM to give a yellow solid (1.20 g).
  • Example 1.2 Preparation of (/f)-l-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-3,4- dihydroisoquinolin-2(l/ )-yl)-2-(l/ -tetrazol-5-yl)ethanone (Compound 1).
  • Example 1.3 Preparation of (/f)-4-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-3,4- dihydroisoquinolin-2(l/ )-yl)-4-oxobutanoic Acid (Compound 2).
  • Example 1.4 Preparation of (/f)-2,2,3,3-tetrafluoro-4-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-4-oxobutanoic acid (Compound 8).
  • Example 1.5 Preparation of (/f)-3,3-Dimethyl-5-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-5-oxopentanoic Acid (Compound 3).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.4 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline dihydrochloride and 4,4-dimethyldihydro-2/f-pyran-2,6(3//)-dione.
  • Example 1.6 Preparation of (/f)-2,2-Dimethyl-5-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-5-oxopentanoic Acid (Compound 4).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.4 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline dihydrochloride and 3,3-dimethyldihydro-2/f-pyran-2,6(3H)-dione.
  • LCMS m/z 463.5 [M+H] + .
  • Example 1.7 Preparation of 3-Methyl-5-(6-(4-(2-((/f)-2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-5-oxopentanoic Acid (Compound 5).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.4 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline dihydrochloride and 4-methyldihydro-2/f-pyran-2,6(3//)-dione.
  • LCMS m/z 449.3 [M+H] + .
  • Example 1.8 Preparation of 3-hydroxy-5-(6-(4-(2-((/f)-2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-5-oxopentanoic acid (Compound 6).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.4 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline dihydrochloride and l ,4-oxathiane-2,6-dione.
  • LCMS m/z 453.5
  • Example 1.10 Preparation of (/f)-2,2-Dimethyl-4-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-4-oxobutanoic Acid (Compound 9).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.4 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline dihydrochloride and 3,3-dimethyldihydrofuran-2,5-dione.
  • LCMS m/z 449.6 [M+H] + .
  • Example 1.11 Preparation of (/f)-2-(2-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-3,4- dihydroisoquinolin-2(l/ )-yl)-2-oxoethoxy)acetic Acid (Compound 10).
  • Example 1.12 Preparation of (/f)-5-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-3,4- dihydroisoquinolin-2(l/ )-yl)-5-oxopentanoic Acid (Compound 11).
  • Example 1.14 Preparation of (S)-Ethyl 2-amino-5-(6-(4-(2-((/f)-2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-5-oxopentanoate (Compound 13).
  • Example 1.15 Preparation of (/f)-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-3,4- dihydroisoquinolin-2(l/ )-yl)(l/ -tetrazol-5-yl)methanone (Compound 14).
  • Example 1.17 Preparation of (fl)-Methyl 2-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-2-oxoacetate (Compound 16).
  • Example 1.18 Preparation of (/f)-2-(l-(2-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)- 3,4-dihydroisoquinolin-2(l/ )-yl)-2-oxoethyl)cyclopentyl)acetic Acid (Compound 17).
  • Example 1.19 Preparation of (/f)-2-(l-(2-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)- 3,4-dihydroisoquinolin-2(l/ )-yl)-2-oxoethyl)cyclohexyl)acetic Acid (Compound 18).
  • Example 1.20 Preparation of (15,25)-2-(6-(4-(2-((K)-2-Methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclohexanecarboxylic Acid (Compound 19).
  • Example 1.22 Preparation of (R) -Methyl l-(6-(4-(2-(2-Methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclopropanecarboxylate (Compound 21).
  • Example 1.23 Preparation of (/f)-2-Methyl-l-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-2-(l/ -tetrazol-5-yl)propan-l-one (Compound 22).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.22 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline dihydrochloride and 2-methyl-2-(l/f-tetrazol-5-yl)propanoic acid.
