Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FATTY ACID AMIDE HYDROLASE (FAAH) CLEAVABLE PRODRUGS OF THYROMIMETICS AND COMBINATION WITH PERIPHERALLY RESTRICTED FAAH INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2022/236133
Kind Code:
A1
Abstract:
Provided herein are fatty acid amide (FAAH) cleavable prodrugs of thyromimetics and pharmaceutical compositions comprising these compounds with at least one pharmaceutically acceptable excipient further comprising a peripherally restricted FAAH inhibitor.

Inventors:
STEARNS BRIAN ANDREW (US)
BACCEI JILL MELISSA (US)
HARRIS JASON RANDALL (US)
Application Number:
PCT/US2022/028187
Publication Date:
November 10, 2022
Filing Date:
May 06, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AUTOBAHN THERAPEUTICS INC (US)
International Classes:
A61K31/44; A61K31/165; A61K31/397; A61K31/40; A61K31/42; A61K31/4965; A61K31/50; A61K31/505; A61K31/5375; A61P25/28; C07C235/06; C07C235/08; C07C235/22; C07C243/28; C07D205/04; C07D213/75; C07D237/20; C07D239/42; C07D241/20; C07D261/14; C07D295/185
Domestic Patent References:
WO2019160980A12019-08-22
WO2017201320A12017-11-23
WO2021108549A12021-06-03
Other References:
MACKENNA DEIDRE A, ET AL.: "085-P ABX-002: A fatty-acid amide hydrolase (FAAH)-activated prodrug enhances functional delivery of a potent TRβ selective thyromimetic to the brain and demonstrates biological activity in models of X-linked Adrenoleukodystrophy ", NEUROLOGY, 13 April 2021 (2021-04-13), pages 2442, XP093004638
MEINIG, J. M. ET AL.: "Targeting fatty-acid amide hydrolase with prodrugs for CNS- selective therapy", ACS CHEMICAL NEUROSCIENCE, vol. 8, no. 11, 17 November 2017 (2017-11-17), pages 2468 - 2476, XP055501373, DOI: 10.1021/acschemneuro.7b00239
D. A. MACKENNA, J. M. BACCEI, M. B. WOERNER, A. ALAVI, M. W. AKHTAR, T. S. SCANLAN, B. A. STEARNS, J. R. HARRIS: "P944.03 Feedback control on the pituitary-thyroid hormone axis by thyroid hormone agonists correlates with peripheral exposure as opposed to central exposure and on-target activity", NEUROSCIENCE 2021 CONFERENCE; 8-11 NOVEMBER 2021, 9 November 2021 (2021-11-09) - 11 November 2021 (2021-11-11), US, pages P944.03, XP009541311
Attorney, Agent or Firm:
MALECHA, James (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A pharmaceutical composition comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F), or a pharmaceutically acceptable salt or solvate thereof:

Formula (F); wherein:

R1 and R2 are independently selected from hydrogen, -OR5, -NR5R6, Ci-C6alkyl, C2- C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3-C6heterocycloalkyl, phenyl, and -Ci-C6alkyl- phenyl, wherein Ci-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3- C6heterocycloalkyl, phenyl, and -Ci-C6alkyl-phenyl are optionally substituted with one or more of halo, cyano, -OR5, -NR5R6, -S(0)2R5, or -S(0)20R5;

R3 and R4 are independently selected from -F, -Cl, -Br, and -I;

R5 and R6 are independently selected from hydrogen and Ci-C6alkyl; and R7 and R8 are independently selected from hydrogen, -F, -Cl, -Br, and -I; and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor.

2. The pharmaceutical composition of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is hydrogen.

3. The pharmaceutical composition of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein R7 is -F.

4. The pharmaceutical composition of any one of claims 1-3, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is hydrogen.

5. The pharmaceutical composition of any one of claims 1-3, or a pharmaceutically acceptable salt or solvate thereof, wherein R8 is -F.

6. A pharmaceutical composition comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (I), or a pharmaceutically acceptable salt or solvate thereof:

Formula (I); wherein:

R1 and R2 are independently selected from hydrogen, -OR5, -NR5R6, Ci-C6alkyl, C2- C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3-C6heterocycloalkyl, phenyl, and -Ci-C6alkyl- phenyl, wherein Ci-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3- C6heterocycloalkyl, phenyl, and -Ci-C6alkyl-phenyl are optionally substituted with one or more of halo, cyano, -OR5, -NR5R6, -S(0)2R5, or -S(0)20R5;

R3 and R4 are independently selected from -F, -Cl, -Br, and -I; and R5 and R6 are independently selected from hydrogen and Ci-C6alkyl; and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor.

7. The pharmaceutical composition of any one of claims 1-6, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is hydrogen.

8. The pharmaceutical composition of any one of claims 1-7, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is Ci-C6alkyl optionally substituted with one or more of halo, cyano, -OR5, -NR5R6, -S(0)2R5, or -S(0)20R5.

9. The pharmaceutical composition of any one of claims 1-8, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is Ci-C6alkyl substituted with one or more of halo, cyano, -OR5, -NR5R6, -S(0)2R5, or -S(0)20R5.

10. The pharmaceutical composition of any one of claims 1-9, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is Ci-C6alkyl substituted with one or more -OH.

11. The pharmaceutical composition of any one of claims 1-9, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is Ci-C6alkyl substituted with one or more of halo.

12. The pharmaceutical composition of any one of claims 1-8, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is unsubstituted Ci-C6alkyl.

13. The pharmaceutical composition of any one of claims 1-7, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is phenyl optionally substituted with one or more of halo, cyano, -OR5, -NR5R6, -S(0)2R5, or -S(0)20R5.

14. The pharmaceutical composition of any one of claims 1-7, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is -Ci-C6alkyl-phenyl optionally substituted with one or more of halo, cyano, -OR5, -NR5R6, -S(0)2R5, or -S(0)20R5.

15. The pharmaceutical composition of any one of claims 1-14, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are independently selected from -F, -Cl, -Br.

16. The pharmaceutical composition of any one of claims 1-15, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are both -Br.

17. The pharmaceutical composition of any one of claims 1-15, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are both -Cl.

18. The pharmaceutical composition of any one of claims 1-15, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 and R4 are both -F.

19. The pharmaceutical composition of claim 1 or claim 6, wherein the fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I) has a structure selected from:

pharmaceutically acceptable salt or solvate thereof.

20. The pharmaceutical composition of claim 1 or claim 6, wherein the fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I) has a structure selected from:

pharmaceutically acceptable salt or solvate thereof.

21. The pharmaceutical composition of claim 1, wherein the fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) has a structure selected from: pharmaceutically acceptable salt or solvate thereof.

22. The pharmaceutical composition of any one of claims 1-21, or a pharmaceutically acceptable salt or solvate thereof, wherein the peripherally restricted FAAH inhibitor is ASP-3652.

23. A method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition of any one of claims 1-22, or a pharmaceutically acceptable salt or solvate thereof.

24. The method of claim 23, wherein the CNS disease or disorder is selected from acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis (AHL or AHLE), adult Refsum disease, infantile Refsum disease, Alexander disease, Alzheimer's disease, Balo concentric sclerosis, Canavan disease, central pontine myelinolysis (CPM), cerebral palsy, cerebrotendineous xanthomatosis, chronic inflammatory demyelinating polyneuropathy (CIDP), Devic's syndrome, diffuse myelinoclastic sclerosis, encephalomyelitis, Guillain-Barre syndrome, idiopathic inflammatory demyelinating disease (HDD), Krabbe disease, Leber hereditary optic neuropathy, leukodystrophy, Marburg multiple sclerosis, Marchiafava-Bignami disease, metachromatic leukodystrophy (MLD), multifocal motor neuropathy (MMN), multiple sclerosis (MS), paraproteinemic demyelinating polyneuropathy, Pelizaeus-Merzbacher disease (PMD), progressive multifocal leukoencephaalopathy (PML), tropical spastic paraparesis (TSP), X-linked adrenoleukodystrophy (X-ALD, ALO, or X-linked ALO), and Zellweger syndrome.

25. The method of claim 24, wherein the CNS disease or disorder is selected from multiple sclerosis and X-linked adrenoleukodystrophy.

26. A compound selected from:

27. A pharmaceutical composition comprising a compound of claim 26, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.

28. The pharmaceutical composition of claim 27, further comprising a peripherally restricted FAAH inhibitor.

29. The pharmaceutical composition of claim 28, wherein the peripherally restricted FAAH inhibitor is ASP-3652.

30. A method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of claim 26, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition of any one of claims 27-29.

31. The method of claim 30, wherein the CNS disease or disorder is selected from acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis (AHL or AHLE), adult Refsum disease, infantile Refsum disease, Alexander disease, Alzheimer's disease, Balo concentric sclerosis, Canavan disease, central pontine myelinolysis (CPM), cerebral palsy, cerebrotendineous xanthomatosis, chronic inflammatory demyelinating polyneuropathy (CIDP), Devic's syndrome, diffuse myelinoclastic sclerosis, encephalomyelitis, Guillain-Barre syndrome, idiopathic inflammatory demyelinating disease (HDD), Krabbe disease, Leber hereditary optic neuropathy, leukodystrophy, Marburg multiple sclerosis, Marchiafava-Bignami disease, metachromatic leukodystrophy (MLD), multifocal motor neuropathy (MMN), multiple sclerosis (MS), paraproteinemic demyelinating polyneuropathy, Pelizaeus-Merzbacher disease (PMD), progressive multifocal leukoencephaalopathy (PML), tropical spastic paraparesis (TSP), X-linked adrenoleukodystrophy (X-ALD, ALO, or X-linked ALO), and Zellweger syndrome.

32. The method of claim 31, wherein the CNS disease or disorder is selected from multiple sclerosis and X-linked adrenoleukodystrophy.

Description:
FATTY ACID AMIDE HYDROLASE (FAAH) CLEAVABLE PRODRUGS OF THYROMIMETICS AND COMBINATION WITH PERIPHERALLY RESTRICTED FAAH INHIBITORS

CROSS-REFERENCE

[0001] This application claims the benefit of U.S. Provisional Application No. 63/185,254 filed on May 6, 2021 and U.S. Provisional Application No. 63/274,856 filed on November 2, 2021, each of which is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

[0002] The blood-brain barrier is composed of tightly linked endothelial cells that limit the passage of pathogens and specific types of small and large molecules from the blood into the brain. This critical protective function also restricts the diffusion of therapeutics into the brain representing a major challenge to the development of new medicines for CNS diseases.

SUMMARY OF THE INVENTION

[0003] In one aspect provided herein is a pharmaceutical composition comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F), or a pharmaceutically acceptable salt or solvate thereof: wherein:

R 1 and R 2 are independently selected from hydrogen, -OR 5 , -NR 5 R 6 , Ci-C 6 alkyl, C2- C 6 alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3-C6heterocycloalkyl, phenyl, and -Ci-C 6 alkyl- phenyl, wherein Ci-C 6 alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3- C 6 heterocycloalkyl, phenyl, and -Ci-C 6 alkyl-phenyl are optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 ;

R 3 and R 4 are independently selected from -F, -Cl, -Br, and -I;

R 5 and R 6 are independently selected from hydrogen and Ci-C 6 alkyl; and

R 7 and R 8 are independently selected from hydrogen, -F, -Cl, -Br, and -I; and a pharmaceutically acceptable excipient; further comprising a peripherally restricted

FAAH inhibitor.

[0004] In some embodiments, R 7 is hydrogen. In some embodiments, R 7 is -F. In some embodiments, R 8 is hydrogen. In some embodiments, R 8 is -F. [0005] In another aspect provided herein is a pharmaceutical composition comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (I), or a pharmaceutically acceptable salt or solvate thereof:

Formula (I); wherein:

R 1 and R 2 are independently selected from hydrogen, -OR 5 , -NR 5 R 6 , Ci-C 6 alkyl, C2- C 6 alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3-C6heterocycloalkyl, phenyl, and -Ci-C 6 alkyl- phenyl, wherein Ci-C 6 alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3- C 6 heterocycloalkyl, phenyl, and -Ci-C 6 alkyl-phenyl are optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 ;

R 3 and R 4 are independently selected from -F, -Cl, -Br, and -I; and

R 5 and R 6 are independently selected from hydrogen and Ci-C 6 alkyl; and a pharmaceutically acceptable excipient; further comprising a peripherally restricted

FAAH inhibitor.

