CN117597122A - Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of thyromimetics and combinations with peripherally restricted FAAH inhibitors - Google Patents

Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of thyromimetics and combinations with peripherally restricted FAAH inhibitors Download PDF

Info

Publication number
CN117597122A
CN117597122A CN202280047554.XA CN202280047554A CN117597122A CN 117597122 A CN117597122 A CN 117597122A CN 202280047554 A CN202280047554 A CN 202280047554A CN 117597122 A CN117597122 A CN 117597122A
Authority
CN
China
Prior art keywords
pharmaceutically acceptable
pharmaceutical composition
disease
solvate
acceptable salt
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280047554.XA
Other languages
Chinese (zh)
Inventor
布莱恩·安德鲁·斯特恩斯
吉尔·梅利莎·巴塞
詹森·兰德尔·哈里斯
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sutong Medical Co
Original Assignee
Sutong Medical Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sutong Medical Co filed Critical Sutong Medical Co
Priority claimed from PCT/US2022/028187 external-priority patent/WO2022236133A1/en
Publication of CN117597122A publication Critical patent/CN117597122A/en
Pending legal-status Critical Current

Links

Abstract

Provided herein are Fatty Acid Amide (FAAH) cleavable prodrugs of thyromimetics, as well as pharmaceutical compositions comprising these compounds and at least one pharmaceutically acceptable excipient, further comprising a peripherally restricted FAAH inhibitor.