  • Example 1.24 Preparation of (fl)-Methyl 3,3-dimethyl-5-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-5-oxopentanoate (Compound 23).
  • HC1 salt of the title compound was prepared in a similar manner to the one described in Example 1.13 using (R)-3,3-dimethyl-5-(6-(4-(2-(2-methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/f)-yl)-5-oxopentanoic acid.
  • LCMS m/z 477.4 [M+H] + .
  • Example 1.25 Preparation of (15,3tf )-Methyl 3-(6-(4-(2-((/f)-2-methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclohexanecarboxylate (Compound 24).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.22 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline bis(2,2,2-trifluoroacetate) and (lR,3S)-3-
  • Example 1.26 Preparation of (lr,4r)-Methyl 4-(6-(4-(2-((/f)-2-Methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclohexanecarboxylate (Compound 25).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.22 using (R)-6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-l,2,3,4- tetrahydroisoquinoline 3 ⁇ 4w(2,2,2-trifluoroacetate) and (l r,4r)-4-
  • Example 1.27 Preparation of (/f)-l-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)- l,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclopropanecarboxylic Acid (Compound 26).
  • Example 1.28 Preparation of (lr,4r)-4-(6-(4-(2-((/f)-2-Methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclohexanecarboxylic Acid (Compound 27).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.27 using (lr,4r)-methyl 4-(6-(4-(2-((R)-2-methylpyrrolidin-l- yl)ethyl)phenyl)- 1 ,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclohexanecarboxylate 2,2,2- trifluoroacetate.
  • Example 1.29 Preparation of (15,3K)-3-(6-(4-(2-((/f)-2-Methylpyrrolidin-l- yl)ethyl)phenyl)-l,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclohexanecarboxylic Acid (Compound 28).
  • TFA salt of the title compound was prepared in a similar manner to the one described in Example 1.27 using (1 S, 3R) -methyl 3-(6-(4-(2-((R)-2-methylpyrrolidin-l- yl)ethyl)phenyl)- 1 ,2,3,4-tetrahydroisoquinoline-2-carbonyl)cyclohexanecarboxylate 2,2,2- trifluoroacetate.
  • LCMS m/z 475.6 [M+H] + .
  • Example 1.30 Preparation of (/f)-4-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-l-oxo- 3,4-dihydroisoquinolin-2(l//)-yl)-4-oxobutanoic Acid (Compound 29).
  • Step A Preparation of (K)-6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-3,4- dihydroisoquinolin-l(2/ )-one.
  • Step B Preparation of (K)-4-(6-(4-(2-(2-Methylpyrrolidin-l-yl)ethyl)phenyl)-l-oxo-
  • Example 1.31 Preparation of (tf)-Ethyl 2-(l-(2-(6-(4-(2-(2-Methylpyrrolidin-l- yl)ethyl)phenyl)-3,4-dihydroisoquinolin-2(l/ )-yl)-2-oxoethyl)cyclohexyl)acetate
  • HC1 salt of the title compound was prepared in a similar manner to the one described in Example 1.13 using (R)-2-(l-(2-(6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)- 3,4-dihydroisoquinolin-2(l//)-yl)-2-oxoethyl)cyclohexyl)acetic acid.
  • LCMS m/z 431.7
  • the histamine receptor binding assay is conducted using standard laboratory procedures as described below.
  • Imetit is used as an assay positive control at varying concentrations.
  • the plate is incubated for 30 min at room temperature.
  • the assay is terminated by rapid filtration through a 96-well glass fiber filtration plate (GF/C) using a cell harvester (Perkin-Elmer). Captured membranes are washed three times with cold assay buffer and the plates are dried at 50 °C. 35 ⁇ of scintillation cocktail is added to each well and membrane-bound radioactivity is recorded using a TopCount 96-well plate scintillation counter (Perkin-Elmer).
  • Compounds that bind to GPCRs can be identified by their ability to displace a radiolabeled or fluorescently labeled tracer ligand from the receptor.
  • the tracer ligand can be a receptor agonist, inverse agonist or a neutral antagonist.
  • Receptor binding assays can be performed using either whole cells or membrane fractions prepared from such cells.