[0006] In some embodiments, R 1 is hydrogen. In some embodiments, R 2 is Ci-C 6 alkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo, cyano, -OR 5 , - NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -OH. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo. In some embodiments, R 2 is unsubstituted Ci-C 6 alkyl. In some embodiments, R 2 is phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is -Ci-C 6 alkyl-phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 3 and R 4 are independently selected from -F, -Cl, and -Br. In some embodiments, R 3 and R 4 are both -Br. In some embodiments, R 3 and R 4 are both -Br. In some embodiments, R 3 and R 4 are both -Cl. In some embodiments, R 3 and R 4 are both -F.

[0007] In some embodiments, the peripherally restricted FAAH inhibitor is ASP-3652.

[0008] In another aspect is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a pharmaceutical composition described herein. In some embodiments, the CNS disease or disorder is selected from acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis (AHL or AHLE), adult Refsum disease, infantile Refsum disease, Alexander disease, Alzheimer's disease, Balo concentric sclerosis, Canavan disease, central pontine myelinolysis (CPM), cerebral palsy, cerebrotendineous xanthomatosis, chronic inflammatory demyelinating polyneuropathy (CIDP), Devic's syndrome, diffuse myelinoclastic sclerosis, encephalomyelitis, Guillain-Barre syndrome, idiopathic inflammatory demyelinating disease (HDD), Krabbe disease, Leber hereditary optic neuropathy, leukodystrophy, Marburg multiple sclerosis, Marchiafava-Bignami disease, metachromatic leukodystrophy (MLD), multifocal motor neuropathy (MMN), multiple sclerosis (MS), paraproteinemic demyelinating polyneuropathy, Pelizaeus-Merzbacher disease (PMD), progressive multifocal leukoencephaalopathy (PML), tropical spastic paraparesis (TSP), X-linked adrenoleukodystrophy (X-ALD, ALO, or X-linked ALO), and Zellweger syndrome. In some embodiments, the CNS disease or disorder is selected from multiple sclerosis and X-linked adrenoleukody strophy .

BRIEF DESCRIPTION OF THE DRAWINGS [0009] The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosures will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the disclosures are utilized, and the accompanying drawings of which:

[0010] Figure 1 depicts the active metabolite of the LL-341070 prodrug, LL-341070A, enhanced oligodendrocyte differentiation in vitro in an oligodendrocyte progenitor cell assay. [0011] Figure 2 depicts thyromimetic treatment enhances 24-OHC synthesis in vivo in the brains of rats following cuprizone-induced demyelination.

[0012] Figure 3 depicts TRp target engagement in brain is demonstrated by increased expression of T3 -responsive target genes in vivo.

[0013] Figure 4 depicts brain and plasma concentration following 21 days of repeat administration of LL-341070 measured 4 hours post-final dose.

[0014] Figure 5 depicts LL-341070 improves in vivo clinical scoring and histology in mouse prophylactic experimental autoimmune encephalitis (EAE) model.

[0015] Figure 6 depicts FAAH expression and specific activity across species and tissue types. [0016] Figure 7 depicts concentrations of ABX-002A in brain, liver, kidney, lung, and heart were measured 1 hour after SC administration of 30 different prodrugs of ABX-002A.

[0017] Figure 8 depicts plasma, liver, and brain concentrations following ABX-002 prodrug treatment with or without peripheral or global FAAH inhibitors. [0018] Figure 9A depicts induction of T3-target genes in brain vs. liver after single administration of ABX-002A.

[0019] Figure 9B depicts induction of T3-target genes in brain vs. liver after single administration of ABX-002.

[0020] Figure 9C depicts induction of T3-target genes in brain vs. liver after single administration of ABX-002 plus FAAH inhibitor.

[0021] Figure 10A depicts gene expression in brain and liver, and effects on T4 after administration of ABX-002A.

[0022] Figure 10B depicts gene expression in brain and liver, and effects on T4 after administration of ABX-002.

[0023] Figure IOC depicts gene expression in brain and liver, and effects on T4 after administration of ABX-002 plus peripheral FAAH inhibitor.

[0024] Figure 10D depicts gene expression in brain and liver, and effects on T4 after administration of ABX-002 plus global FAAH inhibitor.

[0025] Figure 11A depicts T4 inhibition as a function of ABX-002 dose in mice or non-human primate (NHP).

[0026] Figure 11B depicts T4 inhibition as a function of plasma ABX-002 prodrug AUC in mice or non-human primate (NHP).

[0027] Figure 11C depicts T4 inhibition as a function of plasma ABX-002A active metabolite AUC in mice or non-human primate (NHP).

DETAILED DESCRIPTION OF THE INVENTION [0028] Fatty acid amide hydrolase (FAAH) is an integral membrane serine hydrolase that degrades the fatty acid amide family of signaling lipids and can hydrolyze select amide prodrugs. FAAH is highly conserved between species and is expressed in many tissues, including the central nervous system (CNS), to varying degrees. Select carboxylic acids can be converted to more permeable amide prodrugs which are then capable of passing through the blood brain barrier where they can be converted to active molecules through the action of FAAH upon the prodrug. This results in the delivery of higher amounts of the carboxylic acid to the CNS as compared to dosing the parent alone. However, peripherally expressed FAAH simultaneously hydrolyzes the prodrug resulting in a considerable amount of non-productive prodrug conversion. Co-administration of a peripherally restricted FAAH inhibitor with a CNS permeable FAAH convertible prodrug increases the selectivity of prodrug delivery to the CNS.

It also results in lower exposures of the parent molecule in plasma and peripheral tissue than what is observed when dosing the prodrug alone. [0029] Candidates for clinical development may be selected from the compounds disclosed herein based on their in vitro FAAH-mediated hydrolysis, in vitro plasma stability, in vivo tissue distribution, in vitro target selectivity, in vitro target potency, target gene expression, in vivo pharmacological efficacy, or degree of drug-like (rule-of-5 compliant) physiochemical properties, or combinations thereof.

Certain Terminology

[0030] The singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a drug” includes reference to one or more of such drugs, and reference to “an excipient” includes reference to one or more of such excipients. When ranges are used herein, all combinations and sub-combinations of ranges and specific embodiments therein are intended to be included. The term "about" when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range varies between 1% and 15% of the stated number or numerical range.

[0031] The terms “formulation” and “composition,” as used herein, are used interchangeably and refer to a mixture of two or more compounds, elements, or molecules. In some aspects the terms “formulation” and “composition” may be used to refer to a mixture of one or more active agents with a carrier or other excipients.

[0032] The terms “active agent,” “active pharmaceutical agent,” “drug,” “active ingredient,” and variants thereof are used interchangeably to refer to an agent or substance that has measurable specified or selected physiologic activity when administered to a subject in a significant or effective amount.

[0033] "Pharmaceutically acceptable salt" includes both acid and base addition salts. A pharmaceutically acceptable salt of any one of the compounds described herein is intended to encompass any and all pharmaceutically suitable salt forms. Preferred pharmaceutically acceptable salts of the compounds described herein are pharmaceutically acceptable acid addition salts, and pharmaceutically acceptable base addition salts.

[0034] "Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, hydroiodic acid, hydrofluoric acid, phosphorous acid, and the like. Also included are salts that are formed with organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and include, for example, acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, / oluenesulfonic acid, salicylic acid, and the like. Exemplary salts thus include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, nitrates, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, trifluoroacetates, propionates, caprylates, isobutyrates, oxalates, malonates, succinate suberates, sebacates, fumarates, maleates, mandelates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, phthalates, benzenesulfonates, toluenesulfonates, phenylacetates, citrates, lactates, malates, tartrates, methanesulfonates, and the like. Also contemplated are salts of amino acids, such as arginates, gluconates, and galacturonates (see, for example, Berge S.M. etah, "Pharmaceutical Salts," Journal of Pharmaceutical Science, 66:1-19 (1997)). Acid addition salts of basic compounds are prepared by contacting the free base forms with a sufficient amount of the desired acid to produce the salt.

[0035] "Pharmaceutically acceptable base addition salt" refers to those salts that retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. In some embodiments, pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts, and the like. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, for example, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, A f , A f -dibenzyl ethyl enedi amine, chloroprocaine, hydrabamine, choline, betaine, ethylenediamine, ethylenedianiline, N- methylglucamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine,

A -ethyl pi peri dine, polyamine resins, and the like. See Berge et al., supra.

[0036] It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms (solvates). Solvates contain either stoichiometric or non- stoichiometric amounts of a solvent, and are formed during the process of product formation or isolation with pharmaceutically acceptable solvents such as water, ethanol, methanol, methyl tert-butyl ether (MTBE), diisopropyl ether (DIPE), ethyl acetate, isopropyl acetate, isopropyl alcohol, methyl isobutyl ketone (MIBK), methyl ethyl ketone (MEK), acetone, nitromethane, tetrahydrofuran (THF), dichloromethane (DCM), dioxane, heptanes, toluene, anisole, acetonitrile, and the like. In one aspect, solvates are formed using, but not limited to, Class 3 solvent(s). Categories of solvents are defined in, for example, the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), “Impurities: Guidelines for Residual Solvents, Q3C(R3), (November 2005). Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. [0037] The terms “effective amount” or “therapeutically effective amount” as used herein, refer to a sufficient amount of an agent or a compound being administered which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result can be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in a disease. An appropriate “effective” amount in any individual case may be determined using techniques, such as a dose escalation study.

[0038] The terms “subject,” “individual,” and “patient” are used interchangeably herein to refer to a mammal. Mammals include, but are not limited to, murines, simians, humans, farm animals, sport animals, and pets.

[0039] The term “peripherally restricted FAAH inhibitor” as used herein, refers to a fatty acid amide hydrolase (FAAH) inhibitor that inhibits FAAH to a greater extent in the periphery than in the central nervous system from a systemic dose. In some embodiments, the peripherally restricted FAAH inhibitor is 60% peripherally restricted. In some embodiments, the peripherally restricted FAAH inhibitor is 70% peripherally restricted. In some embodiments, the peripherally restricted FAAH inhibitor is 80% peripherally restricted. In some embodiments, the peripherally restricted FAAH inhibitor is 90% peripherally restricted. In some embodiments, the peripherally restricted FAAH inhibitor is 95% peripherally restricted. Target

[0040] Thyroid hormone (TH) is a key signal for oligodendrocyte differentiation and myelin formation during development, and also stimulates remyelination in adult models of multiple sclerosis (MS) (Calza L et al , Brain Res Revs 48:339-346, 2005). However, TH is not an acceptable long-term therapy due to there being virtually no therapeutic window in which remyelination can be achieved while avoiding the cardiotoxicity and bone demineralization associated with chronic hyperthyroidism. Some thyroid hormone analogs can activate thyroid hormone-responsive genes while avoiding the associated downsides of TH by exploiting molecular and physiological features of thyroid hormone receptors (Malm J et al , Mini Rev Med Chem 7:79-86, 2007). These receptors are expressed in two major forms with heterogenous tissue distributions and overlapping but distinct sets of target genes (Yen PM, Physiol Rev 81 : 1097-1142, 2001). TRa is enriched in the heart, brain, and bone while TRP is enriched in the liver (O’Shea PJ et al, Nucl Recept Signal 4:e011, 2006).

[0041] Developing selective thyromimetics has been challenging due to the high sequence homology of thyroid hormone receptor subtypes; namely, only one amino acid residue on the internal surface of the ligand binding domain cavity varies between the al and bΐ forms.

[0042] In some embodiments, the pharmaceutical compositions described herein comprise a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F), wherein the prodrug of Formula (F) is a prodrug of a TRP agonist. In some embodiments, the pharmaceutical compositions described herein comprise a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (I), wherein the prodrug of Formula (I) is a prodrug of a TRP agonist. In some embodiments, the pharmaceutical compositions described herein comprise a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II), wherein the prodrug of Formula (II) is a prodrug of a TRP agonist.

Pharmaceutical Compositions

[0043] In some embodiments described herein is a pharmaceutical composition comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F), or a pharmaceutically acceptable salt or solvate thereof:

Formula (F); wherein:

R 1 and R 2 are independently selected from hydrogen, -OR 5 , -NR 5 R 6 , Ci-C 6 alkyl, C2- C 6 alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3-C6heterocycloalkyl, phenyl, and -Ci-C 6 alkyl- phenyl, wherein Ci-C 6 alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3- C 6 heterocycloalkyl, phenyl, and -Ci-C 6 alkyl-phenyl are optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 ;

R 3 and R 4 are independently selected from -F, -Cl, -Br, and -I;

R 5 and R 6 are independently selected from hydrogen and Ci-C 6 alkyl; and R 7 and R 8 are independently selected from hydrogen, -F, -Cl, -Br, and -I; and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor.