Description

Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of thyromimetics and combinations with peripherally restricted FAAH inhibitors
Cross reference
The present application claims the benefit of U.S. provisional application Ser. No. 63/185,254, filed on 5/6 of 2021, and U.S. provisional application Ser. No. 63/274,856, filed on 11/2 of 2021, each of which is incorporated herein by reference in its entirety.
Background
The blood brain barrier consists 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 limits the diffusion of therapeutic agents into the brain, thus creating a significant challenge for developing new drugs against CNS diseases.
Disclosure of Invention
In one 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:
Wherein:
R 1 and R is 2 Independently selected from hydrogen, -OR 5 、-NR 5 R 6 、C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 Alkyl-phenyl, wherein C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 The alkyl-phenyl group is optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
R 3 And R is 4 Independently selected from-F, -Cl, -Br and-I;
R 5 and R is 6 Independently selected from hydrogen and C 1 -C 6 An alkyl group; and is also provided with
R 7 And R is 8 Independently selected from hydrogen, -F, -Cl, -Br and-I;
a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor.
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.
In another aspect, provided herein is a pharmaceutical composition comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I):
wherein:
R 1 and R is 2 Independently selected from hydrogen, -OR 5 、-NR 5 R 6 、C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 Alkyl-phenyl, wherein C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 The alkyl-phenyl group is optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
R 3 And R is 4 Independently selected from-F, -Cl, -Br and-I; and is also provided with
R 5 And R is 6 Independently selected from hydrogen and C 1 -C 6 An alkyl group;
a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor.
In some embodiments, R 1 Is hydrogen. In some embodiments, R 2 Is C optionally substituted with one or more of 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more of the following 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more-OH groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by one or more halogen groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is unsubstituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is phenyl optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 is-C optionally substituted with one or more of 1 -C 6 Alkyl-phenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 3 And R is 4 Independently selected from-F, -Cl and-Br. In some embodiments, R 3 And R is 4 Are all-Br. In some embodiments, R 3 And R is 4 Are all-Br. In some embodiments, R 3 And R is 4 Are all-Cl. In some embodiments, R 3 And R is 4 Are all-F.
In some embodiments, the peripherally restricted FAAH inhibitor is ASP-3652.
In another aspect is a method of treating a CNS disease or disorder in a patient in need thereof, the method 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 the group consisting of Acute Disseminated Encephalomyelitis (ADEM), acute hemorrhagic white matter encephalitis (AHL or AHLE), adult refsum disease, infant refsum disease, alexander disease (Alexander disease), alzheimer's disease, barocentric sclerosis, canavan disease, pontic central myelination (CPM), cerebral palsy, tendineomatosis, chronic Inflammatory Demyelinating Polyneuropathy (CIDP), devic's syndrome (AHL or AHLE), myelinating lytic diffuse sclerosis, encephalomyelitis, guillain-Barre syndrome (Guillain-Barre syndrome), idiopathic inflammatory demyelinating disease (HDD), krabbe disease (Krabbe disease), leopathies, leukodystrophy, malnutrition, multiple sclerosis, maldevelopment-bixas (bixas-focal), multiple sclerosis (mmfibrous demux-dysmyelin disease (mmfibrous), multiple sclerosis (mmfibrous demux-dyskinesis) (42-dyskinesia), PMD), progressive Multifocal Leukoencephalopathy (PML), tropical Spastic Paraparesis (TSP), X-linked adrenoleukodystrophy (X-ALD, ALO or X-linked ALO), and Ji Weige syndrome (Zellweger syndrome). In some embodiments, the CNS disease or disorder is selected from multiple sclerosis and X-linked adrenoleukodystrophy.
Drawings
The novel features of the disclosure are set forth with particularity in the appended claims. A more complete appreciation of the features and advantages of the present disclosure will be obtained by reference to the following detailed description, which sets forth illustrative embodiments in which the principles of the present disclosure are utilized, and the accompanying drawings in which:
FIG. 1 depicts the enhancement of oligodendrocyte differentiation in an in vitro oligodendrocyte progenitor assay of the active metabolite LL-341070A of the LL-341070 prodrug.
Figure 2 depicts thyromimetic treatment enhancing 24-OHC synthesis in the brain of rats after cyclohexanone dihydrazone (cuprimazole) induced demyelination in vivo.
Figure 3 depicts TR beta target engagement in the brain as evidenced by increased expression of T3 responsive target genes in vivo.
Fig. 4 depicts brain and plasma concentrations measured 4 hours after the final dose after repeated administration of LL-341070 over 21 days.
FIG. 5 depicts LL-341070 improving in vivo clinical scores and histology in a mouse prophylactic Experimental Autoimmune Encephalitis (EAE) model.
Fig. 6 depicts FAAH expression and specific activity in multiple species and tissue types.
FIG. 7 depicts the measurement of the concentration of ABX-002A in brain, liver, kidney, lung and heart 1 hour after SC administration of 30 different prodrugs of ABX-002A.
Figure 8 depicts plasma, liver and brain concentrations after ABX-002 prodrug treatment with or without peripheral or global FAAH inhibitors.
FIG. 9A depicts a comparison of T3 target gene induction in brain with T3 target gene induction in liver after a single administration of ABX-002A.
FIG. 9B depicts a comparison of T3 target gene induction in brain with T3 target gene induction in liver after a single administration of ABX-002.
FIG. 9C depicts a comparison of T3 target gene induction in brain with T3 target gene induction in liver after a single administration of ABX-002 plus FAAH inhibitor.
FIG. 10A depicts gene expression in brain and liver and effect on T4 after administration of ABX-002A.
FIG. 10B depicts gene expression in brain and liver and effect on T4 after administration of ABX-002.
FIG. 10C depicts gene expression in brain and liver and effect on T4 after administration of ABX-002 plus peripheral FAAH inhibitor.
FIG. 10D depicts gene expression in brain and liver and the effect on T4 after administration of ABX-002 plus global FAAH inhibitor.
FIG. 11A depicts T4 inhibition as a function of ABX-002 dose in mice or non-human primate (NHP).
FIG. 11B depicts T4 inhibition as a function of plasma ABX-002 prodrug AUC in mice or non-human primate (NHP).
FIG. 11C depicts T4 inhibition as a function of plasma ABX-002A active metabolite AUC in mice or non-human primate (NHP).
Detailed Description
Fatty Acid Amide Hydrolase (FAAH) is an integral membrane serine hydrolase that degrades the signaling lipids of the fatty acid amide family and can hydrolyze selected amide prodrugs. FAAH is highly conserved among species and expressed to varying degrees in many tissues, including the Central Nervous System (CNS). The selected carboxylic acids can be converted to more permeable amide prodrugs, which can then cross the blood brain barrier where they can be converted to active molecules by the action of FAAH on the prodrug. This results in higher amounts of carboxylic acid being delivered to the CNS than if the parent was administered alone. However, peripherally expressed FAAH hydrolyzes the prodrug simultaneously, resulting in a large number of non-productive prodrug conversions. Co-administration of a peripherally restricted FAAH inhibitor with a CNS permeable FAAH convertible prodrug increases the selectivity of prodrug delivery to the CNS. This also results in lower exposure of the parent molecule to plasma and peripheral tissues than is observed when the prodrug is administered alone.
Candidates for clinical development may be selected from the compounds disclosed herein based on the extent of FAAH-mediated hydrolysis in vitro, plasma stability in vitro, tissue distribution in vivo, target selectivity in vitro, target potency in vitro, target gene expression in vivo, pharmacological efficacy in vivo, or drug-like (compliant with patent 5 rules) physiochemical properties of the compounds disclosed herein, or combinations thereof.
Certain terms
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 such drugs, and reference to "an excipient" includes reference to one or more such excipients. When ranges are used herein, it is intended to include all combinations and subcombinations of the ranges and specific embodiments therein. The term "about" when referring to a numerical value or range of numerical values means that the numerical value or range of numerical values referred to is an approximation within experimental variability (or within statistical experimental error), and therefore, the numerical value or range of numerical values varies between 1% and 15% of the stated numerical value or range of numerical values.
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 excipient.
The terms "active agent," "drug," "active ingredient," and variations thereof are used interchangeably to refer to an agent or substance that has a measurable specified or selected physiological activity when administered to a subject in significant or effective amounts.
"pharmaceutically acceptable salts" include both acid addition salts and base addition salts. Pharmaceutically acceptable salts of any of the compounds described herein are 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.
By "pharmaceutically acceptable acid addition salts" is meant those salts which retain the biological effectiveness and properties of the free base, are not undesirable in biological or other respects, and are formed with mineral 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 formed with organic acids such as aliphatic mono-and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxyalkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, and the like, 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, p-toluenesulfonic acid, salicylic acid, and the like. Thus, exemplary salts include sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogen phosphate, dihydrogen phosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide, acetate, trifluoroacetate, propionate, octanoate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenylacetate, citrate, lactate, malate, tartrate, methanesulfonate, and the like. Salts of amino acids such as arginine salts, gluconate and galacturonate are also contemplated (see, e.g., berge S.M. et al, "Pharmaceutical Salts," Journal of Pharmaceutical Science,66:1-19 (1997)). Acid addition salts of basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt.
By "pharmaceutically acceptable base addition salts" is meant those salts that retain the biological effectiveness and properties of the free acid, and are not undesirable in biological or other respects. These salts are prepared by adding an inorganic or organic base to the free acid. In some embodiments, the pharmaceutically acceptable base addition salts are formed with metals or amines such as alkali metals 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, 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 such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine (caffeine), procaine (procaine), N-diphenylmethyl ethylenediamine, chloroprocaine, hydrabamine, choline, betaine, ethylenediamine, diaminodibenzyl, N-methyl-reduced glucosamine, methyl-reduced glucosamine, theobromine (theobromine), purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like. See Berge et al (supra).
References to pharmaceutically acceptable salts are understood to include solvent addition forms (solvates). Solvates contain a stoichiometric or non-stoichiometric amount of solvent and are formed with pharmaceutically acceptable solvents such as water, ethanol, methanol, methyl tert-butyl ether (MTBE), diisopropyl ether (DIPE), ethyl acetate, isopropyl alcohol, methyl isobutyl ketone (MIBK), methyl Ethyl Ketone (MEK), acetone, nitromethane, tetrahydrofuran (THF), dichloromethane (DCM), dioxane, heptane, toluene, anisole, acetonitrile, and the like during the process of product formation or isolation. In one aspect, the solvate is formed using, but not limited to, one or more group 3 solvents. The types of solvents are described in, for example, international consortium of drug registration technical requirements (International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use) (ICH), "impurities: residual solvent guidelines (impulities: guidelines for Residual Solvents), Q3C (R3), (11 month 2005). When the solvent is water, hydrates are formed, or when the solvent is an alcohol, alcoholates are formed.
The term "effective amount" or "therapeutically effective amount" as used herein refers to a sufficient amount of an agent or compound administered that will alleviate one or more symptoms of the disease or condition being treated to some extent. The result may be a alleviation and/or a 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 use is that amount of a composition comprising a compound as disclosed herein that is required to provide clinically significant relief of a disease. The appropriate "effective" amount in any single instance can be determined using techniques such as dose escalation studies.
The terms "subject," "individual," and "patient" are used interchangeably herein to refer to a mammal. Mammals include, but are not limited to, mice, apes, humans, farm animals, sports animals, and pets.
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 at systemic doses. 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(s)
Thyroid Hormone (TH) is a key signal for oligodendrocyte differentiation and myelin formation during development, and also stimulates remyelination in the adult model of Multiple Sclerosis (MS) (Calz-a L et al, brain Res Revs 48:339-346,2005). However, TH is not an acceptable long-term therapy because remyelination is rarely achieved while avoiding the therapeutic window for cardiotoxicity and bone demineralization associated with chronic hyperthyroidism. Some thyroid hormone analogs can activate the thyroid hormone responsive gene by exploiting the molecular and physiological characteristics of the thyroid hormone receptor, while avoiding the associated drawbacks of TH (Malm J et al Mini Rev Med Chem7:79-86,2007). These receptors are expressed in two major forms, with heterogeneous tissue distribution and overlapping but distinct sets of target genes (Yen PM, physiol Rev 81:1097-1142,2001). Trα is enriched in heart, brain and bone, while trβ is enriched in liver (O' Shea PJ et al Nucl Recept Signal 4:e011, 2006).
Development of selective thyromimetics has been challenging due to the high sequence homology of the thyroid hormone receptor subtype; that is, only one amino acid residue differs between the α1 and β1 forms on the inner surface of the ligand binding domain cavity.
In some embodiments, the pharmaceutical compositions described herein comprise Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of formula (I '), wherein the prodrug of formula (I') is a prodrug of a TR beta agonist. In some embodiments, the pharmaceutical compositions described herein comprise Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of formula (I), wherein the prodrug of formula (I) is a prodrug of a TR beta agonist. In some embodiments, the pharmaceutical compositions described herein comprise Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of formula (II), wherein the prodrug of formula (II) is a prodrug of a TR beta agonist.
Pharmaceutical composition
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:
wherein:
R 1 and R is 2 Independently selected from hydrogen, -OR 5 、-NR 5 R 6 、C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 Alkyl-phenyl, wherein C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 The alkyl-phenyl group is optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
R 3 And R is 4 Independently selected from-F, -Cl, -Br and-I;
R 5 and R is 6 Independently selected from hydrogen and C 1 -C 6 An alkyl group; and is also provided with
R 7 And R is 8 Independently selected from hydrogen, -F, -Cl, -Br and-I;
a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor.
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.
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.
In some embodiments, R 1 Is hydrogen. In some embodiments, R 1 Is C 1-6 An alkyl group.
In some embodiments, R 2 Is C optionally substituted with one or more of 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more of the following 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more halogen groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by a cyano group 1 -C 6 An alkyl group. In some embodiments, R 2 Is one OR more-OR 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by one or more-OH groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by-OH 1 -C 6 An alkyl group. In one placeIn some embodiments, R 2 Is covered by one or more-NR' s 5 R 6 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered with one or more-NH 2 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered with a-NH 2 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 R 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 H substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 OR 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 OH-substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is unsubstituted C 1 -C 6 An alkyl group. In some embodiments, R 2 is-CH 3 . In some embodiments, R 2 is-CH 2 CH 3 . In some embodiments, R 2 is-CH 2 CH 2 CH 3