  • a typical radioligand binding assay cell membranes prepared from cells either endogenously or recombinantly expressing the desired receptor, are prepared and allowed to equilibrate with a mixture of a test ligand and tracer radioligand. Following equilibration, the cell membranes are captured by rapid filtration and washed with cold assay buffer to remove any unbound compounds. A liquid scintillant is typically added to the captured membranes and the samples are then counted on a scintillation counter. Compounds that bind to the receptor and displace the radioligand result in lower counts. For some receptors, the radiolabeled tracer ligand can be replaced by a fluorescently labeled ligand.
  • Homogeneous binding assays have also been developed for some receptors. These can involve the use of either radioactive tracer ligands (e.g., scintillation proximity assays) or fluorescent tracer ligands (fluorescence polarization binding assays).
  • radioactive tracer ligands e.g., scintillation proximity assays
  • fluorescent tracer ligands fluorescence polarization binding assays
  • Example 3.1 Human Histamine H3 Receptor Binding Assay - MDS Pharma Services (Taiwan).
  • Example 3.2 Rat Cortex H3 Receptor Radioligand Binding Assay.
  • Test compounds prepared from isolated, Sprague-Dawley rat cortex, which are known to abundantly express the rat H3 receptor, are plated into 96-well microliter plates at a concentration of 50 ⁇ g total membrane protein per well.
  • Test compounds prepared in assay buffer (50 mM Tris-HCl, pH7.4, with 5 mM EDTA) containing the selective H3 receptor agonist radioligand [ 3 H] iV-methylhistamine (final assay concentration 1.25 nM), are added to each well on the plate.
  • Test compound concentrations typically begin at 2 or 10 ⁇ (final assay concentration) and 1 :5 serial dilutions are prepared in order to generate 10-point dose -response curves for IC 50 and 3 ⁇ 4 determinations.
  • membranes are harvested into a PEI-washed filter plate (Whatman GF/C Unifilter) by rapid filtration (Perkin Elmer harvester) and washed three times with ice cold assay buffer (3 x 150 ⁇ ). Filter plates are then partially dried in a 50°C oven for 1-2 hours. Finally, the plate bottoms are sealed and BetaScint (Perkin Elmer 1205-440; 25 ⁇ per well) is added to each well.
  • IC 50 values can be converted to Kj values using the Cheng-Prusoff equation and the K d value for [ 3 H] N-methylhistamine at the rat H3 receptor (0.4 nM).
  • Example 3.3 Assays for Determination of GPCR Activation or Inhibition.
  • a G protein-coupled receptor When a G protein-coupled receptor is in its active state, either as a result of agonist ligand binding or constitutive activation, the receptor couples to G proteins, stimulating the release of GDP and subsequent binding of GTP to the G protein alpha subunit.
  • the activated G protein alpha subunit acts as a GTPase and slowly hydrolyzes the GTP back to GDP.
  • the non- hydrolyzable GTP analog, [ 35 S]GTPyS can be utilized to monitor binding of GTP to membrane- associated G proteins.
  • test compounds are incubated with receptor-expressing cell membranes in the presence of [ 35 S]GTPyS for 30 to 60 minutes. If the test compound is an agonist or an inverse agonist at the receptor of interest, enhanced or diminished uptake of
  • [ 35 S]GTPyS into the membrane-associated G-proteins will be detected.
  • a neutral antagonist with no intrinsic efficacy at the receptor, can be detected by its ability to prevent agonist- stimulated [ 35 S]GTPyS exchange.
  • the advantage of using [ 35 S]GTPyS binding to measure activation is that: (a) under appropriate assay conditions, it is generically applicable to all G protein-coupled receptors; (b) it is proximal at the membrane surface making it less likely to pick-up molecules which affect the rest of the G protein mediated intracellular signaling cascade.
  • recombinant human H3 receptor-expressing CHO-K1 cell membranes (90 ⁇ g membrane protein per well) are equilibrated in assay buffer (20 rriM HEPES (pH 7.4), 10 mM MgCl 2 , 100 mM NaCl, 1 mM DTT, 1 mM EDTA) containing test compounds and 10 ⁇ GDP for 20 minutes.