[0044] In some embodiments, R 7 is hydrogen. In some embodiments, R 7 is -F. In some embodiments, R 7 is -Cl. In some embodiments, R 7 is -Br. [0045] In some embodiments, R 8 is hydrogen. In some embodiments, R 8 is -F. In some embodiments, R 8 is -Cl. In some embodiments, R 8 is -Br.

[0046] In some embodiments, R 1 is hydrogen. In some embodiments, R 1 is Ci- 6 alkyl.

[0047] In some embodiments, R 2 is Ci-C 6 alkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one cyano. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -OR 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more - OH. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -OH. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -NR 5 R 6 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -MB. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -MB. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 H. In some embodiments, R 2 is Ci- C 6 alkyl substituted with one -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 0H. In some embodiments, R 2 is unsubstituted Ci-C 6 alkyl. In some embodiments, R 2 is -CH3. In some embodiments, R 2 is -CH2CH3. In some embodiments, R 2 is - CH2CH2CH3.

[0048] In some embodiments, R 2 is C2-C6alkenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C2-C6alkenyl.

[0049] In some embodiments, R 2 is C2-C6alkynyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C2-C6alkynyl.

[0050] In some embodiments, R 2 is C3-C6cycloalkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C3-C6cycloalkyl.

[0051] In some embodiments, R 2 is C3-C6heterocycloalkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C3-C6heterocycloalkyl.

[0052] In some embodiments, R 2 is phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is phenyl substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is phenyl substituted with one or more of halo. In some embodiments, R 2 is phenyl substituted with one or more -OR 5 . In some embodiments, R 2 is phenyl substituted with one or more -OH. In some embodiments, R 2 is unsubstituted phenyl.

[0053] In some embodiments, R 2 is -Ci-C 6 alkyl-phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted -Ci-C 6 alkyl-phenyl.

[0054] In some embodiments, R 2 is -OR 5 . In some embodiments, R 2 is -OH. In some embodiments, R 2 is -NR 5 R 6 . In some embodiments, R 2 is -NH2.

[0055] In some embodiments, R 2 is hydrogen.

[0056] In some embodiments, R 3 and R 4 are independently selected from -F, -Cl, and -Br. In some embodiments, R 3 and R 4 are both -Br. In some embodiments, R 3 and R 4 are both -Br. In some embodiments, R 3 and R 4 are both -Cl. In some embodiments, R 3 and R 4 are both -F. In some embodiments, R 3 is -Cl and R 4 is -Br. In some embodiments, R 3 is -F and R 4 is -Br. In some embodiments, R 3 is -F and R 4 is -Cl.

[0057] In some embodiments of the pharmaceutical compositions described herein, the fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) has a structure selected from: pharmaceutically acceptable salt or solvate thereof.

[0058] In some embodiments described herein is a pharmaceutical composition comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (I), or a pharmaceutically acceptable salt or solvate thereof:

Formula (I); wherein:

R 1 and R 2 are independently selected from hydrogen, -OR 5 , -NR 5 R 6 , Ci-C 6 alkyl, C2- C 6 alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3-C6heterocycloalkyl, phenyl, and -Ci-C 6 alkyl- phenyl, wherein Ci-C 6 alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3- C 6 heterocycloalkyl, phenyl, and -Ci-C 6 alkyl-phenyl are optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 ;

R 3 and R 4 are independently selected from -F, -Cl, -Br, and -I; and R 5 and R 6 are independently selected from hydrogen and Ci-C 6 alkyl; and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor.

[0059] In some embodiments, R 1 is hydrogen. In some embodiments, R 1 is Ci- 6 alkyl.

[0060] In some embodiments, R 2 is Ci-C 6 alkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one cyano. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -OR 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more - OH. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -OH. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -NR 5 R 6 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -NH2. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -NH2. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 H. In some embodiments, R 2 is Ci- C 6 alkyl substituted with one -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 0H. In some embodiments, R 2 is unsubstituted Ci-C 6 alkyl. In some embodiments, R 2 is -CH 3 . In some embodiments, R 2 is -CH 2 CH 3 . In some embodiments, R 2 is - CH2CH2CH3.

[0061] In some embodiments, R 2 is C2-C6alkenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C2-C6alkenyl.

[0062] In some embodiments, R 2 is C2-C6alkynyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C2-C6alkynyl.

[0063] In some embodiments, R 2 is C3-C6cycloalkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C3-C6cycloalkyl.

[0064] In some embodiments, R 2 is C3-C6heterocycloalkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C3-C6heterocycloalkyl.

[0065] In some embodiments, R 2 is phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is phenyl substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is phenyl substituted with one or more of halo. In some embodiments, R 2 is phenyl substituted with one or more -OR 5 . In some embodiments, R 2 is phenyl substituted with one or more -OH. In some embodiments, R 2 is unsubstituted phenyl.

[0066] In some embodiments, R 2 is -Ci-C 6 alkyl-phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted -Ci-C 6 alkyl-phenyl.

[0067] In some embodiments, R 2 is -OR 5 . In some embodiments, R 2 is -OH. In some embodiments, R 2 is -NR 5 R 6 . In some embodiments, R 2 is -NH2.

[0068] In some embodiments, R 2 is hydrogen.

[0069] In some embodiments, R 3 and R 4 are independently selected from -F, -Cl, and -Br. In some embodiments, R 3 and R 4 are both -Br. In some embodiments, R 3 and R 4 are both -Br. In some embodiments, R 3 and R 4 are both -Cl. In some embodiments, R 3 and R 4 are both -F. In some embodiments, R 3 is -Cl and R 4 is -Br. In some embodiments, R 3 is -F and R 4 is -Br. In some embodiments, R 3 is -F and R 4 is -Cl.

[0070] In some embodiments of the pharmaceutical compositions described herein, the fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I) has a structure selected from:

pharmaceutically acceptable salt or solvate thereof.

[0071] In some embodiments of the pharmaceutical compositions described herein, the fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I) has a structure selected from: pharmaceutically acceptable salt or solvate thereof. [0072] In some embodiments described herein is a pharmaceutical composition comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II), or a pharmaceutically acceptable salt or solvate thereof:

Formula (II); wherein:

R 1 and R 2 are independently selected from hydrogen, -OR 5 , -NR 5 R 6 , Ci-C 6 alkyl, C2- C 6 alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3-C6heterocycloalkyl, phenyl, and -Ci-C 6 alkyl- phenyl, wherein Ci-C 6 alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-C6cycloalkyl, C3- C 6 heterocycloalkyl, phenyl, and -Ci-C 6 alkyl-phenyl are optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 ; and

R 5 and R 6 are independently selected from hydrogen and Ci-C 6 alkyl; and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor, wherein the peripherally restricted FAAH inhibitor is ASP-3652.

[0073] In some embodiments, R 1 is hydrogen. In some embodiments, R 1 is Ci- 6 alkyl.

[0074] In some embodiments, R 2 is Ci-C 6 alkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more of halo. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one cyano. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -OR 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more - OH. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -OH. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -NR 5 R 6 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one or more -NH2. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -NH2. In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 H. In some embodiments, R 2 is Ci- C 6 alkyl substituted with one -S(0) 2 0R 5 . In some embodiments, R 2 is Ci-C 6 alkyl substituted with one -S(0) 2 0H. In some embodiments, R 2 is unsubstituted Ci-C 6 alkyl. In some embodiments, R 2 is -CH3. In some embodiments, R 2 is -CH2CH3. In some embodiments, R 2 is - CH2CH2CH3.

[0075] In some embodiments, R 2 is C2-C6alkenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C2-C6alkenyl.

[0076] In some embodiments, R 2 is C2-C6alkynyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C2-C6alkynyl.

[0077] In some embodiments, R 2 is C3-C6cycloalkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C3-C6cycloalkyl. [0078] In some embodiments, R 2 is C3-C6heterocycloalkyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted C3-C6heterocycloalkyl.

[0079] In some embodiments, R 2 is phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is phenyl substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is phenyl substituted with one or more of halo. In some embodiments, R 2 is phenyl substituted with one or more -OR 5 . In some embodiments, R 2 is phenyl substituted with one or more -OH. In some embodiments, R 2 is unsubstituted phenyl.

[0080] In some embodiments, R 2 is -Ci-C 6 alkyl-phenyl optionally substituted with one or more of halo, cyano, -OR 5 , -NR 5 R 6 , -S(0) 2 R 5 , or -S(0) 2 0R 5 . In some embodiments, R 2 is unsubstituted -Ci-C 6 alkyl-phenyl.

[0081] In some embodiments, R 2 is -OR 5 . In some embodiments, R 2 is -OH. In some embodiments, R 2 is -NR 5 R 6 . In some embodiments, R 2 is -NH2.

[0082] In some embodiments, R 2 is hydrogen.

[0083] In some embodiments of the pharmaceutical compositions described herein, the fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II) has a structure selected from:

Peripherally restricted FAAH inhibitors [0084] The pharmaceutical compositions described herein comprise a peripherally restricted FAAH inhibitor. In some embodiments, the peripherally restricted FAAH inhibitor is disclosed in US 2008/0306046, which is herein incorporated by reference in its entirety.

[0085] In some embodiments, the peripherally restricted FAAH inhibitor is a compound of Formula (X), or a pharmaceutically acceptable salt thereof:

Formula (X); wherein: ring A is a benzene ring, cyclopentane ring, cyclohexane ring, cycloheptane ring, or a 5- to 7 membered nitrogen-containing hetero ring;

L is a single bond, lower alkylene, lower alkenylene, -N(R 15 )-C(=0)-, -C(=0)-N(R 15 )-, - (lower alkenylene)-C(=0), -0-, or C(=0);

R 15 is H or lower alkyl;

X is CH orN;

R 8 , R 9 , and R 10 are each independently selected from:

(i) a group selected from the group consisting of H, halo, -CN, CF 3 , lower alkyl, and -O- lower alkyl;

(ii) aryl optionally substituted with 1 to 5 groups independently selected from the group consisting of H, halo, -CN, CF 3 , lower alkyl, and -O-lower alkyl;

(iii) nitrogen-containing heteroaryl optionally substituted with 1 to 5 groups independently selected from the group consisting of H, halo, -CN, -CF 3 , lower alkyl, and -O-lower alkyl;

(iv) R 16 -(lower alkenylene)-0-;

(v) R 16 -(lower alkenylene)-N(R 15 )-; or

(vi) R 17 R 18 N-C(=0)-;

R 16 is

(i) aryl optionally substituted with 1 to 5 groups independently selected from the group consisting of H, halo, -CN, -CF 3 , lower alkyl, and -O-lower alkyl;

(ii) nitrogen-containing heteroaryl optionally substituted with 1 to 5 groups independently selected from the group consisting of H, halo, -CN, -CF 3 , lower alkyl, and -O-lower alkyl; or

(iii) 3- to 8-membered cycloalkyl; R 17 and R 18 are each independently selected from H, lower alkyl, and 3- to 8-membered cycloalkyl; or R 17 and R 18 may form, together with the nitrogen atom bonded thereto, a 3- to 8- membered nitrogen-containing hetero ring;

R 11 is selected from H, lower alkyl, and oxo (=0); and one of R 12 , R 13 , and R 14 is -C(=0)-0-(lower alkyl) or -CO2H, and the others are H.

[0086] In some embodiments, the peripherally restricted FAAH inhibitor is 5-(((4-(4-((3- fluorobenzyl)oxy)phenoxy)piperidin-l-yl)carbonyl)oxy)nicotin ic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5-(((4-(2-phenylethyl)piperidin-l- yl)carbonyl)oxy)nicotinic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5-(((4-(4-(2-cyclohexylethoxy)phenoxy)piperidin-l-yl)carbony l)oxy)nicotinic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5-(((4-((E)-2- phenylvinyl)piperidin-l-yl)carbonyl)oxy)nicotinic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5-(((4-(3-(l-(6-methylpyridin-2-yl)piperidin-4-yl)propyl)pip eridin- l-yl)carbonyl)oxy)nicotinic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5-(methoxycarbonyl)pyridin-3-yl 4-(2-phenylethyl)piperazine-l-carboxylate. In some embodiments, the peripherally restricted FAAH inhibitor is ASP-3652. In some embodiments, the peripherally restricted FAAH inhibitor is ASP-3652 which is 5-(((4-(2- phenylethyl)piperidin-l-yl)carbonyl)oxy)nicotinic acid.