In some embodiments, R 2 Is C optionally substituted with one or more of 2 -C 6 Alkenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 2 -C 6 Alkenyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 2 -C 6 Alkynyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 2 -C 6 Alkynyl groups.
In some embodiments, R 2 Is optionally covered byC substituted by one or more of (C) 3 -C 6 Cycloalkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 3 -C 6 Cycloalkyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 3 -C 6 Heterocycloalkyl group: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 3 -C 6 A heterocycloalkyl group.
In some embodiments, R 2 Is phenyl optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is phenyl substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is phenyl substituted with one or more halo groups. In some embodiments, R 2 Is one OR more-OR 5 A substituted phenyl group. In some embodiments, R 2 Is phenyl substituted with one or more —oh. In some embodiments, R 2 Is unsubstituted phenyl.
In some embodiments, R 2 is-C optionally substituted with one or more of 1 -C 6 Alkyl-phenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted-C 1 -C 6 Alkyl-phenyl.
In some embodiments, R 2 is-OR 5 . In some embodimentsWherein R is 2 is-OH. In some embodiments, R 2 is-NR 5 R 6 . In some embodiments, R 2 is-NH 2
In some embodiments, R 2 Is hydrogen.
In some embodiments, R 3 And R is 4 Independently selected from-F, -Cl and-Br. In some embodiments, R 3 And R is 4 Are all-Br. In some embodiments, R 3 And R is 4 Are all-Br. In some embodiments, R 3 And R is 4 Are all-Cl. In some embodiments, R 3 And R is 4 Are all-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.
In some embodiments of the pharmaceutical compositions described herein, the Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') has a structure selected from the group consisting of:
or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments, described herein is a pharmaceutical composition comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I):
wherein:
R 1 and R is 2 Independently selected from hydrogen, -OR 5 、-NR 5 R 6 、C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 Alkyl-phenyl, wherein C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 The alkyl-phenyl group is optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
R 3 And R is 4 Independently selected from-F, -Cl, -Br and-I; and is also provided with
R 5 And R is 6 Independently selected from hydrogen and C 1 -C 6 An alkyl group;
a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor.
In some embodiments, R 1 Is hydrogen. In some embodiments, R 1 Is C 1-6 An alkyl group.
In some embodiments, R 2 Is C optionally substituted with one or more of 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more of the following 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more halogen groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by a cyano group 1 -C 6 An alkyl group. In some embodiments, R 2 Is one OR more-OR 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by one or more-OH groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by-OH 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by one or more-NR' s 5 R 6 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered with one or more-NH 2 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered with a-NH 2 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 R 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 H substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 OR 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 OH-substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is unsubstituted C 1 -C 6 An alkyl group. In some embodiments, R 2 is-CH 3 . In some embodiments, R 2 is-CH 2 CH 3 . In some embodiments, R 2 is-CH 2 CH 2 CH 3
In some embodiments, R 2 Is C optionally substituted with one or more of 2 -C 6 Alkenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 2 -C 6 Alkenyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 2 -C 6 Alkynyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 2 -C 6 Alkynyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 3 -C 6 Cycloalkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 3 -C 6 Cycloalkyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 3 -C 6 Heterocycloalkyl group: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 3 -C 6 A heterocycloalkyl group.
In some embodiments, R 2 Is phenyl optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is phenyl substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is phenyl substituted with one or more halo groups. In some embodiments, R 2 Is one OR more-OR 5 A substituted phenyl group. In some embodiments, R 2 Is phenyl substituted with one or more —oh. In some embodiments, R 2 Is unsubstituted phenyl.
In some embodiments, R 2 is-C optionally substituted with one or more of 1 -C 6 Alkyl-phenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted-C 1 -C 6 Alkyl-phenyl.
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-NH 2
In some embodiments, R 2 Is hydrogen.
In some embodiments, R 3 And R is 4 Independently selected from-F, -Cl and-Br. In some embodiments, R 3 And R is 4 Are all-Br. In some embodiments, R 3 And R is 4 Are all-Br. In some embodiments, R 3 And R is 4 Are all-Cl. In some embodiments, R 3 And R is 4 Are all-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.
In some embodiments of the pharmaceutical compositions described herein, the Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I) has a structure selected from the group consisting of:
/>
/>
or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments of the pharmaceutical compositions described herein, the Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I) has a structure selected from the group consisting of:
/>
or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments, described herein is a pharmaceutical composition comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (II):
wherein:
R 1 and R is 2 Independently selected from hydrogen, -OR 5 、-NR 5 R 6 、C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 Alkyl-phenyl, wherein C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 The alkyl-phenyl group is optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 The method comprises the steps of carrying out a first treatment on the surface of the And is also provided with
R 5 And R is 6 Independently selected from hydrogen and C 1 -C 6 An alkyl group;
a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor, wherein said peripherally restricted FAAH inhibitor is ASP-3652.
In some embodiments, R 1 Is hydrogen. In some embodiments, R 1 Is C 1-6 An alkyl group.
In some embodiments, R 2 Is C optionally substituted with one or more of 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more of the following 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is C substituted by one or more halogen groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by a cyano group 1 -C 6 An alkyl group. In some embodiments, R 2 Is one OR more-OR 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by one or more-OH groups 1 -C 6 An alkyl group. In some embodiments, R 2 Is C substituted by-OH 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by one or more-NR' s 5 R 6 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered with one or more-NH 2 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered with a-NH 2 Substituted C 1 -C 6 An alkyl group. In some implementationsIn embodiments, R 2 Is covered by a-S (O) 2 R 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 H substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 OR 5 Substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is covered by a-S (O) 2 OH-substituted C 1 -C 6 An alkyl group. In some embodiments, R 2 Is unsubstituted C 1 -C 6 An alkyl group. In some embodiments, R 2 is-CH 3 . In some embodiments, R 2 is-CH 2 CH 3 . In some embodiments, R 2 is-CH 2 CH 2 CH 3
In some embodiments, R 2 Is C optionally substituted with one or more of 2 -C 6 Alkenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 2 -C 6 Alkenyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 2 -C 6 Alkynyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 2 -C 6 Alkynyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 3 -C 6 Cycloalkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 3 -C 6 Cycloalkyl groups.
In some embodiments, R 2 Is C optionally substituted with one or more of 3 -C 6 Heterocycloalkyl group: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted C 3 -C 6 A heterocycloalkyl group.
In some embodiments, R 2 Is phenyl optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is phenyl substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is phenyl substituted with one or more halo groups. In some embodiments, R 2 Is one OR more-OR 5 A substituted phenyl group. In some embodiments, R 2 Is phenyl substituted with one or more —oh. In some embodiments, R 2 Is unsubstituted phenyl.
In some embodiments, R 2 is-C optionally substituted with one or more of 1 -C 6 Alkyl-phenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5 . In some embodiments, R 2 Is unsubstituted-C 1 -C 6 Alkyl-phenyl.
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-NH 2
In some embodiments, R 2 Is hydrogen.
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 the group consisting of:
/>
peripherally restricted FAAH inhibitors
The pharmaceutical compositions described herein comprise a peripherally restricted FAAH inhibitor. In some embodiments, peripheral limiting FAAH inhibitors are disclosed in US2008/0306046, which is incorporated herein by reference in its entirety.
In some embodiments, the peripherally restricted FAAH inhibitor is a compound of formula (X):
wherein:
ring a is a benzene ring, a cyclopentane ring, a cyclohexane ring, a cycloheptane ring or a 5 to 7 membered nitrogen containing heterocycle;
l is a single bond, lower alkylene, lower alkenylene, -N (R) 15 )-C(=O)-、-C(=O)-N(R 15 ) -, - (lower alkenylene) -C (=o), -O-, or C (=o);
R 15 is H or lower alkyl;
x is CH or N;
R 8 、R 9 and R is 10 Each independently selected from:
(i) A group selected from: H. halo, -CN, CF 3 Lower alkyl and-O-lower alkyl;
(ii) Optionally by 1 to 5 independentlyAryl substituted with a group selected from: 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: H. halo, -CN, -CF 3 Lower alkyl and-O-lower alkyl;
(iv)R 16 - (lower alkenylene) -O-;
(v)R 16 - (lower alkenylene) -N (R) 15 ) -; or (b)
(vi)R 17 R 18 N-C(=O)-;
R 16 Is that
(i) Aryl optionally substituted with 1 to 5 groups independently selected from: 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: H. halo, -CN, -CF 3 Lower alkyl and-O-lower alkyl; or (b)
(iii) 3 to 8 membered cycloalkyl;
R 17 and R is 18 Each independently selected from the group consisting of H, lower alkyl, and 3 to 8 membered cycloalkyl; or R is 17 And R is 18 Can form a 3-to 8-membered nitrogen-containing heterocyclic ring together with the nitrogen atom to which they are bonded;
R 11 selected from H, lower alkyl, and oxo (=o); and is also provided with
R 12 、R 13 And R is 14 One of them is-C (=O) -O- (lower alkyl) or-CO 2 H, and the remainder are H.
In some embodiments, the peripherally restricted FAAH inhibitor is 5- (((4- (4- ((3-fluorobenzyl) oxy) phenoxy) piperidin-1-yl) carbonyl) oxy) nicotinic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5- (((4- (2-phenylethyl) piperidin-1-yl) carbonyl) oxy) niacin. In some embodiments, the peripherally restricted FAAH inhibitor is 5- (((4- (4- (2-cyclohexylethoxy) phenoxy) piperidin-1-yl) carbonyl) oxy) nicotinic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5- (((4- ((E) -2-phenylvinyl) piperidin-1-yl) carbonyl) oxy) niacin. In some embodiments, the peripherally restricted FAAH inhibitor is 5- (((4- (3- (1- (6-methylpyridin-2-yl) piperidin-4-yl) propyl) piperidin-1-yl) carbonyl) oxy) nicotinic acid. In some embodiments, the peripherally restricted FAAH inhibitor is 5- (methoxycarbonyl) pyridin-3-yl 4- (2-phenylethyl) piperazine-1-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-1-yl) carbonyl) oxy) niacin.
Compounds of formula (I)
The compounds of formula (I'), (I) and (II) described herein are amide prodrugs of TR beta agonists. The amide prodrugs described herein are cleaved by Fatty Acid Amide Hydrolase (FAAH) to produce the active TR beta agonist. In some embodiments are compounds selected from the group consisting of:
/>
or a pharmaceutically acceptable salt or solvate thereof.
In some embodiments is a pharmaceutical composition comprising a pharmaceutically acceptable excipient and a compound selected from the group consisting of:
/>
/>
or a pharmaceutically acceptable salt or solvate thereof.
Method
In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof, the method comprising administering to the patient a pharmaceutical composition described herein comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof, the method comprising administering to the patient a pharmaceutical composition described herein comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a 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, the method comprising administering to the patient a pharmaceutical composition described herein comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is selected from Acute Disseminated Encephalomyelitis (ADEM), acute hemorrhagic encephalomyelitis (AHL or AHLE), adult graves ' disease, infant graves ' disease, alexander disease, alzheimer's disease, baroclavicular sclerosis, canten's disease, pontine central myelination (CPM), cerebral palsy, tendinosis, chronic Inflammatory Demyelinating Polyneuropathy (CIDP), devic's syndrome, myelinating diffuse sclerosis, encephalomyelitis, green-barre syndrome, idiopathic inflammatory demyelinating disease (HDD), krabbe's disease, leber's hereditary optic neuropathy, leukodystrophy, markerberg's multiple sclerosis, markia-binby's disease, metachromatic Leukodystrophy (MLD), multifocal Motor Neuropathy (MMN), multiple sclerosis (parametrics), MS demyelinating disease, alph-focal leukosis (TSP), and pre-spasticity syndrome (alph), pansy (p-focal) and pre-spasticity syndrome (TSP), pansy (alph), pansy (p-focal) and (p-focal) linked) multiple sclerosis (TSP). In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof, the method comprising administering to the patient a pharmaceutical composition described herein comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises 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, the method comprising administering to the patient a pharmaceutical composition described herein comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I), or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is X-linked adrenoleukodystrophy.
In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof, the method 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; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor. In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof, the method 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; the pharmaceutical composition further comprises a 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, the method 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; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is selected from Acute Disseminated Encephalomyelitis (ADEM), acute hemorrhagic encephalomyelitis (AHL or AHLE), adult graves ' disease, infant graves ' disease, alexander disease, alzheimer's disease, baroclavicular sclerosis, canten's disease, pontine central myelination (CPM), cerebral palsy, tendinosis, chronic Inflammatory Demyelinating Polyneuropathy (CIDP), devic's syndrome, myelinating diffuse sclerosis, encephalomyelitis, green-barre syndrome, idiopathic inflammatory demyelinating disease (HDD), krabbe's disease, leber's hereditary optic neuropathy, leukodystrophy, markerberg's multiple sclerosis, markia-binby's disease, metachromatic Leukodystrophy (MLD), multifocal Motor Neuropathy (MMN), multiple sclerosis (parametrics), MS demyelinating disease, alph-focal leukosis (TSP), and pre-spasticity syndrome (alph), pansy (p-focal) and pre-spasticity syndrome (TSP), pansy (alph), pansy (p-focal) and (p-focal) linked) multiple sclerosis (TSP). In some embodiments is a method of treating a CNS disease or disorder in a patient in need thereof, the method 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; the pharmaceutical composition further comprises 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, the method 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; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor, wherein the CNS disease or disorder is X-linked adrenoleukodystrophy.
Excipient
Suitable optional excipients for use in the pharmaceutical compositions described herein include any commonly used excipients in pharmacy and are selected based on compatibility with the active 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. An overview of the excipients described herein can be found, for example, in the following: remington, the Science and Practice of Pharmacy, nineteenth edition (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.editions, pharmaceutical Dosage Forms, marcel Decker, new York, n.y.,1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, seventh edition (Lippincott Williams & Wilkins, 1999), which are incorporated herein by reference in their entirety.