  • SPA beads sinintillation microsphere beads
  • the reaction is then initiated by the addition of 0.3 nM [ 35 S]GTPyS for 30 minutes. Plates are then sealed, centrifuged and counted in a scintillation counter (Packard TopCount).
  • the recombinantly-expressed histamine H3 receptor is known to be constitutively active.
  • test compounds that have either positive (agonist) or negative (inverse agonist) efficacy at the H3 receptor would be detected by their ability to increase or decrease basal levels of [ 35 S]GTPyS binding, respectively.
  • the assay may be modified to include a low dose (typically an EC 8 o-9o concentration) of a selective H3 receptor agonist such as N-methylhistamine.
  • a selective H3 receptor agonist such as N-methylhistamine.
  • antagonists ligands with no intrinsic receptor efficacy
  • GPCRs coupled to either Gs or Gi G-proteins modulate levels of intracellular cAMP and cAMP levels can be determined using a variety of commercially available assay kits. Examples of commonly used cAMP detection assays include FlashPlate® (New England Nuclear), HTRF® (Cisbio), c AMP- Screen® (Applied Biosystems), HitHunter® (Applied
  • GloSensor® Promega and numerous ELISA products. Most of these assays rely on the use of an anti-cAMP antibody to detect cAMP. Homogeneous time -resolved fluorescence assays (HTRF®, Cisbio) detect levels of cAMP in lysed cell preparations using a europium or terbium cryptate-labeled anti-cAMP antibody and fluorophore-labeled cAMP (cAMP-d2). In the absence of exogenous cAMP, the anti-cAMP antibody binds to cAMP-d2.
  • HTRF® Homogeneous time -resolved fluorescence assays
  • Photoexcitation of the cryptate donor results in a combination of cryptate emission at 620 nm and fluorescence resonance energy transfer (FRET) from the cryptate to the acceptor d2 fluorophore, which then fluoresces at 665 nm.
  • FRET fluorescence resonance energy transfer
  • the 620/665 nm emission ratio is monitored.
  • FRET is decreased and the 620/665 nm emission ratio therefore increases, providing a sensitive and accurate means to measure cAMP levels in biological assays.
  • H3R human H3 receptor
  • HEK293 cells expressing the human H3 receptor were suspended in PBS containing 100 ⁇ IBMX and plated into 384-well assay plates (Perkin Elmer Proxiplate 384-Plus; 15,000 cells per well; 5 ⁇ plating volume) and allowed to equilibrate for an hour.
  • Test compounds were serially diluted in 100% DMSO and then further diluted in PBS containing forskolin (2 ⁇ ). Test compounds (5 ⁇ ) were then added to the assay plate and the mixture was incubated for 1 hour.
  • HTRF assay reagents (Cisbio, Dynamic 2 cAMP Kit), cAMP-d2 and cryptate-labeled anti-cAMP antibody, are mixed with cell lysis buffer and added to the assay plate. After 1-hour incubation with these reagents, the assay plate was read on an HTRF-compatible microplate reader (Perkin Elmer EnVision or BMG Pherastar). A cAMP standard curve was included on each assay plate and HTRF emission ratios for test compounds were fit to this curve to generate accurate measures of cAMP levels in each test well. The observed H3R IC 50 values in the HTRF cAMP assay for several compounds of the present invention are shown in Table B.
  • the assay is modified to include a low dose of histamine (typically 20 nM) in the test compound buffer.
  • histamine typically 20 nM
  • the recombinantly-expressed histamine H3 receptor is known to be constitutively active.
  • test compounds that have either positive (agonist) or negative (inverse agonist) efficacies are detected by their ability to decrease or increase forskolin stimulated levels of cAMP, respectively. In this configuration, both inverse agonists and antagonists are efficiently detected.