Compounds

[0087] The compounds of Formula (F), (I), and (II) described herein are amide prodrugs of TRP agonists. The amide prodrugs described herein are cleaved by fatty acid amide hydrolase (FAAH) to give the active TRp agonist. In some embodiments is a compound selected from:

pharmaceutically acceptable salt or solvate thereof. [0088] In some embodiments is a pharmaceutical composition comprising a pharmaceutically pharmaceutically acceptable salt or solvate thereof.

Methods

[0089] In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising the peripherally restricted FAAH inhibitor ASP-3652. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is selected from acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis (AHL or AHLE), adult Refsum disease, infantile Refsum disease, Alexander disease, Alzheimer's disease, Balo concentric sclerosis, Canavan disease, central pontine myelinolysis (CPM), cerebral palsy, cerebrotendineous xanthomatosis, chronic inflammatory demyelinating polyneuropathy (CIDP), Devic's syndrome, diffuse myelinoclastic sclerosis, encephalomyelitis, Guillain-Barre syndrome, idiopathic inflammatory demyelinating disease (HDD), Krabbe disease, Leber hereditary optic neuropathy, leukodystrophy, Marburg multiple sclerosis, Marchiafava-Bignami disease, metachromatic leukodystrophy (MLD), multifocal motor neuropathy (MMN), multiple sclerosis (MS), paraproteinemic demyelinating polyneuropathy, Pelizaeus-Merzbacher disease (PMD), progressive multifocal leukoencephaalopathy (PML), tropical spastic paraparesis (TSP), X-linked adrenoleukodystrophy (X-ALD, ALO, or X-linked ALO), and Zellweger syndrome. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is multiple sclerosis. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (F) or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is X-linked adrenoleukodystrophy.

[0090] In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising the peripherally restricted FAAH inhibitor ASP-3652. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is selected from acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis (AHL or AHLE), adult Refsum disease, infantile Refsum disease, Alexander disease, Alzheimer's disease, Balo concentric sclerosis, Canavan disease, central pontine myelinolysis (CPM), cerebral palsy, cerebrotendineous xanthomatosis, chronic inflammatory demyelinating polyneuropathy (CIDP), Devic's syndrome, diffuse myelinoclastic sclerosis, encephalomyelitis, Guillain-Barre syndrome, idiopathic inflammatory demyelinating disease (HDD), Krabbe disease, Leber hereditary optic neuropathy, leukodystrophy, Marburg multiple sclerosis, Marchiafava-Bignami disease, metachromatic leukodystrophy (MLD), multifocal motor neuropathy (MMN), multiple sclerosis (MS), paraproteinemic demyelinating polyneuropathy, Pelizaeus-Merzbacher disease (PMD), progressive multifocal leukoencephaalopathy (PML), tropical spastic paraparesis (TSP), X- linked adrenoleukodystrophy (X-ALD, ALO, or X-linked ALO), and Zellweger syndrome. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is multiple sclerosis. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof comprising administering to the patient a pharmaceutical composition described herein comprising a fatty acid amide hydrolase (FAAH) cleavable prodrug of Formula (II), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; further comprising a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is X-linked adrenoleukodystrophy.

Excipients

[0091] Suitable optional excipients for use in the pharmaceutical compositions described herein include any commonly used excipients in pharmaceutics and are selected on the basis of compatibility with the active pharmaceutical agent and the release profile properties of the desired dosage form. Excipients include, but are not limited to, binders, fillers, flow aids, disintegrants, lubricants, glidants, polymeric carriers, plasticizers, stabilizers, surfactants, and the like. A summary of excipients described herein, may be found, for example in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington ’s Pharmaceutical Sciences , Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms , Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Norms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins, 1999), herein incorporated by reference in their entirety.

[0092] Binders impart cohesiveness to solid oral dosage form formulations: for powder filled capsule formulation, they aid in plug formation that can be filled into soft or hard shell capsules and for tablet formulation, they ensure the tablet remaining intact after compression and help assure blend uniformity prior to a compression or fill step. Materials suitable for use as binders in the solid dosage forms described herein include, but are not limited to, carboxymethylcellulose, methylcellulose (e.g., Methocel ® ), hydroxypropylmethylcellulose (e.g. Hypromellose USP Pharmacoat-603, hydroxypropylmethylcellulose acetate stearate (Aqoate HS-LF and HS), hydroxyethylcellulose, hydroxypropylcellulose (e.g., Klucel ® ), ethylcellulose (e.g., Ethocel ® ), and microcrystalline cellulose (e.g., Avicel ® ), microcrystalline dextrose, amylose, magnesium aluminum silicate, polysaccharide acids, bentonites, gelatin, polyvinyl pyrrolidone/vinyl acetate copolymer, crospovidone, povidone, starch, pregelatinized starch, tragacanth, dextrin, a sugar, such as sucrose (e.g., Dipac ® ), glucose, dextrose, molasses, mannitol, sorbitol, xylitol (e.g., Xylitab ® ), lactose, a natural or synthetic gum such as acacia, tragacanth, ghatti gum, mucilage of isapol husks, starch, polyvinyl pyrrolidone (e.g., Povidone ® CL, Kollidon ® CL, Polyplasdone ® XL-10, and Povidone ® K-12), larch arabogalactan, Veegum ® , polyethylene glycol, waxes, sodium alginate, and the like.

[0093] Fillers or diluents increase bulk in the pharmaceutical formulation. Such compounds include e.g., lactose; starch; mannitol; sorbitol; dextrose; microcrystalline cellulose such as Avicel®; dibasic calcium phosphate; dicalcium phosphate dihydrate; tricalcium phosphate; calcium phosphate; anhydrous lactose; spray-dried lactose; pregelatinzed starch; compressible sugar, such as Di-Pac® (Amstar); hydroxypropylmethylcellulose; sucrose-based diluents; confectioner's sugar; monobasic calcium sulfate monohydrate; calcium sulfate dihydrate; calcium lactate trihydrate; dextrates; hydrolyzed cereal solids; amylose; powdered cellulose; calcium carbonate; glycine; kaolin; sodium chloride; inositol; bentonite; and the like.

[0094] Glidants improve the flow characteristics of a powder mixtures. Such compounds include, e.g., colloidal silicon dioxide such as Cab-o-sil®; tribasic calcium phosphate, talc, com starch, DL-leucine, sodium lauryl sulfate, magnesium stearate, calcium stearate, sodium stearate, kaolin, and micronized amorphous silicon dioxide (Syloid®) and the like.

[0095] Lubricants are compounds which prevent, reduce, or inhibit adhesion or friction of materials. Exemplary lubricants include, e.g., stearic acid; calcium hydroxide, talc; a hydrocarbon such as mineral oil, or hydrogenated vegetable oil such as hydrogenated soybean oil (Sterotex ® ), Lubritab ® , Cutina ® ; higher fatty acids and their alkali-metal and alkaline earth metal salts, such as aluminum, calcium, magnesium, zinc, stearic acid, sodium stearates, magnesium stearate, glycerol, talc, waxes, Stearowet ® , boric acid, sodium acetate, leucine, a polyethylene glycol or a methoxypolyethylene glycol such as Carbowax™, sodium oleate, glyceryl behenate (Compitrol 888 ® ), glyceryl palmitostearate (Precirol ® ), colloidal silica such as Syloid™, Carb-O-Sil ® , a starch such as corn starch, silicone oil, a surfactant, and the like. Hydrophilic lubricants include, e.g., sodium stearyl fumarate (currently marketed under the trade name PRUV ® ), polyethylene glycol (PEG), magnesium lauryl sulfate, sodium lauryl sulfate (SLS), sodium benzoate, sodium chloride, and the like.

[0096] Disintegrants facilitate breakup or disintegration of the pharmaceutical formulation after administration. Examples of disintegrants include a starch, e.g., a natural starch such as corn starch or potato starch, a pregelatinized starch such as National 1551 or Amijel®, or sodium starch glycolate such as Promogel® or Explotab®; a cellulose such as a wood product, microcrystalline cellulose, e.g., Avicel®, Avicel® PH101, Avicel® PHI 02, Avicel® PHI 05, Elcema® PI 00, Emcocel®, Vivacel®, Ming Tia®, and Solka-Floc®, methylcellulose, croscarmellose, or a cross-linked cellulose, such as cross-linked sodium carboxymethylcellulose (Ac-Di-Sol®), cross-linked carboxymethylcellulose, or cross-linked croscarmellose; a cross- linked starch such as sodium starch glycolate; a cross-linked polymer such as crospovidone; a cross-linked polyvinyl pyrrolidone; alginate such as alginic acid or a salt of alginic acid such as sodium alginate; a clay such as Veegum® HV (magnesium aluminum silicate); a gum such as agar, guar, locust bean, Karaya, pectin, or tragacanth; sodium starch glycolate; bentonite; a natural sponge; a resin such as a cation-exchange resin; citrus pulp; sodium lauryl sulfate; sodium lauryl sulfate in combination starch; and the like.

[0097] Polymeric carriers include compounds such as polyvinyl pyrrolidone, e.g., polyvinylpolyvinyl pyrrolidone K12, polyvinyl pyrrolidone K17, polyvinyl pyrrolidone K25, or polyvinyl pyrrolidone K30, polyvinyl pyrrolidone vinyl acetate (PVPVA 64), hydroxypropylmethylcellulose (HPMC), hydroxypropylmethylcellulose acetyl succinate (HPMC AS), and methylmethacrylate polymers (Eudragit polymers) and the like.

[0098] Stabilizers include compounds such as any anti-oxidation agents, e.g., butylated hydroxytoluene (BHT), sodium ascorbate, and tocopherol; buffers, acids, and the like.

[0099] Surfactants include compounds such as sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, e.g., Pluronic® (BASF), d-a- tocopheryl polyethylene glycol succinate (Vitamin E TPGS); and the like.

[00100] The aforementioned excipients are given as examples only and are not meant to include all possible choices. Other suitable excipient classes include coloring agents, granulating agents, preservatives, anti-foaming agents, plasticizers, and the like. Additionally, many excipients can have more than one role or function, or can be classified in more than one group; the classifications are descriptive only, and are not intended to limit any use of a particular excipient.

[00101] Disclosed pharmaceutical formulations are administered to patients (animals and humans) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy. It will be appreciated that the dose required for use in any particular application will vary from patient to patient, not only with the particular pharmaceutical formulation selected, but also with the nature of the condition being treated, the age and condition of the patient, concurrent medication or special diets then being followed by the patient, and other factors, with the appropriate dosage ultimately being at the discretion of the attendant physician.

[00102] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

EXAMPLES

[00103] The following examples are offered for purposes of illustration and are not intended to limit the scope of the claims provided herein. All literature citations in these examples and throughout this specification are incorporated herein by references for all legal purposes to be served thereby. The starting materials and reagents used for the synthesis of the compounds described herein may be synthesized or can be obtained from commercial sources, such as, but not limited to, Sigma-Aldrich, Acros Organics, Fluka, and Fischer Scientific. In some embodiments, the compounds provided herein are synthesized as described in US 2019/0210950, which is herein incorporated by reference. In some embodiments, the compounds provided herein are synthesized as described in US 2021/0002208, which is herein incorporated by reference. In some embodiments, the compounds provided herein are synthesized as described in WO 2021/108549, which is herein incorporated by reference. In some embodiments, the compounds provided herein are synthesized as described below in Examples 1-33.

EXAMPLE 1: Synthesis of 2-(3,5-dichloro-4-{[4-hydroxy-3-(propan-2- yl)phenyl]methyl}phenoxy)-N-(6-methoxypyridin-3-yl)acetamide (Compound 101)

[00104] Step 1 : To a solution of 2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)acetic acid (Compound 100) (100 mg, 0.3 mmol) in DCM (3 mL) was added DMF (cat). The mixture was cooled to 0°C and oxalyl chloride (57 mg, 0.45 mmol) was added. The mixture was stirred at rt for 30 min, then concentrated in vacuo to afford 2-(3,5- dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)acetyl chloride (110 mg, 95% yield) as a yellow oil.

[00105] Step 2: To solution of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)acety l chloride (110 mg, 0.3 mmol) in DCM (2 mL) was added to a mixture of 6-m ethoxypyri din-3 - amine (37 mg, 0.3 mmol) and triethylamine (61 mg, 0.6 mmol) in DCM (3 mL). The mixture was stirred at rt for lh. Water (15 mL) was added, and the resultant mixture was extracted with DCM (20 mL*3). The combined organic phase was washed with brine (20 mL), dried over Na 2 SC> 4 , concentrated in vacuo and purified by prep-HPLC to afford 2-(3,5-dichloro-4-(4- hydroxy-3-isopropylbenzyl)phenoxy)-N-(6-methoxypyridin-3-yl) acetamide (Compound 101) (30 mg, 21% yield) as a white solid. LCMS: M+H = 475.2.