Binders impart tackiness to solid oral dosage form formulations: for powder filled capsule formulations they help form a plug that can be filled into a soft or hard shell capsule, and for tablet formulations they ensure that the tablet remains intact after compression and help ensure blend uniformity prior to the compression or filling step. Materials suitable for use as binders in the solid dosage forms described herein include, but are not limited to, carboxymethyl cellulose, methyl cellulose (e.g.) Hydroxypropyl methylcellulose (e.g., hydroxypropyl methylcellulose USP Pharmacoat-603), hydroxypropyl methylcellulose acetate stearate HS-LF and HS, hydroxyethyl cellulose, hydroxypropyl cellulose (e.g.,>) Ethylcellulose (e.g) And microcrystalline cellulose (e.g.)>) Microcrystalline dextrose, amylose, magnesium aluminum silicate, polysaccharide acids, bentonite, gelatin, polyvinylpyrrolidone/vinyl acetate copolymer, crospovidone, povidone, starch, pregelatinized starch, tragacanth, dextrin, sugars such as sucrose (e.g.)>) Glucose, dextrose, molasses, mannitol, sorbitol, xylitol (e.g.)>) Lactose, natural or synthetic gums such as acacia, tragacanth, ghatti, seashell mucilage of ezetimibe (isapol), starch, polyvinylpyrrolidone (e.g.)>CL、/>CL、XL-10 and->K-12), larch arabinogalactan,/c>Polyethylene glycol, wax, sodium alginate, and the like.
The filler or diluent may increase the volume of the pharmaceutical formulation. Such compounds include, for example, lactose; starch; mannitol; sorbitol; dextrose; microcrystalline cellulose such asCalcium hydrogen phosphate; dicalcium phosphate dihydrate; tricalcium phosphate; a calcium phosphate; anhydrous lactose; spray-dried lactose; pregelatinized starch; compressible sugar, such as- >(Amstar); hydroxypropyl methylcellulose; sucrose-based diluents; sugar powder; calcium bisulfate monohydrate; calcium sulfate dihydrate; calcium lactate trihydrate; a dextran binder; hydrolyzing the cereal solids; amylose; powdered cellulose; calcium carbonate; glycine; kaolin; sodium chloride; inositol; bentonite; etc.
Glidants improve the flow characteristics of powder mixtures. Such compounds include, for example, colloidal silica such asTricalcium phosphate, talcum, corn starch, DL-leucine, sodium lauryl sulfate, magnesium stearate, calcium stearate, sodium stearate, kaolin and micronized amorphous silica->Etc.
Lubricants are compounds that prevent, reduce, or inhibit material adhesion or friction. Exemplary lubricants include, for example, stearic acid; calcium hydroxide, talc; hydrocarbons, such as mineral oils, or hydrogenated vegetable oils, such as hydrogenated soybean oilHigher fatty acids and their alkali and alkaline earth metal salts, such as aluminum salts, calcium salts, magnesium salts, zinc salts, stearic acid, sodium stearate, magnesium stearate, glycerol, talc, waxes, < >>Boric acid, sodium acetate, leucine, polyethylene glycol or methoxypolyethylene glycol such as Carbowax TM Sodium oleate, glyceryl behenate (Compitrol +)>) Glyceryl palmitostearate->Colloidal silica such as Syloid TMStarches such as corn starch, silicone oil, surfactants, and the like. Hydrophilic lubricants include, for example, sodium stearyl fumarate (currently under the trade name +.>Sales), polyethylene glycol (PEG), magnesium lauryl sulfate, sodium Lauryl Sulfate (SLS), sodium benzoate, sodium chloride, and the like.
Disintegrants promote the breakdown or disintegration of the pharmaceutical formulation after administration. Examples of disintegrants include starches, for example natural starches such as corn starch or potato starch, pregelatinized starches such as National 1551 orOr sodium starch glycolate such as->Or->The cellulose is used as a raw material for the production of cellulose,such as wood products, microcrystalline cellulose, e.g.PH101、/>PH102、/>PH105、/>P100、Ming/>And->Methylcellulose, croscarmellose or croscarmellose, such as croscarmellose sodium +.>Crosslinked carboxymethyl cellulose or crosslinked carboxymethyl cellulose; crosslinked starches, such as sodium starch glycolate; crosslinked polymers such as crospovidone; crosslinked polyvinylpyrrolidone; alginic acid such as alginic acid or a salt of alginic acid such as sodium alginate; clays, such as->HV (magnesium aluminium silicate); gums such as agar, guar gum, locust bean gum, karaya gum, pectin, or tragacanth gum; sodium starch glycolate; bentonite; natural sponge; resins such as cation exchange resins; citrus pulp; sodium lauryl sulfate; combination of sodium lauryl sulfate with starch; etc.
The polymer carrier includes compounds such as the following: polyvinylpyrrolidone, such AS polyvinylpyrrolidone K12, polyvinylpyrrolidone K17, polyvinylpyrrolidone K25 or polyvinylpyrrolidone K30, polyvinylpyrrolidone vinyl acetate (PVPVA 64), hydroxypropyl methylcellulose (HPMC), hydroxypropyl methylcellulose acetyl succinate (HPMC AS), and methyl methacrylate polymer (Eudragit polymer), and the like.
Stabilizers include compounds such as: any antioxidant, such as Butylated Hydroxytoluene (BHT), sodium ascorbate, and tocopherol; buffers, acids, and the like.
Surfactants include compounds such as the following: sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, poloxamers (polaxomers), bile salts, glyceryl monostearate, copolymers of ethylene oxide and propylene oxide, for example(BASF), d-alpha-tocopheryl polyethylene glycol succinate (vitamin E TPGS); etc.
The excipients mentioned above are given by way of example only and are not intended to include all possible choices. Other suitable classes of excipients include colorants, granulating agents, preservatives, defoamers, plasticizers, and the like. In addition, many excipients may have more than one effect or function, or may be categorized in more than one group; the classification is merely descriptive and is not intended to limit any use of a particular excipient.
The disclosed pharmaceutical formulations are administered to patients (animals and humans) in need of such treatment in dosages that provide optimal pharmaceutical efficacy. It will be appreciated that the dosage 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, the concurrent medication or the particular meal to be followed by the patient, and other factors, with the appropriate dosage ultimately being at the discretion of the attendant physician.
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
The following examples are provided for illustrative purposes and are not intended to limit the scope of the claims provided herein. All literature citations in these embodiments and throughout this specification are incorporated herein by reference for all legal purposes served by them. The starting materials and reagents for the synthesis of the compounds described herein may be synthesized or may 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 US2019/0210950, which is incorporated herein by reference. In some embodiments, the compounds provided herein are synthesized as described in US 2021/0002208, which is incorporated herein by reference. In some embodiments, the compounds provided herein are synthesized as described in WO 2021/108549, which is incorporated herein 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)
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 (catalytic amount). The mixture was cooled to 0deg.C and oxalyl chloride (57 mg,0.45 mmol) was added. The mixture was stirred at room temperature for 30min, 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.
Step 2: to a solution of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) acetyl chloride (110 mg,0.3 mmol) in DCM (2 mL) was added a mixture of 6-methoxypyridin-3-amine (37 mg,0.3 mmol) and triethylamine (61 mg,0.6 mmol) in DCM (3 mL). The mixture was stirred at room temperature for 1h. Water (15 mL) was added and the resulting mixture was extracted with DCM (20 mL. Times.3). The combined organic phases were washed with brine (20 mL), and dried over Na 2 SO 4 Dried, concentrated in vacuo and purified by preparative HPLC to afford 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (6-methoxypyridin-3-yl) acetamide (compound 101) as a white solid (30 mg,21% yield). 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyrazin-2-yl) acetamide (compound 102) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (3, 4-dimethylisoxazol-5-yl) acetamide (compound 103) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyridazin-3-yl) acetamide (compound 104) was synthesized according to the procedure 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)
N-cyclohexyl-2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) acetamide (compound 105) was synthesized according to the procedure of example 1 using cyclohexylamine in step 2. LCMS, M-h= 448.2.
Example 6: synthesis of N- (but-2-yn-1-yl) -2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) acetamide (Compound 106)
Step 1: a sealed tube (50 mL) was charged with 1-bromobut-2-yne (400 mg,3.0 mmol) and NH 3 (10 mL,7M in MeOH). The mixture was stirred at 60 ℃ overnight. The mixture was concentrated in vacuo to afford but-2-yn-1-amine hydrobromide as a yellow oil (400 mg,89% yield).
Step 2: n- (but-2-yn-1-yl) -2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) acetamide (compound 106) was synthesized according to the procedure of example 1 using but-2-yn-1-amine hydrobromide in step 2. LCMS, M-h=418.1.
Example 7: synthesis of N- (but-2-yn-1-yl) -2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-methylacetamide (Compound 107)
Step 1: to a mixture of N-methylpropan-2-yn-1-amine (2.0 g,29.0 mmol) in THF (20 mL) was added tert-butyl dicarbonate (18.9 g,87.0 mmol). The mixture was cooled to 40 ℃ and stirred for 2.0h. The mixture was then concentrated in vacuo to afford tert-butyl methyl (prop-2-yn-1-yl) carbamate (4.0 g,82% yield) as a colorless oil.
Step 2: to a solution of tert-butyl methyl (prop-2-yn-1-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 ℃. The mixture was stirred for 1h and methyl iodide was added and stirred for another 1.0h. Water (50 mL) was added and the resulting mixture was extracted with DCM (20 mL. Times.3). The combined organic phases were washed with brine (50 mL), and dried over Na 2 SO 4 Dried and concentrated in vacuo to afford crude but-2-yn-1-yl (methyl) carbamic acid tert-butyl ester (1.0 g,92% yield) as a colorless oil.
Step 3: to a solution of tert-butyl but-2-yn-1-yl (methyl) carbamate (1.0 g,5.5 mmol) in DCM (2 mL) was added TFA (1.3 g,11.0 mmol) and stirred at 0deg.C for 2.0h. The resulting mixture was concentrated in vacuo to afford N-methylbut-2-yn-1-amine (200.0 mg,44% yield) as a colorless oil.
Step 4: n- (but-2-yn-1-yl) -2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-methylacetamide (compound 107) was synthesized according to the procedure of example 1 using N-methylbut-2-yn-1-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' -trimethylacethydrazide (Compound 108)
Step 1: to 1-methylhydrazine-1-carboxylic acid tert-butyl esterTo a mixture of ester (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 room temperature for 2 hours. After this time, sodium cyanoborohydride (860.0 mg,13.7 mmol) was added to the solution. The mixture was stirred at room temperature for 2 hours, then the mixture was quenched with water (20 mL) and extracted with EtOAc (10 ml×2). The organic phase was washed with water (20 mL) and brine (20 mL), and dried over Na 2 SO 4 Dried, concentrated in vacuo and purified by silica gel column (DCM to DCM/meoh=10:1) to afford tert-butyl 1, 2-trimethylhydrazine-1-carboxylate (0.1 g,8.4% yield) as a colorless oil.
Step 2: to a solution of tert-butyl 1, 2-trimethylhydrazine-1-carboxylate (0.1 g, 573.9. Mu. Mol) in DCM (2 mL) was added HCl (1M/diethyl ether) (5.7 mL,5.7 mmol). The mixture was stirred at room temperature for 1 hour, then concentrated in vacuo to afford 1, 2-trimethylhydrazinium dihydrochloride as a white solid (70 mg,71.1% yield).
Step 3: 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N, N ', N' -trimethylacethydrazide (compound 108) was synthesized according to the procedure of example 1 using 1, 2-trimethylhydrazinium 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)
A solution of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyrimidin-5-yl) acetamide (compound 100) (0.1 g, 271. Mu. Mol), pyrimidin-5-amine (25.8 mg, 271. Mu. Mol), HATU (124 mg, 325. Mu. Mol) and DIPEA (112. Mu.L, 2.5 eq, 677. Mu. Mol) in DMF (2 mL) was stirred at room temperature for 5.0h. Water (10 mL) was added and the resulting mixture was extracted with EtOAc (10 mL. Times.3). The combined organic phases were washed with water (15 mL. Times.2), brine (15 mL), and dried over Na 2 SO 4 Dried, concentrated in vacuo and purified by prep. HPLC to afford 2- (3, 5-dichloro-4) as a white solid(4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyrimidin-5-yl) acetamide (compound 109) (20 mg, 44.8. Mu. Mol). 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyrimidin-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- ((1R, 2S) -2-fluorocyclopropyl) acetamide (Compound 111)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- ((1R, 2S) -2-fluorocyclopropyl) acetamide (compound 111) was synthesized according to the procedure for example 9 using (1R, 2S) -2-fluorocyclopropan-1-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)
/>
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (3-fluoropropyl) acetamide (compound 112) was synthesized according to the procedure of example 9 using 3-fluoroprop-1-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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (6-methoxypyridazin-3-yl) acetamide (compound 113) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyridin-3-yl) acetamide (compound 114) was synthesized according to the procedure for example 9. 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyridin-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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (pyridazin-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) -1- (pyrrolidin-1-yl) ethan-1-one (compound 117)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -1- (pyrrolidin-1-yl) ethan-1-one (compound 117) was synthesized according to the procedure of example 9. LCMS, m+h= 422.1.
Example 18: synthesis of 1- (azetidin-1-yl) -2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) ethan-1-one (Compound 118)
1- (azetidin-1-yl) -2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) ethan-1-one (compound 118) was synthesized according to the procedure for example 9. 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)
N- (tert-butyl) -2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) acetamide (compound 119) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-isobutyl-N-methylacetamide (Compound 120) was synthesized according to the procedure of example 9 using N, 2-dimethylpropan-1-amine. LCMS, m+h= 438.2.
Example 21: synthesis of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-isobutylacetamide (Compound 121)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-isobutylacetamide (Compound 121) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-isopropyl-N-methylacetamide (Compound 122) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (2-hydroxyethyl) -N-methylacetamide (Compound 123) was synthesized according to the procedure of example 9 using 2- (methylamino) ethan-1-ol. LCMS, M-h=424.1.
Example 24: synthesis of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N, N' -dimethylacethydrazide (Compound 124)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N, N' -dimethylacethydrazide (Compound 124) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (2-fluoroethyl) -N-methylacetamide (compound 125) was synthesized according to the procedure of example 9 using 2-fluoro-N-methylethyl-1-amine. LCMS, m+h=428.1.
Example 26: synthesis of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-isopropylacetamide (Compound 126)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-isopropylacetamide (compound 126) was synthesized according to the procedure for 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)
N-cyclobutyl-2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) acetamide (compound 127) was synthesized according to the procedure of example 9. 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)
N-allyl-2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-methylacetamide (Compound 128) was synthesized according to the procedure of example 9 using N-methylprop-2-en-1-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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-methyl-N-propylacetamide (compound 129) was synthesized according to the procedure 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-ethyl-N-methylacetamide (compound 130) was synthesized according to the procedure of example 9 using N-methylethylamine. LCMS, m+h=410.2.
Example 31: synthesis of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-methyl-N- (2, 2-trifluoroethyl) acetamide (Compound 131)
To a solution of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) acetic acid (100 mg,0.3 mmol) in DMF (3 mL) was added 2, 2-trifluoro-N-methylethyl-1-amine (134 mg,0.9 mmol), 1- (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 room temperature overnight. Water (20 mL) was added. The mixture was extracted with EtOAc (15 mL. Times.2). The combined organic phases were washed with brine (20 mL), and dried over Na 2 SO 4 Dried, concentrated in vacuo and purified by preparative HPLC to afford 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N-methyl-N- (2, 2-trifluoroethyl) acetamide (compound 131) as a white solid (50 mg,36% yield). 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)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -N- (2, 2-difluoroethyl) -N-methylacetamide (compound 132) was synthesized according to the procedure of example 31 using 2, 2-difluoro-N-methylethyl-1-amine hydrochloride. LCMS, m+h=446.1.
Example 33: synthesis of 2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -1-morpholinoethyl-1-one (Compound 133)
2- (3, 5-dichloro-4- (4-hydroxy-3-isopropylbenzyl) phenoxy) -1-morpholinoethyl-1-one (compound 133) was synthesized according to the procedure of example 31. LCMS, M-h=436.0.
Example 34: FAAH substrate evaluation