  • Melanophores are skin cells found in lower vertebrates. They contain pigmented organelles termed melanosomes. Melanophores are able to redistribute these melanosomes along a microtubule network upon G-protein coupled receptor (GPCR) activation. The result of this pigment movement is an apparent lightening or darkening of the cells. In melanophores, the decreased levels of intracellular cAMP that result from activation of a Gi-coupled receptor such as the H3 receptor cause melanosomes to migrate to the center of the cell, resulting in a dramatic lightening in color.
  • GPCR G-protein coupled receptor
  • cAMP levels are then raised, following activation of a Gs-coupled receptor or addition of an H3 receptor inverse agonist, the melanosomes are re-dispersed and the cells appear dark again.
  • the response of the melanophores takes place within minutes of receptor activation and results in a simple, robust color change. The response can be easily detected using a conventional absorbance microplate reader or a modest video imaging system.
  • the H3 receptor is a constitutively active Gi-coupled receptor, melanophores expressing the H3 receptor will exhibit partial pigment aggregation in the resting state.
  • H3 receptor agonist or inverse agonist will cause either further pigment aggregation or dispersion, respectively.
  • a neutral antagonist at the H3 receptor would be detected by its ability to inhibit pigment aggregation stimulated by a selective H3 receptor agonist
  • IP inositol phosphates
  • diacylglycerol levels of intracellular inositol phosphates
  • IP levels can be determined in cells loaded with [ 3 H]-myo-inositol, resulting in the production of tritiated IP, which can be detected using standard radiometric techniques. IP levels can also be determined using an HTRF IP-One assay (Cisbio) which relies on an antibody to inositol monophosphate to detect IP.
  • Cytosolic calcium can be monitored using membrane-permeable dyes that become fluorescent when bound to calcium.
  • the most widely used instrument for conducting intracellular calcium release assays is the Fluorometric Imaging Plate Reader (FLIPR®,
  • the FLIPR instrument is able to simultaneously add test compounds to all wells on appropriate microplates and take real-time measurements of the fluorescence of calcium bound dye, allowing accurate measurement of intracellular calcium levels. Similar experiments can be performed with a number of alternate, commercially available instruments or by the imaging of single cells or small numbers of cells with a fluorescence microscope.
  • Intracellular calcium levels can also be measured in cells engineered to express calcium sensitive proteins such as aequorin.
  • Aequorin is a photoprotein isolated from jellyfish. While the calcium sensitive dyes used in FLIPR experiments require an excitation source in order to fluoresce, aequorin emits light in the presence of calcium without the need for an excitation source.
  • Gq G-proteins such as the H3 receptor
  • G15 and Gi6 promiscuous G- proteins
  • G15 and Gi6 promiscuous G-proteins
  • chimeric G-proteins may be used. These chimeric proteins typically utilize a Gq alpha protein in which approximately 5 amino acids at the carboxy-terminus are replaced with the corresponding amino acids from Gi alpha subunits. The resulting chimeric alpha subunit will recognize and be activated by Gi-coupled receptors but will signal though the Gq pathway to release intracellular calcium
  • H3 receptor agonists will stimulate and H3 receptor inverse agonists will inhibit, respectively, calcium release.
  • Antagonists and inverse agonists are typically detected by their ability to block the action of a selective H3 receptor agonist.
  • Activation of a GPCR typically results in receptor phosphorylation via a variety of kinases and then recruitment of ⁇ -arrestin from the cytosol.
  • ⁇ -arrestin proteins By monitoring the translocation of ⁇ -arrestin proteins from the cytosol to GPCRs in the cell membrane or quantitating the amount of receptor-arrestin complex formed in the cell, one can determine the extent of receptor activation.