EXAMPLE 2: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (pyrazin-2-yl)acetamide (Compound 102)

Compound 100 Compound 102

[00106] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(p yrazin-2-yl)acetamide (Compound 102) was synthesized according to the method of Example 1 using pyrazin-2-amine in step 2. LCMS: M+H = 446.1.

EXAMPLE 3: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (3,4-dimethylisoxazol-5-yl)acetamide (Compound 103)

[00107] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(3 ,4-dimethylisoxazol-5- yl)acetamide (Compound 103) was synthesized according to the method of Example 1 using 3,4-dimethylisoxazol-5-amine in step 2. LCMS: M+H = 463.1.

EXAMPLE 4: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (pyridazin-3-yl)acetamide (Compound 104)

Compound 100 Compound 104

[00108] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(p yridazin-3- yl)acetamide (Compound 104) was synthesized according to the method of Example 1 using pyridazin-3 -amine in step 2. LCMS: M+H = 446.1.

EXAMPLE 5: Synthesis of N-cyclohexyl-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)acetamide (Compound 105)

Compound 100 Compound 105

[00109] N-Cyclohexyl-2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl) phenoxy)acetamide (Compound 105) was synthesized according to the method of Example 1 using cyclohexanamine in step 2. LCMS: M-H = 448.2.

EXAMPLE 6: Synthesis of N-(but-2-yn-l-yl)-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)acetamide (Compound 106)

[00110] Step 1 : A sealed tube (50 mL) was charged with l-bromobut-2-yne (400 mg, 3.0 mmol) and NH 3 (10 mL, 7 M in MeOH). The mixture was stirred at 60 °C overnight. The mixture was concentrated in vacuo to afford but-2-yn-l -amine hydrobromide (400 mg, 89% yield) as a yellow oil.

[00111] Step 2: N-(But-2-yn-l-yl)-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)acetamide (Compound 106) was synthesized according to the method of Example 1 using but-2-yn-l -amine hydrobromide in step 2. LCMS: M-H = 418.1.

EXAMPLE 7: Synthesis of N-(but-2-yn-l-yl)-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)-N-methylacetamide (Compound 107)

Compound 100 Compound 107

[00112] Step 1: To a mixture of N-methylprop-2-yn-l -amine (2.0 g, 29.0 mmol) in THF (20 mL) was added tert-butyldi carbonate (18.9 g, 87.0 mmol). The mixture was cooled to 40 °C and stirred for 2.0 h. Then the mixture was concentrated in vacuo to afford tert-butyl methyl (prop-2 - yn-l-yl)carbamate (4.0 g, 82% yield) as a colorless oil.

[00113] Step 2: A solution tert-butyl methyl (prop-2-yn-l-yl)carbamate (1.0 g, 5.9 mmol) in DCM (5 mL) was added n-butyllithium (2.5M/THF) (2.8 mL, 7.1 mmol) at -70 °C. The mixture was stirred for lh and was added Iodomethane for another 1.0 h. Water (50 mL) was added, and the resultant mixture was extracted with DCM (20 mL*3). The combined organic phase was washed with brine (50 mL), dried over NaiSCL, concentrated in vacuo to afford crude tert-butyl but-2-yn-l-yl(methyl)carbamate (1.0 g, 92% yield) as a colorless oil.

[00114] Step 3: A solution of tert-butyl but-2-yn-l-yl (methyl)carbamate (1.0 g, 5.5 mmol) in DCM (2 mL) was added TFA (1.3 g, 11.0 mmol) was stirred at 0 °C for 2.0 h. The resulting mixture was concentrated in vacuo to afford N-methylbut-2-yn-l -amine (200.0 mg, 44% yield) as a colorless oil.

[00115] Step 4: N-(But-2-yn-l-yl)-2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbe nzyl)phenoxy)- N-methylacetamide (Compound 107) was synthesized according to the method of Example 1 using N-methylbut-2-yn-l -amine in step 2. LCMS: M+H = 434.1.

EXAMPLE 8: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)- N,N',N'-trimethylacetohydrazide (Compound 108)

Compound 100 Compound 108

[00116] Step 1: To a mixture of tert-butyl 1-methylhydrazine-l-carboxylate (1.0 g, 6.8 mmol) in acetonitrile (10 mL) was added formaldehyde (37 wt. % in water) (5.26 mL, 68.4 mmol). The mixture was stirred at rt 2 hours. After that time, sodium cyanoborohydride (860.0 mg, 13.7 mmol) was added into the solution. The mixture was stirred at rt 2 hours, then the mixture was quenched by water (20 mL) and extracted with EtOAc (10 mL*2). The organic phase was washed by water (20 mL) and brine (20 mL), dried over NaiSCL, concentrated in vacuum and purified by silica gel column (DCM to DCM/MeOH=10: 1) to afford tert-butyl 1,2,2-trimethylhydrazine-l-carboxylate (0.1 g, 8.4% yield) as a colorless oil.

[00117] Step 2: To a solution of tert-butyl 1,2,2-trimethylhydrazine-l-carboxylate (0.1 g, 573.9 pmol) in DCM (2 mL) was added HC1 (lM/ether) (5.7 mL, 5.7 mmol). The mixture was stirred at rt 1 hour, then concentrated in vacuum to afford 1,1,2-trimethylhydrazine dihydrochloride (70 mg, 71.1% yield) as a white solid.

[00118] Step 3: 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N,N' ,N'- trimethylacetohydrazide (Compound 108) was synthesized according to the method of Example 1 using 1,1,2-trimethylhydrazine dihydrochloride in step 2. LCMS: M+H = 425.0.

EXAMPLE 9: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (pyrimidin-5-yl)acetamide (Compound 109)

Compound 100 Compound 109

[00119] A solution of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(p yrimidin- 5-yl)acetamide (Compound 100) (0.1 g, 271 pmol), pyrimidin-5-amine (25.8 mg, 271 pmol), HATU (124 mg, 325 pmol) and DIPEA (112 pL, 2.5 eq., 677 pmol) in DMF (2 mL) was stirred at room temperature for 5.0 h. Water (10 mL) was added, and the result mixture was extracted with EtOAc (10 mL*3). The combined organic phase was washed with water (15 mL*2), brine (15 mL), dried over NaiSCL, concentrated in vacuo and purified by prep-HPLC to afford 2-(3,5- dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(pyrimidi n-5-yl)acetamide (Compound 109) (20 mg, 44.8 pmol) as a white solid. LCMS: M+H = 446.1.

EXAMPLE 10: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (pyrimidin-4-yl)acetamide (Compound 110)

Compound 100 Compound 110

[00120] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(p yrimidin-4- yl)acetamide (Compound 110) was synthesized according to the method of Example 9 using pyrimidin-4-amine. LCMS: M+H = 446.1.

EXAMPLE 11: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- ((lR,2S)-2-fluorocyclopropyl)acetamide (Compound 111)

[00121] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(( lR,2S)-2- fluorocyclopropyl)acetamide (Compound 111) was synthesized according to the method of Example 9 using (lR,2S)-2-fluorocyclopropan-l-amine. LCMS: M+H = 426.1.

EXAMPLE 12: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (3-fluoropropyl)acetamide (Compound 112)

Compound 100 Compound 112

[00122] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(3 - fluoropropyl)acetamide (Compound 112) was synthesized according to the method of Example 9 using 3-fluoropropan-l-amine. LCMS: M-H = 426.1.

EXAMPLE 13: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (6-methoxypyridazin-3-yl)acetamide (Compound 113)

[00123] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(6 -methoxypyridazin-3- yl)acetamide (Compound 113) was synthesized according to the method of Example 9 using 6- methoxypyridazin-3 -amine. LCMS: M+H = 476.2.

EXAMPLE 14: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (pyridin-3-yl)acetamide (Compound 114) [00124] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(p yridin-3-yl)acetamide (Compound 114) was synthesized according to the method of Example 9 using pyri din-3 -amine. LCMS: M+H = 445.1.

EXAMPLE 15: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (pyridin-4-yl)acetamide (Compound 115)

Compound 100 Compound 115

[00125] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(p yridin-4-yl)acetamide (Compound 115) was synthesized according to the method of Example 9 using pyridin-4-amine. LCMS: M+H = 445.2.

EXAMPLE 16: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (pyridazin-4-yl)acetamide (Compound 116)

Compound 100 Compound 116

[00126] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(p yridazin-4- yl)acetamide (Compound 116) was synthesized according to the method of Example 9 using pyridazin-4-amine. LCMS: M+H = 446.2.

EXAMPLE 17: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-l- (pyrrolidin-l-yl)ethan-l-one (Compound 117)

Compound 100 Compound 117

[00127] 2-(3 ,5-Dichloro-4-(4-hydroxy-3 -isopropylbenzyl)phenoxy)- 1 -(pyrrolidin- 1 -yl)ethan- 1 - one (Compound 117) was synthesized according to the method of Example 9 using pyrrolidine. LCMS: M+H = 422.1.

EXAMPLE 18: Synthesis of l-(azetidin-l-yl)-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)ethan-l-one (Compound 118)

Compound 100 Compound 118

[00128] l-(Azetidin-l-yl)-2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbe nzyl)phenoxy)ethan-l- one (Compound 118) was synthesized according to the method of Example 9 using azetidine. LCMS: M+H = 408.1.

EXAMPLE 19: Synthesis of N-(tert-butyl)-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)acetamide (Compound 119)

Compound 100 Compound 119

[00129] N-(tert-Butyl)-2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzy l)phenoxy)acetamide (Compound 119) was synthesized according to the method of Example 9 using 2-methylpropan- 2-amine. LCMS: M+H = 424.1.

EXAMPLE 20: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- isobutyl-N-methylacetamide (Compound 120)

Compound 100 Compound 120

[00130] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-is obutyl-N- methylacetamide (Compound 120) was synthesized according to the method of Example 9 using N,2-dimethylpropan-l -amine. LCMS: M+H = 438.2.

EXAMPLE 21: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- isobutylacetamide (Compound 121)

Compound 100 Compound 121 [00131] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-is obutylacetamide (Compound 121) was synthesized according to the method of Example 9 using 2-methylpropan- 1 -amine. LCMS: M-H = 422.1.

EXAMPLE 22: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- isopropyl-N-methylacetamide (Compound 122)

Compound 100 Compound 122

[00132] 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-is opropyl-N- methylacetamide (Compound 122) was synthesized according to the method of Example 9 using N-methylpropan-2-amine. LCMS: M-H = 422.1.

EXAMPLE 23: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (2-hydroxyethyl)-N-methylacetamide (Compound 123)

Compound 100 Compound 123

[00133] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(2 -hydroxyethyl)-N- methylacetamide (Compound 123) was synthesized according to the method of Example 9 using 2-(methylamino)ethan-l-ol. LCMS: M-H = 424.1.

EXAMPLE 24: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)- N,N'-dimethylacetohydrazide (Compound 124)

Compound 100 Compound 124

[00134] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N,N' - dimethylacetohydrazide (Compound 124) was synthesized according to the method of Example 9 using 1,2-dimethylhydrazine. LCMS: M+H = 411.1.

EXAMPLE 25: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (2-fluoroethyl)-N-methylacetamide (Compound 125)

Compound 100 Compound 125

[00135] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(2 -fluoroethyl)-N- methylacetamide (Compound 125) was synthesized according to the method of Example 9 using 2-fluoro-N-methylethan-l -amine. LCMS: M+H = 428.1.

EXAMPLE 26: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- isopropylacetamide (Compound 126)

Compound 100 Compound 126

[00136] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-is opropylacetamide (Compound 126) was synthesized according to the method of Example 9 using propan-2-amine. LCMS: M+H = 410.1.

EXAMPLE 27: Synthesis of N-cyclobutyl-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)acetamide (Compound 127)

Compound 100 Compound 127

[00137] N-Cyclobutyl-2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl) phenoxy)acetamide (Compound 127) was synthesized according to the method of Example 9 using cyclobutanamine. LCMS: M+H = 422.2.

EXAMPLE 28: Synthesis of N-allyl-2-(3,5-dichloro-4-(4-hydroxy-3- isopropylbenzyl)phenoxy)-N-methylacetamide (Compound 128) [00138] N-Allyl-2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)pheno xy)-N- methylacetamide (Compound 128) was synthesized according to the method of Example 9 using N-methylprop-2-en-l -amine. LCMS: M-H = 420.1.

EXAMPLE 29: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- methyl-N-propylacetamide (Compound 129)

Compound 100 Compound 129

[00139] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-me thyl-N- propylacetamide (Compound 129) was synthesized according to the method of Example 9 using N-methylpropan-1 -amine. LCMS: M-H = 422.1.