Purified recombinant human FAAH (rhFAAH) was purchased from Cayman Chemical (Ann Arbor, MI, USA). The total volume per incubation was 400. Mu.L, containing final 0.5 ng/. Mu.L rhFAAH, 1. Mu.M test compound, 1.25% ethanol or 1. Mu.M PF-3845 (FAAH inhibitor) and 0.1% bovine serum albumin in Tris-EDTA buffer at pH 8.0. The positive control was LL-341001. Incubation was performed at room temperature. At 0, 5, 15, 30 and 60 minutes, 30. Mu.L aliquots of the reaction mixture were removed and mixed with 300. Mu.L of acetonitrile containing 5ng/mL terfenadine and 10ng/mL tolbutamide as internal standard to quench the reaction. The resulting mixture was centrifuged at 4000rpm at 4 ℃ for 15 minutes, and 100 μl of the supernatant was ready for LC-MS/MS analysis to measure the formation of acid metabolites.
LC-MS/MS analysis
The Acquity ultra-efficient LC system from Waters was used for sample analysis. In a reverse phase Kinetex 2.6 μm C column (2.1 x 30mm,) Chromatography was performed thereon. Mobile phase a contained an aqueous solution of 0.1% formic acid and mobile phase B contained an acetonitrile solution of 0.1% formic acid, run time was 2min, flow rate was 0.8mL/min for acid metabolites from positive control, or 1.5min, flow rate was 0.9mL/min for acid metabolites of test compound. Mass spectrometers (API-5500 and APIQ Trap 4000Applied Biosystems/MDS SCIEX Instruments, framingham, MA, USA) were operated in ESI positive or negative ion MRM mode.
Data analysis
The formation of acid metabolites was monitored and quantified using a 1 μm calibration spot. The observed rate constant (ke) of acid metabolite formation was calculated by plotting the metabolite concentration against incubation time, where the slope is ke, and is shown in table 1.
TABLE 1
/>
/>
/>
/>
/>
/>
/>
A=ke is greater than or equal to 0.1; b=ke is less than 0.1 and greater than 0;
c=ke is 0; nt=no test.
Example 35: in vitro stability assessment in mouse plasma
Male CD-1 mouse plasma was purchased from BioIVT (catalog number MSE00PLK2 YNN) and thawed in a 37℃water bath on study day, and the pH was adjusted to 7.4. After a pre-warming period of 15 minutes in a 37 ℃ water bath 398 μl of plasma was added with 2 μl of a stock solution of test compound or positive control (propanepheline) in Dimethylsulfoxide (DMSO) to achieve a final concentration of 1 μΜ with 0.5% DMSO. After thorough mixing, the mixture was returned to a 37 ℃ water bath for incubation. At 0, 15, 30, 60 and 120 minutes, an aliquot of 30. Mu.L of the reaction mixture was removed and mixed with 300. Mu.L of acetonitrile containing 5ng/mL terfenadine and 10ng/mL tolbutamide as internal standard to quench the reaction. The resulting mixture was centrifuged at 4000rpm at 4℃for 15 minutes, and 100. Mu.L of the supernatant was taken and mixed with 100. Mu.L of water for analysis by liquid chromatography tandem mass spectrometry (LC-MS/MS).
LC-MS/MS analysis
The Shimadzu LC 30-AD HPLC system was used for sample analysis. In a reverse phase Kinetex 2.6 μm C column (3.0 x 30mm,) Chromatography was performed thereon. Mobile phase a contained an aqueous solution of 0.1% formic acid and mobile phase B contained an acetonitrile solution of 0.1% formic acid, with a run time of 2min. Mass spectrometers (API-4000 and APIQ Trap 4500Applied Biosystems/MDS SCIEX Instruments, framingham, MA, USA) were operated in electrospray ionization (ESI) positive or negative ion Multiple Reaction Monitoring (MRM) mode.
Data analysis
The percentage of compound remaining at a particular time point was calculated based on the peak area ratio at time 0 (as 100%). By plotting the natural logarithm of the percentage of remaining compound against the incubation time (where the slope is k obs ) To calculate the observation rate constant (k) of the metabolism of the test compound obs ). Half-life (t) was determined according to the following equation 1/2 ):t 1/2 =0.693/k obs
Example 36: in vivo tissue distribution studies in Male CD-1 mice
Male CD-1 mice (n=6/group) of 7-10 weeks of age were acclimatized to the study room for at least 3 days prior to dose administration under study. Test compounds were formulated as 0.1mg/mL clear solutions in Phosphate Buffered Saline (PBS) containing 1% N-methyl-2-pyrrolidone (NMP) and 1% Solutol, and the dose volume was 10mL/kg. The peripheral limiting FAAH inhibitor LL-650021 was formulated at 0.1mg/mL in water containing 0.5% carboxymethylcellulose and the dose volume was 10mL/kg. The concentration of the formulation is determined to meet the acceptance criteria within 20% of the target value.
Test compounds were administered to non-fasted mice at 1mg/kg by Subcutaneous (SC) injection or oral gavage (PO), with or without pretreatment of 1mg/kg LL-650021 1 hour prior to test compound administration. At 1, 4 and 8 hours post-administration, CO was used 2 Inhalation euthanizes animals (n=2/time point). Blood samples (0.3 mL) were collected from saphenous vein or other suitable site to pre-chilled K 2 EDTA tubes were placed in and on wet ice, and brain and liver were collected. The blood samples were centrifuged at 3200g at 4 ℃ for 10 minutes and the plasma samples were transferred to polypropylene tubes, flash frozen with dry ice and kept at-60 ℃ or lower until analysis. The tissue was washed with cold saline, wiped dry, weighed, and then homogenized in 15mM PBS (pH 7.4): methanol=2:1 buffer at a ratio of 1:10 (1 g tissue to 10mL buffer, resulting in a 11-fold dilution). The tissue homogenate is maintained at-60 ℃ or lower until analysis.
Sample extraction
Plasma and tissue homogenates were extracted by protein precipitation. Protein precipitation was performed on 10-50. Mu.L of plasma or 40-50. Mu.L aliquots of tissue homogenates by adding 200-800. Mu.L of acetonitrile containing an internal standard (10 ng/mL LL-120001 and 100ng/mL celecoxib), dexamethasone (dexamethasone), glibenclamide, labetalol, tolbutamide and verapamil), vortexing at 800rpm for 10min and centrifuging at 4000rpm at 4℃for 15 min. The supernatant was transferred to a 96-well plate and centrifuged at 4000rpm at 4℃for 5 minutes followed by feeding for LC-MS/MS analysis, or 200. Mu.L of the supernatant was transferred to a 96-well plate, evaporated to dryness at 25℃under a nitrogen stream, reconstituted with 50. Mu.L of 70% acetonitrile, vortex-mixed at 800rpm for 10 minutes and centrifuged at 4000rpm at 4℃for 5 minutes followed by feeding for LC-MS/MS analysis.
LC-MS/MS analysis
The Acquity ultra-efficient LC system from Waters was used for sample analysis. The separation was carried out on a ACQUITY UPLC BEH C column (50X 2.10mm;1.7 μm) at 50℃at a flow rate of 0.6 mL/min. Mobile phase a consisted of 5:95 methanol in water with 2mM ammonium acetate and mobile phase B consisted of 95:5 acetonitrile in water with 2mM ammonium acetate. Chromatography uses a linear gradient starting with 2% mobile phase B, which ranges from 2% to 90% in 2.6 minutes, is maintained for 0.2 minutes at 90% B wash, and is equilibrated for an additional 0.2 minutes at 2% B. Aliquots of 2-9. Mu.L of sample were used for injection. The mass spectrometer (API-6500+, applied Biosystems/MDS SCIEX Instruments, framingham, mass., USA) was operated in either positive or negative ion MRM mode under ESI.
Example 37: in vitro prodrugs and agonist TR beta receptor selectivity
The potency and selectivity of LL-341070 at the thyroid hormone β receptor (trβ) was evaluated, and LL-341070 is a thyromimetic prodrug of formula (I') described herein that delivers LL-341070a as a potent and selective small molecule agonist of thyroid hormone receptor (TR) β following Fatty Acid Amide Hydrolase (FAAH) mediated transformation. In vitro potency was determined by applying test compounds to a luciferase-based TR reporter cell line using thyroid hormone (T3) as a positive control. Table 2 depicts the potency profiles of the LL-341070 prodrug and LL-341070A active metabolite for TR beta and TR alpha, as measured by half maximal effective concentration (EC 50), with the TRalpha bias for T3 being adjusted in the assay for the selectivity measurement. Both LL-341070 and LL-341070A show enhanced selectivity for TR beta and LL-341070A shows enhanced potency.
TABLE 2
Example 38: LL-341070A enhances oligodendrocyte progenitor differentiation in vitro
To dissect LL-341070A, in vitro Oligodendrocyte Progenitor Cell (OPC) assays were performed on primary OPC cultures generated from the brains of E14.5 PLP-EGFPC57Bl/6 mouse embryos. Thyroid hormone (T3), which is 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 1nM to about 1000 nM. After 5 days of OPC differentiation (n=6/concentration) with or without LL-341070a compound, cells were fixed and stained for Myelin Basic Protein (MBP) normalized to total cell count.
Figure 1 depicts that the active metabolite LL-341070a of the LL-341070 prodrug enhances oligodendrocyte differentiation (ec50=1.4 nM) in an oligodendrocyte progenitor assay in vitro. Enhanced oligodendrocyte differentiation was demonstrated to be relatively constant with changes in LL-341070a treatment concentration.
Example 39: thyromimetic therapy to enhance 24-OHC synthesis in vivo
The ability of thyromimetics to accelerate the remyelination process in vivo was assessed by measuring fractional synthesis of 24-hydroxycholesterol (24-OHC) in the brain of rats following cyclohexanone oxalyl dihydrazone-induced demyelination. As shown in fig. 2, the cyclohexanone oxalyl dihydrazone demyelination model evaluates n=10f rats/group. During active remyelination, the effect of thyromimetic treatment on fractional synthesis of 24OHC in brain and plasma was measured using deuterated water labeling of 24OHC in a cyclohexanone dihydrazone demyelination model following withdrawal of 0.6% of the cyclohexanone dihydrazone diet. At the 3 week withdrawal of the 0.6% bicyclohexanoyl dihydrazone diet, 3 weeks of rats were continuously supplied with deuterated water and LL-341070 was administered at 30 or 100 μg/kg, followed by measurement of 24-OHC deuterium enrichment and labeling patterns in the cortex and callus. LL-341070 induced a dose-dependent increase in deuterated 24-OHC compared to vehicle controls, indicating an increase in the rate of myelin synthesis. As shown in fig. 2, thyromimetic treatment enhanced the Fractional Synthesis Rate (FSR) of 24S-hydroxycholesterol in the brain and showed a strong correlation with 24OHC FSR in plasma. The 24OHC Fractional Synthesis Rate (FSR) is calculated based on data collected from tissues and plasma subjected to alkaline hydrolysis and derivatization for GC/MS or LC/MS analysis of deuterated 24 OHC. Plasma analysis of 24OHC was measured by Ardena Biosciences as fraction of labeled plasma 24OHC relative to total plasma 24OHC by LC/MS. Fractional synthesis was calculated by mass isotope isomer distribution analysis using precursor 2H enrichment in body water from hepatic palmitate 6. And (3) statistics: data were analyzed by one-way ANOVA with the Dukker multiple comparison test (Tukey's multiple comparison test) and expressed as mean +/-SEM. * p <0.05.
The compound potency, pharmacokinetics and target engagement of LL-341070 were confirmed prior to testing efficacy in a myelin regeneration model including in vitro oligodendrocyte precursor cell differentiation and in vivo experimental autoimmune encephalitis.
Example 40: engagement of TR beta in brain increases expression of T3 target genes in vivo
Figure 3 depicts TR beta target engagement in the brain as evidenced by increased expression of T3 responsive target genes in vivo. A single PO administration of LL-341070 (in the range of about 0.1 μg/kg to about 300 μg/kg) or T3 (about 300 μg/kg) in male C57BL/6 mice increased expression of Hr, dio3, klf9 (quantified by QuaniCalex) and complex mean log2 fold changes in the brain. Klf9, a T3 responsive gene associated with in vitro myelin regeneration, was upregulated at various treatment concentrations. This increase in expression was confirmed (quantified by Nanostring) in the brain of the rat bicyclohexanoyl dihydrazone model (as previously discussed) administered either 30 μg/kg or 100 μg/kg of LL-341070 or 300 μg/kg of T3 repeatedly over 21 days. Of interest, dio3 has enhanced expression enhancement upon repeated administration.
Example 41: in vivo tissue distribution demonstrates enhanced brain exposure of the active compound compared to the prodrug
In vivo brain exposure of the active compounds compared to the prodrugs was assessed by Tissue Distribution (TD) assay in the mouse and rat cyclohexanone oxalyldihydrazone model, measured as brain to plasma brain exposure ratio after thyromimetic treatment. As shown in Table 3, in male C57BL/6 mice, a single PO administration of LL-341070 (100 μg/kg) or LL-341070A (100 μg/kg) showed an enhanced brain exposure of active compound LL-341070A compared to prodrug LL-341070, resulting in brain to plasma AUC ratio of LL-341070A >1, wherein AUC is 0-24h, when measured in brain and plasma. The data shows that the AUC of LL-341070A in the brain is about 7-fold higher than that of prodrug LL-341070. Table 3 also depicts brain to plasma AUC ratios. As shown in FIG. 4, repeated administration of LL-341070 (30 μg/kg or 100 μg/kg) or LL-341070A (30 μg/kg or 100 μg/kg) over 21 days showed enhanced brain exposure of active compound LL-341070A compared to prodrug LL-341070 in the rat cyclohexanone dihydrazone model, measured in brain and plasma 4 hours after the final dose.
TABLE 3 Table 3
Example 42: LL-341070 improves in vivo clinical scores and histology in the mouse EAE model
As shown in fig. 5, LL-341070 efficacy was assessed in a mouse prophylactic Experimental Autoimmune Encephalitis (EAE) model, where EAE scores were made on days 7-28 after induction of EAE by MOG35-55/cfa+ptx, with disease onset 8-18 days after induction. The EAE model evaluates n=12fc57bl/6 mice/group, with daily PO administration of LL-341070 (10 μg/kg to 100 μg/kg) or vehicle following EAE induction. LL-341070 given daily in a prophylactic paradigm improved the median disease onset day and reduced the maximum disease severity in a dose-dependent manner. Histological analysis of the spinal cord 28 days after immunization showed reduced inflammatory lesions, reduced apoptotic cell counts, and reduced areas of demyelination according to H & E and MBP staining. LL-341070 improves mean clinical scores and histological endpoints for inflammation and demyelination in the mouse EAE model. Clinical scores were determined by unknowing observers. Histological analysis was performed in spinal cord samples (demyelination score was assessed by% demyelination area in anti-MBP staining, inflammatory lesions refer to a group number of >20 cells per section in H & E staining). Statistics: median day of onset of EAE was compared using Wilcoxon's survival test.
Example 43: enrichment of FAAH expression in brain
As shown in fig. 6, a brain-directed thyromimetic prodrug, such as ABX-002, which is compound 1 described herein, activated to ABX-002A, activated by Fatty Acid Amide Hydrolase (FAAH) was used to elucidate the mechanism by which thyromimetic drugs disrupt the Thyroid Hormone Axis (THA). Strength-changing nail polishThe delivery of prostaglandin drugs to help determine whether feedback control of THA is derived from the central (hypothalamic) or peripheral (pituitary) mechanisms and potentially enhance the therapeutic index of thyromimetics. These studies used recombinant FAAH, tissue-derived S9 fraction, in vivo Tissue Distribution (TD), gene expression in brain and liver, and T as a marker of THA disruption in mice 4 Is performed by the influence of (a) on the surface of the substrate. Northern blot assays confirm that FAAH is expressed in multiple species (rodent and human), and that relative mRNA FAAH expression is enhanced in the brain. FAAH specific activity (AMC lysis assay) of tissue-derived S9 fractions from different organs (liver, brain, small intestine) of multiple species (mouse, rat, non-human primate, human) was demonstrated to increase in the brain of human and non-human primate, calculated as a percentage of liver activity.
Example 44: FAAH expression enhances delivery of ABX-002A to the brain
To assess delivery, the concentration of ABX-002A in brain, liver, kidney, lung and heart was measured 1 hour after SC administration of 30 different prodrugs of ABX-002A. As shown in fig. 7, the brain to plasma ratio of the prodrug was increased relative to ABX-002A, while the tissue to plasma ratio of peripheral organs (liver, kidney, lung and heart) showed a linear (constant) tissue to plasma relationship. The data show that FAAH is highly expressed in the CNS and ABX prodrugs enhance delivery of active metabolites to the brain >30 fold, with brain to plasma ratio >1. In organs other than the brain, the data show that tissue concentration is driven by plasma concentration of the active metabolite ABX-002A.
Example 45: global and peripheral FAAH inhibitors alter metabolite distribution in mice
Global penetration and peripheral limiting FAAH inhibitors (GFI and PFI, respectively) were evaluated for their ability to alter the distribution of ABX-002 and ABX-002A. Table 4 depicts peripheral and global FAAH inhibitors: efficacy profiles (in apparent IC) of LL-650177 (PFI), URB9373 (PFI) and PF-044578454 (GFI) 50 (nM) and the efficacy profile was obtained after a 30min pre-incubation with recombinant human FAAH and 7-amino-4-methylcoumarin (AMC).
TABLE 4 Table 4
FAAH inhibitors Apparent IC 50 (nM) Distribution of
LL-650177 9.1 The outer periphery
URB937 69 The outer periphery
PF-04457845 3.0 Global situation
FIG. 8 shows plasma, liver and brain concentrations after co-administration of prodrug (ABX-002) with or without PFI or GFI. Prodrug levels did not change with FAAH inhibition, or increased slightly with FAAH inhibition. Active metabolite (ABX-002A) levels were reduced in plasma and liver at PFI and in all organs at GFI. Table 5 depicts inhibition of active metabolites (LL-650177 or PF-044578454) in AUC in plasma, liver and brain following co-administration of prodrug (ABX-002). Tissue distribution studies in mice confirm global and peripheral inhibition of FAAH.
TABLE 5
/>
Example 46: induction of T3 regulatory genes by prodrugs and FAAH inhibitors