  • the recruitment of arrestin and the formation of arrestin-GPCR complexes can result from both constitutive receptor activity and the influence of test compounds, with agonists promoting arrestin-receptor complexation.
  • Arrestin translocation assays In a typical arrestin translocation assay, cells expressing the receptor of interest are plated in transparent assay plates and allowed to fully adhere to the bottom of the wells. Test compounds are then added and incubated with the cells for up to an hour. The intracellular location of arrestin may be monitored by the use of cells recombinantly expressing a modified arrestin protein fused to a fluorescent protein such as green fluorescent protein (GFP). Alternatively, cells can be fixed and permeabilized, then treated with a fluorescently labeled anti- ⁇ - arrestin antibody.
  • GFP green fluorescent protein
  • Path Hunter® DiscoveRx
  • Tango® Invitrogen
  • the transcription factor is linked to the receptor via a protease-sensitive amino acid sequence and upon recruitment of protease-tagged arrestin, the transcription factor is cleaved from the receptor and translocates to the cell nucleus to initiate a transcriptional readout.
  • BRET bioluminescence resonance energy transfer
  • Activation of a GPCR typically results in changes in the activities of numerous protein signaling pathways within the cell. Some of these signaling pathways also lead to the modulations of intracellular concentrations of second messenger molecules such as cAMP, inositol phosphates, diacylglycerol and calcium. Many of these signaling pathways can ultimately result in a transcriptional response in the cell nucleus. Reporter gene assays take advantage of this response.
  • cells are engineered to express a reporter gene product such as ⁇ -galactosidase or luciferase with gene expression regulated by a promoter that is sensitive to the type of signaling expected from the receptor of interest.
  • cells expressing a receptor capable of modulating cAMP levels within the cell can be used to determine intracellular cAMP levels.
  • a Gs-coupled receptor or treatment with forskolin leading to the production of cAMP, will increase reporter gene expression.
  • Activation of a Gi coupled receptor such as the H3 receptor leading to reductions in cAMP production, will reduce levels of reporter gene expression.
  • agonists or inverse agonists would be detected by their ability to either decrease or increase forskolin-stimulated reporter gene expression, respectively.
  • a neutral antagonist would be detected by its ability to block the actions of a selective H3 agonist.
  • Example 4 Blockade of RAMH-Induced Drinking Assay.
  • histamine H3 receptor agonists such as (R)-a-methyl- histamine (RAMH)
  • RAMH histamine H3 receptor agonists
  • Blockade of RAMH-induced drinking can therefore be utilized as an in vivo assay for functional histamine H3 receptor inhibition activity.
  • male Sprague Dawley rats 250-350 g are housed three per cage and maintained under a reverse 12 h light cycle (lights off at 1130 h). At 1030 h on the day of test, rats are individually housed in new cages and food is removed. 120 min later, rats are administered test article (vehicle or histamine H3 receptor inhibitor, 0.3 mg kg PO).
  • RAMH vehicle or RAMH 3 mg/kg salt SC
  • 10 min after administration of RAMH weighed water bottles are placed in the cages, and drinking is allowed for 20 min. Water consumption is determined for each animal by weighing each bottle to the nearest 0.1 g. Data is expressed as percentage reduction in water intake according to the following formula:
  • mice Female C57BL/6 mice (5/group) were sensitized two times/week via epicutaneous administration of the allergen DNFB (0.3% in 4: 1 acetone:olive oil) on the rostral back skin.