EXAMPLE 30: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- ethyl-N-methylacetamide (Compound 130)

Compound 100 Compound 130

[00140] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-et hyl-N- methylacetamide (Compound 130) was synthesized according to the method of Example 9 using N-methylethanamine. LCMS: M+H = 410.2.

EXAMPLE 31: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- methyl-N-(2,2,2-trifluoroethyl)acetamide (Compound 131)

Compound 100 Compound 131

[00141] To a solution of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)aceti c acid (100 mg, 0.3 mmol) in DMF (3 mL) was added 2,2,2-trifluoro-N-methylethan-l -amine (134 mg, 0.9 mmol ), l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI) (77 mg, 0.4 mmol), 1-hydroxybenzotriazole (HOBT) (55 mg, 0.4 mmol) and N,N-diisopropylethylamine (105 mg, 0.8 mmol). The mixture was stirred at rt overnight. Water (20 mL) was added. The mixture was extracted with EtOAc (15 mL*2). The combined organic phase was washed with brine (20 mL), dried over NaiSCL, concentrated in vacuo and purified by prep-HPLC to afford 2- (3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-meth yl-N-(2,2,2- trifluoroethyl)acetamide (Compound 131) (50 mg, 36% yield) as a white solid. LCMS: M-H = 462.1.

EXAMPLE 32: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N- (2,2-difluoroethyl)-N-methylacetamide (Compound 132)

Compound 100 Compound 132

[00142] 2-(3,5-Dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-N-(2 ,2-difluoroethyl)-N- methylacetamide (Compound 132) was synthesized according to the method of Example 31 using 2,2-difluoro-N-methylethan-l -amine hydrochloride. LCMS: M+H = 446.1.

EXAMPLE 33: Synthesis of 2-(3,5-dichloro-4-(4-hydroxy-3-isopropylbenzyl)phenoxy)-l- morpholinoethan-l-one (Compound 133)

Compound 100 Compound 133

[00143] 2-(3 , 5 -Diehl oro-4-(4-hy droxy-3 -i sopropylbenzyl)phenoxy)- 1 -morpholinoethan- 1 -one (Compound 133) was synthesized according to the method of Example 31 using morpholine. LCMS: M-H = 436.0.

EXAMPLE 34: FAATT Substrate Evaluation

[00144] Purified recombinant human FAAH (rhFAAH) was purchased from Cayman Chemical (Ann Arbor, MI, USA). The total volume for each incubation was 400 pL containing a final 0.5 ng/pL rhFAAH, 1 pM test compound, 1.25% ethanol or 1 pM PF-3845 (FAAH inhibitor), and 0.1% bovine serum albumin in Tris-EDTA buffer at pH 8.0). The positive control was LL- 341001. The incubation was conducted at the room temperature. At 0, 5, 15, 30 and 60 minutes, an aliquot of 30 pL reaction mixtures was removed and mixed with 300 pL acetonitrile containing 5 ng/mL terfenadine and 10 ng/mL tolbutamide as internal standards to quench the reaction. The resulting mixture was centrifuged at 4000 rpm, 4°C for 15 minutes, and 100 pL supernatant was ready for LC-MS/MS analysis to measure the formation of acid metabolite. LC-MS/MS Analysis [00145] Acquity Ultra Performance LC system from Waters was used for sample analysis. The chromatography was performed on a reverse phase Kinetex 2.6 pm Cl 8 column, 2.1 x 30 mm, 100 A. The mobile phase A comprised of 0.1% formic acid in water and mobile phase B comprised of 0.1% formic acid in acetonitrile with a 2-min run time at the flow rate of 0.8 mL/min for the acid metabolite from positive control or a 1.5 min run time at the flow rate of 0.9 mL/min for the acid metabolite of test compounds. The mass spectrometer (API-5500 and API Q Trap 4000 Applied Biosystems/MDS SCIEX Instruments, Framingham, MA, USA) was operated under ESI positive or negative ion MRM mode.

Data Analysis

[00146] The formation of acid metabolite was monitored and quantified using one calibration point of 1 pM. The observed rate constant (ke) for the acid metabolite formation was calculated by plotting the metabolite concentration versus time of incubation with the slope being ke and is shown in Table 1.

Table 1

A = ke is more than or equal to 0.1; B = ke is less than 0.1 and more than 0; C = ke is 0; NT = not tested.

EXAMPLE 35: In Vitro Stability Evaluation in Mouse Plasma

[00147] Male CD-I mouse plasma is purchased from BioIVT (catalog #MSE00PLK2YNN) and thawed in a 37°C water bath with pH adjusted to 7.4 on Study day. After a pre-warm period of 15 minutes in a 37°C water bath, 398 pL plasma is spiked with an aliquot of 2 pL stock solution of the test compound or positive control (propantheline) in dimethyl sulfoxide (DMSO) to achieve a final concentration of 1 pM with 0.5% DMSO. After a thorough mix, the mixture is placed back to the 37°C water bath for incubations. At 0, 15, 30, 60, and 120 minutes, an aliquot of 30 pL reaction mixtures is removed and mixed with 300 pL acetonitrile containing 5 ng/mL terfenadine and 10 ng/mL tolbutamide as internal standards to quench the reaction. The resulting mixture is centrifuged at 4000 rpm, 4°C for 15 minutes, and 100 pL supernatant is removed and mixed with 100 pL water for liquid chromatography -tandem mass spectrometry (LC-MS/MS) analysis.

LC-MS/MS Analysis

[00148] Shimadzu LC 30-AD HPLC system is used for sample analysis. The chromatography is performed on a reverse phase Kinetex 2.6 pm C18 column, 3.0 x 30 mm, 100 A. The mobile phase A comprises of 0.1% formic acid in water and mobile phase B comprises of 0.1% formic acid in acetonitrile with a 2-min run time. The mass spectrometer (API-4000 and API Q Trap 4500 Applied Biosystems/MDS SCIEX Instruments, Framingham, MA, USA) is operated under electrospray ionization (ESI) positive or negative ion multiple reaction monitoring (MRM) mode.

Data Analysis

[00149] Percent compound remaining at a specific time point is calculated based on the peak area ratios at time 0 (as 100%). The observed rate constant (k 0 bs) for the metabolism of test compounds is calculated by plotting the natural log of percentage compound remaining versus time of incubation with the slope being k 0 bs. The half-life (ti / 2) is determined according to the following equation: ti/2 = 0.693/k O bs.

EXAMPLE 36: In Vivo Tissue Distribution Studies in Male CD-I Mice [00150] Male CD-I mice (n = 6 per group), 7-10 weeks old, are acclimated to the study room for a minimum of 3 days before dose administration in the studies. The test compounds are formulated in 1% N-methyl-2-pyrrolidone (NMP) and 1% Solutol in phosphate buffered saline (PBS) at 0.1 mg/mL clear solution and the dose volume was 10 mL/kg. The peripherally restricted FAAH inhibitor LL-650021 is formulated in 0.5% carboxymethyl cellulose in water at 0.1 mg/mL and the dose volume is 10 mL/kg. The concentrations of the formulation are determined to meet the acceptance criteria of within 20% of the target values.

[00151] The test compounds are administered to non-fasted mice at 1 mg/kg via subcutaneous (SC) injection or oral gavage (PO) with or without pretreatment of 1 mg/kg LL-650021 1 hour prior to test compound administration. At 1, 4, and 8 hours post-dose, the animals (n = 2 per time point) are euthanized using CO2 inhalation. A blood sample (0.3 mL) is collected from saphenous vein or other suitable site into pre-chilled K2EDTA tube and placed on wet ice and brain and liver are harvested. The blood samples are centrifuged at 3200 g, 4°C for 10 minutes and the plasma samples are transferred into polypropylene tubes, quick frozen over dry ice and kept at -60°C or lower until analysis. The tissues are washed with cold saline, wiped dry, weighed, and then homogenized in 15 mM PBS (pH 7.4):methanol = 2:1 buffer at the ratio of 1:10 (1 g tissue with 10 mL buffer resulting in 11 -fold dilution). The tissue homogenates are kept at -60°C or lower until analysis.

Sample Extraction

[00152] The plasma and tissue homogenates are extracted by protein precipitation. An aliquot of 10-50 pL plasma or 40-50 pL tissue homogenates is protein precipitated by adding 200-800 pL acetonitrile containing internal standards (10 ng/mL LL- 120001 and 100 ng/mL of celecoxib, dexamethasone, glyburide, labetalol, tolbutamide, and verapamil), vortex-mixed for 10 min at 800 rpm and centrifuged at 4000 rpm, 4°C for 15 minutes. The supernatant is transferred to the 96-well plate and centrifuged at 4000 rpm, 4°C for 5 minutes before injected for LC-MS/MS analysis, or 200 pL supernatant is transferred to the 96-well plate, evaporated to dryness under a stream of nitrogen at 25°C, reconstituted with 50 pL of 70% acetonitrile, vortex-mixed for 10 min at 800 rpm and centrifuged at 4000 rpm, 4°C for 5 minutes before injected for LC-MS/MS analysis.

LC-MS/MS Analysis

[00153] Acquity Ultra Performance LC system from Waters is used for sample analysis. The separations are performed on a ACQUITY UPLC BEH C18 column (50 x 2.10 mm; 1.7 pm) at 50°C with a flow rate of 0.6 mL/min. Mobile phase A consists of 2 mM ammonium acetate in methanol: water 5:95 and mobile phase B consists of 2 mM ammonium acetate in acetonitrile: water 95:5. Chromatography uses a linear gradient starting at 2% mobile phase B, 2% to 90% mobile phase B over 2.6 minutes, maintained at 90% B wash for 0.2 minutes, and a re-equilibration at 2% B for 0.2 minutes. An aliquot of 2-9 pL sample is injected. The mass spectrometer (API-6500+, Applied Biosystems/MDS SCIEX Instruments, Framingham, MA, USA) is operated under ESI in positive ion or negative ion MRM mode.

EXAMPLE 37: In vitro Prodrug and Agonist TRjl Receptor Selectivity [00154] LL-341070, a thyromimetic prodrug of Formula (F) described herein which delivers LL-341070A, a potent and selective small molecule agonist of thyroid hormone receptor (TR) beta following fatty acid amide hydrolase (FAAH)-mediated conversion, were evaluated for potency and selectivity for the thyroid hormone beta receptor (TRP). In vitro potency was determined via test compounds administered to luciferase-based TR reporter cell lines, using thyroid hormone (T3) as a positive control. Table 2 depicts the potency profiles of LL-341070 prodrug and LL-341070 A active metabolite against TRp and TRa as measured in half maximal effective concentration (EC50), with selectivity measurement adjusted for the TRa-bias of T3 in the assays. Both LL-341070 and LL-341070A show enhanced selectivity for TRP, with LL- 341070 A showing enhanced potency.

Table 2 EXAMPLE 38: LL-341070A Enhances Oligodendrocyte Progenitor Cell Differentiation In Vitro

[00155] To profile LL-341070A, in vitro oligodendrocyte progenitor cell (OPC) assays were performed on primary OPC cultures generated from brains of E14.5 PLP-EGFP C57B1/6 mouse embryos. Thyroid hormone (T3), known to induce OPC differentiation and remyelination, was used as a positive control at 10 ng/mL. Primary OPC cultures were treated with LL-341070A compound concentrations ranging from about InM to about lOOOnM. After OPC differentiation was induced with or without LL-341070A compound for 5 days (N=6/concentration), cells were fixed and stained for myelin basic protein (MBP), normalized to total cell count.

[00156] Figure 1 depicts the active metabolite of the LL-341070 prodrug, LL-341070A, enhanced oligodendrocyte differentiation in vitro in an oligodendrocyte progenitor cell assay (EC50 = 1.4 nM). Enhanced oligodendrocyte differentiation was shown to be relatively constant as a function of LL-341070 A treatment concentration.