Female C57BL/6 mice (n=5/group) of 6-8 weeks old were acclimatized to the study room for at least 3 days prior to dose administration under study. A single dose of PFI or vehicle was orally administered (PO) to non-fasted mice at time = -1 hour on day 0. A single dose was administered at 5mL/kg based on recent body weight, collected once during the entire study. After PFI or vehicle dose, animals were given a single dose of test article at time = 0 hours. One group (n=5) was PO administered 300ug/kg of T3 only at time=0 hours. Animals were humanly euthanized about 4 hours (t=4 hours) after the test article dose, and brain, liver, heart, pituitary, spinal cord and plasma samples were collected.
Sample processing
a. Expression analysis samples-at the end point, multiple organs were collected and tissues were immediately processed as described below.
i. Brain: for each mouse, the cranium was opened and the brain was removed. The cerebellum was resected and the cerebral cortex was sagittal cut in half and the left half was collected. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the cerebral cortex specimen was placed into a tube containing 1.2mL of precooled RNALater and stored at 4 ℃.
Liver: for each mouse, one liver biopsy (100-150 mg) was collected from the left lateral lobe. After rinsing the irrelevant blood from the biopsy with ice-cold 0.9% NaCl, the sample was placed into 1.2mL pre-chilled RNALater and stored at 4 ℃. Left ventricle: for each mouse, left Ventricle (LV) blood was cleared using PBI standard methods, and half of the LV free wall was collected. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the LV free wall was placed into 1.2mL of precooled RNALater and stored at 4 ℃. LV tissue remains in PBI for potential future analysis or until the appropriate genes can be identified, for up to 6 months after the end of the survival phase of the study. Sample handling was confirmed prior to discarding.
Pituitary gland: for each mouse, pituitary glands were collected after brain removal. After rinsing the irrelevant blood from the pituitary with ice-cold 0.9% NaCl, the specimens were placed into 0.15mL of precooled RNALater and stored at 4 ℃. Pituitary tissue remains in PBI for potential future analysis or until the appropriate genes can be identified, for up to 6 months after the end of the survival phase of the study. Sample handling was confirmed prior to discarding.
b. Pharmacokinetic samples-at the endpoint, blood and tissue specimens were immediately processed as described below. Samples for PK analysis remained in PBI at-80 ℃ for up to 90 days after the end of the survival phase of the study.
i. Plasma: whole blood (about 300. Mu.L) was collected by cardiac puncture under isoflurane anesthesia on K3 EDTA. The blood was immediately placed on wet ice. After the end of the disassembly procedure, the blood was centrifuged at 10,000Xg for 10 minutes at 4 ℃. Plasma (approximately 125 μl) was aliquoted into appropriately labeled tubes and flash frozen.
Liver: for each mouse, one liver biopsy (30-50 mg) was collected from the left lateral lobe. After rinsing the irrelevant blood from the biopsy with ice-cold 0.9% NaCl, the sample was placed into a suitably labeled tube and flash frozen in liquid nitrogen.
Brain: for each mouse, midbrain biopsies (30-50 mg) were collected from the right cerebral cortex. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the biopsies were placed into appropriately labeled tubes and flash frozen.
Left ventricle: for each mouse, left Ventricle (LV) blood will be cleared using PBI standard methods, and half of the LV free wall is collected. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the LV free wall was placed into a suitably labeled tube and flash frozen.
Target engagement
The change in expression of the selected gene identified by transcriptomic analysis WAs measured from purified RNA using an in situ RNA quantification method based on hybridization (NanoString, seattle, WA). Briefly, will be newFresh tissue was collected in RNALater catalogue number AM7021 TM The stabilized solution (ThermoFisher Scientific; carlsbad, calif.) was frozen at-20℃until ready for RNA extraction. Whole blood was collected by terminal cardiac puncture in MiniCollet K2EDTA tubes, catalog number 450480, of Greinder Bio-one GmbH (Kremsmunster, austria) and processed into plasma by centrifugation at 2000 Xg for 10 min at 4 ℃. For RNA extraction, the tissue was homogenized in TRIzol reagent (ThermoFisher Scientific) catalog number 15596026 using a bead homogenizer and RNA was extracted according to the manufacturer's protocol and purified using Econospin RNA mini spin column (Ephoch Life Sciences, missouri City, TX, catalog numbers 1940-250) for RNA according to the manufacturer's protocol. Specific gene probes were designed by the NanoString bioinformatics team using defined target sequences based on the NCBI reference sequence (RefSeq) database. Custom probes were synthesized from Integrated DNA Technologies (IDT; coralville, IA). Using the multiplex method in the nCounter PlexSet-12 kit under the catalog number PS-GX-PTK-12 (CSO) according to the manufacturer's protocol (NanoString, inc, seattle, WA) mRNA expression was analyzed on the SPRINT Profiler NanoString system.
Data analysis
After a single administration of the drug, the T3 target gene is increased, wherein the relative activity of the brain with respect to the liver is determined by prodrug and/or FAAH inhibition. In different dosage forms, the relative activity of the brain with respect to the liver (as a sign of peripheral activity) varies>1500 times. Figures 9A, 9B and 9C show induction of T3 regulated genes in brain (blue) and liver (orange) 4h after a single administration of either (a) active metabolite or (B) prodrug alone or (C) prodrug + PFI (URB 937). RNA was analyzed by Nanostring; mean fold changes for multiple genes were calculated on log2 scale and normalized to data obtained for T3 at 300 mg/kg. PFI administration activates the prodrug to T in the liver 3 Reduced potency of the regulated genes<1/10 without affecting activity or exposure in the brain. PFI also reduces efficacy against THA, as opposed to being based onNegative feedback of circulating peripheral metabolites rather than brain exposure was consistent. Thus, the use of PFI allows for in-target brain effects to be separated from the effects on THA.
Example 47: t (T) 4 Corresponding to peripheral activity
Female C57BL/6 mice (n=5/group) of 6-8 weeks old were acclimatized to the study room for at least 3 days prior to dose administration under study. Mice were dosed at 5mL/kg based on recent body weight and collected once during the entire study. Based on recent body weight, mice were placed into a body weight matched treatment dosing cohort, collected once throughout the study. Mice were given a single administration of PFI or vehicle (n=5/group) orally (PO) daily for 7 days at time = -1 hour. Animals were given test articles daily at time=0 hours after PFI (100 μg/kg) or vehicle dose (10 mL/kg, p.o.). The test article was administered at one of eight dosage levels (0.1, 0.3, 1, 3, 10, 30, 100 or 300 μg/kg) on days 1-7 for a total of seven doses. With (a) an active metabolite or (B) a prodrug alone; (C) Prodrug+pfi (LL-650177) or (D) prodrug+gfi was administered to mice PO for 7 days QD. Animals were humanly euthanized using standard procedures about 4 or 8 hours (t=4 hours or t=8 hours) after the test article dose, and brain, liver, and plasma samples were collected. RNA WAs quantified from samples collected 4 hours after the final dose using the hybridization-based in situ RNA quantification method (NanoString, seattle, WA) as described below. RNA from samples collected 8 hours after the final dose was quantified using the hybridization-based in situ RNA quantification method (QuantiGene Plex) as described below. On the last day of administration, mice were dosed according to a schedule to mitigate the effect of circadian effects on thyroid hormone-sensitive gene expression. Thus, at the time of end-point sacrifice, the treatment group was equilibrated with respect to "time of day". Mice were anesthetized 4 or 8 hours after final dosing, blood was collected by retroorbital puncture, and euthanized using standard procedures. Immediately after euthanasia, tissues were collected and processed according to the following procedure.
Sample processing
a. Expression analysis samples-at the end point, multiple organs were collected and tissues were immediately processed as described below.
i. Brain: for each mouse, the cranium was opened and the brain was removed. The cerebellum was resected and the cerebral cortex was sagittal cut in half and the left half was collected. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the cerebral cortex specimen was placed into a tube containing 1.2mL of precooled RNALater and stored at 4 ℃.
Liver: for each mouse, one liver biopsy (100-150 mg) was collected from the left lateral lobe. After rinsing the irrelevant blood from the biopsy with ice-cold 0.9% NaCl, the sample was placed into 1.2mL pre-chilled RNALater and stored at 4 ℃. Left ventricle: for each mouse, left Ventricle (LV) blood was cleared using PBI standard methods, and half of the LV free wall was collected. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the LV free wall was placed into 1.2mL of precooled RNALater and stored at 4 ℃. LV tissue remains in PBI for potential future analysis or until the appropriate genes can be identified, for up to 6 months after the end of the survival phase of the study. Sample handling was confirmed prior to discarding.
Pituitary gland: for each mouse, pituitary glands were collected after brain removal. After rinsing the irrelevant blood from the pituitary with ice-cold 0.9% NaCl, the specimens were placed into 0.15mL of precooled RNALater and stored at 4 ℃. Pituitary tissue remains in PBI for potential future analysis or until the appropriate genes can be identified, for up to 6 months after the end of the survival phase of the study. Sample handling was confirmed prior to discarding.
b. Pharmacokinetic samples-at the endpoint, blood and tissue specimens were immediately processed as described below. Samples for PK analysis remained in PBI at-80 ℃ for up to 90 days after the end of the survival phase of the study.
i. Plasma: whole blood (about 300. Mu.L) was collected by cardiac puncture under isoflurane anesthesia on K3 EDTA. The blood was immediately placed on wet ice. After the end of the disassembly procedure, the blood was centrifuged at 10,000Xg for 10 minutes at 4 ℃. Plasma (approximately 125 μl) was aliquoted into appropriately labeled tubes and flash frozen.
Liver: for each mouse, one liver biopsy (30-50 mg) was collected from the left lateral lobe. After rinsing the irrelevant blood from the biopsy with ice-cold 0.9% NaCl, the sample was placed into a suitably labeled tube and flash frozen in liquid nitrogen.
Brain: for each mouse, midbrain biopsies (30-50 mg) were collected from the right cerebral cortex. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the biopsies were placed into appropriately labeled tubes and flash frozen.
Left ventricle: for each mouse, left Ventricle (LV) blood will be cleared using PBI standard methods, and half of the LV free wall is collected. After rinsing the irrelevant blood from the tissue with ice-cold 0.9% NaCl, the LV free wall was placed into a suitably labeled tube and flash frozen.
Target engagement
Tissue samples for biochemical analysis were prepared by cryogenic powdering under liquid nitrogen and solubilized using standard methods of PBI. The change in expression (mRNA expression) of the selected genes identified by transcriptomic analysis was measured from purified RNA using hybridization-based in situ RNA quantification methods (NanoString or QuantiGene Plex). The target gene expression data is presented as a ratio to the geometric mean of the properly expressed normalized genes. Briefly, fresh tissue was collected in RNALater under accession number AM7021 TM The stabilized solution (ThermoFisher Scientific; carlsbad, calif.) was frozen at-20℃until ready for RNA extraction. Whole blood was collected by terminal cardiac puncture in MiniCollet K2EDTA tubes, catalog number 450480, of Greinder Bio-one GmbH (Kremsmunster, austria) and processed into plasma by centrifugation at 2000 Xg for 10 min at 4 ℃. For RNA extraction, the tissue was homogenized in TRIzol reagent (ThermoFisher Scientific) catalog number 15596026 using a bead homogenizer and RNA was extracted according to the manufacturer's protocol and purified using Econospin RNA mini-spin/column (Ephoch Life Sciences, missouri City, TX, catalog numbers 1940-250) according to the manufacturer's protocol. Specific gene probes NCBI-based reference sequences by the NanoString bioinformatics team And (3) designing a determined target sequence of the (RefSeq) database. Custom probes were synthesized from Integrated DNA Technologies (IDT; coralville, IA). Using the multiplex method in the nCounter PlexSet-12 kit under the catalog number PS-GX-PTK-12 (CSO) according to the manufacturer's protocol (NanoString, inc, seattle, WA)mRNA expression was analyzed on the SPRINT Profiler NanoString system.
T4 analysis
T4 was measured in final plasma samples using ELISA kit (Biovision, inc., thyroxine [ T4] [ mouse/rat ] ELISA kit, catalog number: K7421-100). According to the manufacturer's instructions, the assay is performed with minor modifications based on previous assay validation work. Briefly, for each assay, a seven-point standard curve (25, 15, 10, 5, 2, 1 μg/dL) of T4 diluted in assay buffer is prepared in duplicate. Plasma samples (undiluted), blanks (assay buffer) and standards were added to individual wells of a 96-well plate pre-coated with T4 capture antibody, followed by T4 enzyme conjugates to each well. The plates were then gently shaken (600 rpm) for 20-30s for mixing, then covered with an acetate plate sealing membrane and incubated at Room Temperature (RT) for 1h with gentle shaking (600 rpm). The plate contents were aspirated and washed three times with 1 x wash buffer, followed by blotting on paper towels to remove excess liquid. TMB substrate was then added to each well and the plate was fixed with acetate sealing membrane and incubated at room temperature for 15min in the absence of light. A stop solution was then added to each well and the plate gently shaken to mix the solutions. Absorbance was read at 450nm within 15min after the addition of the stop solution using a Varioskan Lux plate reader (ThermoFisher Scientific, carlsbad, CA). The relative Optical Density (OD) was background corrected against the blank sample and standard curve. The T4 concentration was interpolated using a four parameter curve fitting method. Unknown sample concentrations were determined using GraphPad Prism software (GraphPad Prism 9.0.2,GraphPad Software,San Diego,CA).
Data analysis
FIGS. 10A, 10B, 10C and 10D show that either (A) active metabolite or (B) single prodrug has been used; (C) In mice dosed with prodrug+pfi (LL-650177) or (D) prodrug+gfipo (QD for 7 days), gene expression in brain (blue) and liver (orange) and effect on T4 (grey) 4 or 8h after last dose. After 7 days of treatment, both the prodrug and active metabolite reduced T4 levels. Table 6 reports ED in μg/kg for each treatment type 50 Values.
TABLE 6
Using T 4 As a marker of the effect on THA; compared to CNS activation corresponding to the target gene, T 4 More corresponding to peripheral activity. Thyromimetic drug pair T 4 The negative regulation of (c) does not appear to be predominantly centrally mediated, as the effects on THA and liver gene expression more closely correspond to plasma distribution than to exposure or activity in the CNS, indicating predominantly pituitary driven effects. The combination of a thyromimetic prodrug and PFI can further enhance delivery of the thyromimetic to the brain and maximize central targeting distribution.
Example 48: peripheral exposure of ABX-002A predicts the effect on THA
The relationship between THA effect and plasma ABX-002A was studied in both mice and NHP. Mice: using female C57BL/6 mouse data from example 14 above, exposure to amides and acids was calculated based on PK data from independent experiments such as detailed in example 12. PK was performed in only a single dose, with other doses being calculated proportionally. Non-human primate (NHP): plasma pharmacokinetics and effects on thyroid hormone axis were measured in non-naive cynomolgus monkeys (n=3/group) after daily ABX-002 dosing at 10, 30, 100 or 300ug/kg for 7 days. ABX-002 was formulated in 0.1% NMP/0.1% solutol and applied at 5mL/kg PO. Blood samples were taken at 0, 0.25, 0.5, 1, 2, 4, 8, 12 and 24h on days 1 and 7 and analyzed for ABX-002, LL-340001, T3 and T4 levels by LCMS (as described above). TSH was measured by immunoassay.
Bioassays for T3 and T4 in NHP serum were performed using surrogate matrices, QC, double blanks, and blanks. Standard curve samples were prepared by adding 5 μl WS to 50 μl blank replacement serum. QC samples were prepared by adding 5 μl WS to 50 μl blank replacement serum. To the unknown sample 5 μl DMSO was added. 500. Mu.L of IS in methanol working solution (2.5 ng/mL T3-13C6 and 25ng/mL T4-13C 6) was added to all calibration standards, QC, samples and blank wells, and 500. Mu.L of blank methanol was added to all double blanks. After transferring 400 μl of supernatant, the sample was evaporated under N2 gas and reconstituted with 100 μl of 80% methanol in water.
FIGS. 11A, 11B and 11C show T after 7 days of treatment in either mouse (orange) or non-human primate NHP (blue) 4 Inhibition varies with (a) dose, (B) plasma prodrug AUC or (C) plasma active metabolite AUC. Day 7T 4 levels normalized to day 1 levels for each animal were compared to exposures in these same animals. In summary, the curve depicted on fig. 11C shows that the relationship between THA effect and plasma ABX-002A exists in both mice and NHP, and that peripheral exposure of active metabolites is a better predictor of the effect of THA compared to dose or plasma prodrug exposure.