  • mice On the 14th day after initial DNFB administration, mice were challenged again with epicutaneous DNFB; at 5h post-DNFB challenge, mice were dosed with Compound 1 at 1, 3, 10, and 30 mg/kg, IP. Scratching bouts were counted from SO- SO minutes post-compound dosing (See Figure 4). Compound 1 inhibited pruritus in a dose dependent fashion.
  • Example 6A Pharmacology Study of Compound 2 in male C57BL6 Mouse, Brain to Plasma Ratio Determination.
  • the analyte plasma concentrations were determined by LC/MS/MS.
  • Male C57BL6 Mice were dosed (IP) with Compound 2 at 3.00 and 30.0 mg kg (as a free base).
  • Mouse plasma and brain samples were taken at 0.5 hr (30 mg kg group only) and 1.0 hr post dose.
  • the plasma concentration was 402 + 202 ng/mL at 1 hr; the brain concentration was 5.37 + 2.72 ng/g at 1 hr.; and the brain to plasma ratio was 0.0112 at 1 hr.
  • the plasma concentrations were 259 + 78.0 ng/mL and 407 + 65.0 ng/mL at 0.5 hr and
  • Example 6B Pharmacology Study of Compound 3 in male C57BL6 Mouse, Brain to Plasma Ratio Determination.
  • the analyte plasma concentrations were determined by LC/MS/MS.
  • Male C57BL6 Mice were dosed (IP) with Compound 3 at 3.00 mg/kg (as a free acid).
  • Mouse plasma and brain samples were taken at 0.5 hr and 1.0 hr post dose.
  • Example 7 Histamine-Induced Model for Pruritus.
  • H3R inhibitors Compound 1 and Compound 3, were dissolved in saline at the appropriate concentration and 100 ⁇ L was administered orally (Compound 3) or
  • Example 8 Central Administration of a H3R Antagonist Does not Inhibit Histamine- induced Pruritus.
  • H3R antagonists When administered peripherally, centrally penetrant H3R antagonists have many effects in the rodent. These include increasing cognition, and inhibiting histamine (HA)-induced pruritus.
  • HA histamine
  • a known H3R antagonist was administered directly into the brain, and effects on both HA-induced pruritus and performance in a cognitive task were measured.
  • H3R antagonist used was (R)-2-hydroxy-l-(6-(4-(2-(2-methylpyrrolidin-l-yl)ethyl)phenyl)-3,4- dihydroisoquinolin-2(l//)-yl)ethanone (referred herein as Compound A), see Compound 10 in PCT publication WO2009/105206.
  • mice Male C57BL/6 mice were implanted with chronically indwelling cannulae aimed at the lateral ventricle. Cannulated mice (6-7/group) were dosed with either vehicle (PO) and vehicle (ICV), Compound A (10 mg kg PO) and vehicle (ICV), or vehicle (PO) and Compound A (10 ⁇ g ICV). Scratching bouts were counted from 5-25 minutes post-histamine challenge.
  • cannulated mice were allowed to explore an open-field (50 X 37 X 24 cm) for five minute habituation sessions on two consecutive days. On day three (training day), two identical objects were presented near opposite corners of the open-field (10 cm from walls), and mice allowed to explore the objects for 10 min.
  • mice were removed from the arena and injected ICV with either vehicle or Compound A (10 ⁇ g). 24 h later, subjects were placed back in the open field in which one of the previously presented objects was replaced by a novel object. Subjects were allowed to explore the two objects for a five-minute session during which their behavior was video recorded for subsequent analysis. The difference between exploration times of the novel and familiar objects as a factor of total object exploration time ((N-F)/(N+F)) was taken as a measure of performance (cognitive index).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Dermatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des composés de Formule (Ia) et leurs sels, solvates et hydrates pharmaceutiquement acceptables, qui modulent l'activité du récepteur H3 histaminique. Les composés de la présente invention et les compositions pharmaceutiques à base de ceux-ci concernent des procédés utiles dans le traitement des troubles associés à l'histamine H3.
PCT/US2013/054307 2012-08-13 2013-08-09 Modulateurs du récepteur h3 histaminique et traitement de troubles s'y rapportant WO2014028322A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261682402P 2012-08-13 2012-08-13
US61/682,402 2012-08-13