EXAMPLE 39: Thyromimetic Treatment Enhances 24-OHC Synthesis In Vivo [00157] The ability of thyromimetics to accelerate the remyelination process in vivo was assessed by measuring the fractional synthesis of 24-hydroxycholesterol (24-OHC) in the brains of rats following cuprizone-induced demyelination. As shown in Figure 2, the cuprizone demyelination model assessed n=10F rats/group. The effect of thyromimetic treatment on the fractional synthesis of 240HC was measured in brain and plasma using deuterated water labeling of 240HC in a cuprizone demyelination model following withdrawal from 0.6% cuprizone diet, during the period of active remyelination. At withdrawal from a 3 week 0.6% cuprizone diet, rats were provided deuterated water with administration of LL-341070 at 30 or 100 pg/kg for 3 weeks, then 24-OHC deuterium enrichment and labeling pattern were measured in cortex and corpus callosum. LL-341070 induced a dose-dependent increase in deuterated 24- OHC compared to vehicle controls, suggesting an increased rate of myelin synthesis. As shown in Figure 2, thyromimetic treatment enhances the fractional synthesis rate (FSR) of 248- hydroxy cholesterol in brain and demonstrates a strong correlation with 240HC FSR in plasma. 240HC Fractional Synthesis Rate (FSR) was calculated based on data collected from tissue and plasma underwent alkaline hydrolysis and derivatization for GC/MS or LC/MS analysis of deuterated 240HC. Plasma analysis of 240HC measured by LC/MS as fraction of labeled to total plasma 240HC by Ardena Biosciences. Fractional synthesis was calculated by Mass Isotopomer Distribution Analysis using precursor 2H enrichment in body water from liver palmitate 6. Statistics: Data were analyzed by one-way ANOVA with Tukey’s multiple comparisons test and are represented as mean +/- SEM. *p<0.05. [00158] Compound potency, pharmacokinetics and target engagement were confirmed for LL- 341070 prior to testing efficacy in remyelination models, including oligodendrocyte precursor cell differentiation in vitro and experimental autoimmune encephalitis in vivo.

EXAMPLE 40: Engagement of TRji in Brain Increases Expression of T3-Target Genes In Vivo

[00159] Figure 3 depicts TR-b target engagement in brain is demonstrated by increased expression of T3 -responsive target genes in vivo. Single PO administration of LL-341070 (ranging from about 0.1 pg/kg to about 300 pg/kg) or T3 (about 300 pg/kg) in male C57BL/6 mouse increases expression of Hr, Dio3, Klf9 (quantified by QuaniPlex) and composite average log2 fold change in brain. Klf9, a T3-responsive gene linked to myelin regeneration in vitro, is upregulated at various treatment concentrations. This expression increase was confirmed in the brain of a rat cuprizone model (as previously discussed) with 21 day repeat administration of LL- 341070 at 30 pg/kg or 100 pg/kg, or T3 dosed at 300 pg/kg (quantified by Nanostring). Interestingly, Dio3 has an enhanced expression increase with repeat dosing.

EXAMPLE 41: In Vivo Tissue Distribution Demonstrates Enhanced Brain Exposure of Active Compound Compared to Prodrug

[00160] In vivo brain exposure of active compound compared to prodrug was assessed via tissue distribution (TD) assay in mouse and rat cuprizone model, measured as brain exposure ratio of brain to plasma following thyromimetic treatment. As shown in Table 3, single PO administration of LL-341070 (100 pg/kg) or LL-341070A(100 pg/kg), in male C57BL/6 mouse measured in brain and plasma, demonstrates enhanced brain exposure of active compound LL- 341070A compared to prodrug LL-341070, leading to a brain-to-plasma AUC ratio >1 for LL- 341070A, wherein AUC is 0-24 hr. Data shows AUC of LL-341070A in brain is ~7-fold higher than prodrug LL-341070. Table 3 also depicts brain-to-plasma AUC ratio. As shown in Figure 4, 21 days of repeat administration of LL-341070 (30 pg/kg or 100 pg/kg) or LL-341070A(30 pg/kg or 100 pg/kg), in rat cuprizone model measured in brain and plasma 4 hours post-final dose, demonstrates enhanced brain exposure of active compound LL-341070A compared to prodrug LL-341070.

Table 3

EXAMPLE 42: LL-341070 Improves In Vivo Clinical Scoring and Histology in Mouse EAE Model

[00161] As shown in Figure 5, LL-341070 efficacy was assessed in a mouse prophylactic experimental autoimmune encephalitis (EAE) model wherein following EAE induction by MOG35-55/CFA + PTX, disease onset 8-18 days after induction, with EAE scoring on day 7-28 after induction. The EAE model assessed n=12F C57B1/6 mice/group, administered LL-341070 (10 pg/kg to 100 pg/kg) or vehicle PO daily after EAE induction. LL-341070 administered daily in a prophylactic paradigm dose-dependently improved median day of disease onset and decreased maximum disease severity. Histological analysis of spinal cord 28 days after immunization demonstrated a reduction of inflammatory foci, apoptotic cell count, and reduced area of demyelination by H&E and MBP staining. LL-341070 improves the clinical scoring mean and histological endpoints of inflammation and demyelination in a mouse EAE model. Clinical scores were determined by blinded observer. Histology analyzed in spinal cord sample (demyelination score assessed by % demyelinated area in anti-MBP stain, inflammatory foci refers to # of groups of >20 cells / section in H&E stain). Statistics: Median day of onset in EAE compared using Wilcoxon’s survival test.

EXAMPLE 43: FAAH Expression is Enriched in Brain

[00162] As shown in Figure 6, brain-directed thyromimetic prodrugs (such as ABX-002 which is Compound 1 described herein, activated to ABX-002A) that are activated by fatty acid amide hydrolase (FAAH) were utilized to elucidate mechanisms by which thyromimetics disrupt the thyroid hormone axis (THA). The delivery of potent thyromimetics was altered to help identify whether feedback control on THA derives from central (hypothalamic) or peripheral (pituitary) mechanisms and potentially enhance therapeutic index of thyromimetics. These studies were performed using recombinant FAAH, tissue-derived S9 fractions, in vivo tissue distribution (TD), gene expression in brain and liver, and effects on T4 in mice as a marker of THA disruption. Northern blot assay confirmed FAAH expressed across species (rodent and human), with relative mRNA FAAH expression enhanced in the brain. FAAH specific activity (cleavage of AMC assay) from tissue-derived S9 fractions of different organs (liver, brain, small intestine) across species (mouse, rat, non-human primate, human), calculated as a percentage of liver activity, was shown to be increased in brain of human and non-human primate. EXAMPLE 44: FAATT Expression Enhances Delivery of ABX-002A to Brain [00163] To assess delivery, concentrations of ABX-002A in brain, liver, kidney, lung, and heart were measured 1 hour after SC administration of 30 different prodrugs of ABX-002A. As shown in Figure 7, brain-to-plasma ratios were increased relative to ABX-002A for the prodrugs, while tissue-to-plasma ratios for peripheral organs (liver, kidney, lung, and heart) showed a linear (constant) tissue-to-plasma relationship. Data shows FAAH is highly expressed in the CNS and ABX prodrugs enhance delivery of active metabolite to the brain by >3 Ox with brain-to-plasma ratios >1. In organs other than the brain, data shows tissue concentrations are driven by plasma concentrations of the active metabolite ABX-002A.

EXAMPLE 45: Global and Peripheral FAATT Inhibitors Alter Metabolite Distribution in Mice

[00164] The ability of globally-penetrant and peripherally-restricted FAAH inhibitors (GFI & PFI, respectively) to alter distribution of ABX-002 and ABX-002A was assessed. Table 4 depicts the potency profiles (measured in apparent ICsos (nM)) of peripheral and global FAAH inhibitors: LL-650177 (PFI), URB9373 (PFI), and PF-044578454 (GFI) obtained after 30 min preincubation with recombinant human FAAH and 7-amino-4-methylcourmarin (AMC).

Table 4

[00165] Figure 8 shows plasma, liver, and brain concentrations after co-dosing prodrug (ABX- 002) with or without PFI or GFI. Prodrug levels did not change or slightly increased with FAAH inhibition. Active metabolite (ABX-002A) levels decreased in plasma & liver with PFI & in all organs with GFI. Table 5 depicts the inhibition of active metabolite (LL-650177 or PF- 044578454) in AUC in the plasma, liver, and brain after prodrug (ABX-002) co-dosing. Tissue distribution studies in mice confirm global & peripheral inhibition of FAAH.

Table 5

EXAMPLE 46: Induction of T3-Regulated Genes in View of Prodrug and FAAH Inhibitors

[00166] Female C57BL/6 mice (n=5/group), 6-8 weeks old, were acclimated to the study room for at least 3 days before dose administration in the studies. Non-fasted mice were given a single dose of PFI or vehicle orally (PO) on Day 0 at time = -1 hour. Single dose administered at 5 mL/kg based on most recent body weight, collected once for the duration of the study.

Following the PFI or vehicle dose, animals were given a single dose of test article at time = 0 hour. One group (n=5) was PO administered 300ug/kg of T3 only at time = 0 hours. Approximately 4 hours post test article dose (t = 4 hours), animals were humanely euthanized and brain, liver, heart, pituitary, spinal cord, and plasma samples were harvested.

Sample Processing

[00167] a. Expression Analysis Samples - At endpoint, multiple organs were harvested and tissues processed immediately as described below.

[00168] i. Brain: For each mouse, the cranium was opened and the brain removed. The cerebellum was sectioned away and the cerebral cortex was hemisected sagittally and the left half collected. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the cerebral cortical specimen was placed into a tube containing 1.2 mL pre-chilled RNALater and stored at 4°C.

[00169] ii. Liver: For each mouse, one liver biopsy (100-150 mg) was collected from the left lateral liver lobe. After rinsing extraneous blood from the biopsy with ice cold 0.9% NaCl, the sample was placed into 1.2 mL pre-chilled RNALater and stored at 4°C. iii. Left Ventricle: For each mouse, the left ventricle (LV) blood cleared using PBIs standard methods, and half of LV free wall was collected. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the LV free wall was placed into 1.2 mL pre-chilled RNALater and stored at 4°C. LV tissue was retained at PBI for potential future analyses or until appropriate genes can be identified for up to 6 months following conclusion of the in-life phase of study. Sample disposition was confirmed prior to disposal.

[00170] iv. Pituitary Gland: For each mouse, after removing the brain the pituitary gland was harvested. After rinsing extraneous blood from the pituitary with ice cold 0.9% NaCl, the specimen was placed into 0.15 mL pre-chilled RNALater and stored at 4°C. Pituitary tissue was retained at PBI for potential future analyses or until appropriate genes can be identified for up to 6 months following conclusion of the in-life phase of study. Sample disposition was confirmed prior to disposal.

[00171] b. Pharmacokinetic Samples - At endpoint, blood and tissue specimens were processed immediately as described below. Samples for PK analysis were retained at PBI at -80°C for up to 90 days following conclusion of the in-life phase of study.

[00172] i. Plasma: whole blood (-300 pL) was collected on K3EDTA via cardiac puncture under isoflurane anesthesia. Blood was immediately placed on wet ice. Following the conclusion of takedown procedures, blood was centrifuged at 4°C for 10 minutes at 10,000xg. Plasma (~125pL) was aliquoted to appropriately labelled tubes and flash frozen.

[00173] ii. Liver: For each mouse, one liver biopsy (30-50 mg) was collected from the left lateral liver lobe. After rinsing extraneous blood from the biopsy with ice cold 0.9% NaCl, the sample was placed into an appropriately labelled tube and flash-frozen in liquid nitrogen.

[00174] iii. Brain: For each mouse, a mid-brain biopsy (30-50 mg) was collected from the right cerebral cortex. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the biopsy was placed into an appropriately labelled tube and flash frozen.

[00175] iv. Left Ventricle: For each mouse, the left ventricle (LV) will have blood cleared using PBIs standard methods, and half LV free wall was collected. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the LV free wall was placed into an appropriately labelled tube and flash frozen.

Target Engagement

[00176] Changes in the expression of select genes identified through transcriptomic analysis were measured from purified RNA using a hybridization-based in situ RNA quantification method (NanoString, Seattle, WA). Briefly, fresh tissues were collected in RNALater™ Stabilization Solution, catalog #AM7021 (ThermoFisher Scientific; Carlsbad, CA) and frozen at -20°C until ready for RNA extraction. Whole blood was collected in MiniCollect K2EDTA tubes, catalog #450480 Greinder Bio-one GmbH (Kremsmunster, Austria), via terminal cardiac puncture and processed to plasma by centrifuging at 2000 x g for 10 minutes at 4°C. For RNA extraction, tissues were homogenized using a bead homogenizer in TRIzol Reagent, catalog # 15596026 (ThermoFisher Scientific), and RNA was extracted according to manufacturer’s protocols and purified using Econospin RNA Mini Spin Columns for RNA (Ephoch Life Sciences, Missouri City, TX, catalog #1940-250) following manufacturer’s protocols. Specific gene probes were designed by NanoString Bioinformatics Team using an identified target sequence based on the NCBI Reference Sequence (RefSeq) database. Custom probes were synthesized by Integrated DNA Technologies (IDT; Coralville, IA). mRNA expression was analyzed on an nCounter® SPRINT Profiler NanoString system using a multiplexing approach with nCounter PlexSet-12 Reagent Pack, catalog #PS-GX-PTK-12 (CSO) according to manufacturer’s protocols (NanoString, Inc, Seattle, WA).