Claims (32)

1. A pharmaceutical composition comprising a Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I'), or a pharmaceutically acceptable salt or solvate thereof:
wherein:
R 1 and R is 2 Independently selected from hydrogen, -OR 5 、-NR 5 R 6 、C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 Alkyl-phenyl, wherein C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 The alkyl-phenyl group is optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
R 3 And R is 4 Independently selected from-F, -Cl, -Br and-I;
R 5 and R is 6 Independently selected from hydrogen and C 1 -C 6 An alkyl group; and is also provided with
R 7 And R is 8 Independently selected from hydrogen, -F, -Cl, -Br and-I;
a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises a peripherally restricted FAAH inhibitor.
2. The pharmaceutical composition of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein R 7 Is hydrogen.
3. The pharmaceutical composition of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein R 7 is-F.
4. The pharmaceutical composition of any one of claims 1-3, or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 Is hydrogen.
5. The pharmaceutical composition of any one of claims 1-3, or a pharmaceutically acceptable salt or solvate thereof, wherein R 8 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:
wherein:
R 1 and R is 2 Independently selected from hydrogen, -OR 5 、-NR 5 R 6 、C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 Alkyl-phenyl, wherein C 1 -C 6 Alkyl, C 2 -C 6 Alkenyl, C 2 -C 6 Alkynyl, C 3 -C 6 Cycloalkyl, C 3 -C 6 Heterocycloalkyl, phenyl and-C 1 -C 6 The alkyl-phenyl group is optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
R 3 And R is 4 Independently selected from-F, -Cl, -Br and-I; and is also provided with
R 5 And R is 6 Independently selected from hydrogen and C 1 -C 6 An alkyl group;
a pharmaceutically acceptable excipient; the pharmaceutical composition further comprises 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 R 1 Is hydrogen.
8. The pharmaceutical composition of any one of claims 1-7, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 Is C optionally substituted with one or more of 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
9. The pharmaceutical composition of any one of claims 1-8, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 Is C substituted by one or more of the following 1 -C 6 Alkyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
10. The pharmaceutical composition of any one of claims 1-9, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 Is C substituted by one or more-OH groups 1 -C 6 An alkyl group.
11. The pharmaceutical composition of any one of claims 1-9, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 Is C substituted by one or more halogen groups 1 -C 6 An alkyl group.
12. The pharmaceutical composition of any one of claims 1-8, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 Is unsubstituted C 1 -C 6 An alkyl group.
13. The pharmaceutical composition of any one of claims 1-7, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 Is phenyl optionally substituted with one or more of the following: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
14. The pharmaceutical composition of any one of claims 1-7, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is-C optionally substituted with one or more of 1 -C 6 Alkyl-phenyl: halo, cyano, -OR 5 、-NR 5 R 6 、-S(O) 2 R 5 or-S (O) 2 OR 5
15. The pharmaceutical composition of any one of claims 1-14, or a pharmaceutically acceptable salt or solvate thereof, wherein R 3 And R is 4 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 R 3 And R is 4 Are all-Br.
17. The pharmaceutical composition of any one of claims 1-15, or a pharmaceutically acceptable salt or solvate thereof, wherein R 3 And R is 4 Are all-Cl.
18. The pharmaceutical composition of any one of claims 1-15, or a pharmaceutically acceptable salt or solvate thereof, wherein R 3 And R is 4 Are all-F.
19. The pharmaceutical composition of claim 1 or claim 6, wherein the Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') or (I) has a structure selected from the group consisting of:
or a 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 (I') or (I) has a structure selected from the group consisting of:
or a pharmaceutically acceptable salt or solvate thereof.
21. The pharmaceutical composition of claim 1, wherein the Fatty Acid Amide Hydrolase (FAAH) cleavable prodrug of formula (I') has a structure selected from the group consisting of:
or a 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, the method comprising administering to the patient a therapeutically effective amount of the 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 white matter encephalitis (AHL or AHLE), adult raffmum disease, infant raffmm disease, alexander disease, alzheimer's disease, baroconcentric sclerosis, kandelian disease, pontic central myelination (CPM), cerebral palsy, tenascitic xanthomatosis, chronic Inflammatory Demyelinating Polyneuropathy (CIDP), devic's syndrome, myelin-lytic diffuse sclerosis, encephalomyelitis, guillain-barre syndrome, idiopathic inflammatory demyelinating disease (HDD), kebert's disease, leber's hereditary optic neuropathy, leukodystrophy, maltesian multiple sclerosis, markia-binimedes disease, metachromatic Leukodystrophy (MLD), multifocal Motor Neuropathy (MMN), multiple Sclerosis (MS), paratyphenic disease, demyelinating polyneuropathy (pmol), polymorphic focal fascian disease (TSP), and alp-zeppy disease (alp-X), or fulgorism (alp-38 o), or linked-pansy (alp).
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 the group consisting of:
/>
or a pharmaceutically acceptable salt or solvate thereof.
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, the method 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 white matter encephalitis (AHL or AHLE), adult raffmum disease, infant raffmm disease, alexander disease, alzheimer's disease, baroconcentric sclerosis, kandelian disease, pontic central myelination (CPM), cerebral palsy, tenascitic xanthomatosis, chronic Inflammatory Demyelinating Polyneuropathy (CIDP), devic's syndrome, myelin-lytic diffuse sclerosis, encephalomyelitis, guillain-barre syndrome, idiopathic inflammatory demyelinating disease (HDD), kebert's disease, leber's hereditary optic neuropathy, leukodystrophy, maltesian multiple sclerosis, markia-binimedes disease, metachromatic Leukodystrophy (MLD), multifocal Motor Neuropathy (MMN), multiple Sclerosis (MS), paratyphenic disease, demyelinating polyneuropathy (pmol), polymorphic focal fascian disease (TSP), and alp-zeppy disease (alp-X), or fulgorism (alp-38 o), or linked-pansy (alp).
32. The method of claim 31, wherein the CNS disease or disorder is selected from multiple sclerosis and X-linked adrenoleukodystrophy.
CN202280047554.XA 2021-05-06 2022-05-06 Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of thyromimetics and combinations with peripherally restricted FAAH inhibitors Pending CN117597122A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/185,254 2021-05-06
US202163274856P 2021-11-02 2021-11-02
US63/274,856 2021-11-02
PCT/US2022/028187 WO2022236133A1 (en) 2021-05-06 2022-05-06 Fatty acid amide hydrolase (faah) cleavable prodrugs of thyromimetics and combination with peripherally restricted faah inhibitors