Publications (1)

Publication Number Publication Date
WO2014028322A1 true WO2014028322A1 (fr) 2014-02-20

Family

ID=49118762

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/054307 WO2014028322A1 (fr) 2012-08-13 2013-08-09 Modulateurs du récepteur h3 histaminique et traitement de troubles s'y rapportant

Country Status (1)

Country Link
WO (1) WO2014028322A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022113008A1 (fr) 2020-11-27 2022-06-02 Richter Gedeon Nyrt. Antagonistes/agonistes inverses du récepteur h3 de l'histamine pour le traitement d'un trouble du spectre autistique

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100331312A1 (en) * 2008-02-19 2010-12-30 Arena Pharmaceuticals, Inc. Modulators of the histamine h3 receptor useful for the treatment of disorders related thereto

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100331312A1 (en) * 2008-02-19 2010-12-30 Arena Pharmaceuticals, Inc. Modulators of the histamine h3 receptor useful for the treatment of disorders related thereto

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LAZEWSKA ET AL.: "Recent advances in histamine H3 receptor antagonists/inverse agonists", EXPERT OPIN. THER. PATENTS, vol. 20, no. 9, 1 September 2010 (2010-09-01), pages 1147 - 1169, XP008127038, ISSN: 1354-3776, DOI: 10.1517/13543776.2010.509346 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022113008A1 (fr) 2020-11-27 2022-06-02 Richter Gedeon Nyrt. Antagonistes/agonistes inverses du récepteur h3 de l'histamine pour le traitement d'un trouble du spectre autistique

Similar Documents

Publication Publication Date Title
CN102153562B (zh) 螺-吲哚酮化合物及其作为治疗剂的用途
US11560369B2 (en) Crystalline forms and processes for the preparation of cannabinoid receptor modulators
JP2022521789A (ja) 脳障害を治療するためのアゼピノ-インドール及び他の複素環化合物
JP5072827B2 (ja) ヒスタミンh3受容体薬剤、製剤及び治療的使用
WO2008005338A1 (fr) Modulateurs du récepteur h3 de l'histamine utiles dans le traitement de troubles liés audit récepteur
EP2074086B1 (fr) Modulateurs biphenylsulfonyle et de phenyl-heterorarylsulfonyle du recepteur h3 de l'histamine utiles pour le traitement de troubles associes a celui-ci
CN102036967B (zh) 作为5-ht6拮抗剂的芳基磺酰基吡唑啉甲脒衍生物
WO2009058300A1 (fr) Dérivés de biphényle comme modulateurs du récepteur h-3 de l'histamine utiles pour le traitement de troubles se rapportant à celui-ci
US20100331312A1 (en) Modulators of the histamine h3 receptor useful for the treatment of disorders related thereto
EP3309150B1 (fr) Acid propanoïque (r)-3-(4'-(2-(2-méthylpyrrolidin-1-yl)éthyl)biphenyl-4-yl) en tant que modulateur du récepteur de l'histamine h3 pour le traitement de troubles cognitifs
DE602005006018T2 (de) Arylpiperazinderivate und deren verwendung als für den dopamin-d3-rezeptor selektive liganden
KR20220024476A (ko) 인다졸 유도체, 이의 제조 방법, 및 이의 약제학적 적용
US20100292288A1 (en) Crystalline forms of (r)-1-{2-[4`- (3-methoxy-propane-1- sulfonyl)-biphenyl-4-yl]-ethyl}-2-methyl-pyrrolidine, and compositions, and methods related thereto
WO2014028322A1 (fr) Modulateurs du récepteur h3 histaminique et traitement de troubles s'y rapportant
WO2007061741A2 (fr) Modulateurs du recepteur h3 utiles pour le traitement de troubles associes au dit recepteur
RU2701861C1 (ru) Новое производное 3-азабицикло[3.1.0]гексана и его применение в медицинских целях
WO2020067456A1 (fr) Composé hétérocyclique

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13759333

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13759333

Country of ref document: EP

Kind code of ref document: A1