Data Analysis

[00177] T3-target genes are increased after a single administration of drug with the relative activity in brain vs. liver determined by prodrug and/or FAAH inhibition. Relative activity in brain vs. liver (as a marker of peripheral activity) shifts >1500-fold across the different dosing paradigms. Figures 9A, 9B, and 9C show induction of T3 -regulated genes in brain (blue) & liver (orange) 4 h after single administration of (A) active metabolite or (B) prodrug alone or (C) prodrug + PFI (URB937). RNA analyzed by Nanostring; Mean fold change of multiple genes was calculated on a log2 scale and normalized to data obtained for 300 mg/kg of T3. PFI administration reduced potency of prodrugs on activation of T 3 -regulated genes in the liver by >10x, without affecting activity or exposure in the brain. PFIs also decreased potency on the THA, consistent with negative feedback based on circulating peripheral metabolite rather than brain exposure. Thus, use of a PFI allowed separation of on-target brain effects from those on THA.

EXAMPLE 47: T4 Parallels Peripheral Activity

[00178] Female C57BL/6 mice (n=5/group), 6-8 weeks old, were acclimated to the study room for at least 3 days before dose administration in the studies. Mice were dosed at 5 mL/kg based on most recent body weight, collected once for the duration of the study. Based on most recent body weight, collected once for the duration of the study, mice were placed into weight-matched treatment dosing cohorts. Mice were given a single administration of PFI or vehicle orally (PO) daily (n=5/group) for 7 days at time = -1 hour. Following the PFI (100 pg/kg) or vehicle dose (10 mL/kg, p.o.), animals were given test article daily at time = 0 hour. Test article administration one of eight dose levels (0.1, 0.3, 1, 3, 10, 30, 100 or 300 pg/kg) on Days 1 - 7 for a total of seven doses. Mice were dosed PO, QD for 7 days with (A) active metabolite or (B) prodrug alone; (C) prodrug + PFI (LL-650177) or (D) prodrug + GFI. Approximately 4 or 8 hours post test article dose (t = 4 hours or t = 8 hours), animals were humanely euthanized using standard procedures, and brain, liver, and plasma samples were harvested. RNA from samples harvested 4 hours after final dose was quantified using a hybridization-based in situ RNA quantification method (NanoString, Seattle, WA), as described below. RNA from samples harvested 8 hours after final dose was quantified using a hybridization-based in situ RNA quantification method (QuantiGene Plex), as described below. On the final day of dosing, mice were dosed on a timetable to mitigate the influence of diurnal effects on thyroid hormone sensitive gene expression. Thus, treatment groups were balanced for “time of day” at endpoint sacrifice. Mice were anesthetized 4 or 8 hours after final dosing, have blood collected via retro- orbital puncture, and euthanized using standard procedures. Immediately following euthanasia, tissues were harvested and processed per the following procedures.

Sample Processing

[00179] a. Expression Analysis Samples - At endpoint, multiple organs were harvested and tissues processed immediately as described below.

[00180] i. Brain: For each mouse, the cranium was opened and the brain removed. The cerebellum was sectioned away and the cerebral cortex was hemisected sagittally and the left half collected. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the cerebral cortical specimen was placed into a tube containing 1.2 mL pre-chilled RNALater and stored at 4°C.

[00181] ii. Liver: For each mouse, one liver biopsy (100-150 mg) was collected from the left lateral liver lobe. After rinsing extraneous blood from the biopsy with ice cold 0.9% NaCl, the sample was placed into 1.2 mL pre-chilled RNALater and stored at 4°C. iii. Left Ventricle: For each mouse, the left ventricle (LV) blood cleared using PBIs standard methods, and half of LV free wall was collected. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the LV free wall was placed into 1.2 mL pre-chilled RNALater and stored at 4°C. LV tissue was retained at PBI for potential future analyses or until appropriate genes can be identified for up to 6 months following conclusion of the in-life phase of study. Sample disposition was confirmed prior to disposal.

[00182] iv. Pituitary Gland: For each mouse, after removing the brain the pituitary gland was harvested. After rinsing extraneous blood from the pituitary with ice cold 0.9% NaCl, the specimen was placed into 0.15 mL pre-chilled RNALater and stored at 4°C. Pituitary tissue was retained at PBI for potential future analyses or until appropriate genes can be identified for up to 6 months following conclusion of the in-life phase of study. Sample disposition was confirmed prior to disposal.

[00183] b. Pharmacokinetic Samples - At endpoint, blood and tissue specimens were processed immediately as described below. Samples for PK analysis were retained at PBI at -80°C for up to 90 days following conclusion of the in-life phase of study.

[00184] i. Plasma: whole blood (-300 pL) was collected on K3EDTA via cardiac puncture under isoflurane anesthesia. Blood was immediately placed on wet ice. Following the conclusion of takedown procedures, blood was centrifuged at 4°C for 10 minutes at 10,000xg. Plasma (~125pL) was aliquoted to appropriately labelled tubes and flash frozen. [00185] ii. Liver: For each mouse, one liver biopsy (30-50 mg) was collected from the left lateral liver lobe. After rinsing extraneous blood from the biopsy with ice cold 0.9% NaCl, the sample was placed into an appropriately labelled tube and flash-frozen in liquid nitrogen. [00186] iii. Brain: For each mouse, a mid-brain biopsy (30-50 mg) was collected from the right cerebral cortex. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the biopsy was placed into an appropriately labelled tube and flash frozen.

[00187] iv. Left Ventricle: For each mouse, the left ventricle (LV) will have blood cleared using PBIs standard methods, and half LV free wall was collected. After rinsing extraneous blood from the tissue with ice cold 0.9% NaCl, the LV free wall was placed into an appropriately labelled tube and flash frozen.

Target Engagement

[00188] Tissue samples were prepared for biochemical analysis by cryopowdering on liquid nitrogen, and lysed using PBFs standard methods. Changes in the expression of select genes identified through transcriptomic analysis (mRNA expression) were measured from purified RNA using a hybridization-based in situ RNA quantification method (NanoString or QuantiGene Plex). Target gene expression data was presented as a ratio to the geometric mean of appropriately expressed normalization genes. Briefly, fresh tissues were collected in RNALater™ Stabilization Solution, catalog #AM7021 (ThermoFisher Scientific; Carlsbad, CA) and frozen at -20°C until ready for RNA extraction. Whole blood was collected in MiniCollect K2EDTA tubes, catalog #450480 Greinder Bio-one GmbH (Kremsmunster, Austria), via terminal cardiac puncture and processed to plasma by centrifuging at 2000 x g for 10 minutes at 4°C. For RNA extraction, tissues were homogenized using a bead homogenizer in TRIzol Reagent, catalog # 15596026 (ThermoFisher Scientific), and RNA was extracted according to manufacturer’s protocols and purified using Econospin RNA Mini Spin/ Columns for RNA (Ephoch Life Sciences, Missouri City, TX, catalog #1940-250) following manufacturer’s protocols. Specific gene probes were designed by NanoString Bioinformatics Team using an identified target sequence based on the NCBI Reference Sequence (RefSeq) database contains. Custom probes were synthesized by Integrated DNA Technologies (IDT; Coralville, IA). mRNA expression was analyzed on an nCounter® SPRINT Profiler NanoString system using a multiplexing approach with nCounter PlexSet-12 Reagent Pack, catalog #PS-GX-PTK-12 (CSO) according to manufacturer’s protocols (NanoString, Inc, Seattle, WA).

T4 Analysis

[00189] T4 was measured in terminal plasma samples using an ELISA kit (Biovision, Inc., Thyroxine [T4] [Mouse/Rat] ELISA Kit, Cat #: K7421-100). Assays were performed according to manufacturer’s instructions with minor modifications based on previous assay validation efforts. Briefly, a seven-point standard curve of provided T4 diluted in Assay Buffer (25, 15, 10, 5, 2, 1 pg/dL) was prepared in duplicate for each assay. Plasma samples (undiluted), blanks (Assay Buffer) and standards were added to separate wells of a 96-well plate pre-coated with a T4 capture antibody, followed by addition of T4 Enzyme Conjugate to each well. Plates were then gently shaken (600 rpm) for 20-30 s to mix, and then covered with an acetate plate seal and incubated for 1 h at room temperature (RT) with gentle shaking (600 rpm). Plate contents were aspirated and washed three times with IX Wash Buffer, then blotted on paper towels to remove excess liquid. TMB Substrate was then added to each well and plates were secured with an acetate seal incubated for 15 min at RT, protected from light. Stop Solution was then added to each well and the plates shaken gently to mix the solution. Absorbance was read at 450 nm within 15 min of addition of the Stop Solution using a Varioskan Lux plate reader (ThermoFisher Scientific, Carlsbad, CA). Relative optical densities (ODs) were background- corrected against blank samples and standard curves. T4 concentrations were interpolated using the four-parameter curve-fit method. Unknown sample concentrations were determined using GraphPad Prism software (GraphPad Prism 9.0.2, GraphPad Software, San Diego, CA).

Data Analysis

[00190] Figures 10A, 10B, IOC and 10D show gene expression in brain (blue) & liver (orange), & effects on T4 (gray) 4 or 8 h after last dose in mice that had been dosed PO, QD for 7 days with (A) active metabolite or (B) prodrug alone; (C) prodrug + PFI (LL-650177) or (D) prodrug + GFI. Both prodrug and active metabolite reduce T4 levels after 7 days of treatment. Table 6 reports ED50 values in pg/kg for each treatment type.

Table 6

[00191] Using T4 as a marker for effects on THA; T4 parallels peripheral activity more than CNS activation of target genes. Negative regulation of T4 by thyromimetics does not appear to be predominantly centrally-mediated because the effects on THA and liver gene expression parallel plasma distribution more closely than exposure or activity in the CNS, suggesting a primarily pituitary-driven effect. The combination of a thyromimetic prodrug and a PFI may further enhance delivery of thyromimetics to the brain and maximize centrally-targeted distribution.

EXAMPLE 48: Peripheral Exposure to ABX-002A Predicts Effects on THA [00192] The relationship between THA effects and plasma ABX-002A was studied in both mice and NHP. Mouse: Female C57BL/6 mouse data from Example 14 above was employed to calculate exposure to amide and acid based on PK data from an independent experiment such as that detailed in Example 12. PK was only performed at a single dose, with other doses calculated proportionately. Non-human primate (NHP): Plasma pharmacokinetics and effects on the thyroid hormone axis were measured in non-naive cynomolgus monkeys (n=3/group) after daily dosing of ABX-002 at 10, 30, 100 or 300 ug/kg for 7 days. ABX-002 was formulated in 0.1% NMP/0.1% solutol and administered at 5 mL/kg PO. Blood samples were taken on day 1 and day 7 at 0, 0.25, 0.5, 1, 2, 4, 8, 12, and 24 hrs and analyzed for ABX-002, LL-340001, T3 and T4 levels by LCMS (as described above). TSH was measured by immunoassay.

[00193] Bioanalysis for T3 and T4 in NHP serum was performed using a surrogate matrix,

QCs, double blank and blank. Standard curve samples were prepared by adding 5 pL of WS to 50 pL blank surrogate serum. QC samples were prepared by adding 5 pL of WS to 50 pL blank surrogate serum. Unknow samples were added 5 pi DMSO. All calibrator standard, QCs, sample and blank wells were added 500 pL of IS working solution (2.5 ng/mL T3-13C6 and 25 ng/mL T4-13C6) in methanol, while 500 pL blank methanol was added to all double blanks. Following 400 pL supernatant transfer, sample were evaporated under N2 gas and reconstituted with 100 pL 80% methanol in water.

[00194] Figures 11 A, 11B, and 11C show T 4 inhibition as a function of (A) dose (B) plasma prodrug AUC or (C) plasma active metabolite AUC after 7 days of treatment in mice (orange) or non-human primate, NHP, (blue). Day 7 T4 levels normalized to the day 1 levels for each animal compared with the exposure in those same animals. Overall, the curves traced on Figure llC show the relationship between THA effects and plasma ABX-002A are present in both mice and NHP, and peripheral exposure to the active metabolite is a better predictor of effects on THA than either dose or plasma prodrug exposure.




 
Previous Patent: ARMPIT LINER

Next Patent: ANTI-IL-27 ANTIBODIES AND USES THEREOF