Publications (1)

Publication Number Publication Date
CN117597122A true CN117597122A (en) 2024-02-23

Family

ID=89917082

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280047554.XA Pending CN117597122A (en) 2021-05-06 2022-05-06 Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of thyromimetics and combinations with peripherally restricted FAAH inhibitors

Country Status (1)

Country Link
CN (1) CN117597122A (en)

Similar Documents

Publication Publication Date Title
US10493158B2 (en) Pharmaceutical compositions for the treatment of inflammatory disorders
WO2016153948A1 (en) Combination therapy using enantiopure, oxy-substituted, deuterium-enriched 5-(benzyl)-5-deutero-thiazolidine-2, 4-diones for treatment of medical disorders
KR20120047273A (en) Dp2 antagonist and uses thereof
EP2744778B1 (en) Meta-substituted biphenyl peripherally restricted faah inhibitors
EP3458448B1 (en) Fasn inhibitors for use in treating non-alcoholic steatohepatitis
US7579504B2 (en) ABCA1 elevating compounds
US20240041849A1 (en) Compounds for the treatment of neurological or mitochondrial diseases
KR20180100373A (en) Uses of Ascochlorin Derivatives and AMPK Activators
US20210346391A1 (en) Treatment of pruritus with p2x3 modulators
US20210322438A1 (en) Certain (2s)-n-[(1s)-1-cyano-2-phenylethyl]-1,4-oxazepane-2-carboxamides for treating lupus nephritis
JP2023524169A (en) Crystal Forms of Pharmaceutical Compounds
US10442761B2 (en) Deuterated GFT-505
CN117597122A (en) Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of thyromimetics and combinations with peripherally restricted FAAH inhibitors
US8378112B2 (en) Glycyrrhetinic acid derivative and use thereof
US11845730B2 (en) 1,3-substituted cyclobutyl derivatives and uses thereof
AU2022271304A1 (en) Fatty acid amide hydrolase (faah) cleavable prodrugs of thyromimetics and combination with peripherally restricted faah inhibitors
CN112851626B (en) Levalacyclomorpholine, preparation method, pharmaceutical composition and application thereof
US9650351B2 (en) Inhibition of IL-2 production
CN113747870A (en) Deuterated forms and derivatives of flurbicilin
WO2024054996A1 (en) Methods for the treatment of depression
CN117642381A (en) Fatty Acid Amide Hydrolase (FAAH) cleavable prodrugs of brain targeting actives in combination with peripherally restricted FAAH inhibitors
WO2023213182A1 (en) Carebastine salt and use thereof
US20210139435A1 (en) Prostaglandin analogs and uses thereof
JP2021134211A (en) Combinations for treating nafld/nash and related diseases
WO2024026540A1 (en) Inhibitors and uses therefor

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication